Dithranol targets keratinocytes, their crosstalk with neutrophils and inhibits the IL-36 inflammatory loop in psoriasis

  1. Theresa Benezeder
  2. Clemens Painsi
  3. VijayKumar Patra
  4. Saptaswa Dey
  5. Martin Holcmann
  6. Bernhard Lange-Asschenfeldt
  7. Maria Sibilia
  8. Peter Wolf  Is a corresponding author
  1. Department of Dermatology, Medical University of Graz, Austria
  2. State Hospital Klagenfurt, Austria
  3. Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Austria

Abstract

Despite the introduction of biologics, topical dithranol (anthralin) has remained one of the most effective anti-psoriatic agents. Serial biopsies from human psoriatic lesions and both the c-Jun/JunB and imiquimod psoriasis mouse model allowed us to study the therapeutic mechanism of this drug. Top differentially expressed genes in the early response to dithranol belonged to keratinocyte and epidermal differentiation pathways and IL-1 family members (i.e. IL36RN) but not elements of the IL-17/IL-23 axis. In human psoriatic response to dithranol, rapid decrease in expression of keratinocyte differentiation regulators (e.g. involucrin, SERPINB7 and SERPINB13), antimicrobial peptides (e.g. ß-defensins like DEFB4A, DEFB4B, DEFB103A, S100 proteins like S100A7, S100A12), chemotactic factors for neutrophils (e.g. CXCL5, CXCL8) and neutrophilic infiltration was followed with much delay by reduction in T cell infiltration. Targeting keratinocytes rather than immune cells may be an alternative approach in particular for topical anti-psoriatic treatment, an area with high need for new drugs.

Introduction

With the development and market introduction of biologics, much progress has been made in recent years in the systemic treatment of psoriasis. Currently, targeted therapy with antibodies against IL-17 or IL-23 exhibits high efficacy and allows complete or almost complete clinical clearance of psoriasis lesions in a high percentage of cases (Blauvelt et al., 2017; Langley et al., 2014; Papp et al., 2017; Reich et al., 2018). However, patients with limited body area involvement (i.e. mild forms of psoriasis) who represent the majority of psoriasis patients with up to 90% (Stern et al., 2004) have been neglected. Not much innovation has occurred in the past few years on topical treatment of psoriasis that is mainly prescribed to these patients. Steroids, vitamin D3 and vitamin A analogues are most commonly used as topical agents in such patients, but besides changes in their pharmaceutical formulation, they have not been developed further in recent years (de Korte et al., 2008; Krueger et al., 2001; Langley et al., 2011; Schaarschmidt et al., 2015). Short courses of dithranol (1,8-dihydroxy-9-anthracenone or anthralin) have been successfully used as intermittent topical treatment of psoriasis since 1916 (Nast et al., 2012; Sehgal et al., 2014; Vleuten et al., 1996). Despite its numerous disadvantages like brown staining and irritation of perilesional skin, dithranol has remained one of the most effective topical treatment modalities in psoriasis. Analogous to the most recent generation of biologics (including anti-IL-17 and anti-IL-23 antibodies), dithranol delivers PASI75 rates in 66–82.5% of patients with fast action and clearance of skin lesion within very few weeks (Kemény et al., 1990; Painsi et al., 2015b; Sehgal et al., 2014; van de Kerkhof, 2015). Although dithranol has been used for many years, its exact mechanism of action has remained largely unknown (Holstein et al., 2017; Kemény et al., 1990; Kucharekova et al., 2006; Sehgal et al., 2014).

With the aim of unraveling dithranol’s therapeutic mechanisms and to possibly uncover new targets for topical treatment of psoriasis, we conducted a clinical trial and employed several mouse models including the c-Jun/JunB knockout model (Zenz et al., 2005) and the imiquimod psoriasis model (van der Fits et al., 2009) in order to address this issue and elucidate dithranol's effects. In this study, we demonstrate that dithranol exerts its anti-psoriatic effects by directly targeting keratinocytes and their crosstalk with neutrophils, as well as disrupting the IL-36 inflammatory loop. Consistent with this finding, we observed that dithranol’s therapeutic activity was completely independent of its pro-inflammatory effect mainly on perilesional skin, thus overthrowing the long-believed paradigm that dithranol-induced irritation is crucial for its anti-psoriatic action and unraveling irritation merely as a bystander effect of treatment.

Results

Topical dithranol leads to fast reduction in PASI score linked to decrease in epidermal hyperproliferation and delayed reduction of inflammatory infiltrate in psoriatic skin

As depicted in Figure 1C (and Figure 1—figure supplement 1), dithranol did lead to a fast reduction of psoriatic skin lesions in most of the 15 patients of the study, as determined by psoriasis area and severity index (PASI) and local psoriasis severity index (PSI) of marker lesions. As shown in Table 1, the mean decrease in PASI score was 58% after 2–3 weeks, confirming its high clinical efficacy (Painsi et al., 2015a; Painsi et al., 2015b; Swinkels et al., 2004). Remarkably, dithranol treatment did lead to a visible inflammatory response only at perilesional skin sites, but not within psoriatic plaques and there was no correlation between dithranol-induced erythema and its anti-psoriatic effect (Figure 1—figure supplement 2). The clinical response was confirmed by the results of histological analysis of skin biopsies taken throughout the dithranol treatment course (Figure 1A and B). Dithranol application led to a significant reduction in epidermal hyperplasia (as measured by thickness of epidermis). Intriguingly, there was no significant change in dermal infiltrate score during treatment. At the follow-up visit, hyperplasia of the epidermis was reduced further and cellular infiltrate in the dermis was significantly diminished (Figure 2).

Figure 1 with 2 supplements see all
Dithranol leads to a fast reduction of psoriatic skin lesions as determined by local psoriasis severity index (PSI) of marker lesions.

(A,B) 15 psoriasis patients were treated with dithranol. Skin biopsies were taken from marker lesions at multiple timepoints: 1a = lesional skin at baseline, 2 = lesional skin at day 6, 3 = lesional skin at end of treatment, week 2–3, 4 = lesional skin at follow-up, 4–6 weeks after end of treatment. In addition, non-lesional skin at baseline (1b) was sampled. (C) Representative images of lesional psoriatic skin and local psoriasis severity index (PSI; sum of erythema (0–4), induration (0–4) and scaling (0–4); 0 = none, 1 = mild, 2 = moderate, 3 = severe, 4 = very severe) at different time points.

Histological and immunohistochemical analysis of lesional skin before (day 0), during (day 6), at end of treatment (week 2–3) and 4–6 weeks after ending treatment (follow-up).

(A) Representative H and E images, epidermal thickness and cellular infiltrate scoring. In treated psoriatic lesions, epidermal thickness was significantly decreased at week 2–3 and at follow-up. Infiltrate score (0 = none, 0.5 = none/low, 1 = low, 1.5 = low/moderate, 2 = moderate, 2.5 = moderate/high, 3 = high infiltration of immune cells) was significantly higher in untreated psoriatic lesions compared to non-lesional skin (NL) and healthy controls (HC). In treated psoriatic lesions, infiltrate score was significantly decreased at follow-up. (B) Ki67 staining in epidermis was significantly reduced at week 2–3 and follow-up. (C) CK16 staining in epidermis was significantly reduced at week 2–3 and follow-up. (D–I) Representative IHC images and mean cell counts for epidermis and dermis. Neutrophil cell counts were significantly reduced at week 2–3 and follow-up in epidermis and dermis (D). Langerhans cell (CD1a+) numbers in epidermis were significantly increased at follow-up (E). Epidermal FoxP3 (H) and CD8 (I) cell counts were significantly reduced at week 2–3. Dermal CD3 (F), CD4 (G), and FoxP3 (H) positive cell counts display significant reduction at follow-up. One-way ANOVA with Dunnet’s multiple comparisons test was used for statistics. Bars represent mean ± SD; *p≤0.05; **p≤0.01; ***p≤0.001; ****p≤0.0001; scale bar = 100 µm.

Table 1
Psoriasis area and severity index (PASI) and psoriasis severity index (PSI) from 15 psoriasis patients.
ParameterTime point
BaselineDay 6End of treatmentFollow-up
PASI
mean ± SD13.6 ± 10.39.0 ± 6.3
(p<0.0001)*
5.1 ± 3.8
(p=0.0001)*
5.7 ± 6.7
(p=0.0002)*
%reduction, mean ± SD (range)-32.9 ± 8.0
(16.7–44.3)
57.5 ± 9.5
(41.8–74.2)
56.1 ± 23.3
(3.1–81.0)
Local PSI
mean ± SD6.7 ± 1.13.3 ± 1.6
(p<0.0001)*
2.0 ± 1.5
(p<0.0001)*
1.6 ± 1.3
(p<0.0001)*
%reduction, mean ± SD (range)-52.6 ± 21.9
(14.3–100)
69.0 ± 23.9
(16.7–100)
76.3 ± 18.3
(37.5–100)
  1. *P value was determined using Wilcoxon test comparing indicated value to baseline.

Clinical response to dithranol in psoriasis patients is linked to I) fast upregulation of keratinization genes and downregulation of neutrophil chemotactic genes and neutrophilic infiltration followed by II) delayed downregulation of inflammatory response-related genes and proteins

Dithranol slowly diminished CD3, CD4, FoxP3 and CD8 cell counts in the dermis as evidenced by unaffected cell numbers early on during the treatment course and significant reduction only at the follow-up visit (4–6 weeks after end of treatment) (Figure 2). The response of T cells in the epidermis occurred a little earlier, with significant reduction present already at the end of treatment (week 2–3) (Figure 2). In agreement with other studies (Holstein et al., 2017; Swinkels et al., 2002a; Vleuten et al., 1996; Yamamoto and Nishioka, 2003), we found a fast decrease in epidermal hyperproliferation (epidermal thickness, Ki-67 and CK16 staining) at end of treatment (week 2–3). Neutrophil numbers (as assessed by myeloperoxidase staining) in epidermis and dermis were also significantly reduced at that time point. An increase in the number of Langerhans cells in the epidermis (as indicated by CD1a staining) was detected at the follow-up visit.

Microarray analysis comparing lesional skin at day 6 of treatment with lesional skin samples at baseline revealed that dithranol led to differential expression of 62 genes including 17 genes with reduced and 45 genes with increased expression. Among these differentially expressed genes (DEGs), there was a significant upregulation of genes involved in keratinization and keratinocyte differentiation (e.g. KRT2, LCE1C, KRT73, LCE1A) and establishment of skin barrier (e.g. FLG2, HRNR, FLG). Furthermore, dithranol downregulated genes of antimicrobial peptides (AMPs) such as ß-defensin-2 (DEFB4A, DEFB4B) and chemoattractants for neutrophils (e.g. CXCL5, CXCL8, PPBP1, TREM1) (Table 2).

Table 2
Top 45 differentially regulated genes (p-value<0.05, fold change >1.5) in dithranol-treated lesional skin after 6 days compared to baseline from 15 patients with psoriasis.
Probe set IDGene symbolGene titleDay 6 vs. baseline
(fold change)
16693318FLG2Filaggrin family member 22.89
16693303HRNRHornerin2.64
16903552NEBNebulin2.18
16765017KRT2Keratin 22.04
16693308FLGFilaggrin1.97
16761221CLEC2AC-type lectin domain family 2, member A1.92
16730782ELMOD1ELMO/CED-12 domain containing 11.91
16735288OVCH2Ovochymase 21.83
16990787SPINK7Serine peptidase inhibitor, Kazal type 7 (putative)1.78
16852824SERPINB12Serpin peptidase inhibitor, clade B (ovalbumin), member 121.74
16693341LCE1CLate cornified envelope 1C1.71
16693249THEM5Thioesterase superfamily member 51.70
16921644MIRLET7CMicroRNA let-7c1.69
16670681ANXA9Annexin A91.68
16821186CLEC3AC-type lectin domain family 3, member A1.68
16859090CASP14Caspase 141.68
16765005KRT73Keratin 731.66
16945497COL6A5Collagen, type VI, alpha 51.66
16976615SULT1E1Sulfotransferase family 1E, estrogen-preferring, member 11.65
16898858CD207CD207 molecule, langerin1.63
16861126UPK1AUroplakin 1A1.60
16704475FAM35DPFamily with sequence similarity 35, member A pseudogene1.60
17085015FRMPD1FERM and PDZ domain containing 11.59
16817034CHP2Calcineurin-like EF-hand protein 21.56
16671082LCE1ALate cornified envelope 1A1.55
16671037LCE2DLate cornified envelope 2D1.55
16748835PIK3C2GPhosphatidylinositol-4-phosphate 3-kinase, catalytic subunit1.54
17039517LY6G6CLymphocyte antigen six complex, locus G6C1.53
16673713FMO2Flavin containing monooxygenase 2 (non-functional)1.52
16812344BCL2A1BCL2-related protein A1−1.52
16968213ANXA3Annexin A3−1.54
17104519RNY4P23RNA, Ro-associated Y4 pseudogene 23−1.55
16948835MIR1224MicroRNA 1224−1.55
16815310TNFRSF12ATumor necrosis factor receptor superfamily, member 12A−1.60
17015084SERPINB1Serpin peptidase inhibitor, clade B (ovalbumin), member 1−1.67
17106398SLC6A14Solute carrier family 6 (amino acid transporter), member 14−1.69
16693375SPRR2FSmall proline-rich protein 2F−1.69
17065458DEFB4ADefensin, beta 4A−1.75
17074361DEFB4BDefensin, beta 4B−1.77
16698947RNU5A-8PRNA, U5A small nuclear 8, pseudogene−1.78
16976827CXCL5Chemokine (C-X-C motif) ligand 5−1.81
16976821PPBPPro-platelet basic protein (chemokine (C-X-C motif) ligand 7)−1.82
17019056TREM1Triggering receptor expressed on myeloid cells 1−1.99
17050251SLC26A4Solute carrier family 26 (anion exchanger), member 4−2.19
16967771CXCL8Chemokine (C-X-C motif) ligand 8−3.36

At the end of treatment (week 2–3), dithranol had deregulated a total of 453 genes, with downregulation of 325 DEGs and upregulation of 128 genes. Compared to day 6, expression of various genes involved in keratinocyte differentiation decreased further upon dithranol treatment and other AMPs (e.g. S100A7A, S100A12, DEFB103A) and genes involved in neutrophil-mediated inflammatory responses significantly diminished in their expression. With delay, dithranol also lowered expression of inflammatory response-related genes (e.g. IL1B, IL17, IL22, IL36A, IL36G, IL36RN) only at the end of treatment (week 2–3) (Table 3). Notably, genes involved in T-cell activation were not differentially expressed in the observation period (during dithranol up to week 2–3). To verify a subset of differentially expressed genes from the microarray data, we performed nCounter Nanostring analysis on 80 target and four reference genes. Ratios of microarray target genes strongly correlated with those obtained from Nanostring analysis at the early (day 6) (r = 0.8830) and late time point examined at the end of dithranol treatment (week 2–3) (r = 0.8859) (Supplementary file 2). Comparing day 6 vs. baseline, we found gene ontology (GO) terms related to keratinization (e.g. keratinocyte differentiation, establishment of skin barrier) and neutrophil chemotaxis (e.g. neutrophil migration, regulation of neutrophil migration) among the most significant GO groups (Table 4). Top significantly enriched GO terms at week 2–3 vs. baseline were related to immune response (e.g. inflammatory response, cytokine secretion) and differentiation of keratinocytes (e.g. epidermis development, keratinization) (Table 5). At follow-up (4–6 weeks after end of treatment), 10 of 13 patients showed >50% reduction in epidermal thickness and >75% reduction in Ki67 staining. These 10 patients were classified as histological responders. Microarray gene expression analysis at end of treatment (week 2–3) revealed that 131 genes were differentially expressed in histological responders compared to non-responders, predicting histological outcome for the follow-up time point 4–6 weeks after the end of treatment. Using gene ontology enrichment analysis of these DEGs, we found pathways like keratinocyte differentiation, cornification and keratin filament formation among the top 20 GO terms (Supplementary file 3).

Table 3
Top 45 differentially regulated genes in dithranol-treated lesional skin at end of treatment compared to baseline from 15 patients with psoriasis (p<0.05, fold change >1.5).
Probe set IDGene symbolGene titleEnd of treatment vs. baseline (fold change)
16947045AADACArylacetamide deacetylase3.07
16765005KRT73Keratin 732.31
16976868BTCBetacellulin2.27
16924792CLDN8Claudin 82.11
17125092CNTNAP3Contactin associated protein-like 32.09
17097358SLC46A2Solute carrier family 46, member 22.07
16780133SLITRK6SLIT and NTRK-like family, member 62.00
16834436RAMP2Receptor (G protein-coupled) activity modifying protein 21.96
17123970ZDHHC11BZinc finger, DHHC-type containing 11B1.88
17104313ARAndrogen receptor1.85
16951140MIR548I2MicroRNA 548i-21.85
17063366ATP6V0A4ATPase, H+ transporting, lysosomal V0 subunit a41.84
16974224MIR548I2MicroRNA 548i-21.83
16687914CYP2J2Cytochrome P450, family 2, subfamily J, polypeptide 21.82
17125106CNTNAP3BContactin associated protein-like 3B1.80
16903552NEBNebulin1.80
17125094CNTNAP3BContactin associated protein-like 3B1.80
16770284TMEM116Transmembrane protein 1161.79
16765041KRT77Keratin 771.79
17123972ZDHHC11BZinc finger, DHHC-type containing 11B1.78
16688210MIR3671MicroRNA 36711.78
17125218CNTNAP3Contactin associated protein-like 31.74
16764923KRT6AKeratin 6A−3.69
16767261IL22Interleukin 22−3.87
17065453DEFB103ADefensin, beta 103A−3.94
17074366DEFB103ADefensin, beta 103A−3.94
16671027LCE3CLate cornified envelope 3C−4.14
16743751MMP12Matrix metallopeptidase 12 (macrophage elastase)−4.42
16979444TNIP3TNFAIP3 interacting protein 3−4.48
17106398SLC6A14Solute carrier family 6 (amino acid transporter), member 14−4.60
16686734CYP4Z2PCytochrome P450, family 4, subfamily Z, polypeptide 2−4.89
16671144S100A7AS100 calcium binding protein A7A−5.00
16764907KRT6CKeratin 6C−5.01
16730157HEPHL1Hephaestin-like 1−5.15
16967831EPGNEpithelial mitogen−5.31
16842517NOS2Nitric oxide synthase 2, inducible−5.38
16693409S100A12S100 calcium binding protein A12−6.12
16813112RHCGRh family, C glycoprotein−6.18
16738803TCN1Transcobalamin I (vitamin B12 binding protein, R binder family)−6.45
16924785CLDN17Claudin 17−8.09
16693365SPRR2CSmall proline-rich protein 2C (pseudogene)−8.72
16967771CXCL8Chemokine (C-X-C motif) ligand 8−9.14
17074361DEFB4BDefensin, beta 4B−9.39
16693375SPRR2FSmall proline-rich protein 2F−10.12
16884602IL36AInterleukin 36, alpha−10.50
Table 4
Top 20 significantly enriched pathways as determined by Gene Ontology (GO) enrichment analysis in dithranol-treated lesional skin after 6 days compared to baseline from 15 patients with psoriasis.

(GO was done using Cytoscape software [Bindea et al., 2009; Shannon et al., 2003]; a.o. = among others).

GOIDGO termP-Value% Associated GenesAssociated genes found
GO:0001533Cornified envelope3.8E-1210.45FLG, HRNR, KRT2, LCE1A, LCE1C, LCE2D, SPRR2F
GO:0031424Keratinization7.7E-123.93CASP14, FLG, HRNR, KRT2, KRT73, LCE1A, LCE1C, LCE2D, SPRR2F
GO:0030216Keratinocyte differentiation79.0E-123.02CASP14, FLG, HRNR, KRT2, KRT73, LCE1A, LCE1C, LCE2D, SPRR2F
GO:0018149Peptide cross-linking300.0E-129.84FLG, KRT2, LCE1A, LCE1C, LCE2D, SPRR2F
GO:0070268Cornification12.0E-95.31CASP14, FLG, KRT2, KRT73, LCE1A, SPRR2F
GO:0004168Receptor CXCR2 binds ligands CXCL1 to 71.0E-633.33CXCL5, CXCL8, PPBP
GO:0042379Chemokine receptor binding5.4E-66.25CXCL5, CXCL8, DEFB4A, PPBP
GO:0045236CXCR chemokine receptor binding6.0E-618.75CXCL5, CXCL8, PPBP
GO:0061436Establishment of skin barrier18.0E-613.04FLG, FLG2, HRNR
GO:0033561Regulation of water loss via skin21.0E-612.00FLG, FLG2, HRNR
GO:0004657IL-17 signaling pathway21.0E-64.30CXCL5, CXCL8, DEFB4A, DEFB4B
GO:0007874Keratin filament formation21.0E-64.17FLG, KRT2, KRT73, SPRR2F
GO:0030593Neutrophil chemotaxis21.0E-64.17CXCL5, CXCL8, PPBP, TREM1
GO:1990266Neutrophil migration27.0E-63.85CXCL5, CXCL8, PPBP, TREM1
GO:0071621Granulocyte chemotaxis38.0E-63.31CXCL5, CXCL8, PPBP, TREM1
GO:1902622Regulation of neutrophil migration45.0E-67.50CXCL5, CXCL8, PPBP
GO:0071622Regulation of granulocyte chemotaxis71.0E-65.66CXCL5, CXCL8, PPBP
GO:0030104Water homeostasis130.0E-64.00FLG, FLG2, HRNR
GO:0002690Positive regulation of leukocyte chemotaxis120.0E-63.30CXCL5, CXCL8, PPBP
GO:0004867Serine-type endopeptidase inhibitor activity79.0E-63.00SERPINB1, SERPINB12, SPINK7
Table 5
Top 20 significantly enriched pathways as determined by Gene Ontology (GO) enrichment analysis in dithranol-treated lesional skin at end of treatment compared to baseline from 15 patients with psoriasis.

(GO was done using Cytoscape software; Bindea et al., 2009; Shannon et al., 2003 a.o. = among others).

GOIDGO termP-Value% Associated GenesAssociated genes found
GO:0006954Inflammatory response17.0E-186.93IL17A, IL1B, IL20, IL22, IL36A, IL36G, IL36RN, a.o.
GO:0006952Defense response52.0E-184.60CXCL8, CXCL9, DEFB103A, DEFB4A, IFNG, IL17A, IL1B, IL20, IL22, IL36A, IL36G, IL36RN, IL4R, IRAK2, IRF1, KRT16, a.o.
GO:0051707Response to other organism56.0E-186.02CCL2, CCL20, CCL22, CD24, CD80, COTL1, CXCL13, CXCL16, CXCL8, CXCL9, DDX21, DEFB103A, DEFB4A, a.o.
GO:0050663Cytokine secretion480.0E-1813.78IFNG, IL17A, IL1B, IL26, IL36RN, IL4R, LYN, MMP12, NOS2, PAEP, PANX1, PNP, S100A12, S100A8, S100A9, a.o.
GO:0001816Cytokine production680.0E-186.76IDO1, IFNG, IL17A, IL1B, IL26, IL36A, IL36RN, IL4R, IRF1, LTF, LYN, MB21D1, MMP12, NOS2, a.o.
GO:0012501Programmed cell death1.0E-154.05CASP5, CASP7, CCL2, CCL21, CD24, CD274, CD38, IFNG, IL17A, IL1B, IRF1, IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, KRT74, KRT77, a.o.
GO:0051240Positive regulation of multicellular organismal process1.5E-154.57CXCL17, CXCL8, FBN2, FERMT1, GBP5, GPR68, HPSE, HRH2, IDO1, IFNG, IL17A, IL1B, IL20, IL26, IL36A, S100A8, S100A9, SERPINB3, SERPINB7, a.o.
GO:0001817Regulation of cytokine production2.0E-157.04CCL2, CCL20, CD24, CD274, CD80, CD83, CXCL17, IFNG, IL17A, IL1B, IL26, IL36A, IL36RN, IL4R, IRF1, TNFRSF9, WNT5A, a.o.
GO:0002237Response to molecule of bacterial origin15.0E-159.22S100A8, S100A9, SELE, SOD2, TIMP4, TNFRSF9, TNIP3, WNT5A, ZC3H12A, a.o.
GO:0070268Cornification100.0E-1517.70DSC2, DSG3, IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, KRT74, KRT77, PI3, SPRR2A, SPRR2B, SPRR2D, a.o.
GO:0043588Skin development190.0E-158.17IL20, IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, KRT74, KRT77, LCE3A, LCE3C, LCE3E, a.o.
GO:0008544Epidermis development220.0E-157.63DSC2, DSG3, EPHA2, FERMT1, FOXE1, FURIN, HPSE, IL20, IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, KRT74, KRT77, a.o.
GO:0009617Response to bacterium240.0E-156.63CCL2, CCL20, CD24, CD80, CXCL13, CXCL16, CXCL8, CXCL9, DEFB103A, DEFB4A, S100A12, S100A8, S100A9, TREM1, a.o.
GO:0001819Positive regulation of cytokine production1.4E-127.89CCL2, CCL20, CD274, CD80, CD83, CXCL17, FERMT1, GBP5, HPSE, IDO1, IFNG, IL17A, IL1B, IL26, IL36A, a.o.
GO:0050707Regulation of cytokine secretion2.4E-1213.10AIM2, CASP5, CD274, FERMT1, GBP1, IFNG, IL17A, IL1B, IL26, IL36RN, IL4R, LYN, MMP12, PAEP, PANX1, a.o.
GO:0005125Cytokine activity4.4E-1210.64CCL20, CCL21, CCL22, CCL4L2, CXCL13, CXCL16, CXCL8, CXCL9, FAM3D, IFNG, IL17A, IL1B, IL20, IL22, IL26, a.o.
GO:0031424Keratinization21.0E-1210.48IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, KRT74, KRT77, LCE3A, LCE3C, LCE3E, PI3, SPRR2A, SPRR2B, a.o.
GO:0002790Peptide secretion97.0E-126.25CD274, CD38, DOC2B, FAM3D, FERMT1, GBP1, GBP5, GLUL, GPR68, IFNG, IL17A, IL1B, IL26, IL36RN, IL4R, LYN, MMP12, NOS2, TREM1, WNT5A, a.o.
GO:0032940Secretion by cell140.0E-124.04AIM2, AMPD3, CASP5, IL36RN, IL4R, LCN2, LRG1, LTF, LYN, MMP12, NOS2, NR1D1, NR4A3, OLR1, PAEP, PANX1, PLA2G3, a.o.
GO:0009913Epidermal cell differentiation190.0E-127.91DSC2, DSG3, EPHA2, FURIN, IL20, IVL, KLK13, KRT16, KRT17, KRT31, KRT6A, KRT6C, KRT73, LCE3E, PI3, SLITRK6, SPRR2A, SPRR2B, SPRR2D, SPRR2E, a.o.

Topical application of dithranol ameliorates psoriasis-like skin lesions in c-Jun/JunB knockout mice by directly targeting keratinocyte genes and inhibiting IL36RN

To study the effect of dithranol in a keratinocyte-based psoriatic mouse model, we used c-Jun/JunB knockout mice (treatment protocol shown in Figure 3A). Topical dithranol application strongly reduced psoriatic lesions as measured by macroscopic overall ear thickness and microscopic epidermal thickness in this genetic model of psoriasis based on inducible epidermal deletion of the AP1 transcription factors c-Jun/JunB (Glitzner et al., 2014; Zenz et al., 2005Figure 3B–D). There was no difference in the outcome to dithranol with regard to different concentrations series, therefore, data was pooled for certain analyses as indicated (Figure 3). Gene expression profiling using Clariom S mouse microarray showed that among the top 45 differentially regulated genes, dithranol downregulated genes belonging to the group of late cornified envelope genes (Lce6a, Lce1i, Lce1g, Lce1f), as well as hornerin (Hrnr) (Table 6). Gene ontology enrichment analysis of all differentially expressed genes (fold change >1.5 and p-value<0.05) revealed skin development, epidermis development, epithelial cell differentiation and keratinization among the top 20 significantly enriched pathways (Table 7). Associated genes found were for example Flg2, Ivl, Hrnr, Lor, Krt2, Casp14, Lce1c, Serpinb7 and Serpbinb13, among others. We then compared DEGs from psoriasis patients treated with dithranol to DEGs from dithranol-treated c-Jun/JunB knockout mice (Figure 4). In total, the microarray assay allowed us to compare 300 DEGs in dithranol-treated psoriasis patients (week 2–3 vs. day 0) and 18 DEGs from day 6 vs. day 0 with 336 murine DEGs from dithranol-treated c-Jun/JunB knockout mice. Notably, comparing DEGs from day 6 vs. day 0 with murine DEGs, we found an overlap of 11 genes. Among these genes were CASP14, a non-apoptotic caspase involved in epidermal differentiation and FLG2, HRNR, KRT2, LCE1C and SERPINB12, involved in keratinocyte differentiation and epidermis development (Bergboer et al., 2011; Henry et al., 2011; Henry, 2012; Kuechle et al., 2001; Sandilands et al., 2009; Sivaprasad et al., 2015; Toulza et al., 2007). Even more strikingly, the overlap between DEGs from week 2–3 vs. day 0 and murine DEGs comprised 20 genes including IL36RN, IVL, SERPINB7 and SERPINB13, that were all increased in lesional skin at baseline compared to non-lesional skin and downregulated by dithranol treatment. Findings from microarray analysis were verified by RT-PCR (Figure 4—figure supplement 1). The group of genes encoding serpins, as well as involucrin have been shown to play a role in keratinocyte differentiation (Henry, 2012; Toulza et al., 2007) and have been associated with psoriasis (Roberson and Bowcock, 2010; Suárez-Fariñas et al., 2012; Wolf et al., 2012), belonging to the ‘psoriasis transcriptome’ identified by Tian et al., 2012. In addition, dithranol downregulated expression of elevated IL36A and IL36G in human psoriatic skin and IL36B (Il1f8) in c-Jun/JunB psoriatic skin. To substantiate dithranol’s effect on keratinocytes, we employed the mouse-tail test, a traditional model to quantify the effect of topical anti-psoriatics on keratinocyte differentiation by measuring degree of orthokeratosis versus parakeratosis (Bosman et al., 1992; Sebök et al., 2000; Wu et al., 2015). We found a strong increase in percentage of orthokeratosis (from 18.8 to 63.4%) reflecting dithranol’s keratinocyte differentiation-inducing activity (Figure 3—figure supplement 1), consistent with previous work (Bosman et al., 1992; Hofbauer et al., 1988; Sebök et al., 2000; Wrench and Britten, 1975). Next, we performed RT-PCRs of a selected panel of keratinocyte differentiation markers, AMPs and inflammatory markers (based on our microarray data) of dithranol-treated murine tail skin. We found a strong upregulation of keratinization markers (Flg, Krt16, Serpinb3a) and several AMPs (Lcn2, S100a8, S100a9, Defb3)) (Figure 3—figure supplement 2). Interestingly, dithranol downregulated expression of the antimicrobial peptide Camp/LL37, as well as Cxcl5, a chemotactic factor for neutrophils.

Figure 3 with 3 supplements see all
Topical application of dithranol ameliorates psoriasis-like skin lesions in c-Jun/JunB knockout mice.

(A) Schematic representation of experimental set-up. Red arrows indicate dithranol application in series of increasing concentrations. (Exp. 1 and 2 = experiment 1 and 2) (B) Macroscopic ear thickness on day 1 compared to day 7. Dithranol treatment led to a significant reduction in ear thickness. Controls (n = 11), dithranol group (n = 11); Paired t-test was used for statistics. (C) Representative H and E images of untreated and dithranol-treated ears. (D) Dithranol treatment led to a significant reduction in epidermis thickness. Controls (n = 10), dithranol group (n = 11); unpaired t-test was used for statistics. Data from the two experiments was pooled (D). Bars represent mean ± SD; n.s. = not significant; *p≤0.05; **p≤0.01; ***p≤0.001; ****p≤0.0001; scale bar = 100 µm.

Figure 4 with 1 supplement see all
Venn diagram (created using InteractiVenn Heberle et al., 2015) showing comparison and overlap between differentially expressed genes (DEGs) in dithranol-treated human psoriatic skin at week 2–3 vs. day 0, DEGs in dithranol-treated human psoriatic skin at day 6 vs. day 0 and DEGs in dithranol-treated c-Jun/JunB psoriatic skin.
Table 6
Top 45 differentially regulated genes in dithranol-treated psoriatic skin of c-Jun/JunB knockout mice compared to that of vehicle-treated controls (p<0.05, fold change (FC) >1.5).
Probe set IDGene symbolGene titleFold change
TC0Y00000006.mm.2Eif2s3yEukaryotic translation initiation factor 2, subunit 3, structural gene Y-linked10,37
TC0Y00000233.mm.2UtyUbiquitously transcribed tetratricopeptide repeat gene, Y chromosome6,38
TC0Y00000235.mm.2Ddx3yDEAD (Asp-Glu-Ala-Asp) box polypeptide 3, Y-linked5,40
TC0900000047.mm.2Mmp3Matrix metallopeptidase 33,64
TC0Y00000079.mm.2Ssty2Spermiogenesis specific transcript on the Y 23,31
TC0100001632.mm.2Ifi209Interferon activated gene 2093,23
TC0500002755.mm.2Cxcl9Chemokine (C-X-C motif) ligand 92,85
TC0300003133.mm.2Ifi44Interferon-induced protein 442,69
TC0100003591.mm.2Ifi213Interferon activated gene 2132,53
TC0100001634.mm.2Ifi208Interferon activated gene 2082,49
TC1900000217.mm.2Ms4a4cMembrane-spanning 4-domains, subfamily A, member 4C2,45
TC0300002684.mm.2Chil3Chitinase-like 32,39
TC0100003550.mm.2Slamf7SLAM family member 72,33
TC0500000922.mm.2Cxcl13Chemokine (C-X-C motif) ligand 132,26
TC1900000500.mm.2Ifit2Interferon-induced protein with tetratricopeptide repeats 22,20
TC0500002840.mm.2Plac8Placenta-specific 82,20
TC0700001630.mm.2AdmAdrenomedullin2,15
TC1900000501.mm.2Ifit3Interferon-induced protein with tetratricopeptide repeats 32,13
TC1800000610.mm.2Iigp1Interferon inducible GTPase 12,07
TC0300001446.mm.2Gbp3Guanylate binding protein 32,07
TC0300003134.mm.2Ifi44lInterferon-induced protein 44 like2,05
TC1900000502.mm.2Ifit3bInterferon-induced protein with tetratricopeptide repeats 3B2,02
TC0400003296.mm.2Skint5Selection and upkeep of intraepithelial T cells 5−2,37
TC0300000482.mm.2AadacArylacetamide deacetylase−2,37
TC1100000469.mm.2Fndc9Fibronectin type III domain containing 9−2,39
TC0300002399.mm.2Lce6aLate cornified envelope 6A−2,42
TC0300002409.mm.2Lce1iLate cornified envelope 1I−2,44
TC0300002412.mm.2KprpKeratinocyte expressed, proline-rich−2,44
TC0300000846.mm.2HrnrHornerin−2,48
TC0300002407.mm.2Lce1gLate cornified envelope 1G−2,49
TC1600000324.mm.2FetubFetuin beta−2,53
TC1300001377.mm.2Akr1c18Aldo-keto reductase family 1, member C18−2,56
TC0300002406.mm.2Lce1fLate cornified envelope 1F−2,57
TC1500001714.mm.2Slurp1Secreted Ly6/Plaur domain containing 1−2,57
TC1500002344.mm.2Gsdmc2Gasdermin C2−2,59
TC1000000145.mm.2Il20raInterleukin 20 receptor, alpha−2,66
TC0700000443.mm.2Cyp2b19Cytochrome P450, family 2, subfamily b, polypeptide 19−2,70
TC0100000103.mm.2Ly96Lymphocyte antigen 96−2,89
TC0700000778.mm.2Klk5Kallikrein related-peptidase 5−3,15
TC0300003252.mm.2Clca3a2Chloride channel accessory 3A2−3,20
TC0900002312.mm.2Elmod1ELMO/CED-12 domain containing 1−3,20
TC0300002401.mm.2Lce1a1Late cornified envelope 1A1−3,26
TC0700000484.mm.2FcgbpFc fragment of IgG binding protein−3,83
TC0300002405.mm.2Lce1eLate cornified envelope 1E−4,16
TC1500002274.mm.2Krt2Keratin 2−5,95
Table 7
Top 20 significantly enriched pathways as determined by Gene Ontology (GO) enrichment analysis in dithranol-treated psoriatic skin of c-Jun/JunB knockout mice compared to that of vehicle-treated controls.

(GO was done using Cytoscape software Bindea et al., 2009; Shannon et al., 2003; a.o. = among others).

Go idGO termP-Value% Associated GenesAssociated genes found
GO:0043588Skin development14,0E-1811,11Casp14, Cldn1, Flg2, Gjb3, Hrnr, Ivl, Krt2, Lce1a1, Lce1a2, Lce1l, Lce1m, Lor, Pou2f3, Ptgs1, Scel, Tfap2b, a.o.
GO:0008544Epidermis development73,0E-159,18Acer1, Alox8, Casp14, Cnfn, Cst6, Dnase1l2, Hrnr, Ivl, Krt2, Lce1c, Lce1d, Lce1e, Lce1f, Lce1g, Lce1h, Lce1i, Lce1j, a.o.
GO:0071345Cellular response to cytokine stimulus1,7E-126,26Ccdc3, Ccl2, Ccl5, Ccr9, Cxcl13, Cxcl9, Edn1, Il18, Il1f5, Il1f8, Il1rl1, Il20ra, Stat2, a.o.
GO:0034097Response to cytokine7,1E-125,62Cd38, Chad, Cldn1, Csf3, Edn1, Gbp2, Gbp3, Gbp4, Gbp7, Gbp8, Gbp9, Gm4951, Ifi203, Ifi204, Ifi209, Ifit1, Ifit2, Ifit3, Ifit3b, Xaf1, a.o.
GO:0035456Response to interferon-beta12,0E-1225,00Gbp2, Gbp3, Gbp4, Gm4951, Ifi203, Ifi204, Ifi209, Ifit1, Ifit3, Iigp1, Irgm1, Xaf1
GO:0006952Defense response63,0E-124,11Ccl2, Ccl5, Cd180, Cd59a, Cxcl13, Cxcl9, Cybb, Defb6, Drd1, Herc6, Hp, Il18, Il1f5, Il1f8, Il1rl1, Irgm1, Kalrn, Klk5, a.o.
GO:0030855Epithelial cell differentiation430,0E-125,51Casp14, Cdkn1a, Cldn1, Cnfn, Dlx3, Dnase1l2, Gsdmc2, Gstk1, Hrnr, Ivl, Klf15, Krt2, Lce1a1, Lor, Pou2f3, a.o.
GO:0020005Symbiont-containing vacuole membrane1,4E-966,67Gbp2, Gbp3, Gbp4, Gbp7, Gbp9, Iigp1
GO:0044216Other organism cell5,6E-930,77C4b, Gbp2, Gbp3, Gbp4, Gbp7, Gbp9, Iigp1, Tap1
GO:0044406Adhesion of symbiont to host120,0E-937,50Ace2, Gbp2, Gbp3, Gbp4, Gbp7, Gbp9
GO:0045087Innate immune response390,0E-94,35Ccl2, Ccl5, Cd180, Cfb, Cldn1, Ifit3, Iigp1, Il1f5, Il1f8, Irgm1, Lbp, Ly96, Mx1, Parp9, Sla, Slamf6, Slamf7, Stat2, Tlr7, Trim62, a.o.
GO:0034341Response to interferon-gamma1,0E-610,58Ccl2, Ccl5, Cldn1, Edn1, Gbp2, Gbp3, Gbp4, Gbp7, Gbp8, Gbp9, Parp9
GO:0006954Inflammatory response5,9E-64,29C3, C4b, Ccl2, Ccl5, Cd180, Cd59a, Chil3, Crip2, Ctla2a, Cxcl13, Cxcl9, Cybb, Hp, Il18, Il1f5, Il1f8, Il1rl1, Lbp, Ly96, a.o.
GO:0042832Defense response to protozoan9,4E-619,35Gbp2, Gbp3, Gbp4, Gbp7, Gbp9, Iigp1
GO:0035457Cellular response to interferon-alpha21,0E-636,36Ifit1, Ifit2, Ifit3, Ifit3b
GO:0030414Peptidase inhibitor activity22,0E-66,19C3, C4b, Cst6, Ctla2b, Fetub, R3hdml, Serpinb12, Serpinb13, Serpinb2, Serpinb7, Spink14, Tfap2b, Wfdc12, Wfdc5
GO:0098542Defense response to other organism34,0E-64,05Adm, Ccl5, Cxcl13, Cxcl9, Defb6, Gbp2, Gbp3, Hp, Ifit1, Ifit2, Ifit3, Ifit3b, Iigp1, Il1f5, Klk5, Lbp, Mx1, Oas1f, Plac8, Stat2, Tlr7, Wfdc12, a.o.
GO:0031424Keratinization43,0E-615,00Casp14, Cnfn, Hrnr, Ivl, Krt2, Lor
GO:0044403Symbiosis, encompassing mutualism through parasitism55,0E-64,11Ace2, Acta2, Atg16l2, Ccl5, Cxcl9, Gbp2, Gbp3, Gbp4, Gbp7, Gbp9, Ifit1, Ifit2, Ifit3, Ifit3b, Lbp, Mx1, Oas1f, Pou2f3, Rab9, Stat2, Tap1, Tlr7, Trim62

In contrast to the effects of dithranol in the c-Jun/JunB model and mouse-tail test, this agent had no therapeutic capacity in the immunologically mediated imiquimod (IMQ) mouse model, which is often referred to as a psoriatic-like skin inflammation model (van der Fits et al., 2009). Indeed, dithranol treatment worsened psoriatic lesions in that model. Overall skin thickness was significantly enhanced, consistent with an increase in epidermal thickness and worsened inflammation (as measured by cellular infiltrate score) in mice treated with both IMQ and dithranol compared to IMQ-treated mice (Figure 3—figure supplement 3). Different set-ups (i.e. simultaneous treatment with dithranol and IMQ and pre-treatment with IMQ for 5 days followed by dithranol treatment) were tested, but similar effects were observed (data not shown). Taken together, these results demonstrate that dithranol primarily targets keratinocytes and only has delayed effects on other immune cells such as T cells belonging to the IL-17/IL-23 axis in psoriatic skin.

Discussion

This study demonstrates that topical dithranol directly targets keratinocytes (in particular their differentiation regulators and AMPs), keratinocyte-neutrophil crosstalk and inhibits the IL-36 inflammatory loop in psoriasis, thus unraveling after over 100 years of use, the therapeutic mechanism of one of the most effective topical treatments of psoriasis. Dithranol significantly diminished mRNA expression of pro-psoriatic IL-1 family members (IL36A, IL36G and IL36RN) (Supplementary file 1 and Figure 4). At day 6 after start of dithranol treatment, PASI had decreased by 33%, but at week 2–3 we saw a reduction of 58%, an effect that was paralleled by reduced expression of IL-36-related genes in psoriasis patients. The therapeutic importance of reduction of IL-1 family members in human skin was substantiated by results generated in the keratinocyte-based c-Jun/JunB mouse psoriasis model (Zenz et al., 2005). Expression of IL36RN (murine Il1f5) and IL36B (Il1f8) was significantly reduced in dithranol-treated psoriatic c-Jun/JunB lesions compared to controls (Figure 4) and among the top differentially expressed genes in our human and mouse dataset were genes involved in keratinocyte and epidermal differentiation (Table 4, Table 7). Dithranol strongly reduced mRNA expression of the keratinocyte differentiation regulator involucrin (IVL) and members of the serpin family (SERPBINB7, SERBINB13) both in lesional skin of patients and c-Jun/JunB knockout mice (Figure 4). Moreover, dithranol downregulated expression of AMPs such as ß-defensins (DEFB4A and DEFB4B) produced by keratinocytes (Liu et al., 2002; Schröder and Harder, 1999) within 6 days and chemotactic factors for neutrophils (such as CXCL5 and CXCL8) (Albanesi et al., 2018Table 2) and neutrophilic infiltration (as determined by MPO staining) within 2–3 weeks of treatment in human psoriatic skin (Figure 2).

Surprisingly, there were no significant changes in overall T cell numbers including CD4+ and CD8+ T cells, as well as FoxP3 positivity indicative for regulatory T cells in the skin in the early phase (within 6 days) during dithranol treatment. Reflecting dithranol’s primary effect on the epidermal compartment, the reduction of T cell numbers in the epidermis preceded that in the dermis. Whereas dithranol had decreased T cell counts in the epidermis at week 2–3, an effect on T cell numbers in the dermis was only evident at the follow-up visit, 4–6 weeks after the end of dithranol treatment (Figure 2), in agreement with previous reports favoring dithranol’s effect on keratinocytes (Holstein et al., 2017; Swinkels et al., 2002a; Vleuten et al., 1996; Yamamoto and Nishioka, 2003).

Vleuten et al., 1996 and Swinkels et al., 2002a applied immunohistochemistry to analyze differentiation and proliferation markers as well as T cell numbers in the skin and observed, as we did, a decrease in keratin 16, restoration of filaggrin and a decrease of Ki67 in the epidermis after 2 weeks of dithranol treatment. However, their data on the effect of dithranol on dermal T cell numbers at 4 weeks was controversial (Swinkels et al., 2002a; Vleuten et al., 1996). The group of Eberle recently tested the effects of dithranol using primary keratinocytes, a 3D psoriasis tissue model and some biopsy samples from psoriasis patients and reported on a reduction in Ki67 and keratin 16 positive cells in the epidermis using immunostaining and an inhibition of the antimicrobial peptide DEFB4 using qPCR analysis. However, based on their observations, they concluded that dithranol’s anti-psoriatic effects cannot be explained by direct effects on keratinocyte differentiation or cytokine expression (Holstein et al., 2017).

Our genome-wide expression analysis indicates that dithranol primarily targets keratinocytes and that this is crucial for response to treatment, considering that differentially regulated genes in histological responders compared to non-responders belonged to pathways like keratinocyte differentiation, cornification and keratin filament formation (Supplementary file 3). The importance of dithranol’s direct effect on keratinocytes has been further substantiated by our findings generated using the mouse-tail model, a simple in vivo model to analyze effects of topical preparations on keratinocyte differentiation and parakeratosis (Bosman et al., 1992; Sebök et al., 2000; Wu et al., 2015). Similar to previous studies (Bosman et al., 1992; Hofbauer et al., 1988; Sebök et al., 2000; Wrench and Britten, 1975), we observed a strong increase in orthokeratosis after dithranol application in the mouse-tail test, reflecting its keratinocyte differentiation-inducing activity (Figure 3—figure supplement 1). Our transcriptional analysis indicated that the effect of dithranol in the mouse-tail test was linked to a strong upregulation of keratinocyte differentiation markers and several AMPs, while the pro-psoriatic antimicrobial peptide Camp/LL37 was downregulated, as well as Cxcl5, a chemotactic factor for neutrophils (Figure 3—figure supplement 2), but not other pro-psoriatic AMPs (Fritz et al., 2017; Wang et al., 2018) such as Defb3, S100a8 or S100a9. Although the mouse-tail test has evidently limitations since the disturbed cell differentiation of this model only reflects one of many aspects of psoriasis, it supports the primary effect of dithranol on keratinocytes with induced induction of orthokeratosis.

In contrast to its therapeutic effect in both keratinocyte-driven psoriasis models, the c-Jun/JunB model and mouse-tail test, dithranol did aggravate psoriatic lesions in the imiquimod model that has been solely shown to be immunologically mediated and dependent on the IL-17/IL-23 axis (van der Fits et al., 2009). In the latter model, biologics such as etanercept and anti-IL-17a agents (Liu et al., 2018), topical steroids (Sun et al., 2014) and vitamin d3 analogues (Germán et al., 2019) but also UVB and PUVA (Shirsath et al., 2018) were shown to have a beneficial effect. In contrast, dithranol significantly enhanced overall macroscopic skin thickness, consistent with a slight increase in epidermal hyperplasia and worsened inflammation (as measured by the density of cellular infiltrate in the dermis) (Figure 3—figure supplement 3). Dithranol treatment of healthy murine skin led to similar effects upon irritation, as it increased epidermal thickness and cellular infiltrate of the skin (data not shown). However, the irritant effect of dithranol may remain without functional anti-psoriatic relevance in human psoriasis (as indicated by the clinical results depicted in Figure 1—figure supplement 2 and discussed below) but might be crucial in the agent's therapeutic action in alopecia areata (Nasimi et al., 2019; Ngwanya et al., 2017).

Together, these data unambiguously demonstrate that dithranol directly acts on keratinocytes, their crosstalk with neutrophils and IL-36 signaling, with AMPs being the potential link (Figure 5). This goes also in line with the observation that Langerhans cells as another type of immune cells showed a delayed response to dithranol, with no changes during treatment and an evident increase in numbers only later on (at the follow-up visit 4–6 weeks after end of treatment). These results are consistent with previous studies performed by Swinkels et al., showing that there was no significant change in T cells or Langerhans cells during twelve days of dithranol treatment (Swinkels et al., 2002a).

Proposed model of dithranol’s mechanism of action in psoriasis.

Dithranol decreases expression of keratinocyte differentiation regulators (involucrin and serpins), IL-36-related genes, keratinocyte-derived antimicrobial peptides (AMPs) (S100A7/A12 and ß-defensins) and chemotactic factors for neutrophils (CXCL5, CXCL8). This disrupts the crosstalk between keratinocytes and neutrophils and leads to diminished neutrophilic infiltration, thereby halting the inflammatory feedback loop in psoriasis. Together, this results in clearance of psoriatic lesions.

Our findings on dithranol’s effect on members of the IL-1 family, being beneficial for its anti-psoriatic efficacy, are well in line with recent work on blockade of IL-36 pathway as a novel strategy for the treatment of pustular psoriasis (Bachelez et al., 2019) as well as plaque-type psoriasis (Benezeder and Wolf, 2019; Mahil et al., 2017). Notably, human keratinocytes express IL-1 family members (IL-36α, IL-36β, IL-36γ, IL-36Ra) and their receptor IL-36R (D'Erme et al., 2015; Foster et al., 2014; Johnston et al., 2017). Furthermore, normal human keratinocytes show increased expression of IL-1 group mRNA after treatment with psoriasis-associated cytokines (TNFα, IL-1α, IL-17,IL-22) (Johnston et al., 2011). Genome-wide association studies revealed that deficiency in interleukin-36 receptor antagonist due to IL36RN mutations was associated with generalized pustular psoriasis (GPP) (Marrakchi et al., 2011; Sugiura et al., 2013). Supporting this notion, blocking IL-36 receptor was effective in reducing epidermal hyperplasia and showed comparable effects to etanercept in a psoriatic skin xenotransplantation model (Blumberg et al., 2010). Furthermore, successful treatment of psoriasis with the anti-psoriatic standard treatment etanercept is accompanied by a decrease in IL36A, IL36G and IL36RN expression (Johnston et al., 2011). A recent clinical phase one study provided proof-of-concept for the efficacy of BI 655130, a monoclonal antibody against the interleukin-36 receptor in the treatment of generalized pustular psoriasis (Bachelez et al., 2019). Intriguingly, topical short-contact dithranol therapy is efficacious not only in plaque-type psoriasis, but under certain conditions (i.e. after stabilization of disease activity with bland emollients) reportedly also in pustular psoriasis (Farber and Nall, 1993; Gerritsen, 1999). In this neutrophilic-driven condition, in which one might expect that dithranol worsens a heavy inflammatory state, it may have beneficial capacity by targeting the IL-36 pathway and neutrophils, both playing a most crucial role in this type of psoriasis (Bachelez, 2018; Bachelez et al., 2019; Johnston et al., 2017; Marrakchi et al., 2011; Sugiura et al., 2013).

As outlined above, we observed a strong effect of dithranol on keratinocyte-neutrophil crosstalk. Importantly, early response to anti-IL-17a blockade, as one of the most effective biological treatments currently available, has been linked to inhibition of keratinocyte-neutrophil crosstalk (Reich et al., 2015). Similar to our observation, anti-IL-17a treatment with secukinumab significantly reduced epidermal hyperproliferation after 2 weeks, decreased mRNA expression of keratinocyte-derived chemotactic factors and led to a strong decrease in IL-17a positive neutrophils. Apparently, disrupting the crosstalk between keratinocytes and neutrophils may be a crucial early effect in the clinical efficacy of secukinumab in psoriasis (Reich et al., 2015). In addition, decreased expression of AMPs like ß-defensin and S100 proteins was observed after only 1 week of anti-IL-17a treatment with secukinumab (Krueger et al., 2019) and after 2 weeks of ixekizumab treatment (Krueger et al., 2012), well in line with our observed early effect of dithranol on AMPs. Krueger et al. concluded that clinical efficacy of anti-IL-17a treatment is closely linked to early inhibition of keratinocyte-derived products such as chemokines and AMPs. Together this suggests that dithranol’s direct action on keratinocytes at the molecular level may disrupt IL-17 pathway dysregulation (without directly blocking IL-17 or its receptor), leading in turn to similar downstream effects as treatment with anti-IL-17 antagonists.

Our study also negates the paradigm that dithranol-induced irritation is crucial for its anti-psoriatic action (Gerritsen, 1999; Kucharekova et al., 2005; Prins et al., 1998; van de Kerkhof, 1991; Wiegrebe and Müller, 1995). As depicted in Figure 1—figure supplement 2, there was no correlation between dithranol-induced perilesional as well as lesional erythema and its anti-psoriatic effect, indicating that dithranol’s irritation (Prins et al., 1998; Swinkels et al., 2002c; Swinkels et al., 2002b) is a treatment side-effect unrelated to its therapeutic mechanism. However, this irritant effect of dithranol may be crucial for its action in alopecia areata, a condition, in which it was shown to be greatly effective, leading to hair regrowth in a high percentage of cases (Ngwanya et al., 2017). Similar to other topical treatment options (Nasimi et al., 2019; Yoshimasu and Furukawa, 2016), an initial irritant reaction to dithranol is followed by regrowth of hair within weeks after treatment.

What our work does not answer, is how dithranol exactly acts at the molecular level. Cell culture studies have shown that dithranol targets mitochondria (McGill et al., 2005), alters cellular metabolism (Hollywood et al., 2015) and induces apoptosis in keratinocytes (George et al., 2013; McGill et al., 2005). Dithranol also inhibits human monocytes in vitro, inhibiting secretion of IL-6, IL-8 and TNFα (Mrowietz et al., 1992; Mrowietz et al., 1997). Its effects on neutrophils were also shown in vitro, where dithranol leads to an increase in superoxide generation and a decrease in leukotriene production in neutrophils (Kavanagh et al., 1996; Schröder, 1986). Moreover, the potential receptor of dithranol remains undefined. Anti-psoriatic effects of other topical treatment options have recently been linked to modulation of the aryl hydrocarbon receptor (AhR) (Smith et al., 2017) and AhR may play a role in pathogenesis of psoriasis (Di Meglio et al., 2014). However, it appears that dithranol does not act via modulation of AhR, as we did not observe differences in the response of skin to dithranol comparing AhR knockout mice to controls (data not shown). There is need for further studies investigating how dithranol exactly acts on the molecular level, which receptor it potentially binds to or inhibits or whether it acts through modulation of a specific transcription factor such as NF-kB or STATs. Another question is whether the effect of dithranol is specific compared to other topical treatments such as steroids and vitamin D3 analogues. There seem to be some overlapping mechanisms between dithranol and vitamin D3 analogues such as calcipotriol that has been shown to act on keratinocytes to repress the expression of IL-36α/γ, an effect mediated through keratinocytic vitamin D receptor (Germán et al., 2019). Moreover, similar to dithranol, calcipotriol decreased expression of AMPs such as ß-defensins in keratinocytes of psoriatic plaques (Peric et al., 2009). At the same time, calcipotriol normalized the proinflammatory cytokine milieu and decreased IL-17A, IL-17F and IL-8 transcript abundance in lesional psoriatic skin. Calcipotriol also directly targets Th17 cells (Fujiyama et al., 2016) and CD8+IL-17+ cells (Dyring-Andersen et al., 2015), whereas we found that dithranol only has delayed effects on T cells. Moreover, cathelicidin (LL37) expression was increased by calcipotriol (Peric et al., 2009), juxtaposing the results of dithranol treatment at least in the mouse-tail test of present study. That dithranol and vitamin D3 analogues may have similar basic mechanisms of action is also supported by the notion that depending on the concentration, both dithranol (Nasimi et al., 2019; Ngwanya et al., 2017) and calcipotriol (El Taieb et al., 2019; Fullerton et al., 1998; Molinelli et al., 2020) can be irritant to the skin and induce hair regrowth in alopecia areata. Compared to vitamin D3 analogues topical steroids have an even broader mechanisms of action in psoriasis linked to their anti-inflammatory, antiproliferative, vasoconstrictive (Uva et al., 2012) and immunomodulatory properties, in particular suppressing the IL-23/IL-17 axis, with IL-23 produced by dendritic cells/macrophages and IL-17 produced by Th17 cells/γδ T cells/innate lymphoid cells (Germán et al., 2019).

Another question that this study does not answer is, whether topical dithranol therapy has any effects on systemic psoriatic inflammation. However nowadays, treatment with dithranol is mainly administered in refractory, circumscribed psoriatic lesions in patients who do not have significant systemic inflammation how it may be otherwise the case in patients with moderate to severe forms of psoriasis.

Together our work opens up several avenues for novel topical (and potentially also systemic) treatment strategies in psoriasis. Not only targeting the IL-36 pathway, but also keratinocyte differentiation regulators (e.g. involucrin), keratinocyte-produced AMPs (ß-defensins like DEFB4A, DEFB4B, DEFB103A, S100 proteins like S100A7, S100A12), and neutrophils and their chemotactic factors (CXCL5 and CXCL8) or members of the serpin family (SERPINB7 and SERPINB13), are promising approaches. Such approaches may not only be helpful for chronic plaque-psoriasis, but also for pustular psoriasis, in which a vicious loop between AMPs such as cathelicidin (LL-37) and IL-36 signaling may drive psoriatic disease (Benezeder and Wolf, 2019; Furue et al., 2018; Li et al., 2014; Madonna et al., 2019; Ngo et al., 2018).

Materials and methods

Key resources table
Reagent type
(species) or
resource
DesignationSource or referenceIdentifiersAdditional information
Antibodyanti-CD1a; mouse monoclonalImmunotech, Beckman CoulterClone: O10
RRID:AB_10547704
(undiluted)
Antibodyanti-CD3; mouse monoclonalNovocastra, Leica BiosystemsClone: PS1
RRID:AB_2847892
(1:100)
Antibodyanti-CD4; mouse monoclonalNovocastra, Leica BiosystemsClone: 1F6
RRID:AB_563559
(1:30)
Antibodyanti-CD8; mouse monoclonalDako, AgilentClone: C8/144b
RRID:AB_2075537
(1:25)
Antibodyanti-CK16; rabbit monoclonalAbcamClone: EPR13504
RRID:AB_2847885
(1:1000)
Antibodyanti-FoxP3; mouse monoclonalBio-RadClone: 236A/E7
RRID:AB_2262813
(1:100)
Antibodyanti-Ki-67; mouse monoclonalDako, AgilentClone: MIB-1
RRID:AB_2631211
(1:50)
Antibodyanti-MPO; mouse monoclonalDako, AgilentClone: MPO-7
RRID:AB_578599
(1:100)
Strain, strain background
Mus musculus
BALB/c, wild-typeCharles River LaboratoriesRRID:IMSR_CRL:028
Charles River Strain code#: 028
Strain, strain background
Mus musculus
JunBf/f c-Junf/f K5-Cre-ERTPMID:16163348Obtained from the laboratory of Maria Sibilia (Medical University of Vienna)
Commercial assay or kitmiRNeasy Mini KitQiagenCat #: 217004
Commercial assay or kitiScript Reverse Transcription SupermixBio-RadCat #: 1708841
Commercial assay or kitGoTag qPCR Master MixPromegaCat #: A6001
Commercial assay or kitHuman GeneChip2.0 ST arraysAffymetrix, ThermoFisher ScientificCat #:902113
Commercial assay or kitmouse Clariom S AssayAffymetrix, ThermoFisher ScientificCat #:902919
Commercial assay or kitGeneChip WT PLUS Reagent KitThermoFisher ScientificCat #: 902280
Commercial assay or kitGeneChip WT Terminal Labeling KitThermoFisher
Scientific
Cat #: 900671
Commercial assay or kitGeneChip Hybridization, Wash and Stain KitThermoFisher
Scientific
Cat #: 900720
Commercial assay or kitnCounter GX Custom codesetNanoString TechnologiesCustom codeset (80 target genes, four reference genes
Chemical compound, drugAldara (Imiquimod)5% creamMEDA PharmaceuticalsCat #: 111981
Chemical compound, drugTamoxifenSigma-AldrichCat #: T5648
Chemical compound, drugDithranol (1,8-Dihydroxy-9(10H)-anthracenone)Gatt-Koller GmbH PharmaceuticalsCat #: 8069994Dissolved in vaseline and provided by the pharmacy of the Medical University of Graz, Austria
Software, algorithmGraphPad Prism version 8GraphPadRRID:SCR_002798
https://www.graphpad.com/scientific-software/prism/
Software, algorithmInteracti VennPMID:25994840http://www.interactivenn.net/
Software, algorithmCytoscapePMID:19237447RRID:SCR_003032
https://cytoscape.org/
Software, algorithmTranscriptome Analysis Console (TAC) 4.0.2ThermoFisher
Scientific
https://www.thermofisher.com/at/en/home/life-science/microarray-analysis/microarray-analysis-instruments-software-services/microarray-analysis-software/affymetrix-transcriptome-analysis-console-software.html
Software, algorithmPartek Genomics Suite version 6.6Partek IncRRID:SCR_011860
https://www.partek.com/partek-genomics-suite/
Software, algorithmR Version 3.5.1The R Project for Statistical
Computing
RRID:SCR_001905
https://www.r-project.org/
Software, algorithmnSolver 2.5 SoftwareNanoString Technologieshttps://www.nanostring.com/products/analysis-software/nsolver

Patients

Trial protocol and patient characteristics

Request a detailed protocol

For the clinical dithranol study, inclusion criteria were diagnosis of chronic plaque psoriasis, and age above 18 years. Exclusion criteria were intolerance of dithranol, autoimmune diseases, general poor health status, pregnancy and breast-feeding, topical treatment (steroids, vitamin D3-analogs and/or Vitamin A acid-derivates) within 2 weeks, and phototherapy within 4 weeks prior to study enrollment. None of the patients had received systemic treatment in the past prior to study enrollment. In total, 15 psoriasis patients (11 men, 4 women; median age 40.5 years, range 19.8–76.9 years) were enrolled. Mean duration of psoriasis had been 17.9 years (SD 11.5 years). Mean duration of dithranol treatment was 15.4 days (SD 3.6 days) and treatment was prematurely discontinued in two patients. Samples of normal skin from patients undergoing surgery for removal of benign skin lesions were available from 12 subjects (median age was 36.3 years, range 22.6–46.9 years) for control purposes. The samples were from lesion-adjacent, excised skin of patients who did not suffer from psoriasis, other inflammatory diseases or autoimmune diseases.

Patient treatment

Request a detailed protocol

Dithranol ointment was prepared in the hospital pharmacy with 2% salicylic acid and white vaseline as base. It was administered to the patients daily in increasing concentrations; dosage was adjusted individually to the level of skin irritation. Concentration was usually increased every other day (starting from 0.1%, next 0.16%, 0.2%, 0.4%, 1% and finally 2%) and mean treatment duration was 15.4 days.

Marker lesions and scoring

Request a detailed protocol

At each of the four visits (see Figure 1), i) Psoriasis Area and Severity index (PASI), ii) local psoriasis severity index (PSI) and erythema score of marker lesions and iii) perilesional erythema score were assessed. PSI was composed by rating of erythema, induration and scaling, each on a scale from 0 to 4, resulting in a maximum score of 12. Erythema score was determined by rating intensity of lesional erythema on a scale of 0–4 (0 = none, 1 = mild, 2 = moderate, 3 = severe, 4 = very severe erythema). For perilesional erythema score, intensity of perilesional erythema was rated on a scale of 0–3 (0 = none, 1 = mild, 2 = moderate, 3 = severe perilesional erythema). Per patient, four marker lesions of similar morphology and size and, if possible, from the same body regions or four marker areas (each 5 cm in diameter) of one or more larger psoriatic lesions were defined at study entry and then scored, treated with dithranol and later biopsy-sampled at certain timepoints.

Patient tissue sampling

Request a detailed protocol

Biopsy samples were taken from the psoriasis patients before (day 0), during early treatment at first strong perilesional inflammation between day 4 and 9 (with most biopsies taken at day 6) at end of treatment (approximately after 2–3 weeks) and at a follow-up visit (4–6 weeks after end of therapy). Per patient, a total of five biopsy samples were taken on four study days by means of a punch cylinder (up to 5 mm) under local anesthesia. At the first visit before starting therapy, one biopsy sample was taken additionally from adjacent non-lesional skin, with a distance of at least 5 cm from the edge of psoriatic skin. One part of each biopsy was fixed in 4% neutral-buffered paraformaldehyde and used for histology and immunohistochemistry. The other part was stored in RNAlater solution (Invitrogen, California, USA) at −80°C until RNA extraction for further analysis.

Histology and immunohistochemistry

Analysis of HE stained sections

Request a detailed protocol

Human and murine samples fixed with paraformaldehyde were processed routinely, cut in 4 µm sections and stained with hematoxylin and eosin (HE). Five randomly selected fields per slide were investigated for histological analysis. Thickness of epidermis was measured from basal layer to stratum corneum using an Olympus BX41 microscope (Olympus Life Science Solutions, Hamburg, Germany), cellSens software (Olympus Life Science Solutions) and 20x magnification. Semi-quantitative scoring (0 = none, 0.5 = none/low, 1 = low, 1.5 = low/moderate, 2 = moderate, 2.5 = moderate/high, 3 = high density of infiltrate) was performed at five randomly selected locations per slide and at 20x magnification.

In the mouse-tail model, degree of orthokeratosis was analyzed as described by Bosman et al., 1992 . In brief, five randomly selected scales per sample were examined and the length of the granular layer (A) as well as the total length of the scale (B) were measured using cellSens software (Olympus-lifescience, Hamburg, Germany) and 20x magnification. The proportion of (A/B) x 100 depicts the percentage of orthokeratosis per scale.

Immunohistochemistry stainings and analysis

Request a detailed protocol

Antigen retrieval was performed using either EDTA-buffer (CD1a, CD3, CD4, CK16, CD8), citrate-buffer (FoxP3, Ki-67) or trypsin (MPO). Primary antibodies used were: anti-human CD1a (mouse monoclonal, clone O10, undiluted; Immunotech, Beckman Coulter, Praque, Czech Republic), anti-human CD3 (mouse monoclonal, clone PS1, dilution 1:100; Novocastra, Leica Biosystems, Mannheim, Germany), anti-human CD4 (mouse monoclonal, clone 1F6, dilution 1:30; Leica Biosystems), anti-human CD8 (mouse monoclonal, clone C8/144b, dilution 1:25, Dako Omnis, Agilent, Santa Clara, CA, USA), anti-human CK16 (rabbit monoclonal, clone EPR13504, dilution 1:1000, Abcam, Cambridge, UK), anti-human FoxP3 (clone 236A/E7, AbD Serotec, Bio-Rad, Hercules, CA, USA), anti-human Ki-67 (mouse monoclonal, clone MIB-1, dilution 1:50, Dako Omnis, Agilent) and anti-human MPO (mouse monoclonal, clone MPO-7, dilution 1:100, Dako Omnis, Agilent). Stainings were performed using the Dako REALTM Detection System, Peroxidase/AEC, rabbit/mouse (Dako, Agilent) on the Dako Autostainer Link 48 (Dako, Agilent) according to the manufacturer’s instructions. For quantification of CD1a, CD3, CD4, FoxP3, CD8, and MPO staining, all positively stained cells with visible nucleus in five randomly selected fields (separately for epidermis and dermis) per slide were counted and results were averaged to obtain mean cell counts. To quantify Ki-67 and CK16 staining, area of positive staining was divided by epidermal area as follows: one representative image per slide was taken on an Olympus BX41 microscope (Olympus Life Science Solutions, Hamburg, Germany) at 10x magnification using cellSens software (Olympus Life Science Solutions). TIFF images were imported into ImageJ program and pixels were converted to µm. Using the polygon sections tool, the outline of epidermis was framed, and the total area was measured. In addition, total area of particles (positively stained cells) within the epidermal area was assessed.

Gene expression analyses

RNA extraction

Request a detailed protocol

Total RNA was extracted from frozen biopsies of psoriasis patients and control subjects. To facilitate homogenization, tissues were cut in 20 µm sections using a cryomicrotome and collected in precooled MagNA Lyser Green Beads tubes (Roche, Basel, Switzerland) and disruption of tissue was performed on the MagNA Lyser Instrument (Roche, Basel, Switzerland). After efficient homogenization, total RNA was extracted using the miRNeasy Mini Kit (Qiagen, Hilden, Germany), according to the manufacturer’s instructions. To ensure complete DNA removal, on-column DNase digestion was performed and RNA was eluted in 15–20 µl RNase-free water. Mouse tissues were handled in the same way except that sufficient homogenization of samples was obtained without using a cryomicrotome.

Microarray and pathway analysis

Request a detailed protocol

Isolated RNA was quality checked on a Bioanalyzer BA2100 (Agilent; Foster City, CA) using the RNA 6000 Nano LabChip according to manufacturer’s instructions and samples with RNA integrity numbers (RIN) between 5 to 8 were considered for analysis using Human GeneChip 2.0 ST arrays (Affymetrix, ThermoFisher Scientific, Waltham, MA, USA; Cat.No.: 902113) for the human samples and mouse Clariom S Assay (Affymetrix, ThermoFisher Scientific, Waltham, MA, USA; Cat No. 902919) for the mouse samples. For first and second strand cDNA synthesis 500 ng total RNA were used in the GeneChip WT PLUS Reagent Kit (ThermoFisher) according to manufacturer’s instructions. Fragmentation and terminal labelling was performed using the GeneChipTM WT Terminal Labeling Kit (ThermoFisher) and hybridization, wash and stain of arrays was performed on a GeneChip Fluidics 450 station using the GeneChip Hybridization, Wash and Stain Kit (ThermoFisher) according to manufacturer’s instructions. Arrays were scanned in a GeneChip GCS300 7G Scanner and analysed with the Affymetrix Expression Console Software 1.3.1 (ThermoFisher) for the human array and Transcriptome Analysis Console (TAC) 4.0.2 for the mouse arrays. Samples were processed at the Core Facility Molecular Biology at the Centre of Medical Research at the Medical University of Graz, Austria. Pre-processing of CEL-files for the human arrays was performed with Partek Genomics Suite version 6.6 (Partek Inc, St Louis, MO, USA) using the robust multi-chip analysis (RMA) algorithm, which includes background adjustment, quantile normalisation and median polished probe summarisation. For statistical analysis, paired-sample t-tests were used and genes with p<0.05 and a fold change of at least 1.5 were considered to be significantly deregulated. All microarray data has been deposited at the public repository Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) with accession numbers GSE145126 and GSE145127.

For analysis of histological responders compared to non-responders, as well as mouse arrays, pre-processing and RMA normalization was done with RStudio Version 1.2.1335 (R Version 3.5.1) with MicroArrayPipeline v1.0 Shiny app based on limma Bioconductor package for differential expression analysis and genes with p<0.05 and a fold change of at least 1.5 were considered to be significantly deregulated. Mouse and human DEGs were tested for potential overlap. Probeset IDs of early (day 6) as well as late (week 2–3) DEGs of human microarray were matched with DEGs of mouse microarray using NetAffxTM Expression Array Comparison Tool.

Nanostring nCounter analysis and microarray verification

Request a detailed protocol

For Nanostring analysis, a nCounter GX Custom codeset (80 target genes, four reference genes, see Supplementary file 2) was designed to verify microarray results of selected DEGs. Total RNA (150 ng) with RIN values between 4.7 and 9 was used and samples were processed according to supplier’s instructions (NanoString Technologies, Seattle, WA USA) and scanned on a nCounter Digital Analyzer (NanoString Technologies). RCC files were used for data analysis. Raw data pre-processing and normalization was performed using nSolver 2.5 Software (NanoString Technologies) according to standard procedures (background subtraction, positive and negative controls normalization). Gene counts were then normalized to the geometric mean of the reference genes. Normalized data was uploaded to Partek Genomic Suite Software v6.6 (Partek Inc, St Louis, MO, USA) and paired t-test was used for statistical analysis. Nanostring and microarray fold change values of selected target genes were log-transformed and the two platforms were compared by Pearson correlation of each gene across samples.

Gene ontology enrichment analysis

Request a detailed protocol

For all comparisons genes with a p-value<0.05 and FC ± 1.5 were assigned to GO enrichment analysis using Cytoscape software ver.3.5.1 (Cytoscape Consortium, NY, USA www.cytoscape.org; Bindea et al., 2009). Gene identifiers were loaded into Cytoscape software and ClueGO analysis was used to identify significantly overrepresented GO terms and associated genes. P-values (significance level <0.05) were adjusted using Bonferroni step-down corrections.

RT-qPCR

Request a detailed protocol

Per sample 2 µg of RNA was reverse transcribed into cDNA using iScript Reverse Transcription Supermix (Bio-Rad, Hercules, CA, USA). Relative gene expression was determined using GoTag qPCR Master Mix (Promega, Mannheim, Germany) on a CFX96 Touch Real-Time PCR Detection System (Bio-Rad). The following cycling conditions were used: Hot-start activation (95°C, 2 min), denaturation for 40 cycles (95°C, 15 s) and annealing/extension (60°C, 60 s). Melting curve analysis was done to confirm amplification specificity. For each sample, qPCRs were run in triplicates. Cycle thresholds (Ct) were determined and relative mRNA expression to Ubc or Ywhaz (reference genes) were calculated using the ΔCt method. Primer sequences are listed in Supplementary file 4.

Animals

Mouse strains and housing

Request a detailed protocol

6–9 week-old mice were kept with food and water ad libitum in the conventional animal facility at the Centre for Medical Research, Medical University of Graz or at the Medical University of Vienna, Austria. During experiments, all mice were monitored closely to ensure sufficient health status. BALB/c mice were purchased from Charles River (Sulzfeld, Germany). c-Jun/JunB knockout mice (Zenz et al., 2005) were bred and maintained in the facilities of the Medical University of Vienna. All animal experiments were in accordance with institutional policies and federal guidelines.

Therapeutic agents used in mice

Request a detailed protocol

For all animal experiments, dithranol in different concentrations (dissolved in vaseline) and vehicle (vaseline cream only) was provided by the pharmacy of the Medical University of Graz, Austria. Aldara (IMQ) 5% cream (MEDA Pharmaceuticals, Vienna, Austria) and tamoxifen (Sigma-Aldrich, Missouri, USA) were purchased.

Imiquimod model

Request a detailed protocol

24 hr before starting an experiment, dorsal skin of BALB/c mice was shaved carefully. To induce psoriasis-like dermatitis, imiquimod (IMQ) cream was applied daily for five consecutive days on dorsal skin (approximately 40 mg) and right ear (approximately 20 mg). A daily topical dose of 62.5 mg of IMQ cream was not exceeded; translating into 3.125 mg of the active compound. Mice received IMQ cream in the morning and dithranol treatment in the afternoon, with a time gap of 8 hr. Dithranol was applied topically on dorsal skin (40 mg) and right ear (20 mg) and concentrations were increased every other day (0.01% on day 1–2, 0.03% on day 3–4% and 0.1% on day 5–6). Control mice were treated similarly with Vaseline cream. Double skin fold of dorsal skin and ear thickness was measured daily in triplicates before any application using a micrometer (Mitutoyo, Kanagawa, Japan). 24 hr after the last topical treatment, mice were sacrificed and approximately 1 cm2 of dorsal skin, both ears (treated and untreated as control) were collected.

c-Jun/JunB knockout mouse model

Request a detailed protocol

Mice carrying floxed alleles for the JunB and c-Jun locus and expressing the K5-CreERT transgene (mixed background) received consecutive i.p. injections of tamoxifen (1 mg/day) for a period of 5 days, leading to deletion of both genes in the epidermis by inducible Cre-recombinase activity (Schonthaler et al., 2009; Zenz et al., 2005). One week after the last injection, psoriasis-like lesions on the ears were treated daily with dithranol in series of increasing concentrations (0.01% on day 1–2, 0.03% on day 3–4% and 0.1% on day 5–6 or 0.1% on day 1–2, 0.3% on day 3–4% and 1% on day 5–6) as depicted in Figure 3A. Control mice were treated similarly with vehicle cream. Ear thickness was measured daily by micrometer before any topical application and 24 hr after the last topical treatment mice were sacrificed and ears were collected.

Mouse tail test

Request a detailed protocol

For the mouse-tail model, 40 mg dithranol 1% was applied daily for 14 days to the surface of the proximal part of tails (approx. 2 cm in length starting 1 cm from the body). 24 hr after the last application, mice were sacrificed, and treated parts of tail skin were removed from the underlying cartilage. The mouse-tail test for psoriasis is a traditional model to quantify the effect of topical anti-psoriatics on keratinocyte differentiation by measuring degree of orthokeratosis versus parakeratosis (Bosman et al., 1992; Sebök et al., 2000; Wu et al., 2015).

Statistical analyses

Request a detailed protocol

Statistical analyses for human and murine microarrays were performed as described in the specific sections. All other statistical analyses were determined using GraphPad Prism version 8 (GraphPad software, California, USA). Normality was determined by Shapiro-Wilk test and differences between two groups were assessed by Mann Whitney test, Wilcoxon test or T-test (paired or unpaired) as appropriate. For multiple comparisons, One-way ANOVA with Dunnett’s test or Tukey’s test was used for parametric data and Kruskal Wallis test with Dunn’s test was used for nonparametrical data as indicated in the specific sections. Significance was set at a p-value of ≤0.05. Each animal experiment was performed at least twice. For correlation analysis of clinical scores, as well as comparison of microarray and nanostring ratios, Pearson or Spearman correlation was used as indicated.

Study approval

Request a detailed protocol

A clinical study (Clinical Trials.gov no. NCT02752672) in psoriatic patients treated with dithranol was completed in cooperation with the Department of Dermatology, Klagenfurt State Hospital. Clinical trial procedures were approved by the ethics committee of the federal state of Carinthia, Austria (protocol number A23/15) and all participants gave written informed consent in accordance with the principles of the Declaration of Helsinki. All mouse experiments were approved by the Austrian Government, Federal Ministry for Science and Research (protocol numbers BMWF-66-010/0032-11/3b/2018, 66.009/0200-WF/II/3b/2014) and animal experiments performed in Vienna were additionally approved by the Animal Experimental Ethics Committee of the Medical University of Vienna.

Data availability

All microarray data has been deposited at the public repository Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) with accession numbers GSE145126 and GSE145127.

The following data sets were generated

References

  1. Book
    1. Gerritsen MJP
    (1999)
    Dithranol
    In: van de Kerkhof PCM, editors. Textbook of Psoriasis. Blackwell Science. pp. 160–178.
    1. Krueger G
    2. Koo J
    3. Lebwohl M
    4. Menter A
    5. Stern RS
    6. Rolstad T
    (2001)
    The impact of psoriasis on quality of life: results of a 1998 national psoriasis foundation patient-membership survey
    Archives of Dermatology 137:280–284.
  2. Book
    1. Kucharekova M
    2. Van De Kerkhof PCM
    3. Schalkwijk J
    4. Van Der Valk PGM
    (2006) Dithranol
    In: Chew AL, Maibach HI, editors. Irritant Dermatitis. Springer. pp. 317–322.
    https://doi.org/10.1007/3-540-31294-3_34
    1. Nast A
    2. Boehncke WH
    3. Mrowietz U
    4. Ockenfels HM
    5. Philipp S
    6. Reich K
    7. Rzany B
    (2012) Guidelines on the treatment of psoriasis vulgaris
    Journal Der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG 10:1–95.
    https://doi.org/10.1111/j.1610-0387.2012.07919.x
    1. Schröder JM
    2. Harder J
    (1999) Human beta-defensin-2
    The International Journal of Biochemistry & Cell Biology 31:645–651.
    https://doi.org/10.1016/S1357-2725(99)00013-8
    1. Swinkels OQ
    2. Prins M
    3. Veeniiuis RT
    4. De Boo T
    5. Gerritsen MJ
    6. Van Der Wilt GJ
    7. Van De Kerkhof PC
    8. Van Der Valk PG
    (2004)
    Effectiveness and side effects of UVB-phototherapy, dithranol inpatient therapy and a care instruction programme of short contact dithranol in moderate to severe psoriasis
    European Journal of Dermatology : EJD 14:159–165.
    1. van de Kerkhof PCM
    (1991)
    Dithranol treatment for psoriasis: after 75 years still going strong!
    European Journal of Dermatology 1:79–88.
    1. Wu J
    2. Li H
    3. Li M
    (2015)
    Effects of baicalin cream in two mouse models: 2,4-dinitrofluorobenzene-induced contact hypersensitivity and mouse tail test for psoriasis
    International Journal of Clinical and Experimental Medicine 8:2128–2137.

Article and author information

Author details

  1. Theresa Benezeder

    Department of Dermatology, Medical University of Graz, Graz, Austria
    Contribution
    Conceptualization, Data curation, Formal analysis, Validation, Investigation, Visualization, Methodology, Writing - original draft, Project administration
    Contributed equally with
    Clemens Painsi
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-6218-2792
  2. Clemens Painsi

    State Hospital Klagenfurt, Klagenfurt am Wörthersee, Austria
    Contribution
    Conceptualization, Data curation, Investigation, Project administration
    Contributed equally with
    Theresa Benezeder
    Competing interests
    No competing interests declared
  3. VijayKumar Patra

    Department of Dermatology, Medical University of Graz, Graz, Austria
    Contribution
    Visualization, Methodology
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-7161-7767
  4. Saptaswa Dey

    Department of Dermatology, Medical University of Graz, Graz, Austria
    Contribution
    Methodology
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7532-7858
  5. Martin Holcmann

    Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
    Contribution
    Resources, Methodology
    Competing interests
    No competing interests declared
  6. Bernhard Lange-Asschenfeldt

    State Hospital Klagenfurt, Klagenfurt am Wörthersee, Austria
    Contribution
    Resources, Project administration
    Competing interests
    No competing interests declared
  7. Maria Sibilia

    Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
    Contribution
    Resources, Writing - review and editing
    Competing interests
    No competing interests declared
  8. Peter Wolf

    Department of Dermatology, Medical University of Graz, Graz, Austria
    Contribution
    Conceptualization, Supervision, Funding acquisition, Writing - review and editing
    For correspondence
    peter.wolf@medunigraz.at
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0001-7777-9444

Funding

Austrian Science Fund (W1241)

  • Peter Wolf

Medical University of Graz (PhD Program Molecular Fundamentals of Inflammation (DK-MOLIN))

  • Peter Wolf

Austrian Science Fund (W1212)

  • Maria Sibilia

Vienna Science and Technology Fund (LS16-025)

  • Maria Sibilia

H2020 European Research Council (ERC-2015-AdG TNT-Tumors 694883)

  • Maria Sibilia

Horizon 2020 Framework Programme (766214)

  • Maria Sibilia

Marshallplan-Jubiläumsstiftung

  • Theresa Benezeder

Austrian Society for Dermatology and Venereology

  • Theresa Benezeder

Elisabeth Hirschheiter research grant

  • Clemens Painsi

National Institute of Arthritis and Musculoskeletal and Skin Diseases (P30AR069625)

  • Peter Wolf

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Acknowledgements

The authors would like to thank all patients who made this work possible and Gerlinde Mayer andUlrike Schmidbauer, for technical support and Karin Wagner and Ingeborg Klymiuk for support in Microarray and Nanostring analyses, all Medical University of Graz; Martina Hammer, Medical University of Vienna, for support in animal experimentation; Ahmed Gehad and Rachael Clark for support in animal experimentation and scientific advice, both at Brigham and Women’s Hospital, Harvard Medical School; and Honnovara Ananthaswamy, Houston, TX, for critical reading of the manuscript. The authors would also like to thank Kathrin Eller and Herbert Strobl, who provided valuable guidance as members of the thesis committee of TB. This work was supported by the Austrian Science Fund FWF (W1241) and the Medical University of Graz through the Ph.D. Program Molecular Fundamentals of Inflammation (DK‐MOLIN) to PW; by grants from the Austrian Science Fund (FWF, W1212), the WWTF-project LS16-025, the European Research Council (ERC) Advanced grant (ERC-2015-AdG TNT-Tumors 694883) and the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement No. 766214 (Meta-Can) to MS. TB was supported by the Austrian Marshall Plan Foundation and the Austrian Society for Dermatology and Venereology (Klaus Wolff fellowship). CP was supported by the Elisabeth Hirschheiter research grant. PW was supported by a Visiting Scholar Award of the Human Skin Disease Resource Center (HSDRC) Grant Program (NIH/NIAMS P30AR069625), Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School.

Ethics

Clinical trial registration Clinical Trials.gov no. NCT02752672.

Human subjects: A clinical study in psoriatic patients treated with dithranol was completed in cooperation with the Department of Dermatology, Klagenfurt State Hospital. Clinical trial procedures were approved by the ethics committee of the federal state of Carinthia, Austria (protocol number A23/15) and all participants gave written informed consent in accordance with the principles of the Declaration of Helsinki.

Animal experimentation: All mouse experiments were approved by the Austrian Government, Federal Ministry for Science and Research (protocol numbers BMWF-66-010/0032-11/3b/2018, 66.009/0200-WF/II/3b/2014) and animal experiments performed in Vienna were additionally approved by the Animal Experimental Ethics Committee of the Medical University of Vienna.

Version history

  1. Received: March 17, 2020
  2. Accepted: May 18, 2020
  3. Version of Record published: June 2, 2020 (version 1)

Copyright

© 2020, Benezeder et al.

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 2,067
    views
  • 265
    downloads
  • 27
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Theresa Benezeder
  2. Clemens Painsi
  3. VijayKumar Patra
  4. Saptaswa Dey
  5. Martin Holcmann
  6. Bernhard Lange-Asschenfeldt
  7. Maria Sibilia
  8. Peter Wolf
(2020)
Dithranol targets keratinocytes, their crosstalk with neutrophils and inhibits the IL-36 inflammatory loop in psoriasis
eLife 9:e56991.
https://doi.org/10.7554/eLife.56991

Share this article

https://doi.org/10.7554/eLife.56991

Further reading

    1. Evolutionary Biology
    2. Immunology and Inflammation
    Mark S Lee, Peter J Tuohy ... Michael S Kuhns
    Research Advance

    CD4+ T cell activation is driven by five-module receptor complexes. The T cell receptor (TCR) is the receptor module that binds composite surfaces of peptide antigens embedded within MHCII molecules (pMHCII). It associates with three signaling modules (CD3γε, CD3δε, and CD3ζζ) to form TCR-CD3 complexes. CD4 is the coreceptor module. It reciprocally associates with TCR-CD3-pMHCII assemblies on the outside of a CD4+ T cells and with the Src kinase, LCK, on the inside. Previously, we reported that the CD4 transmembrane GGXXG and cytoplasmic juxtamembrane (C/F)CV+C motifs found in eutherian (placental mammal) CD4 have constituent residues that evolved under purifying selection (Lee et al., 2022). Expressing mutants of these motifs together in T cell hybridomas increased CD4-LCK association but reduced CD3ζ, ZAP70, and PLCγ1 phosphorylation levels, as well as IL-2 production, in response to agonist pMHCII. Because these mutants preferentially localized CD4-LCK pairs to non-raft membrane fractions, one explanation for our results was that they impaired proximal signaling by sequestering LCK away from TCR-CD3. An alternative hypothesis is that the mutations directly impacted signaling because the motifs normally play an LCK-independent role in signaling. The goal of this study was to discriminate between these possibilities. Using T cell hybridomas, our results indicate that: intracellular CD4-LCK interactions are not necessary for pMHCII-specific signal initiation; the GGXXG and (C/F)CV+C motifs are key determinants of CD4-mediated pMHCII-specific signal amplification; the GGXXG and (C/F)CV+C motifs exert their functions independently of direct CD4-LCK association. These data provide a mechanistic explanation for why residues within these motifs are under purifying selection in jawed vertebrates. The results are also important to consider for biomimetic engineering of synthetic receptors.

    1. Genetics and Genomics
    2. Immunology and Inflammation
    Jean-David Larouche, Céline M Laumont ... Claude Perreault
    Research Article

    Transposable elements (TEs) are repetitive sequences representing ~45% of the human and mouse genomes and are highly expressed by medullary thymic epithelial cells (mTECs). In this study, we investigated the role of TEs on T-cell development in the thymus. We performed multiomic analyses of TEs in human and mouse thymic cells to elucidate their role in T-cell development. We report that TE expression in the human thymus is high and shows extensive age- and cell lineage-related variations. TE expression correlates with multiple transcription factors in all cell types of the human thymus. Two cell types express particularly broad TE repertoires: mTECs and plasmacytoid dendritic cells (pDCs). In mTECs, transcriptomic data suggest that TEs interact with transcription factors essential for mTEC development and function (e.g., PAX1 and REL), and immunopeptidomic data showed that TEs generate MHC-I-associated peptides implicated in thymocyte education. Notably, AIRE, FEZF2, and CHD4 regulate small yet non-redundant sets of TEs in murine mTECs. Human thymic pDCs homogenously express large numbers of TEs that likely form dsRNA, which can activate innate immune receptors, potentially explaining why thymic pDCs constitutively secrete IFN ɑ/β. This study highlights the diversity of interactions between TEs and the adaptive immune system. TEs are genetic parasites, and the two thymic cell types most affected by TEs (mTEcs and pDCs) are essential to establishing central T-cell tolerance. Therefore, we propose that orchestrating TE expression in thymic cells is critical to prevent autoimmunity in vertebrates.