Mercurio et al. Cell Death and Disease (2018) 9:1104 DOI 10.1038/s41419-018-1143-3 Cell Death & Disease

ARTICLE Open Access IL-38 has an anti-inflammatory action in psoriasis and its expression correlates with disease severity and therapeutic response to anti-IL-17A treatment

Laura Mercurio1, Martina Morelli1,2, Claudia Scarponi1,ElanZ.Eisenmesser3,NunziannaDoti4, Gianluca Pagnanelli5, Emanuela Gubinelli6, Cinzia Mazzanti5, Andrea Cavani7, Menotti Ruvo 4, Charles A. Dinarello8,9, Cristina Albanesi 1 and Stefania Madonna1

Abstract IL-36 cytokines, a subgroup of IL-1 family, comprise IL-36α, IL-36β, and IL-36γ agonists, abundantly expressed in psoriatic skin, and IL-36RA and IL-38 antagonists. In psoriatic skin, IL-36 cytokines interfere with keratinocyte cornification programs and induce the release of antimicrobial peptides and chemokines active on neutrophils and Th17 lymphocytes. To date, the role of IL-38 antagonist in psoriasis remains to be defined. Here, we demonstrate that skin and circulating IL-38 levels are reduced in psoriatic patients and in other skin diseases characterized by neutrophilic infiltrate. In psoriasis, the balance of IL-36γ agonist/IL-38 antagonist serum levels is in favor of agonists and is closely associated with disease severity. Interestingly, IL-38 is upregulated by anti-IL-17A biological treatment and positively correlates with the therapeutic efficacy of in psoriatic patients. The downregulation of IL-38

1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; expression is strictly related to keratinocyte de-differentiation triggered by the inflammatory cytokines IL-36γ, IL-17, and IL-22. Finally, we demonstrate that administration of recombinant full-length IL-38 counteracts in vitro the biological processes induced by IL-36γ in human keratinocytes and endothelial cells and attenuates in vivo the severity of the psoriasiform phenotype induced by IMQ in mice. Such effects are achieved by restoring the physiological programs of keratinocyte proliferation and differentiation, and reducing the immune cell infiltrates.

Introduction inflammation molecules by keratinocytes3,4. A growing Psoriasis is an immune-mediated skin disease in which number of studies demonstrated that also IL-36 cytokines interferon (IFN)-γ, tumor necrosis factor (TNF)-α, inter- are pathogenic drivers of psoriasis5,6. IL-36s belong to IL- leukin (IL)-17, and IL-22 cytokines, released by Th1 and 1 family and comprise three agonists, IL-36α, IL-36β, and Th17 lymphocytes1,2, have a pathogenic action by pro- IL-36γ, and two receptor antagonists IL-36RA and IL-387. moting hyperproliferation, interfering with the terminal IL-36 agonists are strongly expressed in psoriatic skin of differentiation and inducing the secretion of pro- individuals affected by plaque psoriasis and generalized pustular psoriasis. Here, these cytokines have inflamma- tory effects on many cell targets, mainly keratinocytes, by Correspondence: Stefania Madonna ([email protected]) interfering with their cornification programs and inducing 1Laboratory of Experimental Immunology and Integrated Research Center for PSOriasis (CRI-PSO), Istituto Dermopatico dell‘Immacolata IDI-IRCCS, via Monti the release of antimicrobial peptides and chemokines di Creta, 104, ROME, Italy active on neutrophils and Th17 lymphocytes8. IL-36s also 2 Section of Dermatology, Department of Medicine, University of Verona, P.zza promote proliferation and migration of human dermal Stefani, 1, Verona 37126, Italy Full list of author information is available at the end of the article. microvascular endothelial cell (HDMEC), thus Edited by G. Melino

© The Author(s) 2018 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a linktotheCreativeCommons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 2 of 13

contributing to the dermal capillary dilatation typical of strong cytoplasmic expression of IL-38 was observed psoriatic lesions9. Although human T lymphocytes do not throughout the epidermis of NLS areas (panel i), as well as express the IL-36R receptor (IL-36R), IL-36 cytokines of healthy skin (Fig. S1, panel i), whereas a lower immu- indirectly promote Th17 lymphocyte polarization by noreactivity was detected in dermal endothelial cells and – activating the maturation of dendritic cells10 12. IL-17, fibroblasts. During the transition from Pre-LS to LS T0 area together with TNF-α and IL-22, upregulates IL-36 of the same skin biopsy, IL-38 staining progressively themselves leading to a local auto-amplification loop13. decreased in the epidermis (Fig. 1a, panel ii). This reduction The role of IL-36 agonists in the pathogenesis of psoriasis was more evident in LS areas, where IL-38 expression was has been widely demonstrated. Capon et al. recently moderately detected in the proliferative layer of epidermis, showed that IL-36R blockade by IL-36Ra or a neutralizing but it was absent in suprabasal layers (Fig. 1a, panel iii). IL- IL-36R antibody decreases the inflammation in ex vivo 36Ra and IL-36γ were minimally expressed in NLS (Fig. 1a, and in vivo experimental models of psoriasis14. However, panels i) and healthy specimens (Fig. S1, panel i), whereas the role of IL-36 antagonists, in particular of IL-38, their levels were enhanced in the upper layers of pre-LS and remains yet undefined. Mutations in IL-36Ra have been LS T0 epidermis (Fig. 1a, panels ii and iii). Infiltrating described as a cause of pustular psoriasis, owing to an immune cells were also positive for IL-36γ and IL-38 in pre- impaired inhibitory activity of IL-36Ra on Th17 respon- LS and LS T0 skin, in accordance with IL-36γ staining – ses15 17. In parallel, IL-38 allelic variants have been cor- previously detected in macrophages, dendritic cells, and + related to rheumatic diseases, including psoriatic Langerhans cells24 (mean percentage of IL-36γ cells: 10 ± arthritis18. IL-38 is a 17–18 kDa sharing 40% 1.2% and 38 ± 1.4% in pre-LS and LS T0 skin, respectively; + sequence similarity with IL-1RA and IL-36Ra antagonists IL-38 cells: 15 ± 1.1% and 35 ± 1.9% in pre-LS and LS and elicits its antagonistic effects through binding to IL- T0 skin, respectively, Fig. 1a, panels ii and iii). Contrarily, 36 receptor, as IL-36Ra7,17. IL-38 is dramatically reduced immune cells infiltrating psoriatic dermis were quite in the epidermis of psoriatic lesions as compared with negative for IL-36Ra (Fig. 1a, panels ii and iii). IL-38 epi- uninvolved or healthy skin, in line with its reduced dermal downregulation was observed in other inflammatory expression observed in de-differentiated keratinocytes skin diseases, characterized by a diffuse neutrophilic infil- compared with differentiated cells19,20. IL-38 reduction is trate, where modest levels of IL-38 were observed only in peculiar of chronic psoriatic skin, as its expression is the upper layers of the epidermis (Fig. S1, panels ii, iii, iv). contrarily induced in synovial tissues of patients with Vice versa, a higher, but variable, immunoreactivity for IL- rheumatoid arthritis and in colonic inflamed biopsies of 38 was detected throughout the epidermis of lesional AD patients with Chron’s disease19. Interestingly, IL-38 has (Fig. S1, panel v). Differently from IL-38, IL-36Ra, and IL- anti-inflammatory effects on mouse models of arthritis 36γ were upregulated in the epidermis of Hidradenitis and on a model of retinopathy, where it suppresses the suppurativa(HS),SweetSyndrome(SS),PyodermaGang- secretion of chemokines involved in Th17 pathway and renosum (PG), and Atopic Dermatitis (AD), as compared inhibits the pathological processes of vascularization, with healthy skin, with different localization among dis- respectively21,22. orders. In particular, IL-36Ra and IL-36γ expression was In this study, we analyzed the potential involvement of IL- diffuse throughout the epidermis of PG and AD (Fig. S1, 38 in psoriasis by evaluating its circulating and skin levels in panels iv and v), whereas it was localized in the upper and affected patients before and after the biological inhibition of lower layers of HS and SS epidermis, respectively (panels ii IL-17A with secukinumab. Furthermore, we investigated and iii). In all diseased skin biopsies, a modest IL-38 the effects of IL-38 administration in both in vitro and expression was observed in immune cells infiltrating the in vivo experimental models of psoriasis, such as in human dermis (Fig. S1, panels i–v). Here, with the exception of AD, + + keratinocyte and endothelial cell cultures activated by pro- a relevant percentage of IL-36Ra and IL-36γ cells with a inflammatory cytokines related to psoriasis, as well as in the neutrophil-like morphology was observed in immune cells. IMQ-induced murine model of skin inflammation. IL-38 is upregulated by anti-IL-17A treatment and Results correlates to the therapeutic efficacy to secukinumab Skin levels of IL-38 are reduced in psoriatic patients and in To evaluate the effects of anti-IL-17A treatment on IL- other skin diseases characterized by neutrophilic infiltrate 38, its expression was analyzed in vivo in skin biopsies Aimed at clarifying the controversial IL-38 expression in taken 8 weeks after therapy with secukinumab from the psoriatic and healthy skin19,23,levelsofIL-38,togetherwith same target plaques analyzed at baseline. As shown in IL-36Ra and IL-36γ, were analyzed in psoriatic specimens, Fig. 1a (panel iv), IL-38 positivity increased in skin of including non-lesional (NLS) skin, and skin overlapping secukinumab-treated patients (mean of LS T8: 2.8 ± 0.3) pre-lesional (Pre-LS) and lesional (LS T0) zones of target as compared with levels before treatment (mean of LS T0: plaques by immunohistochemistry. As shown in Fig. 1a, a 1.0 ± 0.2, p ≤ 0.05), becoming similar to those observed in

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 3 of 13

Fig. 1 (See legend on next page.)

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 4 of 13

(see figure on previous page) Fig. 1 IL-38 levels are reduced in skin and serum of psoriatic patients and upregulated by secukinumab treatment. a IHC for IL-38, IL-36Ra, and IL-36γ (all stained in red-brown) was performed on paraffin-embedded sections of biopsies obtained from psoriatic skin (n = 8) including not lesional (NLS) (i), proximal-to-lesion (Pre-LS) (ii), lesional zones of evolving plaques, before (LS T0) (iii) and after 8 weeks (LS T8) (iv) of secukinumab treatment. Sections were counterstained with Mayer’s H&E. One out of eight representative stainings of psoriatic skin biopsies are shown. Bars, 100 μm. Graphs show the mean of four-stage score values for IL-38, IL-36Ra, and IL-36γ ± SD epidermal expression per three different fields of all six sections. *p ≤ 0.05, as assessed by Mann–Whitney U test. b mRNA expression of IL-38, IL-36Ra, and IL-36γ were analyzed by RT-PCR on healthy, NLS, LS T0, and LS T8 biopsies (n = 10) and normalized to HPRT-1 levels. c Cytokine levels were analyzed by ELISA in serum obtained by venous blood of healthy (n = 25) and psoriatic individuals (n = 25) before (Pso T0) and after (Pso T8) secukinumab treatment. b, c The results are shown as individual values and mean. *p ≤ 0.05; p** ≤ 0.01, as assessed by Mann–Whitney U test. d Correlation between the ratio of IL-36γ and IL-38 serum levels and PASI (psoriasis area and severity index) at baseline. e, f Correlations between the ratio of IL-38 or IL-36Ra between T8 and T0 and Δ PASI (Δ PASI, calculated as percentage of PASI reduction) score in psoriatic individuals after secukinumab treatment. Spearman’s test was used for correlation analysis. r indicates the strength of the linear relationship, n indicates the number of values in each plot, and p value shows the probability that the slope of the true relationship is zero. p ≤ 0.05 was considered significant

NLS or healthy skin (mean of NLS: 3.0 ± 0.3) Fig. 1a and IL-38 expression in post-confluent keratinocyte cultures, Fig. S1, respectively, panels i). Contrarily, IL-36Ra and IL- resembling differentiated cells of the epidermal suprabasal 36γ staining was strongly downregulated (IL-36Ra: 0.9 ± layer11,27. Keratinocytes, but not HDMEC, constitutively 0.3 in LS T8 vs 2.9 ± 0.1 in LS, p ≤ 0.05; IL-36γ: 1.0 ± 0.2 in expressed and released IL-38 and, at lower extent, IL- LS T8 vs 3.0 ± 0.1 in LS, p ≤ 0.05), according to previous 36Ra (Fig. 2a, b). IL-38 and IL-36Ra were also expressed observations (Fig. 1a, panels iv)24,25. IL-38 mRNA levels in sub-confluent (proliferating) keratinocyte cultures in also increased in skin after 8-week therapy, compared basal condition, even though at reduced levels as com- with those detected before treatment (means: 215.0 in LS pared with differentiated cultures (approximately fivefolds T8 vs 82.5 in LS T0, p ≤ 0.05), whereas IL-36Ra and IL- lower, data not shown). mRNA expression and release of 36γ mRNA was reduced (IL-36Ra: 295.0 in LS T8 vs 52.5 IL-38 were significantly downregulated by IL-17A in post- in LS T0, p ≤ 0.05; IL-36γ: 215.0 in LS T8 vs 15.5 in LS T0, confluent keratinocyte cultures, whereas they were not p ≤ 0.05) (Fig. 1b). Next, we investigated the potential influenced by TNF-α (Fig. 2a, b). In parallel, IL-17A and systemic relevance of IL-38 by measuring its levels in TNF-α, alone and, at higher extent in combination, sig- serum of psoriatic patients before and after secukinumab. nificantly induced IL-36γ and IL-36Ra (Fig. 2a, b). mRNA At baseline, IL-38 levels were significantly lower in and protein levels of IL-36γ were also significantly psoriatic patients than in healthy donors, and were induced in HDMECs, but at a lower extent compared with upregulated following secukinumab (155.0 in Pso T8 vs keratinocytes (Fig. 2a, b). Interestingly, IL-38 levels were 95.0 in Pso T0, p ≤ 0.05) (Fig. 1c). IL-36Ra and IL-36γ also strongly downregulated by IL-22 and, at a lower serum release was instead increased at baseline of the extent, by IFN-γ and IL-36γ in keratinocytes (Fig. 2c, d). same psoriatic patients, compared with healthy group Vice versa, in line with literature data28, we found that IL- (means of IL-36Ra: 490.0 in Pso T0 vs 355.0 in healthy, 36γ was induced by IL-36γ itself, whereas it was not p ≤ 0.05; IL-36γ: 255.0 in Pso T0 vs 115.0 in healthy, p ≤ influenced by IFN-γ and IL-22 (Fig. 2c, d). IL-36Ra was 0.05), and reduced by anti-IL-17A therapy (means of IL- also upregulated in keratinocytes by IL-36γ, but not 36Ra: 490.0 in Pso T0 vs 150.0 in Pso T8, p ≤ 0.05; IL-36γ: influenced by IFN-γ and IL-22 (Fig. 2c, d). As a whole, 255.0 in Pso T0 vs 135.0 in Pso T8, p ≤ 0.05) (Fig. 1c). As these data demonstrate that keratinocytes are the main shown in Fig. 1d, the ratio of serum IL-36γ/IL-38 levels in producers of IL-38 in resident skin cells, and its expres- psoriatic patients strongly correlated to the disease sion is downregulated by inflammatory cytokines. activity as determined by psoriasis area and severity index (PASI) scores (r = 0.51, p < 0.01). The ratio of serum levels IL-38 counteracts the biological processes induced by IL- of IL-38, but not of IL-36Ra, after and before secukinu- 36γ in human keratinocytes and dermal vascular mab resulted to be closely associated with the resolution endothelial cells of the clinical psoriatic manifestations (r = 0.51, p < 0.01, As shown in Fig. 3a, similarly to IL-36Ra7,29, IL-38 and i = 0.094, p = 0.65, respectively), evaluated as per- inhibited the phosphorylation of P38 MAPK and the centage of PASI reduction determined after and before subunit P65 of NFκB induced by IL-36γ (Fig. 3a). Other treatment (Fig. 1e, f). than inducing the expression of chemokines attracting neutrophils and T cells, IL-36 cytokines interfere with IL-38 and IL-36Ra expression is differently regulated in keratinocyte differentiation in organotypic models by human keratinocytes downregulating filaggrin8,13. We thus analyzed the effects As IL-36 cytokines inhibit keratinocyte terminal differ- of IL-38 on differentiation and inflammatory responses entiation during psoriasis development26, we investigated induced by IL-36γ in keratinocytes. As shown in Fig. 3b,

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 5 of 13

Fig. 2 Regulation of the expression and release of IL-38, IL-36Ra, and IL-36γ cytokines by psoriasis-related cytokines in human keratinocytes and endothelial cells. a, c IL-38, IL-36Ra, and IL-36γ mRNA expression was detected by real-time PCR in keratinocyte cultures undergoing terminal differentiation (4 days post confluency) and HDMEC cultures stimulated with IL-17A (50 ng/ml) and TNF-α (50 ng/ml for keratinocytes and 10 ng/ml for HDMEC), alone or in combination a or in keratinocytes stimulated with IL-22 (50 ng/ml), IFN-γ (200 U/ml, or IL-36γ (50 ng/ml) c, for 6 h. GAPDH mRNA levels were detected for normalization. b, d IL-36 cytokine levels were analyzed by ELISA in supernatants obtained by cell cultures after 24 h cytokine stimulation. All data shown are the mean of three different experiments. *p ≤ 0.01 and **p ≤ 0.001 compared with untreated cultures, as assessed by Mann–Whitney U test post-confluent keratinocyte cultures expressed higher pro-inflammatory chemokines and adhesion molecules, levels of differentiation markers, such as KRT1 and whose levels are reduced by IL-36Ra30. IL-36γ not only KRT10 and loricrin, compared with proliferating cells synergizes with TNF-α in the release of these inflamma- (T0), and their expression was abrogated by IL-36γ. tory mediators, but it induces proliferation of cultured Interestingly, IL-38 dose-responsively upregulated and HDMEC. As shown in Fig. S2, IL-38 inhibited the phos- normalized the levels of KRT1, KRT10, and loricrin, but phorylation of ERK1/2 strongly induced by IL-36γ in not those of ΔNp63, a typical marker of proliferating HDMEC, as well as that of P65 (panel a). Interestingly, keratinocytes. Similarly, IL-36Ra increased KRT1, KRT10, HDMEC proliferation induced by IL-36γ was significantly and loricrin levels in IL-36γ-treated keratinocytes, attenuated by IL-38 (Fig. S2b). IL-38, similarly to IL-36Ra, although these effects were more evident at lower doses, also reverted the IL-36γ- and TNF-α-induced expression compared with IL-38 (Fig. 3b). IL-38 also reduced the IL- of inflammatory molecules in HDMEC cultures 36γ-induced expression of CXCL8, CXCL20, IL-6, and (Fig. S2b–d). vascular endothelial growth factor A (VEGF-A), as well as of antimicrobial peptides HBD-2 and LL-37, although a IL-38 ameliorates the psoriasiform phenotype of IMQ- dose–response was not observed (Fig. 3c). At high doses, treated mice IL-36Ra counteracted the expression of most inflamma- We further investigated IL-38 potential in IMQ-induced tory mediators more efficiently than IL-38 (Fig. 3c). IL-38 psoriasiform dermatitis, an IL-17/IL-22-mediated mouse also downregulated the IL-36γ-induced expression of model of the disease31. Subcutaneous injection of IL-38 intercellular adhesion molecule 1 (ICAM-1), with a sig- for 5 days, concomitantly to topical IMQ application, nificant reduction at higher dose (Fig. 3d). HDMEC also ameliorated the psoriasiform manifestations, with a ~ 40% express IL-36 receptor and, upon IL-36γ, they express reduction of acanthosis and consequent scale thickness, as

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 6 of 13

Fig. 3 IL-38 inhibits IL-36γ-induced P38 and NF-κB signalings, as well as regulates the expression of differentiation markers and inflammatory molecules in human keratinocytes. All experiments were performed on keratinocyte cultures (n = 3 strains) undergoing terminal differentiation and stimulated or not with IL-36γ in presence or absence of the indicated doses of IL-38 or IL-36Ra. a Protein extracts were obtained from keratinocyte cultures stimulated for 24 h and subjected to WB analysis to detect P38 and P65 phosphorylation. Filters were probed with anti-P38 and -P65 Abs. b Keratinocyte cultures were analyzed for KRT1, KRT10, Loricrin and ΔNp63 expression by WB. a, b β-actin was used as loading control and DI ratio indicates the densitometric intensity of the indicated phosphorylated/unphosphorylated shown in one representative of three different WB. c mRNA levels of CXCL8, CCL20, VEGF-A, IL-6, HBD-2, and LL-37 were detected by real-time PCR analysis in keratinocytes stimulated for 6 h and normalized for GAPDH mRNA levels. TNF-α treatment was used as positive control. d ICAM-1 expression was evaluated by flow cytometry analysis on keratinocytes stimulated for 24 h with IL-36γ (50 ng/ml) and shown as mean fluorescence intensity. All data shown are the mean of three different experiments. c *p ≤ 0.05 and p** ≤ 0.01 compared with untreated or IL-36γ-treated cultures, as assessed by Mann–Whitney U test well as of the dermal inflammatory infiltrate (~ 50% also observed in IL-38-treated inflamed skin (Fig. 5a, b). decrease) (Fig. 4a–e). As shown in Fig. 5, epidermal dif- In addition, the epidermis of IL-38-treated mice showed a ferentiation markers, such as KRT10, which are normally reduced expression of the pro-angiogenic VEGF-A cyto- expressed in the suprabasal layers of epidermis, displayed kine, compared with controls (Fig. 5a, b). IL-38 treatment + a broad staining throughout keratinocyte layers of IMQ- also decreased the number of infiltrating CD3 T lym- + induced skin lesions (Fig. 5a, b). Interestingly, adminis- phocytes and Ly6G neutrophils (panels iii), compared tration of IL-38 normalized the expression of KRT10, with IMQ-treated mice group (panels ii), whereas it did + which compartmentalized to the upper granular layers, as not influence the number of CD11c cells (panels ii and typically observed in normal epidermis (Fig. 5a). A iii). Both IL-36Ra and IL-38 administration determined a + reduced number of proliferating Ki67 keratinocytes was significant reduction of the endogenous levels of IL-36Ra

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 7 of 13

Fig. 4 Ameliorative effects of IL-38 administration on pathological changes of IMQ-induced psoriasiform murine skin lesions. a Macroscopic views of back skin from mice left untreated (IMQ-) (i), IMQ-treated (IMQ + ) (ii) or co-treated with IMQ and IL-38 (IMQ + /IL-38) (iii) or IMQ and IL-36Ra (IMQ + /IL-36Ra) (iv), both subcutaneously injected, after 5 days. b Representative H&E staining of untreated (i), treated with IMQ cream (ii), in presence of IL-38 (iii) or IL-36Ra (iv). Bars, 500 μm. Subcutaneous injection of IL-38 or IL-36Ra reverted the phenotypic skin changes determined by IMQ. The quantifications of epidermal c, scale thickness d, and immune cell influx e were analyzed as parameters of skin acanthosis and inflammation. Graphs show means of microns of epidermis and stratum corneum thickness, and mean of number of infiltrating immune cells per section (n = 3 sections) ± S.D. per group (n = 8 mice). p* ≤ 0.01 and **p ≤ 0.001 compared with untreated or IMQ-treated groups, as assessed by unpaired Student‘s t test

(~ 30% decrease of relative mRNA expression) and IL-36γ those of IL-36Ra, a well-characterized antagonist of IL-36 (~ 35% decrease), whereas they did not influence endo- cytokines strongly induced in psoriatic lesions32.We genous IL-38 expression (data not shown). In parallel, also found that the balance of IL-36γ agonist/IL-38 antagonist CCL20, CXCL8, and IL-6 mRNA expression was down- levels, but not of IL-36γ/IL-36Ra, in serum of psoriatic regulated in the skin of IL-38-treated mice group (data patients is closely associated with disease severity. Inter- not shown). Finally, similarly to IL-38, IL-36Ra amelio- estingly, IL-38 expression is responsive to IL-17A inhibi- rated the psoriasiform phenotype, with positive effects on tion, as its levels significantly increase in skin and in all pathological markers analyzed (Fig. 5, panels iv). serum of affected patients after treatment with secuki- numab, a selective anti-IL-17A antibody used in clinical Discussion practise33, whereas IL-36γ and IL-36Ra levels decrease in In this study, we identify IL-38 as a new responsive line with previous studies24,25. We show that the ratio of biomarker for psoriasis, and demonstrate its anti- IL-38 serum levels after and before secukinumab corre- inflammatory properties in in vitro and in vivo experi- lates to the therapeutic efficacy in psoriatic patients, thus mental models of psoriasis. The interest toward IL-38 in identifying IL-38 as a potential responsive biomarker for psoriatic context arises from our and earlier observations psoriasis. However, these findings need to be further that its expression is reduced in the epidermis of skin confirmed on a larger cohort of psoriatic patients treated lesions and in the serum of psoriatic patients, as com- by secukinumab and other biologicals, such as antibodies pared with uninvolved or healthy skin, and is inverse to blocking TNF-α, a strong inducer of IL-36 agonists.

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 8 of 13

Fig. 5 IL-38 reverts the pathological markers in IMQ-induced murine model of psoriasis. a (i), IMQ-treated (n = 8) (ii) and co-treated with IMQ and IL-38 (IMQ + /IL-38) (n = 8) (iii) or IL-36Ra (IMQ + /IL-36Ra) (n = 8) (iv) shows an upregulated and normalized expression of KRT10 in the epidermis after IL-38 and IL-36Ra treatment, as well as a reduction of VEGF-A expression, and of positive Ki67, Ly6G, CD3, and CD11c cells. Sections were counterstained with Mayer’s H&E and were visually evaluated by a pathologist experienced in dermatology. One out of eight representative stainings is shown. Bars, 500 μm. b Graphs show the mean of number of positive cells or of four-stage score values for KRT10 ± SD per three sections per experimental group (six of eight mice were analyzed). *p ≤ 0.01, **p ≤ 0.001 compared with untreated or IMQ-treated groups, as assessed by unpaired Student‘s t test Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 9 of 13

Taken together, our findings unveil a relevant role of IL- induced mice, whereas that of IL-36Ra is induced and 38 in psoriasis, supporting the hypothesis that the dysre- correlates with IL-36γ upregulation, thus reflecting the gulation of its expression could be pathogenic in psoriasis. expression pattern of these cytokines in psoriatic skin IL-38 and IL-36Ra antagonists have anti-inflammatory lesions. Interestingly, we demonstrate that the adminis- properties by antagonizing the IL-36 pathways through a tration of IL-38 significantly attenuates the severity of direct binding to IL-36 receptor with similar affinities5,7. psoriasiform phenotype induced by IMQ, by restoring the However, the upregulation of IL-36Ra alone might be not physiological proliferation and differentiation programs in sufficient to unleash IL-36-dependent inflammatory keratinocytes, and decreasing the expression of VEGF-A, responses in psoriatic lesions, where several cytokines a cytokine involved in the altered angiogenic processes inducing IL-36 are active on resident skin and immune occurring in psoriasis41. IL-38 reduces infiltration of cells. Owing to the different in vitro and in vivo expres- T cells and neutrophils in IMQ model, accordingly with a sion of IL-38 and IL-36Ra, IL-38, and IL-36Ra can be significant reduction of psoriasis-related chemokines considered as not redundant cytokines, having IL-38 involved in T-cell and neutrophil recruitment and acti- additional functions closely related to the keratinocyte vation. The ameliorative impact of IL-38 on the psoriatic differentiation. In fact, the loss of IL-38 in the suprabasal clinical phenotype is also associated with the decrease of layers of psoriatic epidermis is associated with the IL-36 local expression, and was comparable to that impaired differentiation processes occurring in the affec- observed with IL-36Ra, whose anti-inflammatory prop- ted skin, to which IL-17 and IL-22 cytokines markedly erties have been recently demonstrated also in an ex vivo contribute1,34. In support of this hypothesis, our results model of human psoriasis16. Recently, Palomo et al.42 show that keratinocytes undergoing terminal differentia- demonstrated that the depletion of IL-38, differently from tion release higher levels of IL-38 compared with IL-36Ra. IL-36Ra, does not affect the severity of the psoriasiform However, IL-17, IL-22, and IL-36γ, known to inhibit the reactions in IMQ-treated mice. These results are not terminal keratinocyte differentiation8,20, have inhibitory surprising if we consider that endogenous IL-38 is per se effects on IL-38 expression, although they induce IL-36Ra absent in skin of this mouse model, whereas IL-36Ra is antagonist. A predictive analysis of potential transcrip- upregulated. In our opinion, IL-38 reinforcement could be tional factor-binding sites on IL-38 and IL-36Ra promoter the better experimental approach to evaluate its function sequences revealed the presence in the promoter of IL-38, in inflammatory skin conditions, and the ameliorative but not of IL-36Ra, of elements likely binding to cAMP- effects of the IL-38 administration in IMQ model confirm responsive repressors, such as E4BP4 and ICER, and our thesis. Kruppel-like factors, known to be induced by inflamma- In our study, contrarily to what observed in vivo in tory cytokines and related to keratinocyte differentia- IMQ-induced model, the anti-inflammatory effects of IL- – tion35 37. These factors could be responsible for IL-38 38 and IL-36Ra are not entirely comparable in vitro, being downregulation in keratinocytes subjected to de- IL-36Ra able to inhibit, at the same doses, the inflam- differentiative processes triggered by IL-36γ, IL-17A, matory responses more efficiently than IL-38. This dis- and IL-22. IL-38 expression varies in different disease crepancy can be explained considering that we use a full- settings, especially in disorders with dysregulated IL-1- length IL-38, whereas IL-36Ra was employed in a more dependent responses, such as spondylitis ankylopoetica, active, N-terminally cleaved, form7,43. As two N- rheumatoid arthritis, and HS. In our study, we found that terminally cleaved forms of IL-38 with an increased the local decline of IL-38, and the concomitant induction inhibitory activity have been identified in tumor cells44, of IL-36γ and IL-36Ra in psoriatic epidermis, are common we hypothesize that IL-38 is processed in our in vivo to other inflammatory skin diseases characterized by a experimental system, likely owing to extracellular neu- prominent neutrophilic infiltrate, including not only HS, trophil proteases abundantly released in the IMQ-treated but also Sweet’s syndrome and PG, where an altered skin31,45,46. Finally, the absence of a dose–response effect epidermal architecture has been reported38,39. Contrarily, of IL-38 (as well as of IL-36Ra) on in vitro expression of a modest IL-38 expression has been found in skin affected inflammatory mediators support the hypothesis of by AD, an immune-mediated skin disease characterized Dinarello et al. that IL-38 and IL-36Ra do not behave as by a prominent Th2 lymphocyte infiltrate and lacking of classic receptor antagonists5. Upon binding to IL-36R, IL- neutrophil circuits40, thus, supporting the involvement of 38, and IL-36Ra might recruit an inhibitory co-receptor at the IL-36 cytokine axis in cutaneous skin disorders low concentrations, but a signaling co-receptor at higher determined by neutrophil presence. Furthermore, we concentrations, similarly to what observed for IL-375.In investigated the effects of IL-38 reinforcement in in vitro summary, IL-38 has strong anti-inflammatory effects on and in vivo models of psoriasis. In line with data reported IL-36-induced responses in the psoriatic skin context, by Boutet et al., we found that the endogenous expression making it an attractive cytokine to explore also in other of IL-38 is significantly reduced in the skin of IMQ- Th17-related diseases characterized by dysregulated IL-1-

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 10 of 13

driven responses. Clinical trials with an IL-36R blocking basal medium (EBM, Lonza) containing 2% fetal bovine antibody (ANB019) are ongoing in generalized pustular serum (starvation medium). When necessary, both cell psoriasis and palmoplantar pustular psoriasis. These stu- types were treated with recombinant human IL-38 or IL- dies represent the first attempt to block inflammatory 36Ra (mature form Val2-Asp155; R&D Systems) con- responses executed by epidermal keratinocytes, the pri- comitantly to cytokine stimulation. mary source of IL-36 cytokines. However, the reinforce- ment of IL-38 could represent an alternative therapeutic Recombinant expression and purification of IL-38 strategy for inhibiting IL-36-induced responses in psor- A single Gibson block (gblock, IDT technologies) was iasis, although understanding the functional biology of IL- produced encoding for an N-terminal 6xHis tag, a 38 in skin context will be needed to reveal its role in new thrombin cleavage site, and full-length IL-38 (152 resi- mechanisms of disease and to evaluate the potential dues). This gblock was PCR amplified for insertion into impact of its manipulation in psoriasis treatment. pET21 using a single NdeI site and subsequently expres- sed in BL21/DE3 cells in loria broth supplemented with Materials and methods 100 mg/ml ampicillin. Cells were grown until an optical Human subjects density (600 nm) of ~ 0.6 and induced for 3–4 h prior to Twenty-five patients with mild-to-severe chronic plaque harvesting. The full-length protein was also frown in psoriasis (4 ≤ PASI ≥ 45), and 25 healthy subjects under- M9 minimal media to obtain 15N-labeled protein in order going plastic surgery were enrolled in this study (age to confirm that IL-38 was well-folded (data not shown). range 18–70 years). Psoriatic patients had similar char- For IL-38 purification, cell pellets were sonicated on ice acteristics, including age of onset and disease duration in lysis buffer (50 mM Na2HPO4, pH 7.5, 500 mM NaCl, and, after an induction phase, they received subcutaneous 10 mM imidazole), centrifuged to remove cellular debris, injections of 300 mg Secukinumab once a week (Cosentyx, and applied to a Ni-affinity column (GE Healthcare Life Novartis). Sciences. IL-38 was eluted using lysis buffer supplemented Biopsies were taken from skin plaques at sites over- with 400 mM imidazole. Fractions comprising IL-38 were lapping LS before treatment (T = 0) and the adjacent Pre- dialyzed against “Q buffer” (50 mM tris, pH 8.0, 50 mM LS. Biopsies at LS sites after 8-week treatment (T = 8) and NaCl, 10 mM imidazole, 1 mM dithiothreitol), applied to at NLS, 3 cm distant from the evolving plaques, were also a Q-sepharose fast flow column (GE Healthcare Life taken from the same psoriatic patients. In parallel, skin Sciences). Finally, fractions comprising IL-38 were con- biopsies were also taken by healthy volunteers undergoing centrated, cleaved with thrombin at room temperature plastic surgery, and from lesional skin areas of patients overnight to remove the 6xHis tag, and applied to a with HS, SS, PG, and AD. Among individuals, 8 were Superdex-75 (GE Healthcare Life Sciences) in “final buf- selected for immunohistochemical and 10 for RT-PCR fer” (10 mM HEPES, pH 70, 150 mM NaCl). IL-38 was studies, whereas all 25 were analyzed for IL-36 serum concentrated and stored at − 80 °C until further use. quantification. This study was approved by the Ethical Committee of the IDI-IRCCS Hospital, Rome (registra- IMQ-induced psoriasiform model tion no.: IDI-IMM-IL36pso) and performed accordingly Eight-week-old female BALB/cJ mice (Harlan Labora- to the Declaration of Helsinki. Informed consent was tories, San Pietro al Natisone, Italy) treated for 5 con- signed by all study subjects. secutive days with 5% (62.5 mg) IMQ (ALDARA cream, Meda AB, Solna, Sweden)49 received daily subcutaneous Cell cultures and treatments injections (1 μg/mouse) of human recombinant full- Normal human keratinocytes were established from length IL-38 (n = 8), Val2-Asp155 IL-36Ra (n = 8), or sun-protected skin of healthy individuals (n = 4) and control vehicle (PBS) (n = 8), starting on day 0 of IMQ cultured as previously reported47. Cells were plated at administration. A mice group (n = 3), not receiving IMQ, 5000/cm2 and maintained to 4-d post confluency. High- was used as negative control. On day 5, full thickness skin confluence cultures were stimulated with recombinant biopsies of the treated area were collected with an eight- human IL-22 (50 ng/ml), IFN-γ (200 U/ml), IL-17A mm biopsy puncher. Skin was either snap frozen in liquid (50 ng/ml), TNF-α (50 ng/ml), or IL-36γ (Ser18-Asp169; N2 for total RNA preparation, or fixed in neutral buffered 50 ng/ml) (R&D Systems, Minneapolis, MN, USA) in formalin (Sigma-Aldrich, St. Louis, MO, USA) for histo- keratinocyte basal medium (Clonetics). pathological analysis. The whole experiment was repeated HDMEC previously isolated from foreskin of three dif- twice with similar results. All mouse procedures were ferent donors48 were retrieved from the Cell Bank of the carried out in accordance with institutional standard Laboratory of Experimental Immunology of IDI-IRCCS guidelines. The experimental design has been authorized and stimulated with TNF-α (10 ng/ml), IL-17A (50 ng/ by the Italian Health Minister (protocol No. SA-IDI-13- ml), IFN-γ (200 U/ml), or IL-36γ (50 ng/ml) in endothelial CA-1).

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 11 of 13

Immunohistochemistry Fluorescence intensity was analyzed by the ABI PRISM SDS Paraffin-embedded sections were obtained from biop- 7000 PCR Instrument (Applied Biosystems, Branchburg, NJ, sies of psoriatic skin including LS at baseline (T = 0) and USA), using SYBR Green PCR reagents or Taqman PCR after 8-week Cosentyx treatment, Pre-LS and NLS zones Master Mix. The values obtained from triplicate experi- of evolving plaques, as well as of healthy skin. Paraffin- ments were averaged, and data are presented as means ± SD. embedded sections obtained from patients affected by HS, AD, SS, or PG syndromes were retrieved by Histo- Cytokine quantification by ELISA pathology Unit at IDI-IRCCS hospital. Concentrations of IL-38, IL-36Ra, and IL-36γ were Immunohistochemistry analyses were performed with measured in serum obtained by venous blood of patients primary antibodies against IL-38 (InVitrogen, Carlsbad, enrolled in this study, as well as in cell-free supernatants CA, USA), IL-36Ra (AbCam, Cambridge, UK) and IL-36γ from untreated or cytokine-treated keratinocyte and (AbCam). Secondary biotinylated mAbs and staining kits endothelial cell cultures, using specific ELISA kits (BD (Vector Laboratories, Burlinagame, CA, USA) were used Pharmingen). The plates were analyzed in an ELISA to develop IL-38 and IL-36Ra immunoreactivities, reader mod.3550 UV (Bio-Rad Hercules, CA, USA). whereas biotinylated secondary Ab and staining from Scytek (Logan, Utah, USA) was used for IL-36γ detection. Western blotting analysis Paraffin-embedded sections were obtained from murine Total protein extracts by cell cultures and immunoblot- skin tissues and analyzed for epidermal and scale thick- ting were performed as previously reported36. The Abs ness, as well as cell infiltrate number. Average epidermal employed for the study were as follows: anti-phospho-Erk1/ and scale thickness was quantified by a researcher blind to 2 (E-4), anti-Erk1/2 (C16), anti-β-actin (C-11), all provided the experimental groups who took five measurements per by Santa Cruz Biotechnology (Santa Cruz, CA, USA). Anti- three sections for each mouse. Cells infiltrating dermis phospho-P65, anti-P65, anti-phospho-P38, and ant-P38 were also counted in three skin sections for each mouse. were from Cell Signaling Technology (Denvers, MA, Immunohistochemistry analyses were performed with USA), whereas anti-KRT1, anti-KRT10, and anti-loricrin primary Abs against CD3 (Dako, Glostruk, Denmark), were from Covance (Emeryville, CA, USA). Anti-ΔNp63 Ab CD11c (Dako, Glostruk, Denmark), Ly6G (BD Bios- was provided by Santa Cruz Biotechnology. Filters were ciences, Franklin Lakes, NJ USA), Ki67 (Novocastra, properly developed with anti-mouse, anti-goat, or anti- Newcastle upon Tyne, UK), KRT10 (Covance, Princeton, rabbit Ig Abs conjugated to horseradish peroxidase using NJ, USA), VEGF-A (Santa Cruz Biotechnology) and the ECL-plus detection system (Amersham, Dubendorf, immunoreactivities developed with secondary biotiny- Switzerland), or, otherwise, the SuperSignal West Femto kit lated mAbs and staining kits (Vector Laboratories, Bur- (Pierce, Rockford, IL, USA). Immunoblots were subjected to lingame, CA, USA). All sections were counterstained with densitometry using an Imaging Densitometer model GS- Mayer’s Hematoxylin and eosin and positivity was eval- 670 (Bio-Rad) supported by the Molecular Analyst software, uated in five adjacent fields at a magnification × 200. and band intensities were evaluated in three independent A semiquantitative, four-stage scoring system was experiments. Data are expressed as densitometric intensity applied, ranging from negative immunoreactivity (0) to (DI) ± SD. strong immunoreactivity (4+) for IL-36 cytokines in human skin, as well as for KRT10 and VEGF-A in murine HDMEC proliferation skin. IL-36 cytokine-positive cells were counted blindly by In total, 6 × 104 HDMEC were seeded in 12-well plates two observers with an eyepiece graticule at a magnifica- in endothelial growth medium in triplicate for each con- tion of × 200. For each skin specimen, two sections were dition. 1 day after, the cells were starved in EBM and analyzed for each staining and positive cells were eval- treated with IL-36γ alone or in presence of recombinant uated in five adjacent fields. IL-38 or IL-36Ra and cultured for 24 and 48 h. The number of viable cells was determined by a trypan blue RNA isolation and real-time RT-PCR exclusion test. Total RNA from keratinocytes and endothelial cells was extracted using the TRIzol reagent (InVitrogen), whereas Flow cytometry analysis total RNA from human and mouse skin biopsies by RNeasy Keratinocyte and HDMEC membrane expression of Lipid Tissue Kit (Qiagen, Chatsworth, CA, USA). mRNA ICAM-1 was evaluated using allophycocyanin (APC)- was reverse-transcribed into complementary DNA and conjugated anti-CD54 monoclonal Ab (clone 84H10; analyzed by real-time PCR. GAPDH or β-2 microglobulin Immunotech, Marseille, France), whereas VCAM were used as housekeeping for human and murine expression on HDMEC membrane was detected by using mRNA, respectively. Primer pairs used in PCR reactions are APC-conjugated anti-human VCAM-1 (clone 51-10C9, listed in the table reported in Supplementary Table S1. BD Pharmingen). Cells were analyzed by a FACScan

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 12 of 13

equipped with Cell Quest software (Becton Dickinson, 9. Jo, S. et al. Reconstitution of ST2 (IL-1R4) specific for IL-33 activity; no sup- Mountain View, CA, USA). pression by IL-1Ra though a common chain IL-1R3 (IL-1RAcP) shared with IL-1. Cytokine 83,33–40 (2016). 10. Tortola, L. et al. Psoriasiform dermatitis is driven by IL-36-mediated DC- Statistical analysis keratinocyte crosstalk. J. Clin. Invest. 122,3965–3976 (2012). Differences between groups were evaluated by the 11. Foster, A. M. et al. IL-36 promotes myeloid cell infiltration, activation, and – U ‘ t fi inflammatory activity in skin. J. Immunol. 192,6053–6061 (2014). Mann Whitney or unpaired Student s test as speci ed 12. Dietrich, D. et al. -36 potently stimulates human M2 macrophages, in the figure legends using GraphPad prism Software (La Langerhans cells and keratinocytes to produce pro-inflammatory cytokines. Jolla, CA, USA). Spearman’s test was used to correlation Cytokine 84,88–98 (2016). 13. Gabay, C. & Towne, J. E. Regulation and function of interleukin-36 analysis. cytokines in homeostasis and pathological conditions. J. Leukoc. Biol. 97, 645–652 (2015). Acknowledgements 14. Mahil, S. K. et al. An analysis of IL-36 signature genes and individuals with This work was supported by a grant founded by the Italian Ministry of Health IL1RL2 knockout mutations validates IL-36 as a psoriasis therapeutic target. Sci. (Young Researcher Project Grant GR-2013-02355700 of Dr. S. Madonna). Transl. Med. 9, 10.1126 (2017). 15. Marrakchi, S. et al. Interleukin-36-receptor antagonist deficiency and general- Author details ized pustular psoriasis. N. Engl. J. Med. 365,620–628 (2011). 1Laboratory of Experimental Immunology and Integrated Research Center for 16. Onoufriadis,A.etal.MutationsinIL36RN/IL1F5 are associated with the severe PSOriasis (CRI-PSO), Istituto Dermopatico dell‘Immacolata IDI-IRCCS, via Monti episodic inflammatory skin disease known as generalized pustular psoriasis. di Creta, 104, ROME, Italy. 2Section of Dermatology, Department of Medicine, Am.J.Hum.Genet.89,432–437 (2011). University of Verona, P.zza Stefani, 1, Verona 37126, Italy. 3Department of 17. van de Veerdonk, F. L. et al. IL-38 binds to the IL-36 receptor and has biological Biochemistry & Molecular Genetics, School of Medicine, University of Colorado effects on immune cells similar to IL-36 receptor antagonist. Proc. Natl Acad. Denver, Anschutz Campus, Aurora 80045 CO, USA. 4Istituto di Biostrutture e Sci. USA 109, 3001–3005 (2012). Bioimmagini-CNR and CIRPEB, Via Mezzocannone, 16, Naples 80134, Italy. 51st 18. Rahman, P. et al. Association between the interleukin- 1 family cluster Division of Dermatology and CRI-PSO, Istituto Dermopatico dell‘Immacolata and psoriatic arthritis. Arthritis Rheum. 54,2321–2325 (2006). IDI-IRCCS, via Monti di Creta, 104, Rome 00167, Italy. 6CRI-PSO Istituto 19. Boutet, M. A. et al. Distinct expression of IL-36 alpha, beta, gamma, their Dermopatico dell’Immacolata, IDI-IRCCS, via Monti di Creta, 104, Rome 00167, antagonist IL-36Ra and IL-38 in psoriasis, rheumatoid arthritis and Crohn’s Italy. 7INMP/NIHMP, via di S.Gallicano, 25, Rome 00153, Italy. 8Department of disease. Clin.Exp.Immunol.184,159–173 (2015). Medicine, Radboud University Medical Center, 6525 HP Nijmegen, The 20. Rabeony, H. et al. Inhibition of keratinocyte differentiation by the synergistic Netherlands. 9Department of Medicine, School of Medicine, University of effect of IL-17A, IL-22, IL-1α,TNFα and oncostatin M. PLoS ONE 9, e101937 Colorado, Denver, Anschutz Campus, Aurora, CO, USA (2014). 21. Boutet, M. A. et al. IL-38 overexpression induces anti-inflammatory effects in Conflict of interests mice arthritis models and in human macrophages in vitro. Ann. Rheum. Dis. The authors declare no conflict of interests. 76,1304–1312 (2017). 22. Zhang, J. et al. The effect of interleukin 38 on angiogenesis in a model of oxygen-induced retinopathy. Sci. Rep. 7,2756–2761 (2017). 23. Keermann, M. et al. Expression of IL-36 family cytokines and IL-37 but not IL-38 Publisher’s note is altered in psoriatic skin. J. Dermatol. Sci. 80,150–152 (2015). Springer Nature remains neutral with regard to jurisdictional claims in 24. Kolbinger,F.etal.β-Defensin 2 is a responsive biomarker of IL-17A-driven skin published maps and institutional affiliations. pathology in patients with psoriasis. J. Allergy Clin. Immunol. 139,923–932 (2017). Supplementary Information accompanies this paper at (https://doi.org/ 25. D’Erme,A.M.etal.IL-36γ (IL-1F9) is a biomarker for psoriasis skin lesions. J. 10.1038/s41419-018-1143-3). Invest. Dermatol. 135,1025–1032 (2017). 26. Krueger, J. G. & Brunner, P. M. Interleukin-17 alters the biology of many cell Received: 1 June 2018 Revised: 10 September 2018 Accepted: 8 October types involved in the genesis of psoriasis, systemic inflammation and asso- 2018 ciated comorbidities. Exp. Dermatol. 27,115–123 (2018). 27. Sestito, R. et al. STAT3-dependent effects of IL-22 in human keratinocytes are counterregulated by sirtuin 1 through a direct inhibition of STAT3 acetylation. FASEB J. 25, 916–927 (2011). 28. Carrier, Y. et al. Inter-regulation of Th17 cytokines and the IL-36 cytokines References in vitro and in vivo: implications in psoriasis pathogenesis. J. Invest. Dermatol. 1. Albanesi, C. & Pastore, S. Pathobiology of chronic inflammatory skin diseases: 131, 2428–2437 (2011). interplay between keratinocytes and immune cells as a target for anti- 29. Swindell, W. R. et al. RNA-Seq analysis of IL-1B and IL-36 responses in epi- inflammatory drugs. Curr. Drug. Metab. 11,210–227 (2010). dermal keratinocytes identifies a shared MyD88-dependent gene signature. 2. Zheng, Y. et al. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced Front. Immunol. 29, 80 (2018). dermal inflammation and acanthosis. Nature 445, 648–651 (2007). 30. Bridgewood,C.etal.IL-36γ has proinflammatory effects on human endo- 3. Ogawa, E., Sato, Y., Minagawa, A. & Okuyama, R. Pathogenesis of psoriasis and thelial cells. Exp. Dermatol. 26,402–408 (2017). development of treatment. J. Dermatol. 45,264–272 (2018). 31. van der Fits, L. et al. Imiquimod-induced psoriasis-like skin inflammation in 4. Madonna, S., Scarponi, C., Pallotta, S., Cavani, A. & Albanesi., C. Anti-apoptotic mice is mediated via the IL-23/IL-17 axis. J. Immunol. 182,5836–5845 (2009). effects of suppressor of cytokine signaling 3 and 1 in psoriasis. Cell Death Dis. 32. Furue, K. et al. Highlighting interleukin-36 signalling in plaque psoriasis and 3, e334 (2012). pustular psoriasis. Acta Derm. Venereol. 98,5–13 (2018). 5. vandeVeerdonk,F.L.,deGraaf,D.M.,Joosten,L.A.&Dinarello,C.A.Biology 33. Gisondi, P., Del Giglio, M., Girolomoni,G.Treatmentapproachestomoderate of IL-38 and its role in disease. Immunol. Rev. 281, 191–196 (2018). to severe psoriasis. Int. J. Mol. Sci. 18, 10.3390 (2017). 6. Ding,L.,Wang,X.,Hong,X.,Lu,L.&Liu,D.IL-36cytokinesinautoimmunity 34. Lowes, M. A., Suárez-Fariñas, M. & Krueger, J. G. Immunology of psoriasis. Annu. and inflammatory disease. Oncotarget 9, 2895–2901 (2017). Rev. Immunol. 32,227–255 (2014). 7. Bassoy, E. Y., Towne, J. E. & Gabay, C. Regulation and function of interleukin-36 35. Mead, J. R., Hughes, T. R., Irvine, S. A., Singh, N. N. & Ramji, D. P. Interferon- cytokines. Immunol. Rev. 281,169–178 (2018). gamma stimulates the expression of the inducible cAMP early repressor in 8. Pfaff,C.M.,Marquardt,Y.,Fietkau,K.,Baron,J.M.&Lüscher,B.Thepsoriasis- macrophages through the activation of casein kinase 2. A potentially novel associated IL-17A induces and cooperates with IL-36 cytokines to control pathway for interferon-gamma-mediated inhibition of gene transcription. J. keratinocyte differentiation and function. Sci. Rep. 7, 15892–15897 (2017). Biol. Chem. 278, 17741–17751 (2003).

Official journal of the Cell Death Differentiation Association Mercurio et al. Cell Death and Disease (2018) 9:1104 Page 13 of 13

36. Madonna, S. et al. The IFN-gamma-dependent suppressor of cytokine sig- 43. Towne, J. E. et al. Interleukin- 36 (IL-36) ligands require processing for full naling 1 promoter activity is positively regulated by IFN regulatory factor-1 agonist (IL- 36alpha, IL- 36beta, and IL- 36 gamma) or antagonist (IL-36Ra) and Sp1 but repressed by growth factor independence-1b and Krüppel-like activity. J. Biol. Chem. 286, 42594–42602 (2011). factor-4, and it is dysregulated in psoriatic keratinocytes. J. Immunol. 185, 44. Mora, J. et al. Interleukin- 38 is released from apoptotic cells to limit inflam- 2467–2478 (2010). matory macrophage responses. J. Mol. Cell Biol. 8,426–438 (2016). 37. Feinberg, M. W. et al. Kruppel-like factor 4 is a mediator of proinflammatory 45. Costa, S. et al. Role of MyD88 signaling in the imiquimod-induced mouse signaling in macrophages. J. Biol. Chem. 280, 38247–38258 (2005). model of psoriasis: focus on innate myeloid cells. J. Leukoc. Biol. 102, 791–803 38. Emtestam, L. & Sartorius, K. Interleukin-36 cytokine family signalling in hidra- (2017). denitis suppurativa. Br. J. Dermatol. 178,591–592 (2018). 46. Chamcheu, J. C. et al. Dual inhibition of PI3K/Akt and mTOR by the dietary 39. Wallach, D. & Vignon-Pennamen, M. D. Pyoderma gangrenosum and sweet antioxidant, delphinidin, ameliorates psoriatic features in vitro and in an syndrome: the prototypic neutrophilic dermatoses. Br. J. Dermatol. 178, imiquimod-induced psoriasis-like disease in mice. Antioxid. Redox Signal. 26, 595–602 (2018). 49–69 (2017). 40. Guttman-Yassky,E.&Krueger,J.G.Atopicdermatitisandpsoriasis:twodif- 47. Scarponi, C. et al. Inhibition of inflammatory and proliferative ferent immune diseases or one spectrum? Curr. Opin. Immunol. 48,68–73 responses of human keratinocytes exposed to the sesquiterpene (2017). lactones dehydrocostuslactone and costunolide. PLoS. ONE 9, 41. Wu,H.H.,Xie,W.L.,Zhao,Y.K.,Liu,J.H.&Luo,D.Q.Imiquimodincreases e107904 (2014). cutaneous VEGF expression in imiquimod-induced psoriatic mouse model. 48. Orecchia, A. et al. Sirtinol treatment reduces inflammation in human dermal Curr. Vasc. Pharmacol. 14,275–279 (2016). microvascular endothelial cells. PLoS. ONE 6, e24307 (2011). 42. Palomo, J., Troccaz, S., Talabot-Ayer, D., Rodriguez, E. & Palmer, G. The severity 49. Palombo, R. et al. Luteolin-7-glucoside inhibits IL-22/STAT3 pathway, reducing of imiquimod-induced mouse skin inflammation is independent of endo- proliferation, acanthosis, and inflammation in keratinocytes and in mouse genous IL-38 expression. PLoS ONE 13, e0194667 (2018). psoriatic model. Cell Death Dis. 7, e2344 (2016).

Official journal of the Cell Death Differentiation Association