Open access peer-reviewed chapter

Pathogenic Role of Cytokines and Effect of Their Inhibition in Psoriasis

Written By

Jitlada Meephansan, Urairack Subpayasarn, Mayumi Komine and Mamitaro Ohtsuki

Submitted: 11 November 2016 Reviewed: 09 March 2017 Published: 05 July 2017

DOI: 10.5772/intechopen.68421

From the Edited Volume

An Interdisciplinary Approach to Psoriasis

Edited by Anca Chiriac

Chapter metrics overview

2,746 Chapter Downloads

View Full Metrics

Abstract

The pathogenesis of psoriasis is complex, and cytokines play an important role in mediating cell-cell interactions that result in abnormal structures and functions of many cell types in psoriasis, such as abnormal proliferation and differentiation of keratinocytes, abnormal proliferation of blood vessels, stimulation of immune cells, and driving abnormal immune reactions. In this chapter, we summarize the roles and functions of inflammatory cytokines that play a crucial role in psoriasis such as tumor necrosis factor (TNF)-α, interleukin (IL)-12/IL-23, and IL-17, as well as their inhibitors that are used to treat psoriasis.

Keywords

  • psoriasis
  • inflammation
  • cytokine
  • biologic drugs
  • pathogenesis

1. Introduction

Psoriasis is a common chronic inflammatory disease characterized by abnormal proliferation of keratinocytes, increased dermal vascularity, and multiple inflammatory cell infiltration. It is an immune-mediated skin disease influenced by genetic and epigenetic variations, which can be triggered by environmental factors. Psoriasis affects approximately 2% of people worldwide [1, 2].

Psoriasis typically presents as indurated scaly erythematous plaques and is easily diagnosed; however, variable clinical manifestations may be presented. As a result, psoriasis remains a clinical diagnosis defined by morphologic findings and appearances. The major clinical manifestations include characteristic cutaneous lesions, including whitish scaly erythematous plaques and/or pustular or guttate lesions. There are several clinical forms of psoriasis, including plaque psoriasis, psoriasis guttate, psoriasis arthropathica, pustular psoriasis, psoriasis erythroderma, and inverse psoriasis. The most common type of psoriasis is psoriasis vulgaris, which accounts for 85–90% of all cases [1, 3].

Histologically, psoriasis is characterized by hyperproliferation and abnormal differentiation of keratinocytes; dilated, hyperplastic blood vessels; and inflammatory infiltration of lymphocytes mainly into the dermis. The skin patches are typically erythematous and scaly, which, in addition to the physical appearance, may result in psychological stress and poor quality of life. Like other systemic inflammatory diseases, psoriasis affects far more organs than the skin and often presents with chronic inflammatory responses in joints, nails, and other organs.

Immunological dysfunction in psoriasis involves cross talk between immune cells and nonimmune cells with cytokines. Several important types of immune cells in psoriasis have been found to play a role in pathogenesis, including Th1, Th17, and regulatory T cells. Corresponding cytokines that may be involved include interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-23, and IL-17. More recently, IL-9-secreting Th9 cells have been identified, and the inflammatory responses of keratinocytes, γδT cells, T regulatory cells, and other cell types in psoriasis have been explored. Emerging evidence indicates that new genetic variations and epigenetic modifications are associated with psoriatic disease [4, 5].

Advertisement

2. Immunological changes in psoriasis

Psoriasis is characterized by keratinocyte hyperproliferation and the abnormal infiltration of effector T cells, dendritic cells, neutrophils, and macrophages [6]. The effect of multiple cell types involved in psoriasis is mediated by a complex network of cytokines and their interactions.

Advertisement

3. Role of inflammatory cytokines in psoriasis

3.1. Interferons (IFN)

Type I interferons (IFNs), IFN-α and IFN-β, can suppress viral replication and stimulate immune reactions in response to viral infections; thus, they are potential mediators of antiviral host defense. Activated plasmacytoid dendritic cells (pDCs) preferentially produce type I IFNs following interactions between intracellular TLR7 and TLR9 with viral RNA and DNA [7, 8]. Type I IFN-α and IFN-β are not expressed in the normal skin but are produced in virally infected skin where pDCs are present, as well as in skin wounds where mechanical injury stimulates infiltration of pDCs and in lesional psoriatic skin where pDC-derived type I IFNs are sustained [9]. This stimulates myeloid DC phenotypic maturation and activation, enabling T-cell priming. Several studies have demonstrated that these cytokines are most relevant in the early phase of psoriasis, as demonstrated by the IFN-α signature in primary psoriatic plaques. Albanesi et al. [10] found that pDC infiltration in psoriatic skin correlates with the expression of markers typical of early stage of disease, whereas it is notably absent in chronic lesions. In this regard, blocking of type I IFN signaling may prevent the upregulation of T cells and development of non-lesional to lesional skin [9]. For downstream inflammatory pathways, type I IFNs modulate the production of IFN-γ and IL-17 and are involved in the differentiation and activation of T cells, particularly Th1 and Th17 cells [11] (Figure 1).

Figure 1.

The scheme of cytokine involvement in the pathogenesis of psoriasis and the mechanism of action of biologics.

Th1 cells are a potential source of IFN-γ, a type II interferon. Previously, the Th1 pathway was proposed to be the predominant pathogenic path for psoriasis [12]. Th1 cells, producing IFN-γ, are increased in the psoriatic lesional skin and peripheral blood and can be decreased by effective therapy. However, the potential role of IFN-γ became less important after the identification of a new key cytokine, Th17-producing IL-17 [13]. Selective blockage of IL-23-induced IL-17 leads to full recovery of psoriasis based on clinical, histological, and molecular markers [14].

IFN-γ acts on psoriatic keratinocytes and endothelial cells, leading to the activation and production of antimicrobial peptides (e.g., LL-37 cathelicidin and β-defensins). IFN-γ induces the cross phosphorylation of Janus kinase 1 (JAK1) and JAK3, resulting in the downstream activation of STAT3. Subsequent activation of STAT transcription factors is important for cell growth and is efficient for regulating many genes expressed in psoriatic lesions [15]. IFN-γ promotes the release of cytokines (IL-23, IL-1) and chemokines (CXCL10, CXCL11), as well as the expression of adhesion molecules from DCs, T cells, keratinocytes, and endothelial cells [16], thus promoting the recruitment of inflammatory cells to lesional plaques. Studies suggest that IFN-γ can be used as a biomarker for determining psoriasis severity and therapy evaluation because of the positive correlation between serum IFN-γ levels and PASI scores [17].

However, direct blockage of IFN-γ with a neutralizing antibody in patients with psoriasis was shown to have little or no therapeutic effect, indicating that IFN-γ does not directly participate the psoriasis phenotype [18]. It has been suggested that the IL-12/IFN-γ axis acts to suppress IL-17-modulated tissue injury [19, 20]. Consequently, continued expression of the IL-12/IFN-γ axis in disease while Th17 circuits are inhibited through IL-23 or IL-17 blockage may lead to better suppression and improvement of psoriasis [5].

3.2. TNF-α

TNF-α is involved in many inflammatory cutaneous diseases, including psoriasis. Several different cells can produce TNF-α in the context of skin inflammation, including keratinocytes, macrophages, T cells (Th1, Th17, and Th22 cells), and psoriatic DCs (particularly TIP-DCs) [5, 21, 22]. Several studies showed that circulating levels of TNF-α (in addition to IFN-γ and IL-12) are elevated in psoriasis patients and correlate with severity of disease [23, 24], although different studies have shown varying results [25].

The key effects of TNF-α are regulating the antigen-presenting ability of DCs and stimulation of T-cell infiltration. It has a variety of effects because there are two types of TNF receptors (TNFR), TNFR1 and TNFR2. TNFR1 is expressed on nearly all cell types, whereas TNFR2 is present predominantly on endothelial cells and hematopoietic cells. TNF-α acts in part by increasing the elevated level of active, phosphorylated NF-κB, a crucial transcription factor involved in psoriatic pathogenesis [26]. TNF-α possesses proinflammatory properties; it activates the expression of C-reactive protein (which is a part of the acute phase response) and several cytokines such as IL-6 (which induces keratinocyte hyperproliferation and T-cell proliferation) and IL-23 (which is a potential mediator synthesized from DCs in psoriasis to stimulate IL-17 production). TNF-α also induces several chemokines including CXCL8/IL-8 (which recruits neutrophil infiltration) and CCL20 (which recruits myeloid DCs and Th17 cells). Therefore, TNF-α is an important regulator of the IL-23/Th17 axis in psoriasis. The multifaceted role of TNF-α has been evaluated in clinical trials of TNF-α antagonists in psoriasis patients, revealing their clinical efficacy [27].

TNF-α-targeting agents were approved for rheumatoid arthritis treatment years before being approved for psoriasis therapy. Inhibition of TNF-α signaling has been broadly used in targeted biological treatment of psoriasis. The three biologics currently approved for the treatment of moderate to severe psoriasis are infliximab, adalimumab, and etanercept (Table 1). Effective treatment with TNF antagonists downregulates T-cell and DC numbers and decreases their cytokine levels [27, 28]. Infliximab, a chimeric monoclonal antibody, suppresses TNF-α biologic activity by neutralizing both soluble and membrane-bound forms of TNF-α [29]. Blocking this cytokine activity with infliximab has been demonstrated to rapidly normalize keratinocyte differentiation and reduce the number of epidermal thickness, epidermal T-cell infiltration, and intracellular adhesion molecules of psoriatic plaques, such as e-selectin and VCAM [3032]. Adalimumab is a fully humanized IgG1 monoclonal antibody [33] that binds with high specificity and affinity to human TNF-α. Adalimumab has been suggested as an effective treatment for moderate to severe chronic plaque psoriasis for up to 12 weeks of therapy [34]. Upon binding to this cytokine, adalimumab neutralizes biologic activities by blocking its interaction with the p55 and p75 cell-surface TNF receptors to inhibit TNF-involved biologic responses [35]. Etanercept, a fusion protein consisting of the extracellular ligand-binding domain of TNF-α receptors and Fc portion of human immunoglobulin G, performs its immune function by neutralizing soluble TNF-α and TNF-β (or known as lymphotoxin-α) [36], which also reduces IL-23, and by suppressing Th17 downstream molecules, including IL-17, IL-22, CC chemokine ligand (CCL) 20, and β-defensin 4 [27]. Particularly, successful treatment was found to be associated with the suppression of genes related to the differentiation and function of Th17 cells. Moreover, inhibition of the IL-23 and Th17 axis led to downregulation of IFN-γ-related genes associated with psoriasis resolution [27, 28]. Furthermore, etanercept can decrease lesional DC expression of co-stimulatory molecules in vitro, impairing DC-T-cell interactions and allogenic T-cell activation [27].

Drug nameDrug targetAgent typeAdministrationEfficacy (% with PASI 75)ReferencesStage of development*
Infliximab (Remicade)TNF-αChimeric TNF-α monoclonal antibody2-h i.v. infusion (5 mg/kg) at weeks 0, 2, and 6 and then every 8 weeks75–88 at 10 weeksGottlieb et al. [31]; Reich et al. [29]; Menter et al. [106]Approved
Adalimumab (Humira)TNF-αHumanized TNF-α monoclonal antibodys.c. 80 mg at week 0 and then 40 mg every 2 weeks53–80 at 12 weeksGordon et al. [34]; Menter et al. [35]; Saurat et al. [107]Approved
Etanercept (Enbrel)TNF-αSoluble TNF-α receptor-igg fusion proteins.c 50 mg every 2 weeks for 3 months and then 50 mg weekly47–49 at 12 weeksLeonardi et al. [108]; Papp et al. [36]; Tyring et al. [109]Approved
Ustekinumab (Stelara, CNTO1275)p40 subunit of IL-12/IL-23Humanized p40 monoclonal antibodys.c. (1) 45 mg or (2) 90 mg weekly for 12 weeks(1) 66.7–67.1 or (2) 66.4–75.7 at 12 weeksLeonardi et al. [51]; Papp et al. [50]Approved
Briakinumab (ABT-874)p40 subunit of IL-12/IL-23Humanized p40 monoclonal antibodys.c. 200 mg at weeks 0 and 4 and then 100 mg at week 880.6–81.9 at 12 weeksGordon et al. [52]; Gottlieb et al. [53]; Strober et al. [54]Terminated
Tildrakizumab (MK-3222)p19 subunit of IL-23Humanized p19 IgG1 monoclonal antibodys.c (1) 5 mg or (2) 25 mg or (3) 100 or (4) 200 mg at weeks 0 and 4 and then every 12 weeks thereafter(1) 33.3 or (2) 64.4 or (3) 66.3 or (4) 74.4 at 16 weeksPapp et al. [58]Phase III studies ongoing
Guselkumab (CNTO1959)p19 subunit of IL-23Humanized p19 IgG1 monoclonal antibodys.c (1) 5 mg at weeks 0 and 4 and then every 12 weeks thereafter or (2) 15 mg every 8 weeks or (3) 50 at weeks 0 and 4 and then every 12 weeks thereafter or (4) 100 mg at weeks 0 and 4 and then every 12 weeks there after (5) 200 mg at weeks 0 and 4 and then every 12 weeks thereafter(1) 44 or (2) 76 or (3) 81 or (4) 79 (5) 81 at 16 weeksGordon et al. [59]Finished phase III trial
Secukinumab (Cosentyx, AIN457)IL-17AHumanized IL-17A IgG1 monoclonal antibodys.c. (1) 150 mg or (2) 300 mg at weeks 0, 1, 2, 3, and 4 and every 4 weeks(1) 67–71.6 or (2) 77.1–81.6 at 12 weeksLangley et al. [68]Approved

Table 1.

Biologic drugs for moderate to severe psoriasis at 10–16 weeks.

*State of development in the United States, as of January 2017.


The clinical advantage of TNF-α suppression is related to blockage of the IL-23/Th17 axis. Furthermore, TNF-α and Th17 have been suggested to synergistically stimulate the production of several keratinocyte proinflammatory mediators involved in psoriasis [37]. Therefore, blocking either or both TNF-α and IL-23-Th17 pathways may affect immunopathogenic molecules involved in psoriasis.

Anti-TNF-α therapies for psoriasis are very effective. However, the diverse roles of this cytokine cause various drug-associated adverse effects. Patients treated with these biologic agents show an increased incidence of reactivating latent tuberculosis [38] and emerging serious infections (such as sepsis and opportunistic infections) [39]. Additionally, some studies have linked these anti-TNF drugs, particularly when used in combination with other drugs, to an increased risk of malignancies such as lymphoma [4042].

3.3. IL-12/IL-23

IL-12 and IL-23 are heterodimeric pleiotropic proteins that share a common p40 subunit (encoded by IL12B) and are thought to be essential for controlling the differentiation of Th1 and Th17 cells, respectively. The second distinct subunit of IL-12 is the p35 subunit, and the second unique subunit of IL-23 is the p19 subunit (encoded by IL23A). Expression of the p19 and p40 subunits was found to be significantly increased in psoriatic skin lesions, while the p35 subunit was not [43, 44], suggesting that IL-23 is important in the pathogenesis of psoriasis. Further support from clinical trials revealed that the expression of IL-12/IL-23 was decreased following psoriasis treatment [4547]. IL-23 and IL-12 are primarily secreted by DCs and macrophages and play a crucial role in psoriatic pathogenesis by regulating Th17 and Th1 cells, including the activation and differentiation of effector T cells, stimulation of keratinocytes, and upregulation of TNF-α expression in psoriatic plaques [43, 48]. IL-23 binds to IL-23R, which is correlated with Jak2 and Tyk2. Binding of its receptor stimulates a signaling circuit via STAT3 activation.

Anti-IL-12/IL-23 and anti-IL-23 drugs are highly effective treatments for psoriasis (Table 1) [49]. Recently, the only published results from clinical trials describe two agents of the p40 subunit inhibitors, ustekinumab and briakinumab. Ustekinumab is a human IgG1 monoclonal antibody that neutralizes the shared p40 subunit of IL-12 and IL-23. The agent prevents the interaction of IL-23 and IL-12 with their cell-surface receptors, blocking the Th17 and Th1 signaling cascades. It has been demonstrated to be efficacious for moderate to severe psoriasis [50, 51]. Clinical trials showed that another fully human anti-IL-12/IL-23p40 monoclonal antibody, briakinumab, was also efficacious for the disease [5254]. However, after phase III trials, safety results concerning a possible increased risk of major adverse cardiovascular events (myocardial infarction, cerebrovascular accident, and cardiac death) with the use of briakinumab let to cessation of its development and withdrawal of the application in 2011 [55, 56].

The structurally related p19 subunit of IL-23 has recently emerged as an attractive target for moderate to severe psoriasis treatment, although these drugs have not been FDA approved [57]. Several agents targeting the p19 subunit are under investigation in clinical trials. Tildrakizumab is a humanized IgG1κ that binds to the unique p19 subunit of IL-23 [58]. This agent was effective in treating moderate to severe plaque psoriasis in a phase IIb clinical trial. Phase III studies are currently underway. Similarly, phase III trials of another fully human IgG1λ monoclonal p19 antibody, guselkumab, are currently a success [59].

3.4. IL-17

IL-17, the main cytokine effector of Th17 cells, is an important cytokine in the pathogenesis of psoriasis. Neutrophils, mast cells, and natural killer (NK) cells also produce IL-17. It is thought to be a proximal regulator of psoriatic cutaneous inflammation and plays a key role in bridging the innate and adaptive immune responses. The IL-17 family comprises six subsets of homo- and heterodimeric cytokines: IL-17A, IL-17B, IL-17C, IL-D, IL-17E, and IL-17F. IL-17A and IL-17F are regarded as the most relevant subtypes in psoriasis. IL-17 is widely thought to be a direct regulator that stimulates keratinocyte proliferation and inhibits keratinocyte differentiation via the antimicrobial protein REG3A, a mediator with antimicrobial functions involved in wound repair [60]. IL-17 mRNA and protein levels are upregulated in lesional psoriatic plaques and/or blood samples from patients [61, 62]. Lowes et al. [63] demonstrated that psoriatic T cells generate large amounts of IL-17 ex vivo, but T cells from the normal healthy skin did not produce IL-17 under the same conditions.

Keratinocytes are the main target of IL-17A in psoriasis. The IL-17 receptor (IL-17R; consisting of two IL-17RA subunits complexed with one IL-17RC subunit) is expressed on the surface of keratinocytes throughout the epidermis and on scattered dermal cells (DCs, dermal fibroblasts, and endothelial cells) in the psoriatic skin [64]. The interaction between IL-17A and its receptor leads to the production of antimicrobial peptides (AMPs); proinflammatory cytokines such as IL-1, IL-6, IL-23, and IL-19; chemokines; and mediators of tissue injury [62].

IL-17A stimulates the expression of AMPs, including β-defensin and S100A family members, and thus activates the innate immune system [64]. A previous study demonstrated that IL-17A activates the production of multiple chemokines, including CCL20, CXCL1, CXCL2, CXCL3, CXCL5, and CXCL8/IL-8 [37, 6466]. In addition to stimulating the recruitment and activation of neutrophils, IL-8 acts as a chemotactic factor for NK cells and T cells. CCL20 from human keratinocytes may direct the recruitment of CCR6-positive cells to the skin. Most Th17 cells express CCR6. Therefore, keratinocytes activate Th17-cell recruitment and increase the production of IL-17, promoting a positive feedback loop that maintains the inflammatory disease response [65, 67]. Moreover, CCL20 combined with ICAM-1 can facilitate the recruitment of DCs and T cells in psoriasis. IL-17A and TNF-α act synergistically on psoriatic keratinocytes, causing further production of TNF-α and other proinflammatory mediators.

Several drugs are available or under development that target IL-17 and its pathway. The potential role of the IL-17 pathway has been revealed in psoriatic clinical trials, which showed dramatic improvement. Secukinumab and ixekizumab are humanized IgG1κ and IgG4 monoclonal antibodies that bind and neutralize the IL-17A cytokine. [68, 69]. Secukinumab was approved by the FDA for moderate-to-severe treatment psoriasis in January 2015. In phase III studies, double-blind, 52-week trials, this agent achieved a PASI75 in 67.0–71.6% of patients at a 150-mg dose and 77.1–81.6% of patients with a 300-mg dose (Table 1) [68]. The common adverse effects associated with this agent are headache, nasopharyngitis, and upper respiratory tract infections, which are similar to those of other biologics. Ixekizumab, a monoclonal antibody specific for IL-17A, has been shown to be effective for treating psoriasis [70]. This biologic inhibits the expression of cytokines and chemokines involved in the IL-17 pathway [71]. Ixekizumab complete responses were observed in 30.8–35.0% of psoriasis patients and PASI90 in 59.7–65.3% after 12 weeks [69]. These results agree with recent findings that IL-17-producing cells are clearly present in the inflammatory infiltrate. Similar to prior biologic agents, the most commonly reported side effects were nasopharyngitis and injection site reactions. No serious adverse effects were observed [72]. Brodalumab, a unique fully human monoclonal antibody targeting the IL-17 receptor A (IL-17RA), is the newest biologic that has been FDA approved for psoriasis treatment. It binds with high affinity to IL-17RA and blocks the biological activity of IL-17A, IL-17F, and IL-25 (IL-17E), suppressing the downstream effect of IL-17 [73, 74]. In phase III trials to treat psoriasis, brodalumab achieved PASI75 in 83–86% of patients with a 210-mg dose and 60–69% of patients with a 140-mg dose and PASI 90 in 69–70% after 12 weeks [75, 76]. The most common side effects were nasopharyngitis, upper respiratory infection, and injection site erythema [74]. Based on published results, drugs targeting IL-17 appear to have highly positive effects in moderate-to-severe psoriasis patients with no serious major side effects. Nevertheless, longer-term studies are needed.

3.5. IL-22

IL-22 belongs to the IL-10 family of cytokines, and its receptor (IL-22R) is a complex of two chains (IL-10R and IL-22RA1), which are exclusively expressed on epithelial cells such as keratinocytes [77]. Elevated levels of IL-22 mRNA in the lesional skin of psoriasis and serum IL-22 have been observed [7880]. Its expression is also decreased after treatment with anti-psoriatic agents [80]. IL-22, produced from Th22 and Th17 cells, induces keratinocyte hyperproliferation, differentiation, migration, and dermal infiltration through STAT3 activation in vivo and in vitro [81]. It also mediates proinflammatory cytokine and AMP production [78, 82].

IL-22 in the human skin can stimulate keratinocytes in various ways. Combined with IL-17, IL-22 can induce AMP production by keratinocytes [83] and parakeratosis and acanthosis by increasing keratinocyte proliferation and inhibit keratinocyte differentiation during part of the tissue-remodeling phase of wound repair, which are observed in psoriasis [84].

Zheng et al. [81] reported that IL-23-induced epidermal hyperplasia in a murine model of psoriasis was dependent on IL-22, and blocking IL-22 in vivo or genetic deletion resulted in downregulation of IL-23-mediated epidermal hyperplasia. Therefore, the important association between the IL-23/Th17 axis and IL-22/Th22 is supported by these studies. However, trials of a human monoclonal antibody targeted against IL-22, fezakinumab, were discontinued because initial processes revealed that the efficacy endpoint could not be achieved [85]. The negative data from these studies suggest that this cytokine is not as critical to psoriasis immunopathogenesis as had initially been considered in earlier studies.

3.6. IL-9

IL-9 is a member of the IL-2 cytokine family. Singh et al. demonstrated markedly elevated expression of IL-9 in the lesional skin of psoriasis patients compared to control subjects. They found increased IL-9R and IL-9 expression in the psoriatic skin and observed a Th17-related inflammatory response after intradermal IL-9 injection in a mouse model [86]. IL-9 is a proinflammatory cytokine that stimulates the production of IL-17, IL-13, IFN-γ, and TNF-α in psoriasis. Both Th9 and Th17 cells are sources of IL-9.

3.7. IL-33

Interleukin-33 is a recently discovered mediator of the IL-1 family [87]. IL-33 mRNA is constitutively expressed in several tissues but is predominantly distributed in epithelial cells, keratinocytes, fibroblasts, DCs, smooth muscle cells, and macrophages. Interestingly, IL-33 specifically localizes to the nucleus of endothelial cells along the vessels and epithelial cells of tissue exposed to the environment [8890].

The IL-33 receptor is selectively expressed on various cell types, including T-helper-cell (Th) type 2, mast cells, eosinophils, basophils, dendritic cells, group 2 innate lymphoid cells, keratinocytes, and invariant NKT cells [87, 9194].

IL-33 can act both as a released cytokine, activating ST2L, and as a nuclear-binding factor, regulating gene transcription [95, 96]. Balato et al. [97] showed that IL-33 is present in both the nucleus and cytoplasm of psoriatic keratinocytes. The structure of IL-33 has been determined, and it exhibits the ability to act both as an extracellular cytokine stimulating the ST2L receptor and an intracellular factor controlling gene transcription. However, the role of IL-33 in psoriasis remains unclear.

Many recent studies have shown that IL-33 expression is increased in the lesional skin of psoriasis compared to the normal skin [91, 97, 98]. Hueber et al. [99] demonstrated that IL-33 and ST2 expression are upregulated in human lesional psoriatic plaques compared to the perilesional and normal healthy skin. Moreover, IL-33 is strongly expressed in the nuclei of keratinocytes in psoriasis, which is considered to be a Th1- and Th17-mediated disease, compared with atopic dermatitis which is a Th2-related disease and lichen planus which is related to Th1 cells [96]. We previously demonstrated that IL-17A induces IL-33 expression in normal human epidermal keratinocytes, suggesting that IL-17 in the lesional skin of psoriasis can induce IL-33 expression in the epidermis [91].

Relatively few studies have demonstrated the pathogenic association between IL-33 and psoriasis. Suttle et al. [100] reported decreased IL-33 immunostaining in biopsies in Koebner-positive psoriasis patients, which can reflect the release of IL-33 after skin injury by tape stripping. Interestingly, the proinflammatory cytokine TNF-α dose- and time-dependently activated IL-33 mRNA expression in normal skin cultures ex vivo. Similarly, TNF-α may stimulate the gene expression of IL-33 in normal human epidermal sheets and psoriatic skin [101]. Moreover, the levels of IL-33 were significantly reduced after TNF-α inhibitor therapy [101, 102].

Furthermore, Mitsui et al. [103] recently found that serum IL-33 levels are significantly elevated in patients with psoriasis and are particularly correlated with serum TNF-α levels; this elevation was decreased after anti-TNF-α treatment. They suggested that IL-33 is a general indicator of inflammation in psoriasis. In contrast, Tamagawa-Mineoka et al. [104], Balato et al. [97], and Talabot-Ayer et al. [105] did not detect serum IL-33 expression in psoriasis patients.

References

  1. 1. Nestle FO, Kaplan DH, Barker J. Psoriasis. New England Journal of Medicine. 2009;361(5):496–509
  2. 2. Perera GK, Di Meglio P, Nestle FO. Psoriasis. Annual Review of Pathology. 2012;7:385–422
  3. 3. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370(9583):263–271
  4. 4. Sabat R, Philipp S, Hoflich C, Kreutzer S, Wallace E, Asadullah K, et al. Immunopathogenesis of psoriasis. Experimental Dermatology. 2007;16(10):779–798
  5. 5. Kim J, Krueger JG. The immunopathogenesis of psoriasis. Dermatologic Clinics. 2015;33(1):13–23
  6. 6. Schon MP, Boehncke WH. Psoriasis. New England Journal of Medicine. 2005;352(18):1899–1912
  7. 7. Ganguly D, Chamilos G, Lande R, Gregorio J, Meller S, Facchinetti V, et al. Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. Journal of Experimental Medicine. 2009;206(9):1983–1994
  8. 8. Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature. 2007;449(7162):564–569
  9. 9. Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M, Boyman O, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. Journal of Experimental Medicine. 2005;202(1):135–143
  10. 10. Albanesi C, Scarponi C, Bosisio D, Sozzani S, Girolomoni G. Immune functions and recruitment of plasmacytoid dendritic cells in psoriasis. Autoimmunity. 2010;43(3):215–219
  11. 11. Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, Lauerma A, et al. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. Journal of Experimental Medicine. 2010;207(13):2921–2930
  12. 12. Lew W, Bowcock AM, Krueger JG. Psoriasis vulgaris: Cutaneous lymphoid tissue supports T-cell activation and “Type 1” inflammatory gene expression. Trends in Immunology. 2004;25(6):295–305
  13. 13. Lowes MA, Russell CB, Martin DA, Towne JE, Krueger JG. The IL-23/T17 pathogenic axis in psoriasis is amplified by keratinocyte responses. Trends in Immunology. 2013;34(4):174–181
  14. 14. Sofen H, Smith S, Matheson RT, Leonardi CL, Calderon C, Brodmerkel C, et al. Guselkumab (an IL-23-specific mAb) demonstrates clinical and molecular response in patients with moderate-to-severe psoriasis. Journal of Allergy and Clinical Immunology. 2014;133(4):1032–1040
  15. 15. Johnson-Huang LM, Suarez-Farinas M, Pierson KC, Fuentes-Duculan J, Cueto I, Lentini T, et al. A single intradermal injection of IFN-gamma induces an inflammatory state in both non-lesional psoriatic and healthy skin. Journal of Investigative Dermatology. 2012;132(4):1177–1187
  16. 16. Madonna S, Scarponi C, Sestito R, Pallotta S, Cavani A, Albanesi C. The IFN-gamma-dependent suppressor of cytokine signaling 1 promoter activity is positively regulated by IFN regulatory factor-1 and Sp1 but repressed by growth factor independence-1b and Kruppel-like factor-4, and it is dysregulated in psoriatic keratinocytes. Journal of Immunology. 2010;185(4):2467–2481
  17. 17. Abdallah MA, Abdel-Hamid MF, Kotb AM, Mabrouk EA. Serum interferon-gamma is a psoriasis severity and prognostic marker. Cutis. 2009;84(3):163–168
  18. 18. Harden JL, Johnson-Huang LM, Chamian MF, Lee E, Pearce T, Leonardi CL, et al. Humanized anti-IFN-gamma (HuZAF) in the treatment of psoriasis. Journal of Allergy and Clinical Immunology. 2015;135(2):553–556
  19. 19. Zhang J. Yin and yang interplay of IFN-gamma in inflammation and autoimmune disease. Journal of Clinical Investigation. 2007;117(4):871–873
  20. 20. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421(6924):744–748
  21. 21. Zaba LC, Fuentes-Duculan J, Eungdamrong NJ, Johnson-Huang LM, Nograles KE, White TR, et al. Identification of TNF-related apoptosis-inducing ligand and other molecules that distinguish inflammatory from resident dendritic cells in patients with psoriasis. Journal of Allergy and Clinical Immunology. 2010;125(6):1261–1268. e9
  22. 22. Lowes MA, Chamian F, Abello MV, Fuentes-Duculan J, Lin SL, Nussbaum R, et al. Increase in TNF-alpha and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proceedings of the National Academy of Sciences of the United States of America. 2005;102(52):19057–19062
  23. 23. Arican O, Aral M, Sasmaz S, Ciragil P. Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediators of Inflammation. 2005;2005(5):273–279
  24. 24. Abanmi A, Al Harthi F, Al Agla R, Khan HA, Tariq M. Serum levels of proinflammatory cytokines in psoriasis patients from Saudi Arabia. International Journal of Dermatology. 2005;44(1):82–83
  25. 25. Jacob SE, Nassiri M, Kerdel FA, Vincek V. Simultaneous measurement of multiple Th1 and Th2 serum cytokines in psoriasis and correlation with disease severity. Mediators of Inflammation. 2003;12(5):309–313
  26. 26. Goldminz AM, Au SC, Kim N, Gottlieb AB, Lizzul PF. NF-kappaB: An essential transcription factor in psoriasis. Journal of Dermatological Science. 2013;69(2):89–94
  27. 27. Zaba LC, Cardinale I, Gilleaudeau P, Sullivan-Whalen M, Suarez-Farinas M, Fuentes-Duculan J, et al. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. Journal of Experimental Medicine. 2007;204(13):3183–3194
  28. 28. Zaba LC, Suarez-Farinas M, Fuentes-Duculan J, Nograles KE, Guttman-Yassky E, Cardinale I, et al. Effective treatment of psoriasis with etanercept is linked to suppression of IL-17 signaling, not immediate response TNF genes. Journal of Allergy and Clinical Immunology. 2009;124(5):1022–1030.e1–395
  29. 29. Reich K, Nestle FO, Papp K, Ortonne JP, Evans R, Guzzo C, et al. Infliximab induction and maintenance therapy for moderate-to-severe psoriasis: A phase III, multicentre, double-blind trial. Lancet. 2005;366(9494):1367–1374
  30. 30. Gottlieb AB, Masud S, Ramamurthi R, Abdulghani A, Romano P, Chaudhari U, et al. Pharmacodynamic and pharmacokinetic response to anti-tumor necrosis factor-alpha monoclonal antibody (infliximab) treatment of moderate to severe psoriasis vulgaris. Journal of the American Academy of Dermatology. 2003;48(1):68–75
  31. 31. Gottlieb AB, Evans R, Li S, Dooley LT, Guzzo CA, Baker D, et al. Infliximab induction therapy for patients with severe plaque-type psoriasis: A randomized, double-blind, placebo-controlled trial. Journal of the American Academy of Dermatology. 2004;51(4):534–542
  32. 32. Chaudhari U, Romano P, Mulcahy LD, Dooley LT, Baker DG, Gottlieb AB. Efficacy and safety of infliximab monotherapy for plaque-type psoriasis: A randomised trial. Lancet. 2001;357(9271):1842–1847
  33. 33. Calabrese LH. Molecular differences in anticytokine therapies. Clinical and Experimental Rheumatology. 2003;21(2):241–248
  34. 34. Gordon KB, Langley RG, Leonardi C, Toth D, Menter MA, Kang S, et al. Clinical response to adalimumab treatment in patients with moderate to severe psoriasis: Double-blind, randomized controlled trial and open-label extension study. Journal of the American Academy of Dermatology. 2006;55(4):598–606
  35. 35. Menter A, Tyring SK, Gordon K, Kimball AB, Leonardi CL, Langley RG, et al. Adalimumab therapy for moderate to severe psoriasis: A randomized, controlled phase III trial. Journal of the American Academy of Dermatology. 2008;58(1):106–115
  36. 36. Papp KA, Tyring S, Lahfa M, Prinz J, Griffiths CE, Nakanishi AM, et al. A global phase III randomized controlled trial of etanercept in psoriasis: Safety, efficacy, and effect of dose reduction. British Journal of Dermatology. 2005;152(6):1304–1312
  37. 37. Chiricozzi A, Guttman-Yassky E, Suárez-Fariñas M, Nograles KE, Tian S, Cardinale I, et al. Integrative responses to IL-17 and TNF-&α in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. Journal of Investigative Dermatology. 2011;131(3):677–687
  38. 38. Mankia S, Peters JE, Kang S, Moore S, Ehrenstein MR. Tuberculosis and anti-TNF treatment: Experience of a central London hospital. Clinical Rheumatology. 2011;30(3):399–401
  39. 39. Galloway JB, Hyrich KL, Mercer LK, Dixon WG, Fu B, Ustianowski AP, et al. Anti-TNF therapy is associated with an increased risk of serious infections in patients with rheumatoid arthritis especially in the first 6 months of treatment: Updated results from the British Society for Rheumatology Biologics Register with special emphasis on risks in the elderly. Rheumatology (Oxford). 2011;50(1):124–131
  40. 40. Mariette X, Tubach F, Bagheri H, Bardet M, Berthelot JM, Gaudin P, et al. Lymphoma in patients treated with anti-TNF: Results of the 3-year prospective French RATIO registry. Annals of the Rheumatic Diseases. 2010;69(2):400–408
  41. 41. Lakatos PL, Miheller P. Is there an increased risk of lymphoma and malignancies under anti-TNF therapy in IBD? Current Drug Targets. 2010;11(2):179–186
  42. 42. Herrinton LJ, Liu L, Weng X, Lewis JD, Hutfless S, Allison JE. Role of thiopurine and anti-TNF therapy in lymphoma in inflammatory bowel disease. American Journal of Gastroenterology. 2011;106(12):2146–2153
  43. 43. Lee E, Trepicchio WL, Oestreicher JL, Pittman D, Wang F, Chamian F, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. Journal of Experimental Medicine. 2004;199(1):125–130
  44. 44. Tonel G, Conrad C, Laggner U, Di Meglio P, Grys K, McClanahan TK, et al. Cutting edge: A critical functional role for IL-23 in psoriasis. Journal of Immunology. 2010;185(10):5688–5691
  45. 45. Chamian F, Lowes MA, Lin SL, Lee E, Kikuchi T, Gilleaudeau P, et al. Alefacept reduces infiltrating T cells, activated dendritic cells, and inflammatory genes in psoriasis vulgaris. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(6):2075–2080
  46. 46. Gottlieb AB, Chamian F, Masud S, Cardinale I, Abello MV, Lowes MA, et al. TNF inhibition rapidly down-regulates multiple proinflammatory pathways in psoriasis plaques. Journal of Immunology. 2005;175(4):2721–2729
  47. 47. Piskin G, Tursen U, Sylva-Steenland RM, Bos JD, Teunissen MB. Clinical improvement in chronic plaque-type psoriasis lesions after narrow-band UVB therapy is accompanied by a decrease in the expression of IFN-gamma inducers – IL-12, IL-18 and IL-23. Experimental Dermatology. 2004;13(12):764–772
  48. 48. Zhou L, Ivanov, II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nature Immunology. 2007;8(9):967–974
  49. 49. Gandhi M, Alwawi E, Gordon KB. Anti-p40 antibodies ustekinumab and briakinumab: Blockade of interleukin-12 and interleukin-23 in the treatment of psoriasis. Seminars in Cutaneous Medicine and Surgery. 2010;29(1):48–52
  50. 50. Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeilding N, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371(9625):1675–1684
  51. 51. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet. 2008;371(9625):1665–1674
  52. 52. Gordon KB, Langley RG, Gottlieb AB, Papp KA, Krueger GG, Strober BE, et al. A phase III, randomized, controlled trial of the fully human IL-12/23 mAb briakinumab in moderate-to-severe psoriasis. Journal of Investigative Dermatology. 2012;132(2):304–314
  53. 53. Gottlieb AB, Leonardi C, Kerdel F, Mehlis S, Olds M, Williams DA. Efficacy and safety of briakinumab vs. etanercept and placebo in patients with moderate to severe chronic plaque psoriasis. British Journal of Dermatology. 2011;165(3):652–660
  54. 54. Strober BE, Crowley JJ, Yamauchi PS, Olds M, Williams DA. Efficacy and safety results from a phase III, randomized controlled trial comparing the safety and efficacy of briakinumab with etanercept and placebo in patients with moderate to severe chronic plaque psoriasis. British Journal of Dermatology. 2011;165(3):661–668
  55. 55. Langley RG, Papp K, Gottlieb AB, Krueger GG, Gordon KB, Williams D, et al. Safety results from a pooled analysis of randomized, controlled phase II and III clinical trials and interim data from an open-label extension trial of the interleukin-12/23 monoclonal antibody, briakinumab, in moderate to severe psoriasis. Journal of the European Academy of Dermatology and Venereology. 2013;27(10):1252–1261
  56. 56. Ryan C, Leonardi CL, Krueger JG, Kimball AB, Strober BE, Gordon KB, et al. Association between biologic therapies for chronic plaque psoriasis and cardiovascular events: A meta-analysis of randomized controlled trials. The Journal of the American Medical Association. 2011;306(8):864–871
  57. 57. Kofoed K, Skov L, Zachariae C. New drugs and treatment targets in psoriasis. Acta Dermato-Venereologica. 2015;95(2):133–139
  58. 58. Papp K, Thaci D, Reich K, Riedl E, Langley RG, Krueger JG, et al. Tildrakizumab (MK-3222), an anti-interleukin-23p19 monoclonal antibody, improves psoriasis in a phase IIb randomized placebo-controlled trial. British Journal of Dermatology. 2015;173(4):930–939
  59. 59. Gordon KB, Duffin KC, Bissonnette R, Prinz JC, Wasfi Y, Li S, et al. A phase 2 trial of guselkumab versus adalimumab for plaque psoriasis. New England Journal of Medicine. 2015;373(2):136–144
  60. 60. Lai Y, Li D, Li C, Muehleisen B, Radek KA, Park HJ, et al. The antimicrobial protein REG3A regulates keratinocyte proliferation and differentiation after skin injury. Immunity. 2012;37(1):74–84
  61. 61. Johansen C, Usher PA, Kjellerup RB, Lundsgaard D, Iversen L, Kragballe K. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. British Journal of Dermatology. 2009;160(2):319–324
  62. 62. Lynde CW, Poulin Y, Vender R, Bourcier M, Khalil S. Interleukin 17A: Toward a new understanding of psoriasis pathogenesis. Journal of the American Academy of Dermatology. 2014;71(1):141–150
  63. 63. Lowes MA, Kikuchi T, Fuentes-Duculan J, Cardinale I, Zaba LC, Haider AS, et al. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. Journal of Investigative Dermatology. 2008;128(5):1207–1211
  64. 64. Nograles KE, Zaba LC, Guttman-Yassky E, Fuentes-Duculan J, Suarez-Farinas M, Cardinale I, et al. Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways. British Journal of Dermatology. 2008;159(5):1092–1102
  65. 65. Harper EG, Guo C, Rizzo H, Lillis JV, Kurtz SE, Skorcheva I, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: Implications for psoriasis pathogenesis. Journal of Investigative Dermatology. 2009;129(9):2175–2183
  66. 66. Homey B, Dieu-Nosjean MC, Wiesenborn A, Massacrier C, Pin JJ, Oldham E, et al. Up-regulation of macrophage inflammatory protein-3 alpha/CCL20 and CC chemokine receptor 6 in psoriasis. Journal of Immunology. 2000;164(12):6621–6632
  67. 67. Ramirez-Carrozzi V, Sambandam A, Luis E, Lin Z, Jeet S, Lesch J, et al. IL-17C regulates the innate immune function of epithelial cells in an autocrine manner. Nature Immunology. 2011;12(12):1159–1166
  68. 68. Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, et al. Secukinumab in plaque psoriasis – results of two phase 3 trials. New England Journal of Medicine. 2014;371(4):326–338
  69. 69. Griffiths CE, Reich K, Lebwohl M, van de Kerkhof P, Paul C, Menter A, et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): Results from two phase 3 randomised trials. Lancet. 2015;386(9993):541–551
  70. 70. Gordon KB, Blauvelt A, Papp KA, Langley RG, Luger T, Ohtsuki M, et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque psoriasis. New England Journal of Medicine. 2016;375(4):345–356
  71. 71. Krueger JG, Fretzin S, Suarez-Farinas M, Haslett PA, Phipps KM, Cameron GS, et al. IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis. Journal of Allergy and Clinical Immunology. 2012;130(1):145–154.e9
  72. 72. Leonardi C, Matheson R, Zachariae C, Cameron G, Li L, Edson-Heredia E, et al. Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. New England Journal of Medicine. 2012;366(13):1190–1199
  73. 73. Farahnik B, Beroukhim K, Abrouk M, Nakamura M, Zhu TH, Singh R, et al. Brodalumab for the treatment of psoriasis: A review of phase III trials. Dermatology and Therapy (Heidelb). 2016;6(2):111–124
  74. 74. Papp KA, Leonardi C, Menter A, Ortonne JP, Krueger JG, Kricorian G, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. New England Journal of Medicine. 2012;366(13):1181–1189
  75. 75. Lebwohl M, Strober B, Menter A, Gordon K, Weglowska J, Puig L, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. New England Journal of Medicine. 2015;373(14):1318–1328
  76. 76. Papp K, Menter A, Strober B, Kricorian G, Thompson EH, Milmont CE, et al. Efficacy and safety of brodalumab in subpopulations of patients with difficult-to-treat moderate-to-severe plaque psoriasis. Journal of the American Academy of Dermatology. 2015;72(3):436–439.e1
  77. 77. Mashiko S, Bouguermouh S, Rubio M, Baba N, Bissonnette R, Sarfati M. Human mast cells are major IL-22 producers in patients with psoriasis and atopic dermatitis. Journal of Allergy and Clinical Immunology. 2015;136(2):351–359.e1
  78. 78. Wolk K, Witte E, Wallace E, Docke WD, Kunz S, Asadullah K, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: A potential role in psoriasis. European Journal of Immunology. 2006;36(5):1309–1323
  79. 79. Coimbra S, Oliveira H, Reis F, Belo L, Rocha S, Quintanilha A, et al. Interleukin (IL)-22, IL-17, IL-23, IL-8, vascular endothelial growth factor and tumour necrosis factor-alpha levels in patients with psoriasis before, during and after psoralen-ultraviolet A and narrowband ultraviolet B therapy. British Journal of Dermatology. 2010;163(6):1282–1290
  80. 80. Boniface K, Lecron JC, Bernard FX, Dagregorio G, Guillet G, Nau F, et al. Keratinocytes as targets for interleukin-10-related cytokines: A putative role in the pathogenesis of psoriasis. European Cytokine Network. 2005;16(4):309–319
  81. 81. Zheng Y, Danilenko DM, Valdez P, Kasman I, Eastham-Anderson J, Wu J, et al. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature. 2007;445(7128):648–651
  82. 82. Eyerich S, Eyerich K, Pennino D, Carbone T, Nasorri F, Pallotta S, et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. Journal of Clinical Investigation. 2009;119(12):3573–3585
  83. 83. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, Collins M, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. Journal of Experimental Medicine. 2006;203(10):2271–2279
  84. 84. Boniface K, Bernard FX, Garcia M, Gurney AL, Lecron JC, Morel F. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. Journal of Immunology. 2005;174(6):3695–3702
  85. 85. Gudjonsson JE, Johnston A, Ellis CN. Novel systemic drugs under investigation for the treatment of psoriasis. Journal of the American Academy of Dermatology. 2012;67(1):139–147
  86. 86. Singh TP, Schon MP, Wallbrecht K, Gruber-Wackernagel A, Wang XJ, Wolf P. Involvement of IL-9 in Th17-associated inflammation and angiogenesis of psoriasis. PLoS One. 2013;8(1):e51752
  87. 87. Schmitz J, Owyang A, Oldham E, Song Y, Murphy E, McClanahan TK, 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. 2005;23(5):479–490
  88. 88. Carriere V, Roussel L, Ortega N, Lacorre DA, Americh L, Aguilar L, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(1):282–287
  89. 89. Moussion C, Ortega N, 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. 2008;3(10):e3331
  90. 90. Kuchler AM, Pollheimer J, Balogh J, Sponheim J, Manley L, Sorensen DR, et al. Nuclear interleukin-33 is generally expressed in resting endothelium but rapidly lost upon angiogenic or proinflammatory activation. American Journal of Pathology. 2008;173(4):1229–1242
  91. 91. Meephansan J, Komine M, Tsuda H, Karakawa M, Tominaga S, Ohtsuki M. Expression of IL-33 in the epidermis: The mechanism of induction by IL-17. Journal of Dermatological Science. 2013;71(2):107–114
  92. 92. Suzukawa M, Iikura M, Koketsu R, Nagase H, Tamura C, Komiya A, et al. An IL-1 cytokine member, IL-33, induces human basophil activation via its ST2 receptor. The Journal of Immunology. 2008;181(9):5981–5989
  93. 93. Cherry WB, Yoon J, Bartemes KR, Iijima K, Kita H. A novel IL-1 family cytokine, IL-33, potently activates human eosinophils. Journal of Allergy and Clinical Immunology. 2008;121(6):1484–1490
  94. 94. Smithgall MD, Comeau MR, Yoon BR, Kaufman D, Armitage R, 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. International Immunology. 2008;20(8):1019–1030
  95. 95. Ali S, Mohs A, Thomas M, Klare J, Ross R, Schmitz ML, et al. The dual function cytokine IL-33 interacts with the transcription factor NF-kappaB to dampen NF-kappaB-stimulated gene transcription. Journal of Immunology. 2011;187(4):1609–1616
  96. 96. Meephansan J, Tsuda H, Komine M, Tominaga S, Ohtsuki M. Regulation of IL-33 expression by IFN-gamma and tumor necrosis factor-alpha in normal human epidermal keratinocytes. Journal of Investigative Dermatology. 2012;132(11):2593–2600
  97. 97. Balato A, Lembo S, Mattii M, Schiattarella M, Marino R, De Paulis A, et al. IL-33 is secreted by psoriatic keratinocytes and induces pro-inflammatory cytokines via keratinocyte and mast cell activation. Experimental Dermatology. 2012;21(11):892–894
  98. 98. Theoharides TC, Zhang B, Kempuraj D, Tagen M, Vasiadi M, Angelidou A, et al. IL-33 augments substance P-induced VEGF secretion from human mast cells and is increased in psoriatic skin. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(9):4448–4453
  99. 99. Hueber AJ, Alves-Filho JC, Asquith DL, Michels C, Millar NL, Reilly JH, et al. IL-33 induces skin inflammation with mast cell and neutrophil activation. European Journal of Immunology. 2011;41(8):2229–2237
  100. 100. Suttle MM, Enoksson M, Zoltowska A, Chatterjee M, Nilsson G, Harvima IT. Experimentally induced psoriatic lesions associate with rapid but transient decrease in interleukin-33 immunostaining in epidermis. Acta Dermato-Venereologica. 2015;95(5):536–541
  101. 101. Balato A, Di Caprio R, Canta L, Mattii M, Lembo S, Raimondo A, et al. IL-33 is regulated by TNF-alpha in normal and psoriatic skin. Archives of Dermatological Research. 2014;306(3):299–304
  102. 102. Vageli DP, Exarchou A, Zafiriou E, Doukas PG, Doukas S, Roussaki-Schulze A. Effect of TNF-alpha inhibitors on transcriptional levels of pro-inflammatory interleukin-33 and Toll-like receptors-2 and -9 in psoriatic plaques. Experimental and Therapeutic Medicine. 2015;10(4):1573–1577
  103. 103. Mitsui A, Tada Y, Takahashi T, Shibata S, Kamata M, Miyagaki T, et al. Serum IL-33 levels are increased in patients with psoriasis. Clinical and Experimental Dermatology. 2016;41(2):183–189
  104. 104. Tamagawa-Mineoka R, Okuzawa Y, Masuda K, Katoh N. Increased serum levels of interleukin 33 in patients with atopic dermatitis. Journal of the American Academy of Dermatology. 2014;70(5):882–888
  105. 105. Talabot-Ayer D, McKee T, Gindre P, Bas S, Baeten DL, Gabay C, et al. Distinct serum and synovial fluid interleukin (IL)-33 levels in rheumatoid arthritis, psoriatic arthritis and osteoarthritis. Joint, Bone, Spine. 2012;79(1):32–37
  106. 106. Menter A, Feldman SR, Weinstein GD, Papp K, Evans R, Guzzo C, et al. A randomized comparison of continuous vs. intermittent infliximab maintenance regimens over 1 year in the treatment of moderate-to-severe plaque psoriasis. J Am Acad Dermatol. 2007;56(1):31.e1–15.
  107. 107. Saurat JH, Stingl G, Dubertret L, Papp K, Langley RG, Ortonne JP, et al. Efficacy and safety results from the randomized controlled comparative study of adalimumab vs. methotrexate vs. placebo in patients with psoriasis (CHAMPION). Br J Dermatol. 2008;158(3):558–66.
  108. 108. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R, Wang A, et al. Etanercept as monotherapy in patients with psoriasis. N Engl J Med. 2003;349(21):2014–22.
  109. 109. Tyring S, Gottlieb A, Papp K, Gordon K, Leonardi C, Wang A, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006;367(9504):29–35.

Written By

Jitlada Meephansan, Urairack Subpayasarn, Mayumi Komine and Mamitaro Ohtsuki

Submitted: 11 November 2016 Reviewed: 09 March 2017 Published: 05 July 2017