Advertisement
Original Article Inflammation| Volume 143, ISSUE 3, P431-443.e19, March 2023

The p300/CBP Inhibitor A485 Normalizes Psoriatic Fibroblast Gene Expression In Vitro and Reduces Psoriasis-Like Skin Inflammation In Vivo

Open AccessPublished:September 26, 2022DOI:https://doi.org/10.1016/j.jid.2022.09.004
      Psoriasis is a chronic inflammatory skin disease that often recurs at the same locations, indicating potential epigenetic changes in lesional skin cells. In this study, we discovered that fibroblasts isolated from psoriatic skin lesions retain an abnormal phenotype even after several passages in culture. Transcriptomic profiling revealed the upregulation of several genes, including the extra domain A splice variant of fibronectin and ITGA4 in psoriatic fibroblasts. A phenotypic library screening of small-molecule epigenetic modifier drugs revealed that selective CBP/p300 inhibitors were able to rescue the psoriatic fibroblast phenotype, reducing the expression levels of extra domain A splice variant of fibronectin and ITGA4. In the imiquimod-induced mouse model of psoriasis-like skin inflammation, systemic treatment with A485, a potent CBP/p300 blocker, significantly reduced skin inflammation, immune cell recruitment, and inflammatory cytokine production. Our findings indicate that epigenetic reprogramming might represent a new approach for the treatment and/or prevention of relapses of psoriasis.

      Graphical abstract

      Abbreviations:

      BRD (bromodomain), DC (dendritic cell), EC (endothelial cell), ECM (extracellular matrix), EDA FN (extradomain A fibronectin), FN (fibronectin), H3K27ac (acetylation of lysine 27 on histone 3), HAT (histone acetyltransferase), IMQ (imiquimod)

      Introduction

      Psoriasis is an immune-mediated chronic inflammatory skin disease characterized by hyperproliferation of epidermal keratinocytes; immune cell infiltration; remodeling of the dermal vasculature; and activation of fibroblasts, which secrete inflammatory cytokines and altered extracellular matrix (ECM) proteins. Up to 30% of patients with psoriasis also suffer from comorbidities such as psoriatic arthritis, metabolic syndrome, and cardiovascular disorders (
      • Oliveira M.F.S.P.
      • Rocha B.O.
      • Duarte G.V.
      Psoriasis: classical and emerging comorbidities.
      ). Currently, psoriasis cannot be cured but can only be treated with biologicals such as anti‒IL-17 and anti‒IL-23 antibodies (
      • Hawkes J.E.
      • Yan B.Y.
      • Chan T.C.
      • Krueger J.G.
      Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis.
      ). Of note, psoriatic skin lesions frequently recur after successful treatment, often at the same site that previously recovered (
      • Clark R.A.
      Gone but not forgotten: lesional memory in psoriatic skin.
      ). Although genetic alterations in psoriasis have been well-characterized by GWASs (
      • Ellinghaus E.
      • Ellinghaus D.
      • Stuart P.E.
      • Nair R.P.
      • Debrus S.
      • Raelson J.V.
      • et al.
      Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2.
      ;
      • Nair R.P.
      • Duffin K.C.
      • Helms C.
      • Ding J.
      • Stuart P.E.
      • Goldgar D.
      • et al.
      Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways.
      ;
      • Tang H.
      • Jin X.
      • Li Y.
      • Jiang H.
      • Tang X.
      • Yang X.
      • et al.
      A large-scale screen for coding variants predisposing to psoriasis.
      ), there is also evidence that an epigenetic memory of skin cells might contribute to the recurrences of the disease (
      • Chandra A.
      • Senapati S.
      • Roy S.
      • Chatterjee G.
      • Chatterjee R.
      Epigenome-wide DNA methylation regulates cardinal pathological features of psoriasis.
      ;
      • Clark R.A.
      Gone but not forgotten: lesional memory in psoriatic skin.
      ;
      • Gudjonsson J.E.
      • Krueger G.
      A role for epigenetics in psoriasis: methylated cytosine-guanine sites differentiate lesional from nonlesional skin and from normal skin.
      ;
      • Pollock R.A.
      • Abji F.
      • Gladman D.D.
      Epigenetics of psoriatic disease: a systematic review and critical appraisal.
      ).
      Fibroblasts represent the major type of stromal cells with a structural role through the synthesis of ECM proteins (e.g., fibronectin [FN], vimentin, and collagens) as well as the production of cytokines, chemokines, and GFs (
      • Kalluri R.
      • Zeisberg M.
      Fibroblasts in cancer.
      ). Fibroblasts play a pivotal role in maintaining the homeostasis of adjacent cells and in coordinating inflammatory responses, rendering them indispensable for tissue development, differentiation, and repair. Under chronic inflammatory conditions, fibroblasts can become activated and manifest enhanced production of ECM proteins, including type I collagen, tenascin C, SPARC (secreted protein acidic and rich in cysteine), and de novo expression of the extra domain A FN (EDA FN) splice variant (
      • Coentro J.Q.
      • Pugliese E.
      • Hanley G.
      • Raghunath M.
      • Zeugolis D.I.
      Current and upcoming therapies to modulate skin scarring and fibrosis.
      ;
      • Kalluri R.
      • Zeisberg M.
      Fibroblasts in cancer.
      ;
      • Van Linthout S.V.
      • Miteva K.
      • Tschöpe C.
      Crosstalk between fibroblasts and inflammatory cells.
      ). The activation is mediated by TGF-β, ECM proteases, and chemokines (
      • Kalluri R.
      • Zeisberg M.
      Fibroblasts in cancer.
      ). Interestingly, not only psoriatic lesional (
      • Verma D.
      • Ekman A.K.
      • Bivik Eding C.B.
      • Enerbäck C.
      Genome-wide DNA methylation profiling identifies differential methylation in uninvolved psoriatic epidermis.
      ) but also nonlesional skin showed high expression levels of EDA FN, which was correlated with the degree of inflammation and also with epigenetic modifications, such as DNA methylation (
      • Roberson E.D.O.
      • Liu Y.
      • Ryan C.
      • Joyce C.E.
      • Duan S.
      • Cao L.
      • et al.
      A subset of methylated CpG sites differentiate psoriatic from normal skin.
      ). These findings indicate that a potential epigenetic memory of skin cells might contribute to the relapse of psoriatic skin lesions.
      Epigenetics describes heritable or reversible changes that can affect gene expression without any alteration in the DNA sequence. Epigenetic changes include alterations of the DNA accessibility by chromatin remodeling, DNA modification, or histone modification (
      • Pinborg A.
      • Loft A.
      • Romundstad L.B.
      • Wennerholm U.B.
      • Söderström-Anttila V.
      • Bergh C.
      • et al.
      Epigenetics and assisted reproductive technologies.
      ). Epigenetic changes are dynamically reversible, and several inhibitor drugs targeting DNA methyltransferases or histone deacetylases have been shown to restore normal epigenetic patterns in a number of diseases (
      • Andreoli F.
      • Jorge Moura Barbosa A.
      • Daniele Parenti M.
      • Del Rio A.
      Modulation of epigenetic targets for anticancer therapy: clinicopathological relevance, structural data and drug discovery perspectives.
      ;
      • Fardi M.
      • Solali S.
      • Farshdousti Hagh M.F.
      Epigenetic mechanisms as a new approach in cancer treatment: an updated review.
      ). Epigenetic writers, readers, and erasers that antagonize or inhibit specific domains of epigenetic protein complexes have been previously used for drug screenings in cancers (
      • Ganesan A.
      • Arimondo P.B.
      • Rots M.G.
      • Jeronimo C.
      • Berdasco M.
      The timeline of epigenetic drug discovery: from reality to dreams.
      ;
      • Wu B.
      • Pan X.
      • Chen X.
      • Chen M.
      • Shi K.
      • Xu J.
      • et al.
      Epigenetic drug library screening identified an LSD1 inhibitor to target UTX-deficient cells for differentiation therapy.
      ). However, despite several epigenetic studies demonstrating the involvement of DNA methylation, histone modification, and noncoding RNA regulation in the pathogenesis of psoriasis (
      • Chandra A.
      • Senapati S.
      • Roy S.
      • Chatterjee G.
      • Chatterjee R.
      Epigenome-wide DNA methylation regulates cardinal pathological features of psoriasis.
      ;
      • Gupta R.
      • Ahn R.
      • Lai K.
      • Mullins E.
      • Debbaneh M.
      • Dimon M.
      • et al.
      Landscape of long noncoding RNAs in psoriatic and healthy skin.
      ;
      • Ovejero-Benito M.C.
      • Reolid A.
      • Sánchez-Jiménez P.
      • Saiz-Rodríguez M.
      • Muñoz-Aceituno E.
      • Llamas-Velasco M.
      • et al.
      Histone modifications associated with biological drug response in moderate-to-severe psoriasis.
      ), no comprehensive epigenetic drug screening has been performed in psoriasis.
      We aimed to establish and apply assays for epigenetic drug screens in cultured fibroblasts derived from psoriatic plaques. We chose fibroblasts because they represent a major population of dermal cells and their contributions to psoriasis pathogenesis have been indicated in several studies (
      • Arasa J.
      • Terencio M.C.
      • Andrés R.M.
      • Marín-Castejón A.
      • Valcuende-Cavero F.
      • Payá M.
      • et al.
      Defective induction of COX-2 expression by psoriatic fibroblasts promotes pro-inflammatory activation of macrophages.
      ;
      • Krueger G.G.
      • Jorgensen C.M.
      Defined system to assess the in vitro induction of a psoriasis phenotype on normal keratinocytes by fibroblasts from psoriatic subjects.
      ;
      • Priestley G.C.
      • Adams L.W.
      Hyperactivity of fibroblasts cultured from psoriatic skin: I. Faster proliferation and effect of serum withdrawal.
      ). In particular, previous studies have shown that cultured fibroblasts from psoriatic skin lesions maintain phenotypic differences over several passages in vitro (
      • Arasa J.
      • Terencio M.C.
      • Andrés R.M.
      • Marín-Castejón A.
      • Valcuende-Cavero F.
      • Payá M.
      • et al.
      Defective induction of COX-2 expression by psoriatic fibroblasts promotes pro-inflammatory activation of macrophages.
      ;
      • Krueger G.G.
      • Jorgensen C.M.
      Defined system to assess the in vitro induction of a psoriasis phenotype on normal keratinocytes by fibroblasts from psoriatic subjects.
      ;
      • Priestley G.C.
      • Adams L.W.
      Hyperactivity of fibroblasts cultured from psoriatic skin: I. Faster proliferation and effect of serum withdrawal.
      ). In line with this, we also found that psoriatic fibroblasts maintained increased expression of several genes, including EDA FN and ITGA4, even after several passages in vitro. An epigenetic drug screen using 48 small molecules revealed that treatment with selective p300/CBP inhibitors reduced the expression of these genes. In a mouse model of psoriasis-like skin inflammation, systemic treatment with the specific p300/CBP histone acetyltransferase (HAT) domain inhibitor, A485, strongly reduced imiquimod (IMQ)-induced skin inflammation. These findings might have potential implications for future strategies to treat and/or prevent the relapse of psoriatic skin lesions.

      Results

      Distinct transcriptomic profiles of fibroblasts derived from psoriatic skin

      Fibroblasts derived from psoriatic lesions were reported to retain an activated phenotype (
      • Arasa J.
      • Terencio M.C.
      • Andrés R.M.
      • Marín-Castejón A.
      • Valcuende-Cavero F.
      • Payá M.
      • et al.
      Defective induction of COX-2 expression by psoriatic fibroblasts promotes pro-inflammatory activation of macrophages.
      ;
      • Krueger G.G.
      • Jorgensen C.M.
      Defined system to assess the in vitro induction of a psoriasis phenotype on normal keratinocytes by fibroblasts from psoriatic subjects.
      ;
      • Priestley G.C.
      • Adams L.W.
      Hyperactivity of fibroblasts cultured from psoriatic skin: I. Faster proliferation and effect of serum withdrawal.
      ). We isolated psoriatic and healthy dermal fibroblasts from chronic psoriatic plaques and healthy skin from patients admitted to plastic surgery, respectively. We developed a standardized workflow for isolating fibroblasts (Figure 1a). The skin tissue was mechanically and enzymatically digested after removing the epidermal layer. Fibroblasts from the dermis were isolated after negative selection with CD31 Dynabeads and showed the expression of PDGFRa, considered a fibroblast marker (Figure 1b and c).
      Figure thumbnail gr1
      Figure 1RNA sequencing of fibroblasts from psoriatic and healthy skin. (a) Overview of the experimental workflow of the study. (b, c) Total RNA was extracted from healthy (n = 6) and psoriatic (n = 3) fibroblasts and subjected to RNA-Seq. Gene expression (TPM) of (b) CD90 and (c) PDGFRa in healthy (n = 6) and psoriatic fibroblasts (n = 3). (d) Representative flow cytometry histograms of PDGFRa protein levels in healthy (blue) and psoriatic (red) fibroblasts. DFs (green) and FMO (gray) control were used as positive control and negative control, respectively, and quantification (MFI) of PDGFRa was expressed as FC over healthy fibroblasts. (e) PCA from RNA-Seq showing a distinct transcriptomic landscape of healthy and psoriatic fibroblasts. (f) Heatmap showing differential gene expression between healthy and psoriatic fibroblasts (log2 FC ≥ 0.6 and P < 0.05). (g) GO analysis of biological processes using PANTHER for the top 10 upregulated terms in psoriatic fibroblasts expressed as ‒Log10(P-value). ∗P < 0.05 and ∗∗∗P < 0.001. (h) ITGA4 and (i) EDA FN expression levels evaluated by qPCR (n = 2 lines per group in 2‒3 biological replicates). Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. ∗∗∗∗P < 0.0001. (j) GSEA showing that genes downregulated in psoriatic fibroblasts were enriched for chromatin silencing, gene expression epigenetics, and histone exchange. Running ES score and positions of gene set members on the rank-ordered list (NES) based on the significance value from GSEA. The GSEA results are listed in . DF, dermal fibroblast; EDA FN, extra domain A of fibronectin; ES, enrichment score; FC, fold change; FMO, fluorescence minus one; GO, gene ontology; GSEA, gene set enrichment analysis; MFI, mean fluorescent intensity; NES, normalized enrichment score; PC, principal component; PCA, principal component analysis; RNA-Seq, RNA sequencing; TPM, transcript per million.
      RNA sequencing was performed in six healthy and three psoriatic fibroblast lines at passages 2‒4 (Supplementary Table S1). Robust expression of the fibroblast-related markers CD90 and PDGFRa but not of keratinocyte (CD49f) or endothelial cell (EC) (CD31) markers confirmed the purity of the samples that were used for sequencing (Figure 1b‒d and Supplementary Figure S1a). Principal component analysis showed that the samples clustered according to healthy and psoriatic fibroblasts (Figure 1e). Differential gene expression analysis, using a log2 fold change ≥ 0.6 and a P < 0.05 as a threshold, revealed major changes at the transcriptional level, with 637 genes being upregulated and 657 genes being downregulated in psoriatic fibroblasts (Figure 1f). Gene ontology analysis using PANTHER highlighted the integrin signaling pathway as the most enriched biological process in psoriatic fibroblasts (Figure 1g and Supplementary Table S2). Interestingly, among the other enriched terms (P < 0.05), there was angiogenesis related to vascular expansion, both found by PANTHER and in gene ontology biological processes, which is a characteristic feature of psoriatic skin lesions (
      • Huggenberger R.
      • Detmar M.
      The cutaneous vascular system in chronic skin inflammation.
      ), indicating a potential contribution of psoriatic fibroblasts to EC activation (Supplementary Figure S1b).
      Remarkably, we found that ITGA4 was significantly increased in psoriatic fibroblasts (log2 fold change = 1.2) (Figure 1f). This was confirmed by qPCR, which showed an upregulation of ITGA4 expression (fold change = 5.3, P < 0.0001) in psoriatic fibroblasts (Figure 1h). ITGA4 pairs exclusively with ITGB1 and ITGB7 to bind to FN (
      • Altevogt P.
      • Hubbe M.
      • Ruppert M.
      • Lohr J.
      • Hoegen P von
      • Sammar M.
      • et al.
      The alpha 4 integrin chain is a ligand for alpha 4 beta 7 and alpha 4 beta 1.
      ). Interestingly, ECM‒receptor interaction was identified by Kyoto Encyclopedia of Genes and Genomes as the most enriched gene ontology term in psoriatic fibroblasts, confirming the important role of psoriatic fibroblasts in ECM molecule production (Supplementary Figure S1b). The EDA FN is an alternatively spliced form of FN expressed in cancer and chronic inflammation but absent under physiological conditions (
      • Hemmerle T.
      • Zgraggen S.
      • Matasci M.
      • Halin C.
      • Detmar M.
      • Neri D.
      Antibody-mediated delivery of interleukin 4 to the neo-vasculature reduces chronic skin inflammation.
      ;
      • Tacconi C.
      • Schwager S.
      • Cousin N.
      • Bajic D.
      • Sesartic M.
      • Sundberg J.P.
      • et al.
      Antibody-mediated delivery of VEGFC ameliorates experimental chronic colitis.
      ;
      • Villa A.
      • Trachsel E.
      • Kaspar M.
      • Schliemann C.
      • Sommavilla R.
      • Rybak J.N.
      • et al.
      A high-affinity human monoclonal antibody specific to the alternatively spliced EDA domain of fibronectin efficiently targets tumor neo-vasculature in vivo.
      ). EDA FN has been reported to be still present in nonlesional psoriatic skin, as a sign of dormant chronic inflammation (
      • Gubán B.
      • Vas K.
      • Balog Z.
      • Manczinger M.
      • Bebes A.
      • Groma G.
      • et al.
      Abnormal regulation of fibronectin production by fibroblasts in psoriasis.
      ). We analyzed the expression of EDA FN by qPCR and found a significant upregulation (fold change = 2.4, P < 0.0001) in psoriatic fibroblasts (Figure 1i).
      These results reveal that fibroblasts isolated from psoriatic skin possess a distinct gene expression profile, which is maintained on at least four passages in culture.

      Screening of a library of small molecular epigenetic modifier drugs identifies the CBP/p300 inhibitor A485 as a repressor of ITGA4 and EDA FN expression

      Gene set enrichment analysis of the transcriptional profiles pinpointed terms related to chromatin silencing (normalized enrichment score = ‒2.48, P < 0.0001), negative regulation of gene expression in epigenetic (normalized enrichment score = 2.42, P < 0.0001), and histone exchange (normalized enrichment score = 2.37, P < 0.0001) in psoriatic fibroblasts (Figure 1j and Supplementary Figure S2a and b and Supplementary Table S3). This suggests that psoriatic fibroblasts can maintain their transcriptional aberrations at least partially through epigenetic modifications. We next screened a library of 48 small-molecule epigenetic modifier drugs for their ability to reduce ITGA4 and EDA FN expression in psoriatic fibroblasts from two different donors (Figure 2a). As shown in Figure 2b and c, (+)-JQ1, an inhibitor of the bromodomain (BRD) and extraterminal domain family (
      • Filippakopoulos P.
      • Qi J.
      • Picaud S.
      • Shen Y.
      • Smith W.B.
      • Fedorov O.
      • et al.
      Selective inhibition of BET bromodomains.
      ), and A485, an inhibitor of CBP/p300 HAT domain activity (
      • Lasko L.M.
      • Jakob C.G.
      • Edalji R.P.
      • Qiu W.
      • Montgomery D.
      • Digiammarino E.L.
      • et al.
      Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours [published correction appears in Nature 2018;558:E1].
      ), significantly decreased gene expression of both ITGA4 and EDA FN, whereas I-CBP 112 and SGC-CBP30, which block the BRD activity of CBP/p300 (
      • Hammitzsch A.
      • Tallant C.
      • Fedorov O.
      • O’Mahony A.
      • Brennan P.E.
      • Hay D.A.
      • et al.
      CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses.
      ;
      • Picaud S.
      • Fedorov O.
      • Thanasopoulou A.
      • Leonards K.
      • Jones K.
      • Meier J.
      • et al.
      Generation of a selective small molecule inhibitor of the CBP/p300 bromodomain for leukemia therapy.
      ), only reduced the expression levels of EDA FN.
      Figure thumbnail gr2
      Figure 2Screening of 48 small-molecule epigenetic modifiers for their potential to downregulate the expression of ITGA4 and EDA FN in psoriatic fibroblasts. (a) Schematic workflow showing the experimental design used for the epigenetic drug screening. An epigenetic drug library was screened in psoriatic fibroblasts (n = 2 lines in duplicates) for either 3 or 7 days depending on the drug used (). After the treatment, cells were analyzed by qPCR for ITGA4 and EDA FN expression changes. (b, c) The variation of the expression of (b) ITGA4 and (c) EDA FN under 48 small-molecule epigenetic modifier drugs in psoriatic fibroblasts was quantified by qPCR (n = 2 lines in duplicates per treatment group). Gene expression was normalized to GAPDH using the 2-ΔCT method and expressed as fold changes to the DMSO vehicle control. Red bars indicate the most efficient and consistent (relative value < 0.5) epigenetic modifier drugs selected for further studies. (d) MUH cell viability assay was performed with the indicated treatment for 72 hours (n = 2 lines with ≥ 6 replicates per group), expressed as fold changes to the DMSO vehicle control. (e) MUH cell viability assay after treatment with SGC-CBP30, I-CBP112, and A485 in both psoriatic and healthy fibroblasts (n = 2 lines each in ≥ 6 replicates per treatment group). (f) Schematic illustration of the action of I-CBP112, SGC-CBP30, and A485 epigenetic modifier compounds on blocking the CBP/p300 BRD or the HAT domain. (g) ITGA4 and (h) EDA FN expression in psoriatic and healthy fibroblasts (n = 1 line each in triplicates per group) under the treatments of I-CBP112, SGC-CBP30, A485, or DMSO vehicle control. Gene expression was normalized to GAPDH and shown with the 2-ΔCT method. (i) Western blot images and quantification of ITGA4 protein levels in psoriatic and healthy fibroblasts treated with I-CBP112, A485, or DMSO control (n = 2 lines each per group in three independent experiments). ß-Actin served as the loading control. Values are shown as a ratio normalized to the DMSO vehicle control. (j) Representative images of western blot for EDA FN protein levels in psoriatic and healthy fibroblasts treated with I-CBP112, A485, or DMSO control (n = 2 lines each per group in three independent experiments) and their quantifications. (k) Western blot image and analysis of H3K27ac levels in vehicle- or A485-treated psoriatic and healthy fibroblasts. Total histone H3 served as the loading control (n = 2 lines each in triplicates per treatment group). All data represent mean ± SD. Significance was determined by unpaired or paired (western blots of vehicle treatment for ik) Student’s t-test (ns P > 0.05). ∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001, and ∗∗∗∗P < 0.0001. BRD, bromodomain; EDA FN, extra domain A of fibronectin; H3K27ac, acetylation of lysine 27 on histone 3; HAT, histone acetyltransferase; IC90, the 90% maximal inhibitory concentration; ns, not significant.
      We next tested the effects of (+)-JQ1, A485, I-CBP 112, and SGC-CBP30 on fibroblast proliferation using the MUH cell viability assay. (+)-JQ1 treatment resulted in significant inhibition of psoriatic fibroblast proliferation and was therefore excluded from further experiments (Figure 2d). By contrast, A485, I-CBP 112, and SGC-CBP30 did not inhibit fibroblast proliferation (Figure 2e). Interestingly, I-CBP 112, SGC-CBP30, and A485 target the activity of the same protein, namely CBP/p300. CBP/p300 is a transcriptional coactivator that can acetylate several lysine sites, for instance, on histone H3K27 (
      • Xue Y.
      • Wen H.
      • Shi X.
      CBP/p300: intramolecular and intermolecular regulations.
      ). As shown in Figure 2f, p300 and CBP contain a BRD and a HAT domain that can be selectively inhibited by the small molecules I-CBP 112, SGC-CBP30, and A485.
      We next compared the effects of I-CBP 112, SGC-CBP30, and A485 on ITGA4 and EDA FN expression in both psoriatic and healthy fibroblasts in more detail. As shown in Figure 2g and h, the expression levels of ITGA4 and EDA FN were higher in psoriatic fibroblasts than in healthy fibroblasts, consistent with the results of transcriptomic profiling (Figure 1f, h, and i). Treatment with all three drugs significantly reduced EDA FN expression in healthy fibroblasts, and A485 and I-CBP 112 reduced its expression in psoriatic fibroblasts. Only A485 treatment reduced ITGA4 transcriptional expression levels. Although A485 and I-CBP112 treatment exhibited no significant effects on ITGA4 protein levels overall (Figure 2i), A485 significantly reduced EDA FN protein levels in both psoriatic and healthy fibroblasts, whereas the effects of I-CBP112 were less pronounced (Figure 2j). Given the significantly higher level of acetylation of lysine 27 on histone 3 (H3K27ac) in psoriatic compared with that in healthy fibroblasts, we next studied the effects of A485 on H3K27ac levels. Treatment with A485 strongly diminished total H3K27ac levels in psoriatic fibroblasts, with only minor effects on healthy fibroblasts (Figure 2k). To assess the occupancy of p300/CBP and H3K27ac at the gene regions of FN1 and ITGA4 in fibroblasts, we performed sequential chromatin immunoprecipitation followed by qPCR. The occupancy of p300/CBP and H3K27ac was significantly reduced on A485 treatment compared with that of vehicle control (Supplementary Figure S2c and d). In addition, another inhibitor of the CBP/p300 HAT domain, iP300w, inhibited the expression levels of EDA FN and ITGA4 significantly in the same manner as A485 (Supplementary Figure S2e and f), validating the effect of H3K27ac on the expression of EDA FN and ITGA4. It is of interest that we found that the H3K27ac expression in psoriatic skin was significantly higher than in healthy skin tissue, especially in the dermis, confirming increased levels of H3K27 acetylation in the skin of patients with psoriasis (Supplementary Figure S3a and b).

      The specific CBP/p300 HAT inhibitor A485 alleviates IMQ-induced skin inflammation and decreases relative EDA Fn expression in vivo

      To study the potential therapeutic efficacy of A485 in a mouse model of psoriasis-like skin inflammation, we topically applied 5% IMQ cream on mouse ear skin and administered it daily with intraperitoneal injections of DMSO vehicle or A485 for 7 consecutive days (Figure 3a). Because A485 selectively inhibits the HAT domain of CBP/p300, which is responsible for the H3K27ac, we first evaluated the levels of H3K27ac in inflamed ear tissue after the treatment. A485 treatment efficiently reduced CBP/p300 HAT activity as shown by lower levels of H3K27ac over total H3 in the inflamed ear tissue of A485-treated mice (Figure 3b).
      Figure thumbnail gr3
      Figure 3The p300/CBP HAT inhibitor A485 reduces psoriasis-like skin inflammation. (a) Schematic of the treatment schedule with IMQ to induce psoriasis-like inflammation in mouse ear skin. Mice were treated once per day with A485 or DMSO vehicle control (vehicle) by intraperitoneal injections and euthanized on day 7. (b) Western blot image and analysis of H3K27ac protein levels in inflamed ear tissue lysates from vehicle- or A485-treated mice. Total histone H3 served as the loading control (n = 3 mice per group). (c) Ear thickness expressed as a change compared with ear thickness before IMQ challenge (n = 6–8 mice per group). (d) PASI score resulting from the visual evaluation of ear skin redness, scaling, and ear thickness (n = 6–8 mice per group). (e) The weights of auricular draining LNs on day 7 after the first IMQ application (n = 6‒8 mice per group). (f) Representative images of H&E-stained ear skin sections and quantification of the epidermal thickness (n = 6‒8 mice per group). Bars = 200 μm. (g) Representative immunofluorescence images of inflamed ears of mice that received DMSO vehicle or A485, stained for K6 (green) and Hoechst (blue) and their quantification (n = 6‒8 mice per group). Values were normalized to BM length. Bars = 50 μm. (h) Representative immunofluorescence images of inflamed ears of mice that received DMSO vehicle or A485-stained for Ki-67 (red) and Hoechst (blue) and their quantification (n = 6‒8 mice per group). Values were normalized by field. Bars = 50 μm. (i‒l) The gene expression level of (i) EDA Fn, (j) Fn1, (k) normalized EDA Fn, and (l) Itga4 in inflamed ear tissues under vehicle or A485 treatment. mRNA levels of EDA Fn were normalized on total fibronectin in k, whereas (i) EDA Fn, (j) Fn1, and Itga4 (l) were normalized to Gapdh (n = 6‒8 mice per group). All data represent mean ± SD. Significance was determined by two-way ANOVA test with Sidak’s correction for multiple comparisons in c and d or by unpaired Student’s t-test for b and e‒l. ∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001, and ∗∗∗∗P < 0.0001. BM, basement membrane; EDA Fn, extra domain A of fibronectin; Fn, fibronectin; H3K27ac, acetylation of lysine 27 on histone 3; HAT, histone acetyltransferase; IMQ, imiquimod; i.p., intraperitoneal; K6, keratin 6; LN, lymph node.
      The ear thickness and PASI score, a combined evaluation of redness, scaling, and tissue swelling, were significantly reduced after A485 treatment (Figure 3c and d). In line with reduced inflammation, the weight of the draining auricular lymph nodes was significantly lighter in A485-treated mice (Figure 3e). Mice under A485 treatment showed a moderate 8% body weight loss compared with control mice after 7 days of treatment (Supplementary Figure S4a) without any other obvious adverse effects, which is in agreement with the findings of a previous study (
      • Lasko L.M.
      • Jakob C.G.
      • Edalji R.P.
      • Qiu W.
      • Montgomery D.
      • Digiammarino E.L.
      • et al.
      Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours [published correction appears in Nature 2018;558:E1].
      ) in tumor-bearing mice, which showed that A485 induced a moderate 9% body weight loss and that the mice recovered rapidly on completion of the A485 treatment. Histological analysis of IMQ-treated ears showed a significant reduction in epidermal thickness after A485 treatment (Figure 3f). Immunostainings for the proliferation-associated keratin 6 revealed a significantly reduced keratin 6‒positive area in the epidermis of A485-treated mice (Figure 3g). In agreement, also the number of Ki-67‒positive proliferating epidermal keratinocytes was significantly reduced (Figure 3h). No major differences in lymphatic and blood vessel numbers or the relative area covered by these vessels were found in the ears of A485-treated and control mice (Supplementary Figure S5a‒e). We found no major changes in overall EDA Fn or Fn1 expression levels (Figure 3i and j) but a significant reduction in the expression of EDA Fn normalized to total Fn1 (Figure 3k) in the ear skin of A485-treated mice, whereas Itga4 expression was slightly reduced (Figure 3l). When we normalized the EDA Fn and Itga4 mRNA expression to the expression of the fibroblast markers vimentin and Pdgfra, since fibroblasts are only one of the cell types in human skin, and since the qPCR was done with RNA extracted from whole skin samples, we found a down-regulation of Fn1, EDA Fn, and Itga4 expression by A485 in vivo although the results did not reach statistical significance (Supplementary Figure S4d). These findings indicate that A485, by inhibiting the acetylation of H3K27, can normalize the expression of EDA FN, an important ECM protein elevated in chronic inflammatory conditions (
      • Gubán B.
      • Vas K.
      • Balog Z.
      • Manczinger M.
      • Bebes A.
      • Groma G.
      • et al.
      Abnormal regulation of fibronectin production by fibroblasts in psoriasis.
      ;
      • Schwager S.
      • Renner S.
      • Hemmerle T.
      • Karaman S.
      • Proulx S.T.
      • Fetz R.
      • et al.
      Antibody-mediated delivery of VEGF-C potently reduces chronic skin inflammation.
      ;
      • Tacconi C.
      • Schwager S.
      • Cousin N.
      • Bajic D.
      • Sesartic M.
      • Sundberg J.P.
      • et al.
      Antibody-mediated delivery of VEGFC ameliorates experimental chronic colitis.
      ), also in vivo.

      A485 treatment reduces inflammatory cell infiltration

      We next investigated the effects of A485 treatment on different immune cell populations. Ears of A485-treated or vehicle control‒treated mice were collected on day 7 after IMQ challenge and processed for flow cytometry. Live cells were gated for CD45 positivity, and each subpopulation of immune cells was identified (Supplementary Figure S6a) and quantified. We observed a significant decrease in CD45+ leukocytes (Figure 4a). Macrophages (Figure 4b), including both major histocompatibility complex II high and negative populations (Supplementary Figure S6b and c), as well as dendritic cells (DCs) and Langerhans cells were also significantly decreased on A485 treatment (Supplementary Figure S6d and e). Although there was a slight reduction in CD4 T cells, including T regulatory cells, as well as CD8 T cells, γδ T cells in particular were significantly decreased on A485 treatment (Supplementary Figure S6f‒i). We next evaluated immune cell populations by immunofluorescence stainings. We found a significant decrease in the CD45-positive area (Figure 4c) as well as in the CD68-positive area (Figure 4d), whereas the CD4- or CD8-positive areas were not strongly affected by A485 treatment (Supplementary Figure S6j and k). We also found a statistically not significant reduction of CD4-positive IL17A-producing cells under A485 treatment (Supplementary Figure S4b and c), which is in agreement with our results that A485 mainly attenuated IMQ-induced Il23 mRNA expression but only slightly reduced Il17a mRNA expression (Figure 4e and f). There was a trend toward a reduced area of positive TCR staining, which marks γδ T cells (Supplementary Figure S6l).
      Figure thumbnail gr4
      Figure 4Inflammatory cell infiltration is reduced by A485 treatment. (a) Representative flow cytometry plots of CD45+ cells in inflamed ears of mice that received DMSO vehicle (left) or A485 (right) and their quantification shown as the percentage of total single living cells (n = 3‒4 mice per group). (b) Representative flow cytometry plots of CD45+F4/80+CD11c‒ macrophages in inflamed ears of mice that received DMSO vehicle (left) or A485 (right) and their quantification shown as the percentage of total single living cells (n = 3‒4 mice per group). (c) Representative immunofluorescence images of inflamed ear skin sections of mice that received DMSO vehicle or A485 stained for CD45 (green) and Hoechst (blue) and quantification thereof (n = 6‒8 mice per group). Values are normalized to BM length. (d) Representative immunofluorescence images of inflamed ear skin sections of mice that received DMSO vehicle or A485-stained for CD68 (red) and Hoechst (blue) and quantification thereof (n = 6‒8 mice per group). Values are normalized to BM length. Bars = 100 μm. (e‒h) Gene expression levels of (e) Il17a, (f) Il23p19, (g) Tnf, and (h) Stat3 in inflamed ear tissues under vehicle or A485 treatment (n = 5‒6 mice per group). Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. ∗P < 0.05 and ∗∗P < 0.01. BM, basement membrane; SSC-A, side scatter area; STAT3, signal transducer and activator of transcription 3.
      We next evaluated changes in the expression of Il17a, Il23p19, Tnf, and signal transducer and activator of transcription 3 gene Stat3 because these genes have been shown to play pivotal roles in psoriasis (
      • Ogdie A.
      • Coates L.C.
      • Gladman D.D.
      Treatment guidelines in psoriatic arthritis.
      ). Il23p19 expression showed a significant decrease under A485 treatment. Il17a showed a trend toward reduced expression after treatment, whereas Tnf and signal transducer and activator of transcription 3 showed no major changes (Figure 4e‒h). IL23 is predominantly expressed by macrophages and DCs (
      • Lee E.
      • Trepicchio W.L.
      • Oestreicher J.L.
      • Pittman D.
      • Wang F.
      • Chamian F.
      • et al.
      Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris.
      ;
      • Nestle F.O.
      • Kaplan D.H.
      • Barker J.
      Psoriasis.
      ), which is in line with the flow cytometry results.
      Overall, these results indicate that A485 alleviates skin inflammation by reducing keratinocyte proliferation and normalizing EDA FN production as well as immune cell infiltration and Il23 expression.

      Discussion

      Despite the currently available treatments for psoriasis, including biologics and topical treatments, the disease often relapses and occurs at the same sites where the inflammation was previously present. One possible explanation for the reoccurrence of psoriasis might be an epigenetic memory of skin cells contributing to the pathogenesis of psoriatic skin lesions. Indeed, recent studies suggest that the acquisition of chemoresistance during the recurrences of cancers, such as leukemia (
      • Bhatla T.
      • Wang J.
      • Morrison D.J.
      • Raetz E.A.
      • Burke M.J.
      • Brown P.
      • et al.
      Epigenetic reprogramming reverses the relapse-specific gene expression signature and restores chemosensitivity in childhood B-lymphoblastic leukemia.
      ) and glioblastoma (
      • Romani M.
      • Pistillo M.P.
      • Banelli B.
      Epigenetic targeting of glioblastoma.
      ), can be partially attributed to epigenetic mechanisms. Previous studies have reported that changes in DNA methylation reverted to baseline under treatment (
      • Gu X.
      • Nylander E.
      • Coates P.J.
      • Fahraeus R.
      • Nylander K.
      Correlation between reversal of DNA methylation and clinical symptoms in psoriatic epidermis following narrow-band UVB phototherapy.
      ), indicating the dynamics of epigenetic modifications in psoriasis. New therapeutic approaches using epigenetic modifier drugs are currently being developed against a multitude of diseases, in particular cancers. Recently, epigenetic modifications have also been suggested to occur in chronic inflammatory diseases, including psoriasis (
      • Gu X.
      • Nylander E.
      • Coates P.J.
      • Fahraeus R.
      • Nylander K.
      Correlation between reversal of DNA methylation and clinical symptoms in psoriatic epidermis following narrow-band UVB phototherapy.
      ;
      • Ovejero-Benito M.C.
      • Reolid A.
      • Sánchez-Jiménez P.
      • Saiz-Rodríguez M.
      • Muñoz-Aceituno E.
      • Llamas-Velasco M.
      • et al.
      Histone modifications associated with biological drug response in moderate-to-severe psoriasis.
      ;
      • Pollock R.A.
      • Abji F.
      • Gladman D.D.
      Epigenetics of psoriatic disease: a systematic review and critical appraisal.
      ;
      • Stylianou E.
      Epigenetics: the fine-tuner in inflammatory bowel disease?.
      ). In this study, we identified the selective EP300/CBP inhibitor, A485, through an epigenetic modifier drug library screening performed on fibroblasts derived from psoriasis plaques and confirmed its therapeutic potential in a mouse model of psoriasis-like skin inflammation.
      The finding that distinct transcriptomic patterns of psoriatic fibroblasts were maintained in culture suggests the preservation of their epigenetic memory. Several genes, including EDA FN and ITGA4, remained significantly upregulated in cultured fibroblasts derived from psoriatic skin. In line with our data, previous studies have shown that upregulation of EDA FN is typically observed in cancer and in inflammatory conditions, including psoriatic skin lesions (
      • Kwon A.
      • Chae I.H.
      • You E.
      • Kim S.H.
      • Ahn S.Y.
      • Lee O.J.
      • et al.
      Extra domain A-containing fibronectin expression in Spin90-deficient fibroblasts mediates cancer-stroma interaction and promotes breast cancer progression.
      ;
      • Schwager S.
      • Renner S.
      • Hemmerle T.
      • Karaman S.
      • Proulx S.T.
      • Fetz R.
      • et al.
      Antibody-mediated delivery of VEGF-C potently reduces chronic skin inflammation.
      ;
      • Tacconi C.
      • Schwager S.
      • Cousin N.
      • Bajic D.
      • Sesartic M.
      • Sundberg J.P.
      • et al.
      Antibody-mediated delivery of VEGFC ameliorates experimental chronic colitis.
      ) and nonlesional skin (
      • Gubán B.
      • Vas K.
      • Balog Z.
      • Manczinger M.
      • Bebes A.
      • Groma G.
      • et al.
      Abnormal regulation of fibronectin production by fibroblasts in psoriasis.
      ). The implications of EDA FN and ITGA4 upregulations for psoriasis pathogenesis remain incompletely understood at present. VLA-4, composed of the integrin α4 and β1 chain, is a known receptor of EDA FN, mediating adhesion to the ECM and also to activated endothelium (
      • Elices M.J.
      • Osborn L.
      • Takada Y.
      • Crouse C.
      • Luhowskyj S.
      • Hemler M.E.
      • et al.
      VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronectin binding site.
      ;
      • Wayner E.A.
      • Garcia-Pardo A.
      • Humphries M.J.
      • McDonald J.A.
      • Carter W.G.
      Identification and characterization of the T lymphocyte adhesion receptor for an alternative cell attachment domain (CS-1) in plasma fibronectin.
      ). EDA FN and α4β7 integrin also interact with innate immune receptors to stimulate inflammation and fibrosis (
      • Kelsh-Lasher R.M.
      • Ambesi A.
      • Bertram C.
      • McKeown-Longo P.J.
      Integrin α4β1 and TLR4 Cooperate to Induce fibrotic Gene Expression in Response to fibronectin’s EDA Domain.
      ). A previous study reported that remodeling of the FN matrix in diseased tissue elicits an extra domain A‒dependent inflammatory response in dermal fibroblasts (
      • Kelsh-Lasher R.M.
      • Ambesi A.
      • Bertram C.
      • McKeown-Longo P.J.
      Integrin α4β1 and TLR4 Cooperate to Induce fibrotic Gene Expression in Response to fibronectin’s EDA Domain.
      ), and it was found (
      • Kohan M.
      • Muro A.F.
      • White E.S.
      • Berkman N.
      EDA-containing cellular fibronectin induces fibroblast differentiation through binding to alpha4beta7 integrin receptor and MAPK/Erk 1/2-dependent signaling.
      ) that EDA FN modulates fibroblast differentiation through a mechanism that involves binding of EDA FN to the ITGAα4/β7, which is followed by the activation of focal adhesion kinase and MAPK signaling pathways.
      Psoriatic skin undergoes several epigenetic modifications in terms of DNA methylation, histone modification, and noncoding RNA expression. For example, PBMCs from patients with psoriasis have reduced histone H3 and H4 acetylation levels and increased H3K4 methylation (
      • Zhang P.
      • Su Y.
      • Zhao M.
      • Huang W.
      • Lu Q.
      Abnormal histone modifications in PBMCs from patients with psoriasis vulgaris.
      ). A recent study found that histone modifications were associated with the responsiveness to biologics in patients with psoriasis. Although methylation of H3K27 showed comparable patterns in healthy controls and in patients with psoriasis who did not receive any treatment, there was a significant increase after the treatment only in patients who responded to the treatments (
      • Ovejero-Benito M.C.
      • Reolid A.
      • Sánchez-Jiménez P.
      • Saiz-Rodríguez M.
      • Muñoz-Aceituno E.
      • Llamas-Velasco M.
      • et al.
      Histone modifications associated with biological drug response in moderate-to-severe psoriasis.
      ). This suggests that epigenetics states are dynamically changed during psoriasis pathogenesis, in agreement with our gene set enrichment analysis showing that the downregulated genes in psoriatic fibroblasts were significantly enriched in ontologies related to chromatin and histone modifications such as HIST1H, SIRT2 (which inhibits transcriptional activation by deacetylating EP300, the target protein of A485), HDACs (histone deacetylases), SMARCA, and HAT1. A very recent study showed that demethylation of H3K27 in CD4 T cells was involved in the generation of effector T cells in autoimmune diseases and drove T helper 17 differentiation (
      • Cribbs A.P.
      • Terlecki-Zaniewicz S.
      • Philpott M.
      • Baardman J.
      • Ahern D.
      • Lindow M.
      • et al.
      Histone H3K27me3 demethylases regulate human Th17 cell development and effector functions by impacting on metabolism.
      ), which is vital for the production of IL-23 and IL-17. Thus, alterations of H3K27 methylation and acetylation may also contribute to immune modulation and disease control in response to treatment.
      We designed the screening of epigenetic inhibitor molecules to evaluate their efficacy in reducing the elevated expression of EDA FN and ITGA4 in psoriatic fibroblasts. The epigenetic library contained different kinds of inhibitors, each targeting different epigenetic writers, readers, erasers, or transcriptional regulators. Inhibition of EP300/CBP proteins showed consistent effectiveness in the downregulation of the target genes, in line with a recent study that revealed that CBP/p300 mediated ECM organization and remodeling in synovial fibroblasts (
      • Krošel M.
      • Gabathuler M.
      • Maciukiewicz M.
      • Moser L.
      • Lee G.I.
      • Marks M.
      • et al.
      Individual functions of the histone acetyl transferases CBP and p300 in regulating the inflammatory response of synovial fibroblasts.
      ). Although EP300 (p300) and CBP did not show differential expression levels between psoriatic and healthy fibroblasts, their enzymatic activity on histone acetylation could be different under inflammatory condition. The most potent and consistent effect was observed with (+)-JQ1, A485, I-CBP112, and SGC-CBP30. The inhibitory effect of (+)-JQ1 on fibroblast proliferation is compatible with the findings from previous studies reporting that (+)-JQ1‒mediated bromodomain inhibition triggers cell cycle arrest (
      • Jostes S.
      • Nettersheim D.
      • Fellermeyer M.
      • Schneider S.
      • Hafezi F.
      • Honecker F.
      • et al.
      The bromodomain inhibitor JQ1 triggers growth arrest and apoptosis in testicular germ cell tumours in vitro and in vivo.
      ), and (+)-JQ1 was hence excluded from further experiments. The p300/CBP inhibitors tested reduced the gene expression of our target genes, EDA FN and ITGA4, without impacting cell proliferation, indicating their possible suitability for in vivo usage. In comparison, I-CBP112 and SGC-CBP30 treatment resulted in a less pronounced reduction in EDA FN levels compared with those of A485.
      I-CBP112, SGC-CBP30, and A485 act on the same target, the p300/CBP proteins (
      • Xue Y.
      • Wen H.
      • Shi X.
      CBP/p300: intramolecular and intermolecular regulations.
      ), suggesting the importance of CBP/P300 activity in controlling the expression of the target genes in fibroblasts. CBP/p300 contains multiple domains that are biologically relevant; hence, specific inhibition of specific domains of CBP/p300 by small molecules has been used as a therapeutic strategy in several diseases (
      • Farria A.
      • Li W.
      • Dent S.Y.R.
      KATs in cancer: functions and therapies.
      ;
      • Valor L.M.
      • Viosca J.
      • Lopez-Atalaya J.P.
      • Barco A.
      Lysine acetyltransferases CBP and p300 as therapeutic targets in cognitive and neurodegenerative disorders.
      ). I-CBP112 (
      • Picaud S.
      • Fedorov O.
      • Thanasopoulou A.
      • Leonards K.
      • Jones K.
      • Meier J.
      • et al.
      Generation of a selective small molecule inhibitor of the CBP/p300 bromodomain for leukemia therapy.
      ;
      • Taylor A.M.
      • Côté A.
      • Hewitt M.C.
      • Pastor R.
      • Leblanc Y.
      • Nasveschuk C.G.
      • et al.
      Fragment-based discovery of a selective and cell-active benzodiazepinone CBP/EP300 bromodomain inhibitor (CPI-637).
      ;
      • Wu Q.
      • Heidenreich D.
      • Zhou S.
      • Ackloo S.
      • Krämer A.
      • Nakka K.
      • et al.
      A chemical toolbox for the study of bromodomains and epigenetic signaling.
      ) and SGC-CBP30 (
      • Taylor A.M.
      • Côté A.
      • Hewitt M.C.
      • Pastor R.
      • Leblanc Y.
      • Nasveschuk C.G.
      • et al.
      Fragment-based discovery of a selective and cell-active benzodiazepinone CBP/EP300 bromodomain inhibitor (CPI-637).
      ;
      • Wu Q.
      • Heidenreich D.
      • Zhou S.
      • Ackloo S.
      • Krämer A.
      • Nakka K.
      • et al.
      A chemical toolbox for the study of bromodomains and epigenetic signaling.
      ) are inhibitors of the BRD activity of CBP/p300 proteins. The BRD binds preferentially to diacetylated and triacetylated histones H4 and H2B. The BRD is crucial for CBP/p300-mediated histone acetylation because its deletion impairs the ability to acetylate histones (
      • Xue Y.
      • Wen H.
      • Shi X.
      CBP/p300: intramolecular and intermolecular regulations.
      ). On the other hand, A485 is an inhibitor targeting the HAT domain of the CBP/p300 proteins. A485 treatment did not alter p300 or CBP protein levels but selectively inhibited HAT (
      • Michaelides M.R.
      • Kluge A.
      • Patane M.
      • Van Drie J.H.V.
      • Wang C.
      • Hansen T.M.
      • et al.
      Discovery of Spiro oxazolidinediones as selective, orally bioavailable inhibitors of p300/CBP histone acetyltransferases.
      ). In general, CBP/p300 preferentially acetylates H3K18/K27, whereas A485 inhibits that acetylation and leads to a marked decrease in acetylation within the CBP/p300 activation loop (
      • Lasko L.M.
      • Jakob C.G.
      • Edalji R.P.
      • Qiu W.
      • Montgomery D.
      • Digiammarino E.L.
      • et al.
      Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours [published correction appears in Nature 2018;558:E1].
      ).
      To assess the therapeutic efficacy of A485 in vivo, we used a mouse model of IMQ-induced psoriasis-like skin inflammation. After applying IMQ cream to mouse ear skin, the ears developed characteristic features of psoriasis, including epidermal thickening, erythema, scaling, immune cell infiltration, and upregulation of proinflammatory cytokines such as IL-17 and IL-23 (
      • Van Belle A.B.V.
      • Heusch M de
      • Lemaire M.M.
      • Hendrickx E.
      • Warnier G.
      • Dunussi-Joannopoulos K.
      • et al.
      IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice.
      ;
      • Fits van der L.
      • Mourits S.
      • Voerman J.S.A.
      • Kant M.
      • Boon L.
      • Laman J.D.
      • et al.
      Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis.
      ). Systemic treatment with A485 significantly reduced the development of psoriatic skin lesions. Thus, our results show that targeting acetylation of H3K27 by A485 not only suppresses EDA FN and ITGA4 expression in psoriatic fibroblasts but also inhibits skin inflammation, immune cell infiltration, and the expression of key genes of psoriasis pathogenesis. These findings of the efficacy of an epigenetic modifier drug in a psoriasis model are in agreement with those of a previous study reporting the beneficial effects of another epigenetic modifier drug (+)-JQ1, a BET1 domain inhibitor, in IMQ-induced skin inflammation in mice. Although (+)-JQ1 attenuated IMQ-induced IL-17A but not IL-23 levels, A485 reduced the level of Il23 but not those of Il17a, suggesting that (+)-JQ1 and A485 may act through different mechanisms (
      • Nadeem A.
      • Al-Harbi N.O.
      • Al-Harbi M.M.
      • El-Sherbeeny A.M.
      • Ahmad S.F.
      • Siddiqui N.
      • et al.
      Imiquimod-induced psoriasis-like skin inflammation is suppressed by BET bromodomain inhibitor in mice through RORC/IL-17A pathway modulation.
      ). Taken together, remodeling of H3K27 methylation, as shown in a recent study (
      • Cribbs A.P.
      • Terlecki-Zaniewicz S.
      • Philpott M.
      • Baardman J.
      • Ahern D.
      • Lindow M.
      • et al.
      Histone H3K27me3 demethylases regulate human Th17 cell development and effector functions by impacting on metabolism.
      ), and acetylation through epigenetic modifier drug treatments may contribute to immune modulation and disease control of psoriasis. H3K27 acetylation as well as methylation may also have a potential role as future biomarkers for psoriasis. The level of H3K27 acetylation in psoriatic skin was significantly higher than in healthy skin tissue, which is in agreement with the findings of a previous study (
      • Masalha M.
      • Ben-Dov I.Z.
      • Ram O.
      • Meningher T.
      • Jacob-Hirsch J.
      • Kassem R.
      • et al.
      H3K27Ac modification and gene expression in psoriasis.
      ), which reported that the H3K27ac pattern was different between psoriatic and uninvolved or healthy skin by showing the correlation of overexpressed genes with enrichment of H3K27ac levels in psoriasis.
      The resolution of the psoriatic phenotype after A485 treatment was likely due to direct effects on fibroblasts as well as potentially on several other skin cell types, including keratinocytes and ECs. The reduced number of leukocytes in inflamed mouse ears, including macrophages, DCs, Langerhans cells, and γδ T cells, indicates the potential effects of A485 on immune cells as well. These effects could have been direct and also indirect by reducing the production of inflammatory mediators by dermal fibroblasts. A potential pathogenic role of macrophages is supported by previous findings of a strong production of TNF-α by macrophages in the IL-23‒induced psoriasis-like inflammation mouse model, where the mRNA expression levels of Tnf, Il1ß, Il17a, and ß-defensin were strongly decreased in the ear skin on prophylactic depletion of macrophages (
      • Wang Y.
      • Edelmayer R.
      • Wetter J.
      • Salte K.
      • Gauvin D.
      • Leys L.
      • et al.
      Monocytes/Macrophages play a pathogenic role in IL-23 mediated psoriasis-like skin inflammation.
      ). γδ T cells play a vital role in psoriasis pathogenesis in different disease models (
      • Cai Y.
      • Shen X.
      • Ding C.
      • Qi C.
      • Li K.
      • Li X.
      • et al.
      Pivotal Role of Dermal IL-17-Producing γδ T cells in Skin Inflammation.
      ). Dermal γδ T cells constitutively express CCR6 and, together with T helper 17 cells, produce a multitude of cytokines, such as IL-23 and IL-17. Ccr6-knockout mice were resistant to IL-23‒induced skin inflammation, underpinning the indispensability of CCR6 in response to IL-23 stimulation (
      • Hedrick M.N.
      • Lonsdorf A.S.
      • Shirakawa A.K.
      • Richard Lee C.C.
      • Liao F.
      • Singh S.P.
      • et al.
      CCR6 is required for IL-23–induced psoriasis-like inflammation in mice.
      ). Although TCRα-deficient mice responded to IL-23 in the same way as wild-type mice, TCRδ-deficient mice showed significantly reduced epidermal thickening, neutrophil infiltration, and IL-17 production in the model of IMQ-induced psoriasis-like inflammation (
      • Cai Y.
      • Shen X.
      • Ding C.
      • Qi C.
      • Li K.
      • Li X.
      • et al.
      Pivotal Role of Dermal IL-17-Producing γδ T cells in Skin Inflammation.
      ), suggesting that γδ T cells are more crucial than CD4 or CD8 T cells in this model.
      The strong reduction in DCs, including Langerhans cells, with a significant decrease in Il23p19 by A485 treatment, is particularly noteworthy. DCs are the primary source of IL-23, a key cytokine in the establishment and development of psoriasis. Blocking of the IL-23 subunits IL-12/23p40 or IL-23p19 showed more prominent therapeutic efficacy than conventional treatments in moderate-to-severe psoriasis, implicating that IL-23 production from inflammatory DCs is critical to psoriasis pathogenesis (
      • Chan T.C.
      • Hawkes J.E.
      • Krueger J.G.
      Interleukin 23 in the skin: role in psoriasis pathogenesis and selective interleukin 23 blockade as treatment.
      ). Intradermal injection of IL-23 into mouse skin induced erythema, inflammatory cell infiltrating, and epidermal hyperplasia (
      • Chan J.R.
      • Blumenschein W.
      • Murphy E.
      • Diveu C.
      • Wiekowski M.
      • Abbondanzo S.
      • et al.
      IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2–dependent mechanisms with implications for psoriasis pathogenesis.
      ), whereas Il23p19‒deficient mice showed alleviated psoriatic skin features (
      • Fits van der L.
      • Mourits S.
      • Voerman J.S.A.
      • Kant M.
      • Boon L.
      • Laman J.D.
      • et al.
      Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis.
      ), suggesting that IL-23‒producing DC subsets might define the inflammatory DCs in psoriasis pathogenesis. Collectively, our findings indicate that A485 exerts its anti-inflammatory effects primarily through macrophages, γδ T cells, and Langerhans cells. It will be of great interest to examine, in future studies, more specifically the distinct effects of A485 in vivo on fibroblasts, epithelial cells, and ECs, in particular with regard to their EDA FN and ITGA4 expression levels. Overall, our findings suggest that epigenetic modifications play an important role in psoriasis pathogenesis and that epigenetic modifier drugs might represent, to our knowledge, a previously unreported therapeutic strategy for the treatment of psoriasis.

      Materials and Methods

      More materials and methods can be found online as Supplementary Materials and Methods.

      Isolation and culture of human dermal fibroblasts

      The epidermis was removed after overnight incubation with 0.25% trypsin (Sigma-Aldrich, St. Louis, MO) in Dulbecco's PBS (Gibco, Waltham, MA) at 4 °C. The dermis was digested mechanically and enzymatically with 1,000 U/ml collagenase type I (Worthington Industries, Columbus, OH) and 40 μg/ml DNase (Roche, Basel, Switzerland) in RPMI medium supplemented with 10% fetal bovine serum, 20 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, and 1% antibiotics (all from Gibco) at 37 °C for 40 minutes. Cell suspensions obtained from the digested tissue were filtered and plated on cell culture dishes. Cells were grown to confluency with regular exchange of the medium to remove nonadherent cells and cultured in FN (Merck Millipore, Darmstadt, Germany) -coated plates with EGM-2 medium (Lonza, Basel, Switzerland). To exclude ECs, CD31 Dynabeads (Invitrogen, Waltham, MA) were used according to the manufacturer’s instructions, and adherent CD31-negative cells were cultured as fibroblasts. Cells were cultured in DMEM supplemented with high glucose (4.5 g/l D-glucose), L-glutamine, pyruvate, 1% antibiotics, and 10% fetal bovine serum (all Gibco) and used for all in vitro assays until passage 6. Cells were passaged every 3‒4 days when they reached 80% confluence. Cells were cultured about 10 days at passage 0 after their isolation from the tissue and were routinely tested for mycoplasma contamination (Mycoscope, Genlantis, San Diego, CA).

      Screening of epigenetic modifier drug library

      Human dermal healthy and psoriatic fibroblasts (20,000 cells/well) were seeded into 24-well plates, and the next day, they were treated with the indicated epigenetic modifier drug or 0.1% DMSO; used as vehicle control; and diluted in DMEM supplemented with high glucose (4.5 g/l D-glucose), L-glutamine, pyruvate, 1% antibiotics, and 10% fetal bovine serum (all Gibco). Used epigenetic modifiers are listed in Supplementary Table S4 with information about the used concentration and the duration of the treatment. The compound library used for the epigenetic screening consisted of 44 epigenetic modifiers (SGC epigenetic chemical probe library, Tocris Bioscience, Bristol, United Kingdom) and 5-azacytidine, trichostatin A (Sigma-Aldrich), tasquinimod, TMP 269 (SelleckChem, Houston, TX), and iP300w (Tocris Bioscience).

      IMQ-induced psoriasis-like skin inflammation model

      To induce skin inflammation, a 5% IMQ-containing cream (Aldara, 3M, Saint Paul, MN) was applied daily for 7 consecutive days to the ear skin of female C57BL/6 mice aged 8 weeks obtained from the Janvier Labs (Le Genest-Saint-Isle, France). Each mouse received daily intraperitoneal injections of 10% DMSO in corn oil as the vehicle control or 100 mg/kg of A485 (MedChemExpress, Monmouth Junction, NJ) diluted in 10% DMSO in corn oil. Ear thickness and PASI score were measured daily, and mice were euthanized on day 7. The mice used in this study were housed in the animal facility of the Swiss Federal Institute of Technology (Zurich, Switzerland) under specific pathogen-free conditions. All applicable international, national, and/or institutional guidelines for the care and use of animals were followed. Special care was used to limit animal discomfort throughout the whole study.

      Study approval

      Surplus human samples were obtained from the plastic surgery department and the Department of Dermatology, University Hospital of Zürich with the assistance of the SKINTEGRITY.CH biobank. Only material from patients who had signed written informed consent was used. The use of material for research purposes was approved by the cantonal ethic commission (Ethics Committee of the Canton Zurich, number 2017-00687). Mouse experiments were performed in accordance with licenses ZH212/16 approved by Kantonales Veterinäramt Zürich.

      Data availability statement

      RNA-sequencing datasets can be found at https://www.ebi.ac.uk/arrayexpress/experiments/E-MTAB-11098, hosted at ArrayExpress under accession number E-MTAB-11098. Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Michael Detmar ([email protected]).

      ORCIDs

      Conflict of Interest

      MPL received research funding for unrelated projects from Roche, Novartis, Molecular Partners, and Oncobit AG. The remaining authors state no conflict of interest.

      Acknowledgments

      We would like to thank Cornelia Halin, Lothar Dieterich, and Eliane Sibler (Swiss Federal Institute of Technology, Zurich, Switzerland) for their valuable input regarding experimental design and results. We also thank Jeannette Scholl (Swiss Federal Institute of Technology), Catharine Aquino (Functional Genomics Center Zurich, Zurich, Switzerland), and Emilio Yángüez (Functional Genomics Center Zurich) for excellent technical assistance. This research was funded by Swiss National Science Foundation grant 310030B_185392, European Research Council grant LYVICAM, by University Medicine Zurich (Flagshipproject SKINTEGRITY) and by the ETH Zurich (Open ETH project SKINTEGRITY.CH).

      Author Contributions

      Conceptualization: JK, CT, MD; Methodology: JK, CT, LD, MD; Investigation: JK, YH, ST, PK, LD, CT; Visualization: JK, YH, CT, MD; Writing - Original Draft Preparation: JK, CT, MD; Writing - Review and Editing: JK, YH, CT, MD.

      Supplementary Materials and Methods

      RNA extraction and RNA deep sequencing

      RNA was extracted from healthy and psoriatic fibroblasts using the NucleoSpin RNA kit (Macherey-Nagel, Duren, Germany) according to the manufacturer’s instructions. RNA quantity and quality were assessed using a bioanalyzer (Agilent, Santa Clara, CA). cDNA libraries were generated from 100 ng of high-quality RNA samples (RNA integrity number > 9) and subjected to paired-end (150 bp) sequencing for 30 million reads per sample on average using the Illumina Hiseq4000 system in collaboration with the Functional Genomics Center Zurich (Zurich, Switzerland). Reads were aligned to the Ensembl human genome assembly GRCh38 using STAR, version 2.4.2a (
      • Dobin A.
      • Davis C.A.
      • Schlesinger F.
      • Drenkow J.
      • Zaleski C.
      • Jha S.
      • et al.
      STAR: ultrafast universal RNA-seq aligner.
      ), and blacklisted regions were removed by BEDTools, version 2.25.0 (
      • Quinlan A.R.
      • Hall I.M.
      BEDTools: a flexible suite of utilities for comparing genomic features.
      ). Mapped reads were assigned to expression counts using featureCounts from the Rsubread package (version 1.26.1) (
      • Liao Y.
      • Smyth G.K.
      • Shi W.
      The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads.
      ). Differential gene expression analysis was done using DESeq2 (version .16.1) (
      • Love M.I.
      • Huber W.
      • Anders S.
      Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.
      ), and the cutoff was defined as log2 fold change > 0.6 and P < 0.05. The heatmaps were generated with ggplot2 (version 3.2.0), and gene expression was normalized and shown as the percent of maximum gene expression for each gene. All bioinformatical analyses were performed in R (version 3.4.0). Gene ontology analyses were performed using the PANTHER classification system (
      • Thomas P.D.
      • Campbell M.J.
      • Kejariwal A.
      • Mi H.
      • Karlak B.
      • Daverman R.
      • et al.
      Panther: A library of protein families and subfamilies indexed by function.
      ), Kyoto encyclopedia of genes and genomes database (Kanehisa et al., 2020), Enrichr (
      • Xie Z.
      • Bailey A.
      • Kuleshov M.V.
      • Clarke D.J.B.
      • Evangelista J.E.
      • Jenkins S.L.
      • et al.
      Gene set knowledge discovery with Enrichr.
      ), and gene set enrichment analysis (
      • Mootha V.K.
      • Lindgren C.M.
      • Eriksson K.F.
      • Subramanian A.
      • Sihag S.
      • Lehar J.
      • et al.
      PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes.
      ;
      • Subramanian A.
      • Tamayo P.
      • Mootha V.K.
      • Mukherjee S.
      • Ebert B.L.
      • Gillette M.A.
      • et al.
      Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles.
      ). The dot plot was generated with ggplot2 (version 3.3.5). Raw data are available at ArrayExpress under accession number E-MTAB-11098.

      The 4-methylumbelliferyl heptanoate proliferation assay

      Proliferation assays were performed as described previously (
      • Hirakawa S.
      • Hong Y.K.
      • Harvey N.
      • Schacht V.
      • Matsuda K.
      • Libermann T.
      • et al.
      Identification of vascular lineage-specific genes by transcriptional profiling of isolated blood vascular and lymphatic endothelial cells.
      ). Briefly, human dermal healthy and psoriatic fibroblasts (2,500 cells/well) were seeded into black 96-well plates (Corning Costar, Raleigh, NC) and cultured in DMEM supplemented with high glucose (4.5 g/l D-glucose), L-glutamine, pyruvate, 1% antibiotics, and 10% fetal bovine serum (all Gibco, Waltham, MA). At the indicated timepoints, 100 μg/ml of 4-methylumbelliferyl heptanoate (Sigma-Aldrich, St. Louis, MO) in Dulbecco's PBS was added, and the fluorescence intensity was measured on a SpectraMax reader (Molecular Devices, San Jose, CA) at 355 nm excitation and 460 nm emission. For each condition, at least quintuplicates were analyzed.

      Quantitative PCR

      RNA was isolated from cultured cells as described in the materials and methods or from imiquimod-inflamed ear tissues lysed with a TissueLyser II (Qiagen, Hilden, Germany) and extracted with the Nucleospin RNA kit (Macherey-Nagel). RNA was measured using a NanoDrop ND-1000 spectrophotometer (Witec, Ulm, Germany) and retrotranscribed with the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Waltham, MA). Gene expression was measured by qPCR with the PowerUp SYBR green master mix (Thermo Fisher Scientific, Waltham, MA) on a QuantStudio Real-Time PCR System, version 1.3 (Applied Biosystems). Expression data were normalized on the basis of the expression levels of GAPDH, which was used as an internal housekeeping gene. Primer sequences for human cells were GAPDH forward 5′-GGAATCCCATCACCATCTTCCAGG-3′ and GAPDH reverse 5′-GAGCCCCAGCCTTCTCCATG-3′, extradomain A fibronectin (EDA FN) forward 5′-CATTCACTGATGTGGATGTC-3′ and EDA FN revers 5′-CAGTGTCTTCTTCACCATCA- 3′, and ITGA4 forward 5′-AACACGCTGTTCGGCTACTC-3′ and ITGA4 reverse 5′-TGGAAAGTGTGACCCCCAAC-3′. Primer sequences for mouse tissues were Gapdh forward 5′-CCTGGAAAACCTGCCAAGTATG-3′ and Gapdh reverse 5′-AGAGTGGGAGTTGCTGTTGAAGTC-3′, EDA Fn forward CCCTAAAGGACTGGCATTCA and EDA Fn reverse CATCCTCAGGGCTCGAGTAG, and Fn1 forward CACATGAGACTGGTGGCTACA and Fn1 reverse TTCTGGAGGTACAGGTGATGC, Itga4 forward CTGGAGGAGAGGGATAACC and Itga4 reverse CCCACAAGTCACGATAGAG, Il17a forward 5′-TCAGCGTGTCCAAACACTGAG-3′ and Il17a reverse 5′-CGCCAAGGGAGTTAAAGACTT-3′, Tnf forward 5′-GCGGAGTCCGGGCAGGTCTA-3′ and Tnf reverse 5′-GGGGGCTGGCTCTGTGAGGA-3′, Il23p19 forward 5′-GGAGCAACTTCACACCTCC-3′ and Il23p19 reverse 5′-GGCAGCTATGGCCAAAAAGG-3′, signal transducer and activator of transcription 3 gene Stat3 forward 5′-CCCCCGTACCTGAAGACCAAG-3′ and signal transducer and activator of transcription 3 gene Stat3 reverse 5′-TCCTCACATGGGGGAGGTAG-3′, Pdgfra forward 5′-TTTACATCTATGTACCAGACCC-3′ and Pdgfra reverse 5′-CATCCTCTTCCACGATGAC-3′, and vimentin forward 5′-CCCTTAAAGGCACTAACGAG-3′ and vimentin reverse 5′-GGTAGTTAGCAGCTTCAAGG-3′.

      Western blot

      Cells or ear skin tissue were lysed with lysis buffer (25 mM 4-[2-hydroxyethyl]-1-piperazineethanesulfonic acid, 5 mM EDTA, 1% Triton-X, 150 mM sodium chloride, 10% glycerol, complete protease inhibitor cocktail or 50 mM Tris, 5 mM EDTA, 1% Triton-X, 150 mM sodium chloride, 1 mM sodium orthovanadate, 25 mM sodium fluoride, 10% glycerol, 1 mM phenylmethylsulfonyl fluoride, complete protease inhibitor cocktail) and then centrifuged at 10,000g for 5 minutes at 4 °C to collect the supernatant. The protein concentration in lysates was determined using the Microplate BCA protein assay kit (Thermo Fisher Scientific), according to the manufacturer’s instructions. Proteins were separated by SDS-PAGE using 4–12% Bis-Tris gels (NuPAGE, Life Technologies, Carlsbad, CA) and transferred to polyvinylidene fluoride membranes (Invitrogen, Waltham, MA). Membranes were blocked with 5% BSA diluted in Tris-buffered saline 0.1% Tween and incubated with anti‒EDA FN (1:2,000, ab6328, Abcam, Cambridge, United Kingdom) IST-9 antibody, anti‒β-actin antibody (1:1,000, ab8227, Abcam), anti‒acetylation of lysine 27 on histone 3 antibody (1:1,000, ab4729, Abcam), and anti-H3 antibody (1:1,000, ab18521, Abcam), followed by horseradish peroxidase‒conjugated anti-mouse (1:10,000, P0161, Dako, Santa Clara, CA) or anti-rabbit (1:10,000, P0448, Dako) secondary antibodies. To develop the membrane, the Clarity Western ECL Substrate (Bio-Rad Laboratories, Hercules, CA) was used, and chemiluminescence signals were visualized by a ChemiDoc (Bio-Rad Laboratories).

      Sequential chromatin immunoprecipitation followed by qPCR after A485 treatment

      Sequential chromatin immunoprecipitation‒qPCR experiments were performed as previously described (
      • Ducoli L.
      • Agrawal S.
      • Sibler E.
      • Kouno T.
      • Tacconi C.
      • Hon C.C.
      • et al.
      LETR1 is a lymphatic endothelial-specific lncRNA governing cell proliferation and migration through KLF4 and SEMA3C.
      ;
      • Furlan-Magaril M.
      • Rincón-Arano H.
      • Recillas-Targa F.
      Sequential chromatin immunoprecipitation protocol: ChIP-reChIP.
      ;
      • Wienert B.
      • Wyman S.K.
      • Yeh C.D.
      • Conklin B.R.
      • Corn J.E.
      CRISPR off-target detection with DISCover-seq.
      ) with modifications. A total of 1 × 107 fibroblasts were treated with 0.8 μM of A485 or vehicle control for 3 days and harvested. After the fixation, sonication of fibroblast nuclei was performed using a Covaris S220 system (Covaris, Woburn, MA; peak power: 140, duty factor: 5.0%, cycles per burst: 200) for 12 on/off cycles of 1 minute. Antibody prebinding was performed as follows: 1.25 mg Dynabeads protein A magnetic beads (Thermo Fisher Scientific) were washed twice with 1 ml of blocking solution (0.5% BSA in Dulbecco's PBS). Then, Dynabeads were incubated with rabbit anti-CBP/p300 antibody (1:50, Cell Signaling Technology, Danvers, MA) for 3 hours at 4 °C by rotating. As a control antibody, rabbit IgG control was used (Sigma-Aldrich). After washing once with 1 ml of blocking solution, antibody-bound beads were added to fibroblast nuclear lysates and incubated overnight at 4 °C with slow rotation. Aliquots of 6 μl nuclear lysate were used as input DNA. The next day, antibody-bound beads were washed twice with 1 ml RIPA buffer (50 mM 4-[2-hydroxyethyl]-1-piperazineethanesulfonic acid-potassium hydroxide, pH 7.5; 500 mM LiCl; 1 mM EDTA, pH 8; 1% igepal CA-630; and 0.7% sodium deoxycholate) and once with Tris-buffered solution (20 mM Tris-hydrogen chloride pH 8, 150 mM sodium chloride). To eluate immunocomplexes from the beads, 10 mM dithiothreitol was added and incubated for 30 minutes at 37 °C, and the supernatant was collected. In the meantime, antibody prebinding was performed as described earlier using acetylation of lysine 27 on histone 3 (1:100, Abcam) and rabbit IgG control antibodies. Antibody-bound beads were mixed with the supernatant and incubated overnight at 4 °C with slow rotation. After the elution of DNA, the MiniElute PCR purification kit (Qiagen) was used for DNA isolation. qPCR was performed as described earlier. Target region Ct values were normalized to DNA input, and then enrichment against IgG isotype control was calculated. Primers were designed on the basis of acetylation of lysine 27 on histone 3 chromatin immunoprecipitation sequencing peaks of normal human dermal fibroblasts (ENCSR000APN) from the ENCODE project. The primers for chromatin immunoprecipitation‒qPCR were as follows: FN1 forward 5′-CGCTGAGAAGGGAAGAAGTC-3′ and FN1 reverse 5′- CTGACTCGGGACTCCCTTATT-3′ and ITGA4 forward 5′-AGCATCAGATGGTGGTTGATA-3′ and ITGA4 reverse 5′-CCCAAGGTCACACCACTTGT-3′.

      Immunofluorescence staining and analysis of frozen mouse ear sections

      Optical cutting temperature‒embedded ear samples of imiquimod-treated mice, human psoriasis, and healthy skin biopsies were frozen on liquid nitrogen. A total of 7 μm cryostat sections were fixed with acetone, dehydrated in 80% methanol, and incubated with blocking solution (5% donkey serum, 0.1% Triton-X, and 1% BSA in PBS) for 1 hour. Tissue sections were incubated with the primary antibodies in a blocking solution at 4 °C overnight. Primary antibodies against CD31 (BD550274, 1:200, BD, Franklin Lakes, NJ), LYVE1 (11-034, 1:600, Angiobio, San Diego, CA), keratin 6 (PRB-169P, 1:200, Covance, Princeton, NJ), Ki-67 (14-5698, 1:200, Invitrogen), CD45 (AF114, 1:100, R&D Systems, San Diego, CA), CD3 (MCA1477T, 1:100, Bio-Rad Laboratories), CD4-biotin (BD553045, 1:100, BD Biosciences, San Jose, CA), CD4 (553647, 1:100, BD Pharmingen, San Diego, CA), CD8a (BD553029, 1:100, BD Biosciences), TCR (ab118868, 1:50, Abcam), CD68 (ab53444, 1:200, Abcam), IL17A (ab79056, 1:400, Abcam), and acetylation of lysine 27 on histone 3(ab4729, 1:1,000, Abcam) were used. After abundant washes in PBS, Alexa Fluor 488‒, Alexa Fluor 594‒, or Alexa streptavidin‒conjugated secondary antibodies and Hoechst 33342 (all purchased from Invitrogen; Alexa secondary antibodies; 1:200, Hoechst 33342; 1:1,000) were added for 2 hours at room temperature. Slides were mounted with Mowiol mounting medium. Mouse ear skin or human skin sections were imaged with an Axioskop2 mot plus microscope (Carl Zeiss, Jena, Germany) with an AxioCam MRc camera (Carl Zeiss). For each sample, at least four images were obtained and quantified using ImageJ (National Institutes of Health, Bethesda, MD) in a blind fashion.

      Flow cytometry

      For the purity check of isolated fibroblasts, fibroblasts (100,000 cells) from psoriatic and healthy tissue were collected after CD31 dynabeads-negative selection. Fibroblasts were washed with FACS buffer (Dulbecco's PBS with 2% fetal bovine serum and 1 mM EDTA) and stained with phycoerythrin/Cy7-conjugated mouse anti-human PDGFRa (323508, 10 μg/ml, BioLegend, San Diego, CA), phycoerythrin-conjugated mouse anti-human CD31 (BD555446, 1:20, BD Pharmingen), and allophycocyanin-conjugated rat anti-human CD49f (17-0495-82, 0.4 μg/ml, eBioscience, San Diego, CA) antibodies diluted in FACS buffer for 30 minutes at 4 °C. After a wash with FACS buffer, cells were fixed with 1% paraformaldehyde. Fibroblasts were defined as PDGFRa-positive and CD31- and CD49f-negative cells. Human endothelial cells (CD31 positive), human keratinocytes (CD49f positive), and dermal fibroblasts (PDGFRa positive) were used as the respective positive controls. Human endothelial cells were obtained from the CD31-positive selection described earlier. Human keratinocytes and dermal fibroblasts were obtained from plastic surgery as described previously (
      • Sandu C.
      • Dumas M.
      • Malan A.
      • Sambakhe D.
      • Marteau C.
      • Nizard C.
      • et al.
      Human skin keratinocytes, melanocytes, and fibroblasts contain distinct circadian clock machineries.
      ). Samples were acquired on a Cytoflex S (Beckman Coulter, Brea, CA) using CytoExpert software. Analysis was performed using FlowJo, version 10.5 (BD Biosciences).
      To evaluate immune cell infiltrate in mouse inflamed skin, ears were harvested on day 7 after imiquimod challenge. After mechanical disruption of the tissue, samples were digested under rotation in DMEM (Gibco) containing 4 mg/ml collagenase IV (Gibco) and 40 μg/ml DNase (Roche, Basel, Switzerland) at 37 °C for 40 minutes. Samples were filtered through 40 μm strainers (Falcon, Mexico City, Mexico), centrifuged, and resuspended in FACS buffer containing 2% fetal bovine serum (Gibco) and 2 mM EDTA (Fluka Chemie, Buchs, Switzerland) in Dulbecco's PBS. Samples were incubated with anti-mouse CD16/32 antibody (1:100, BioLegend) on ice for 20 minutes and subsequently stained on ice for 30 minutes using CD11b-BV605 (101237, 1:100, BioLegend), F4/80-AF647 (AbD MCA497A647, 1:100), CD45-PB (103126, 1:400, BioLegend), GDTCR-PerCP710 (46-5711-82, 1:400, eBioscience), CD11c-PE-Cy7 (117318, 1:400, BioLegend), major histocompatibility complex II‒AF700 (107622, 1:800, BioLegend), CD4-FITC (100405, 1:100, BioLegend), CD8-allophycocyanin-Cy7 (100714, 1:100, BioLegend), and langerin-biotin (13-2075-82, 1:100, eBioscience). Cells were incubated with streptavidin-BV650 for 20 minutes. Live/dead cell staining with Zombie Aqua (423102, 1:500, BioLegend) was done together with the antibody incubation. To quantify T regulatory cells, intracellular staining was performed with Foxp3-phycoerythrin (12-5773-80, 1:100, eBioscience) using an intracellular staining kit (72-5775, eBioscience) following the manufacturer’s instructions. Samples were acquired on an LSRFortessa (BD Biosciences) using FACSDiva software. The analysis was performed using FlowJo, version 10.5 (BD Biosciences) in a blinded fashion.

      Statistics

      Statistical analysis was performed with GraphPad Prism 8.0 software (GraphPad Software, San Diego, CA) using unpaired or paired two-tailed Student’s t-test or two-way ANOVA, as indicated. Data are shown as mean plus SD, and differences were considered statistically significant when P < 0.05, as indicated by asterisks with ∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001, and ∗∗∗∗P < 0.0001.
      Figure thumbnail fx2
      Supplementary Figure S1The purity of fibroblasts and enriched terms in psoriatic fibroblasts. (a) Flow cytometry analysis of CD49f and CD31 in dermal fibroblasts from both healthy skin and psoriatic skin lesions. FMO was used as a staining control. (b) Dot plot of gene ontology enrichment analysis using KEGG (
      • Kanehisa M.
      • Furumichi M.
      • Sato Y.
      • Ishiguro-Watanabe M.
      • Tanabe M.
      KEGG: integrating viruses and cellular organisms.
      ), GO:BP (
      • Xie Z.
      • Bailey A.
      • Kuleshov M.V.
      • Clarke D.J.B.
      • Evangelista J.E.
      • Jenkins S.L.
      • et al.
      Gene set knowledge discovery with Enrichr.
      ), and PANTHER (
      • Thomas P.D.
      • Campbell M.J.
      • Kejariwal A.
      • Mi H.
      • Karlak B.
      • Daverman R.
      • et al.
      Panther: A library of protein families and subfamilies indexed by function.
      ) showing the most upregulated terms in psoriatic fibroblasts expressed as gene ratio (overlap) and ‒log10(P-value). EC, endothelial cells; ECM, extracellular matrix; FMO, fluorescence minus one; FSC-H, forward scatter height; GO:BP, gene ontology biological process; KC, keratinocyte; KEGG, kyoto encyclopedia of genes and genomes; SSC-A, side scatter area.
      Figure thumbnail fx3
      Supplementary Figure S2Distinct gene expression in psoriatic fibroblasts and suppression of target genes by p300/CBP HAT inhibitors. (a) Heatmap and (b) enrichment plots from GSEA (
      • Mootha V.K.
      • Lindgren C.M.
      • Eriksson K.F.
      • Subramanian A.
      • Sihag S.
      • Lehar J.
      • et al.
      PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes.
      ;
      • Subramanian A.
      • Tamayo P.
      • Mootha V.K.
      • Mukherjee S.
      • Ebert B.L.
      • Gillette M.A.
      • et al.
      Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles.
      ) showing that genes downregulated in psoriatic fibroblasts are enriched for chromatin assembly or disassembly, chromatin organization, and remodeling. Running ES score and positions of Gene set members on the rank-ordered list based on the significance value from GSEA. (psoriatic, n = 3 and healthy fibroblasts, n = 6). (c, d) Enrichment for CBP/p300 and H3K27ac at (c) FN1 and (d) ITGA4 genomic loci displayed as fold change against IgG control after vehicle or A485 treatment in fibroblasts. (e, f) Downregulation of (e) EDA FN and (f) ITGA4 after vehicle or iP300 treatment (0.3 μM) for 72 hours in fibroblasts. Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. ∗∗P < 0.01 and ∗∗∗P < 0.001. EDA FN, extra domain A of fibronectin; ES, enrichment score; FDR, false discovery rate; GSEA, gene set enrichment analysis; H3K27ac, acetylation of lysine 27 on histone 3; HAT, histone acetyltransferase; NES, normalized enrichment score.
      Figure thumbnail fx4
      Supplementary Figure S3The level of H3K27ac in human healthy and psoriatic skin. (a) Representative immunofluorescence images of healthy and psoriatic skin (n = 4 per group) stained for nuclei (Hoechst; blue) and H3K27ac (red). Bars = 100 μm. (b) Quantification of the H3K27ac-positive area in dermis and epidermis (n = 4 per group). Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. ∗P < 0.05 and ∗∗P < 0.01. H3K27ac, acetylation of lysine 27 on histone 3.
      Figure thumbnail fx5
      Supplementary Figure S4The effect of systemic administration of A485. (a) Average body weight of mice treated with A485 or vehicle control. The mean body weights of the mice in each group (n = 6‒8) were measured. (b) Representative immunofluorescence images of inflamed ears of mice that received DMSO vehicle or A485 (n = 3 mice per group) stained for CD4 (green), IL17A (red), and Hoechst (blue). Bars = 100 μm. (c) Quantification of CD4+IL17A+ cells per total CD4+ immune cells in inflamed ears of mice that received DMSO vehicle or A485, shown as the percentage of CD4+IL17A+ cells of total CD4+ cells (n = 3 mice per group). (d) The mRNA expression levels of Fn1, EDA Fn, and Itga4 normalized per vimentin and Pdgfra mRNA levels in mice treated with A485 or vehicle control (n= 6‒8 mice per group). All data represent mean ± SD. Significance was determined by two-way ANOVA test with Sidak’s correction for multiple comparisons in a or by unpaired Student’s t-test for c and d.P < 0.05 and ∗∗P < 0.01. EDA FN, extra domain A of fibronectin; ns, not significant.
      Figure thumbnail fx6
      Supplementary Figure S5A485 treatment does not affect lymphatic or blood vessels. (a) Representative immunofluorescence images of inflamed ears of mice that received DMSO vehicle or A485 (n = 6‒8 mice per group) stained for LYVE1 (green), CD31 (red), and Hoechst (blue). Bar = 100 μm. (b) The number of CD31-positive LYVE1-positive lymphatic vessels and (c) quantification of lymphatic vessel area in inflamed ears of mice that received the indicated treatment (n = 6‒8 mice per group). (d) The number of CD31-positive LYVE1-negative blood vessels and (e) quantification of blood vessel area in inflamed ears of mice that received the indicated treatment (n = 6‒8 mice per group). The numbers of vessels are normalized to BM length, and the area of vessels is expressed as the percentage of the analyzed area. Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. BM, basement membrane.
      Figure thumbnail fx7
      Supplementary Figure S6Inflammatory cell infiltration in inflamed ears. (a) FACS gating strategy for immune cell subpopulations in inflamed ear tissues (n = 3‒4 mice per group). Alive single cells were selected for CD45 positivity. All gates were set on the basis of FMO controls (except for CD45). (b‒i) Quantification of MHCII-high and -negative macrophages, DCs, Langerhans cells, CD4, CD8, Tregs, γδ T cells in inflamed ears of mice that received DMSO vehicle or A485 by flow cytometry and shown as the percentage of total single living cells (n = 3‒4 mice per group). (j‒l) Quantification of CD4, CD8, and TCR stainings in inflamed ears of mice that received DMSO vehicle or A485 (n = 6‒8 mice per group). Data represent mean ± SD. Significance was determined by unpaired Student’s t-test. ∗P < 0.05, and ∗∗P < 0.01. DC, dendritic cell; FMO, fluorescence minus one; FSC-A, forward scatter area; FSC-H, forward scatter height; K, thousand; MHCII, major histocompatibility complex II; SSC-A, side scatter area; Treg, regulatory T cell.
      Supplementary Table S1Information on Three Patients with Psoriasis and Six Healthy Donors in the Study
      SampleStatusSexAgeLocationPASI
      Patient with psoriasis 1PsoriaticFemale37Abdomen10.3
      Patient with psoriasis 2PsoriaticFemale53Abdomen4.2
      Patient with psoriasis 3PsoriaticFemale62Foot1.5
      Healthy donor 1HealthyFemaleNANA
      Healthy donor 2HealthyFemale37Abdomen
      Healthy donor 3HealthyFemale51Breast
      Healthy donor 4HealthyFemale43Thigh
      Healthy donor 5HealthyFemale72Breast
      Healthy donor 6HealthyFemale45Breast
      Abbreviation: NA, not applicable.
      Supplementary Table S2List of Enriched GO Terms among the Upregulated Genes in Psoriatic Fibroblasts
      TermP-ValueGenes
      Integrin signaling pathway Homo sapiens P000344.78E-04COL27A1;COL13A1;ITGA4;LAMA2;CAV1;COL12A1;NTN4;PIK3R3;ASAP1;RND2;RND3;RAP2B;ITGA8;COL4A5
      p53 pathway Homo sapiens P000590.02545604KAT6B;AKT2;PERP;CCNG1;PIK3R3;DDB2
      Endothelin signaling pathway Homo sapiens P000190.03222586AKT2;PRKCE;PLA2G4A;PIK3R3;ECE1;PRKG1
      Angiogenesis Homo sapiens P000050.03812389EFNB2;WNT2B;WNT5B;AKT2;PRKCE;WNT5A;PDGFC;PIK3R3;PLA2G4A
      Beta2 adrenergic receptor signaling pathway Homo sapiens P043780.05843051PRKX;CACNA1D;VAMP2
      Beta1 adrenergic receptor signaling pathway Homo sapiens P043770.05843051PRKX;CACNA1D;VAMP2
      5HT1 type receptor-mediated signaling pathway Homo sapiens P043730.06368835HTR1B;PRKX;VAMP2
      Axon guidance mediated by netrin Homo sapiens P000090.06915638NTN4;PIK3R3;NFATC2
      Metabotropic glutamate receptor group II pathway Homo sapiens P000400.06915638BDNF;PRKX;VAMP2
      Alzheimer disease-presenilin pathway Homo sapiens P000040.09629226WNT2B;WNT11;WNT5B;WNT5A;LRP1B;CD44
      Abbreviation: GO, gene ontology.
      Supplementary Table S3List of significantly enriched terms by GSEA among the downregulated genes in psoriatic fibroblasts (FDR q-value cutoff < 0.05)
      NameESNESNOM P-valueFDR q-value
      GO_DNA_REPLICATION‒0.66‒2.620.000.00
      GO_DNA_PACKAGING‒0.67‒2.620.000.00
      GO_CHROMOSOME_SEGREGATION‒0.63‒2.600.000.00
      GO_CHROMATIN_ASSEMBLY_OR_DISASSEMBLY‒0.66‒2.580.000.00
      GO_NUCLEAR_CHROMOSOME_SEGREGATION‒0.64‒2.570.000.00
      GO_DNA_CONFORMATION_CHANGE‒0.62‒2.540.000.00
      GO_SISTER_CHROMATID_SEGREGATION‒0.65‒2.520.000.00
      GO_DNA_DEPENDENT_DNA_REPLICATION‒0.70‒2.510.000.00
      GO_SISTER_CHROMATID_COHESION‒0.69‒2.510.000.00
      GO_PROTEIN_DNA_COMPLEX_SUBUNIT_ORGANIZATION‒0.62‒2.490.000.00
      GO_CHROMATIN_SILENCING‒0.68‒2.480.000.00
      GO_CENTROMERE_COMPLEX_ASSEMBLY‒0.76‒2.460.000.00
      GO_TELOMERE_ORGANIZATION‒0.66‒2.430.000.00
      GO_NEGATIVE_REGULATION_OF_GENE_EXPRESSION_EPIGENETIC‒0.66‒2.420.000.00
      GO_ORGANELLE_FISSION‒0.56‒2.400.000.00
      GO_CELL_CYCLE_PHASE_TRANSITION‒0.59‒2.390.000.00
      GO_MITOTIC_NUCLEAR_DIVISION‒0.57‒2.380.000.00
      GO_HISTONE_EXCHANGE‒0.73‒2.370.000.00
      GO_GENE_SILENCING‒0.59‒2.370.000.00
      GO_DOUBLE_STRAND_BREAK_REPAIR‒0.61‒2.360.000.00
      GO_DNA_REPLICATION_INDEPENDENT_NUCLEOSOME_ORGANIZATION‒0.71‒2.340.000.00
      GO_REGULATION_OF_GENE_EXPRESSION_EPIGENETIC‒0.58‒2.330.000.00
      GO_DNA_REPLICATION_DEPENDENT_NUCLEOSOME_ORGANIZATION‒0.76‒2.330.000.00
      GO_CHROMOSOME_ORGANIZATION‒0.53‒2.330.000.00
      GO_DNA_RECOMBINATION‒0.59‒2.330.000.00
      GO_MITOTIC_CELL_CYCLE‒0.54‒2.330.000.00
      GO_STRAND_DISPLACEMENT‒0.81‒2.320.000.00
      GO_PROTEIN_HETEROTETRAMERIZATION‒0.76‒2.310.000.00
      GO_CHROMATIN_SILENCING_AT_RDNA‒0.74‒2.300.000.00
      GO_DNA_REPLICATION_INITIATION‒0.79‒2.300.000.00
      GO_CELL_CYCLE_G1_S_PHASE_TRANSITION‒0.61‒2.290.000.00
      GO_ATP_DEPENDENT_CHROMATIN_REMODELING‒0.66‒2.290.000.00
      GO_CELL_DIVISION‒0.54‒2.290.000.00
      GO_MITOTIC_SISTER_CHROMATID_SEGREGATION‒0.63‒2.280.000.00
      GO_DNA_METABOLIC_PROCESS‒0.53‒2.280.000.00
      GO_POSITIVE_REGULATION_OF_GENE_EXPRESSION_EPIGENETIC‒0.64‒2.270.000.00
      GO_MEIOTIC_CELL_CYCLE‒0.58‒2.260.000.00
      GO_CELL_CYCLE_PROCESS‒0.51‒2.250.000.00
      GO_DNA_STRAND_ELONGATION‒0.76‒2.250.000.00
      GO_PROTEIN_LOCALIZATION_TO_CHROMOSOME‒0.74‒2.250.000.00
      GO_DNA_STRAND_ELONGATION_INVOLVED_IN_DNA_REPLICATION‒0.77‒2.230.000.00
      GO_DNA_BIOSYNTHETIC_PROCESS‒0.61‒2.230.000.00
      GO_DNA_REPAIR‒0.52‒2.230.000.00
      GO_GENE_SILENCING_BY_RNA‒0.59‒2.220.000.00
      GO_MITOTIC_RECOMBINATION‒0.71‒2.220.000.00
      GO_TELOMERE_MAINTENANCE_VIA_RECOMBINATION‒0.73‒2.200.000.00
      GO_INNATE_IMMUNE_RESPONSE_IN_MUCOSA‒0.84‒2.190.000.00
      GO_MEIOSIS_I‒0.62‒2.190.000.00
      GO_CELL_CYCLE_G2_M_PHASE_TRANSITION‒0.57‒2.190.000.00
      GO_CHROMOSOME_LOCALIZATION‒0.65‒2.190.000.00
      GO_MEIOTIC_CELL_CYCLE_PROCESS‒0.57‒2.180.000.00
      GO_CHROMATIN_ORGANIZATION‒0.50‒2.180.000.00
      GO_DNA_SYNTHESIS_INVOLVED_IN_DNA_REPAIR‒0.63‒2.170.000.00
      GO_POSITIVE_REGULATION_OF_MITOTIC_CELL_CYCLE‒0.59‒2.160.000.00
      GO_NON_RECOMBINATIONAL_REPAIR‒0.63‒2.160.000.00
      GO_CHROMATIN_REMODELING‒0.56‒2.150.000.00
      GO_REGULATION_OF_CELL_DIVISION‒0.52‒2.120.000.00
      GO_REGULATION_OF_NUCLEAR_DIVISION‒0.54‒2.120.000.00
      GO_REGULATION_OF_DNA_DEPENDENT_DNA_REPLICATION‒0.67‒2.120.000.00
      GO_CELL_CYCLE_CHECKPOINT‒0.53‒2.110.000.00
      GO_RECOMBINATIONAL_REPAIR‒0.61‒2.110.000.00
      GO_MICROTUBULE_ORGANIZING_CENTER_ORGANIZATION‒0.59‒2.100.000.00
      GO_REGULATION_OF_CENTROSOME_CYCLE‒0.67‒2.090.000.00
      GO_CENTROSOME_DUPLICATION‒0.70‒2.080.000.00
      GO_DICARBOXYLIC_ACID_TRANSPORT‒0.60‒2.080.000.00
      GO_NEGATIVE_REGULATION_OF_CELL_DIVISION‒0.63‒2.080.000.00
      GO_CENTROSOME_CYCLE‒0.65‒2.080.000.00
      GO_MICROTUBULE_CYTOSKELETON_ORGANIZATION‒0.50‒2.070.000.00
      GO_MITOTIC_SPINDLE_ORGANIZATION‒0.60‒2.070.000.00
      GO_POSITIVE_REGULATION_OF_MITOTIC_NUCLEAR_DIVISION‒0.65‒2.070.000.00
      GO_POSITIVE_REGULATION_OF_CELL_CYCLE_PROCESS‒0.51‒2.070.000.00
      GO_RNA_3_END_PROCESSING‒0.56‒2.060.000.00
      GO_POSITIVE_REGULATION_OF_NUCLEAR_DIVISION‒0.62‒2.060.000.00
      GO_REGULATION_OF_GENE_SILENCING‒0.62‒2.040.000.00
      GO_CELLULAR_RESPONSE_TO_DNA_DAMAGE_STIMULUS‒0.47‒2.040.000.00
      GO_PROTEIN_HETEROOLIGOMERIZATION‒0.55‒2.040.000.00
      GO_METAPHASE_PLATE_CONGRESSION‒0.64‒2.040.000.00
      GO_AMINO_ACID_TRANSMEMBRANE_TRANSPORT‒0.60‒2.030.000.00
      GO_SPINDLE_ASSEMBLY‒0.58‒2.030.000.00
      GO_REGULATION_OF_MICROTUBULE_BASED_PROCESS‒0.50‒2.030.000.00
      GO_POSITIVE_REGULATION_OF_CELL_CYCLE‒0.49‒2.020.000.00
      GO_CYTOKINESIS‒0.57‒2.020.000.00
      GO_ORGAN_OR_TISSUE_SPECIFIC_IMMUNE_RESPONSE‒0.72‒2.010.000.00
      GO_PLATELET_DEGRANULATION‒0.54‒2.010.000.00
      GO_PROTEIN_TETRAMERIZATION‒0.53‒2.010.000.00
      GO_MALE_MEIOSIS‒0.67‒2.010.000.00
      GO_MICROTUBULE_BASED_PROCESS‒0.47‒2.000.000.00
      GO_CYTOSKELETON_DEPENDENT_CYTOKINESIS‒0.63‒2.000.000.00
      GO_HOMOLOGOUS_CHROMOSOME_SEGREGATION‒0.65‒2.000.000.00
      GO_POSITIVE_REGULATION_OF_CELL_CYCLE_PHASE_TRANSITION‒0.58‒1.990.000.00
      GO_SPINDLE_CHECKPOINT‒0.70‒1.990.000.00
      GO_POSITIVE_REGULATION_OF_CELL_DIVISION‒0.53‒1.990.000.00
      GO_G2_DNA_DAMAGE_CHECKPOINT‒0.67‒1.980.000.00
      GO_RECIPROCAL_DNA_RECOMBINATION‒0.66‒1.980.000.00
      GO_REGULATION_OF_DNA_REPLICATION‒0.51‒1.980.000.00
      GO_RNA_PHOSPHODIESTER_BOND_HYDROLYSIS_EXONUCLEOLYTIC‒0.65‒1.980.000.00
      GO_DNA_GEOMETRIC_CHANGE‒0.55‒1.980.000.00
      GO_NEGATIVE_REGULATION_OF_CELL_CYCLE_PROCESS‒0.50‒1.970.000.00
      GO_NUCLEUS_ORGANIZATION‒0.52‒1.970.000.00
      GO_POSITIVE_REGULATION_OF_DNA_BIOSYNTHETIC_PROCESS‒0.58‒1.970.000.00
      GO_L_ALPHA_AMINO_ACID_TRANSMEMBRANE_TRANSPORT‒0.68‒1.970.000.00
      GO_NEGATIVE_REGULATION_OF_MEGAKARYOCYTE_DIFFERENTIATION‒0.74‒1.960.000.00
      GO_NUCLEAR_EXPORT‒0.51‒1.960.000.00
      GO_REGULATION_OF_CELL_CYCLE_PROCESS‒0.46‒1.960.000.00
      GO_NCRNA_3_END_PROCESSING‒0.72‒1.960.000.00
      GO_REGULATION_OF_CHROMOSOME_SEGREGATION‒0.55‒1.960.000.00
      GO_RNA_SECONDARY_STRUCTURE_UNWINDING‒0.62‒1.950.000.00
      GO_CHROMOSOME_ORGANIZATION_INVOLVED_IN_MEIOTIC_CELL_CYCLE‒0.63‒1.950.000.00
      GO_EXTRACELLULAR_MATRIX_DISASSEMBLY‒0.56‒1.950.000.00
      GO_TELOMERE_CAPPING‒0.66‒1.950.000.00
      GO_RIBONUCLEOPROTEIN_COMPLEX_LOCALIZATION‒0.53‒1.950.000.00
      GO_MITOTIC_CYTOKINESIS‒0.64‒1.940.000.00
      GO_NEGATIVE_REGULATION_OF_HEMATOPOIETIC_PROGENITOR_CELL_DIFFERENTIATION‒0.68‒1.940.000.00
      GO_PROTEIN_LOCALIZATION_TO_CYTOSKELETON‒0.66‒1.940.000.00
      GO_DNA_DEPENDENT_DNA_REPLICATION_MAINTENANCE_OF_FIDELITY‒0.73‒1.940.000.00
      GO_MEIOTIC_CHROMOSOME_SEGREGATION‒0.59‒1.930.000.00
      GO_AMINO_ACID_TRANSPORT‒0.51‒1.930.000.00
      GO_L_AMINO_ACID_TRANSPORT‒0.58‒1.930.000.00
      GO_MITOTIC_SPINDLE_ASSEMBLY‒0.62‒1.930.000.00
      GO_REGULATION_OF_MICROTUBULE_POLYMERIZATION_OR_DEPOLYMERIZATION‒0.49‒1.920.000.00
      GO_NEGATIVE_REGULATION_OF_CHROMOSOME_SEGREGATION‒0.68‒1.920.000.00
      GO_DNA_DAMAGE_RESPONSE_DETECTION_OF_DNA_DAMAGE‒0.62‒1.920.000.00
      GO_REGULATION_OF_CENTROSOME_DUPLICATION‒0.64‒1.920.000.00
      GO_CHROMATIN_MODIFICATION‒0.45‒1.910.000.00
      GO_REGULATION_OF_TRANSCRIPTION_INVOLVED_IN_G1_S_TRANSITION_OF_MITOTIC_CELL_CYCLE‒0.67‒1.910.000.00
      GO_ANATOMICAL_STRUCTURE_HOMEOSTASIS‒0.46‒1.910.000.00
      GO_PYRIMIDINE_DEOXYRIBONUCLEOTIDE_METABOLIC_PROCESS‒0.74‒1.910.000.00
      GO_REGULATION_OF_SIGNAL_TRANSDUCTION_BY_P53_CLASS_MEDIATOR‒0.49‒1.910.000.00
      GO_T_HELPER_1_TYPE_IMMUNE_RESPONSE‒0.74‒1.900.000.00
      GO_REGULATION_OF_CELL_CYCLE_PHASE_TRANSITION‒0.46‒1.900.000.00
      GO_NUCLEOTIDE_EXCISION_REPAIR_DNA_GAP_FILLING‒0.68‒1.900.000.00
      GO_RNA_PHOSPHODIESTER_BOND_HYDROLYSIS‒0.51‒1.900.000.00
      GO_REGULATION_OF_DNA_RECOMBINATION‒0.58‒1.900.000.00
      GO_NCRNA_PROCESSING‒0.45‒1.890.000.00
      GO_REGULATION_OF_MEGAKARYOCYTE_DIFFERENTIATION‒0.66‒1.890.000.00
      GO_NEGATIVE_REGULATION_OF_CYTOSKELETON_ORGANIZATION‒0.47‒1.890.000.00
      GO_DNA_INTEGRITY_CHECKPOINT‒0.49‒1.890.000.00
      GO_REGULATION_OF_DNA_BINDING‒0.52‒1.880.000.00
      GO_RNA_LOCALIZATION‒0.48‒1.880.000.00
      GO_NUCLEAR_ENVELOPE_ORGANIZATION‒0.53‒1.880.000.00
      GO_REGULATION_OF_MITOTIC_CELL_CYCLE‒0.44‒1.880.000.00
      GO_TRNA_METABOLIC_PROCESS‒0.48‒1.870.000.00
      GO_NEGATIVE_REGULATION_OF_PROTEIN_COMPLEX_DISASSEMBLY‒0.48‒1.870.000.00
      GO_DNA_TEMPLATED_TRANSCRIPTION_TERMINATION‒0.51‒1.870.000.00
      GO_REGULATION_OF_CHROMOSOME_ORGANIZATION‒0.46‒1.870.000.00
      GO_NUCLEIC_ACID_PHOSPHODIESTER_BOND_HYDROLYSIS‒0.46‒1.870.000.00
      GO_CENTRIOLE_ASSEMBLY‒0.72‒1.870.000.00
      GO_MITOTIC_CELL_CYCLE_CHECKPOINT‒0.50‒1.870.000.00
      GO_PROTEIN_HOMOTRIMERIZATION‒0.72‒1.870.000.00
      GO_EXTRACELLULAR_STRUCTURE_ORGANIZATION‒0.45‒1.870.000.00
      GO_BLASTOCYST_GROWTH‒0.74‒1.870.000.01
      GO_REGULATION_OF_CELLULAR_RESPONSE_TO_HEAT‒0.52‒1.870.000.01
      GO_REGULATION_OF_CELL_CYCLE‒0.43‒1.860.000.01
      GO_NCRNA_METABOLIC_PROCESS‒0.44‒1.860.000.01
      GO_NEGATIVE_REGULATION_OF_CELL_CYCLE_PHASE_TRANSITION‒0.49‒1.860.000.01
      GO_CELLULAR_MACROMOLECULAR_COMPLEX_ASSEMBLY‒0.43‒1.860.000.01
      GO_DNA_LIGATION‒0.73‒1.860.000.01
      GO_AORTA_MORPHOGENESIS‒0.66‒1.850.000.01
      GO_XENOPHAGY‒0.50‒1.850.000.01
      GO_PEPTIDYL_LYSINE_MODIFICATION‒0.45‒1.850.000.01
      GO_NITROGEN_COMPOUND_TRANSPORT‒0.44‒1.850.000.01
      GO_ACUTE_PHASE_RESPONSE‒0.59‒1.850.000.01
      GO_SYNAPSIS‒0.63‒1.850.000.01
      GO_MEMBRANE_DISASSEMBLY‒0.58‒1.850.000.01
      GO_PROTEIN_LOCALIZATION_TO_CENTROSOME‒0.73‒1.840.000.01
      GO_RIBOSOME_BIOGENESIS‒0.45‒1.840.000.01
      GO_REGULATION_OF_SISTER_CHROMATID_SEGREGATION‒0.54‒1.840.000.01
      GO_ADAPTIVE_IMMUNE_RESPONSE_BASED_ON_SOMATIC_RECOMBINATION_OF_IMMUNE_RECEPTORS_BUILT_FROM_IMMUNOGLOBULIN_SUPERFAMILY_DOMAINS‒0.49‒1.840.000.01
      GO_POSITIVE_REGULATION_OF_DNA_METABOLIC_PROCESS‒0.47‒1.840.000.01
      GO_NEGATIVE_REGULATION_OF_NUCLEAR_DIVISION‒0.59‒1.840.000.01
      GO_RESPONSE_TO_IONIZING_RADIATION‒0.48‒1.840.000.01
      GO_CHROMOSOME_CONDENSATION‒0.64‒1.840.000.01
      GO_DETECTION_OF_TEMPERATURE_STIMULUS‒0.73‒1.840.000.01
      GO_MITOCHONDRIAL_RNA_METABOLIC_PROCESS‒0.65‒1.840.000.01
      GO_ANDROGEN_RECEPTOR_SIGNALING_PATHWAY‒0.58‒1.840.000.01
      GO_REGULATION_OF_DNA_BIOSYNTHETIC_PROCESS‒0.50‒1.830.000.01
      GO_RESPONSE_TO_ESTRADIOL‒0.48‒1.830.000.01
      GO_MUSCLE_HYPERTROPHY‒0.64‒1.830.000.01
      GO_INTERSTRAND_CROSS_LINK_REPAIR‒0.58‒1.830.000.01
      GO_POSTREPLICATION_REPAIR‒0.56‒1.830.000.01
      GO_POSITIVE_REGULATION_OF_WOUND_HEALING‒0.58‒1.820.000.01
      GO_RRNA_METABOLIC_PROCESS‒0.45‒1.820.000.01
      GO_SOMATIC_DIVERSIFICATION_OF_IMMUNE_RECEPTORS‒0.57‒1.820.000.01
      GO_POSITIVE_REGULATION_OF_COAGULATION‒0.65‒1.820.000.01
      GO_PEPTIDE_CROSS_LINKING‒0.58‒1.820.000.01
      GO_TRNA_TRANSPORT‒0.61‒1.820.000.01
      GO_REGULATION_OF_TELOMERE_MAINTENANCE‒0.52‒1.820.000.01
      GO_TRNA_PROCESSING‒0.49‒1.820.000.01
      GO_REGULATION_OF_DNA_METABOLIC_PROCESS‒0.44‒1.820.000.01
      GO_POSITIVE_REGULATION_OF_CHROMOSOME_ORGANIZATION‒0.47‒1.810.000.01
      GO_LACTATION‒0.59‒1.810.000.01
      GO_PTERIDINE_CONTAINING_COMPOUND_METABOLIC_PROCESS‒0.60‒1.810.000.01
      GO_REGULATION_OF_CYTOKINESIS‒0.54‒1.810.000.01
      GO_TELOMERE_MAINTENANCE_VIA_TELOMERE_LENGTHENING‒0.65‒1.810.000.01
      GO_PROTEIN_SUMOYLATION‒0.48‒1.810.000.01
      GO_ANTEROGRADE_AXONAL_TRANSPORT‒0.65‒1.810.000.01
      GO_BASE_EXCISION_REPAIR‒0.58‒1.810.000.01
      GO_REGULATION_OF_CELL_CYCLE_G2_M_PHASE_TRANSITION‒0.53‒1.810.000.01
      GO_NUCLEOBASE_CONTAINING_COMPOUND_TRANSPORT‒0.46‒1.810.000.01
      GO_MAMMARY_GLAND_LOBULE_DEVELOPMENT‒0.70‒1.810.000.01
      GO_NUCLEAR_IMPORT‒0.47‒1.800.000.01
      GO_REGULATION_OF_CELL_CYCLE_CHECKPOINT‒0.62‒1.800.000.01
      GO_COPPER_ION_TRANSPORT‒0.66‒1.800.000.01
      GO_POSITIVE_REGULATION_OF_LEUKOCYTE_MEDIATED_IMMUNITY‒0.52‒1.800.000.01
      GO_NEGATIVE_REGULATION_OF_ORGANELLE_ORGANIZATION‒0.43‒1.800.000.01
      GO_POSITIVE_REGULATION_OF_NUCLEOTIDE_CATABOLIC_PROCESS‒0.70‒1.800.000.01
      GO_POSITIVE_REGULATION_OF_CELL_CYCLE_G2_M_PHASE_TRANSITION‒0.69‒1.800.000.01
      GO_RNA_PROCESSING‒0.41‒1.800.000.01
      GO_POSITIVE_REGULATION_OF_DNA_REPLICATION‒0.51‒1.800.000.01
      GO_NEGATIVE_REGULATION_OF_MITOTIC_CELL_CYCLE‒0.45‒1.790.000.01
      GO_ACIDIC_AMINO_ACID_TRANSPORT‒0.66‒1.790.000.01
      GO_REGULATION_OF_CYCLIN_DEPENDENT_PROTEIN_KINASE_ACTIVITY‒0.50‒1.790.000.01
      GO_MISMATCH_REPAIR‒0.60‒1.790.000.01
      GO_NEGATIVE_REGULATION_OF_MITOTIC_NUCLEAR_DIVISION‒0.58‒1.780.000.01
      GO_REGULATION_OF_PROTEIN_COMPLEX_DISASSEMBLY‒0.45‒1.780.000.01
      GO_MRNA_3_END_PROCESSING‒0.51‒1.780.000.01
      GO_PYRIMIDINE_NUCLEOTIDE_METABOLIC_PROCESS‒0.55‒1.770.000.01
      GO_DEFENSE_RESPONSE_TO_OTHER_ORGANISM‒0.42‒1.770.000.01
      GO_ACUTE_INFLAMMATORY_RESPONSE‒0.52‒1.770.000.01
      GO_PROTEIN_K11_LINKED_UBIQUITINATION‒0.62‒1.770.000.01
      GO_NEGATIVE_REGULATION_OF_DNA_REPLICATION‒0.53‒1.770.000.01
      GO_TRANSITION_METAL_ION_HOMEOSTASIS‒0.48‒1.770.000.01
      GO_ANAPHASE_PROMOTING_COMPLEX_DEPENDENT_CATABOLIC_PROCESS‒0.50‒1.770.000.01
      GO_RIBONUCLEOPROTEIN_COMPLEX_BIOGENESIS‒0.42‒1.760.000.02
      GO_RNA_METHYLATION‒0.54‒1.760.000.02
      GO_CELLULAR_TRANSITION_METAL_ION_HOMEOSTASIS‒0.51‒1.760.000.02
      GO_REGULATION_OF_DEFENSE_RESPONSE_TO_VIRUS‒0.44‒1.760.000.02
      GO_ORGANELLE_ASSEMBLY‒0.41‒1.760.000.02
      GO_NUCLEAR_TRANSCRIBED_MRNA_CATABOLIC_PROCESS_EXONUCLEOLYTIC‒0.59‒1.750.000.02
      GO_GLUTAMATE_SECRETION‒0.60‒1.750.000.02
      GO_SOMATIC_CELL_DNA_RECOMBINATION‒0.59‒1.750.000.02
      GO_RESPONSE_TO_ESTROGEN‒0.44‒1.750.000.02
      GO_REGULATION_OF_LEUKOCYTE_MEDIATED_IMMUNITY‒0.46‒1.750.000.02
      GO_REGULATION_OF_RESPONSE_TO_INTERFERON_GAMMA‒0.63‒1.750.000.02
      GO_OOCYTE_MATURATION‒0.68‒1.750.000.02
      GO_TRANSITION_METAL_ION_TRANSPORT‒0.48‒1.750.000.02
      GO_POSITIVE_REGULATION_OF_DNA_BINDING‒0.55‒1.750.000.02
      GO_MULTI_MULTICELLULAR_ORGANISM_PROCESS‒0.44‒1.740.000.02
      GO_CYCLIC_NUCLEOTIDE_CATABOLIC_PROCESS‒0.67‒1.740.000.02
      GO_REGULATION_OF_DEFENSE_RESPONSE_TO_VIRUS_BY_HOST‒0.46‒1.740.000.02
      GO_B_CELL_MEDIATED_IMMUNITY‒0.50‒1.740.000.02
      GO_POSITIVE_REGULATION_OF_NUCLEOSIDE_METABOLIC_PROCESS‒0.63‒1.740.000.02
      GO_HISTONE_H4_ACETYLATION‒0.55‒1.740.000.02
      GO_RESPIRATORY_SYSTEM_DEVELOPMENT‒0.44‒1.740.000.02
      GO_SENSORY_PERCEPTION_OF_TEMPERATURE_STIMULUS‒0.65‒1.740.000.02
      GO_REGULATION_OF_TELOMERE_MAINTENANCE_VIA_TELOMERE_LENGTHENING‒0.53‒1.730.000.02
      GO_REGULATED_EXOCYTOSIS‒0.43‒1.730.000.02
      GO_DNA_DOUBLE_STRAND_BREAK_PROCESSING‒0.65‒1.730.010.02
      GO_FOLIC_ACID_CONTAINING_COMPOUND_METABOLIC_PROCESS‒0.59‒1.730.000.02
      GO_SOMATIC_DIVERSIFICATION_OF_IMMUNOGLOBULINS‒0.59‒1.730.000.02
      GO_PROTEIN_IMPORT‒0.45‒1.730.000.02
      GO_POSITIVE_REGULATION_OF_TRANSCRIPTION_FROM_RNA_POLYMERASE_II_PROMOTER_IN_RESPONSE_TO_STRESS‒0.63‒1.730.000.02
      GO_MALE_GENITALIA_DEVELOPMENT‒0.64‒1.730.000.02
      GO_ISOTYPE_SWITCHING‒0.68‒1.730.010.02
      GO_HUMORAL_IMMUNE_RESPONSE_MEDIATED_BY_CIRCULATING_IMMUNOGLOBULIN‒0.54‒1.730.000.02
      GO_TRNA_METHYLATION‒0.64‒1.730.000.02
      GO_REGULATION_OF_HUMORAL_IMMUNE_RESPONSE‒0.53‒1.730.000.02
      GO_REGULATION_OF_PROTEIN_UBIQUITINATION_INVOLVED_IN_UBIQUITIN_DEPENDENT_PROTEIN_CATABOLIC_PROCESS‒0.46‒1.730.000.02
      GO_RNA_MODIFICATION‒0.47‒1.730.000.02
      GO_MITOCHONDRIAL_TRANSPORT‒0.45‒1.720.000.02
      GO_POSITIVE_REGULATION_OF_PROTEIN_MODIFICATION_BY_SMALL_PROTEIN_CONJUGATION_OR_REMOVAL‒0.43‒1.720.000.02
      GO_NEUROMUSCULAR_JUNCTION_DEVELOPMENT‒0.56‒1.720.000.02
      GO_POSITIVE_REGULATION_OF_CELL_CYCLE_G1_S_PHASE_TRANSITION‒0.58‒1.720.000.02
      GO_REGULATION_OF_IMMUNE_EFFECTOR_PROCESS‒0.41‒1.720.000.02
      GO_ORGANELLE_TRANSPORT_ALONG_MICROTUBULE‒0.52‒1.720.000.02
      GO_ANTIMICROBIAL_HUMORAL_RESPONSE‒0.56‒1.720.000.02
      GO_NEGATIVE_REGULATION_OF_HEMOPOIESIS‒0.46‒1.720.000.02
      GO_REGULATION_OF_VASCULAR_ENDOTHELIAL_GROWTH_FACTOR_PRODUCTION‒0.57‒1.720.000.02
      GO_NEGATIVE_REGULATION_OF_CELL_CYCLE‒0.41‒1.720.000.02
      GO_MULTI_ORGANISM_LOCALIZATION‒0.50‒1.720.000.02
      GO_REGULATION_OF_DOUBLE_STRAND_BREAK_REPAIR_VIA_HOMOLOGOUS_RECOMBINATION‒0.65‒1.720.010.02
      GO_MATURATION_OF_5_8S_RRNA_FROM_TRICISTRONIC_RRNA_TRANSCRIPT_SSU_RRNA_5_8S_RRNA_LSU_RRNA_‒0.63‒1.720.010.02
      GO_RRNA_CATABOLIC_PROCESS‒0.65‒1.720.000.02
      GO_COVALENT_CHROMATIN_MODIFICATION‒0.41‒1.710.000.02
      GO_POSITIVE_REGULATION_OF_INTERFERON_BETA_PRODUCTION‒0.58‒1.710.000.02
      GO_NEGATIVE_REGULATION_OF_CHROMOSOME_ORGANIZATION‒0.47‒1.710.000.02
      GO_POSTTRANSCRIPTIONAL_GENE_SILENCING‒0.57‒1.710.000.02
      GO_CELLULAR_RESPONSE_TO_REACTIVE_NITROGEN_SPECIES‒0.68‒1.710.010.02
      GO_REGULATION_OF_TELOMERASE_ACTIVITY‒0.54‒1.710.000.02
      GO_REGULATION_OF_CYTOSKELETON_ORGANIZATION‒0.40‒1.710.000.02
      GO_REGULATION_OF_TRANSCRIPTION_REGULATORY_REGION_DNA_BINDING‒0.56‒1.710.000.02
      GO_ORGANIC_ACID_TRANSMEMBRANE_TRANSPORT‒0.47‒1.710.000.02
      GO_CELL_PROLIFERATION‒0.40‒1.710.000.02
      GO_POSITIVE_REGULATION_OF_LIGASE_ACTIVITY‒0.46‒1.710.000.02
      GO_REGULATION_OF_AMINO_ACID_TRANSPORT‒0.64‒1.700.000.03
      GO_POSITIVE_REGULATION_OF_LYMPHOCYTE_MEDIATED_IMMUNITY‒0.51‒1.700.000.03
      GO_REGULATION_OF_PROTEASOMAL_UBIQUITIN_DEPENDENT_PROTEIN_CATABOLIC_PROCESS‒0.44‒1.700.000.03
      GO_IMMUNOGLOBULIN_PRODUCTION‒0.54‒1.700.000.03
      GO_REGULATION_OF_LIGASE_ACTIVITY‒0.45‒1.700.000.03
      GO_REGULATION_OF_HEMATOPOIETIC_PROGENITOR_CELL_DIFFERENTIATION‒0.54‒1.700.000.03
      GO_REGULATION_OF_PROTEIN_ACTIVATION_CASCADE‒0.55‒1.700.000.03
      GO_NEGATIVE_REGULATION_OF_MYELOID_CELL_DIFFERENTIATION‒0.48‒1.700.000.03
      GO_BETA_CATENIN_TCF_COMPLEX_ASSEMBLY‒0.52‒1.700.000.03
      GO_IN_UTERO_EMBRYONIC_DEVELOPMENT‒0.41‒1.690.000.03
      GO_MACROMOLECULE_METHYLATION‒0.43‒1.690.000.03
      GO_POSITIVE_REGULATION_OF_VASCULAR_ENDOTHELIAL_GROWTH_FACTOR_PRODUCTION‒0.59‒1.690.010.03
      GO_REGULATION_OF_RESPONSE_TO_BIOTIC_STIMULUS‒0.42‒1.690.000.03
      GO_NEGATIVE_REGULATION_OF_CELLULAR_PROTEIN_CATABOLIC_PROCESS‒0.49‒1.690.000.03
      GO_DEVELOPMENTAL_PROCESS_INVOLVED_IN_REPRODUCTION‒0.39‒1.690.000.03
      GO_CELLULAR_IRON_ION_HOMEOSTASIS‒0.54‒1.680.000.03
      GO_DNA_TEMPLATED_TRANSCRIPTION_ELONGATION‒0.46‒1.680.010.03
      GO_IRON_ION_HOMEOSTASIS‒0.49‒1.680.000.03
      GO_REGULATION_OF_ACUTE_INFLAMMATORY_RESPONSE‒0.49‒1.680.000.03
      GO_REGULATION_OF_CELLULAR_EXTRAVASATION‒0.63‒1.680.010.03
      GO_NUCLEOSIDE_MONOPHOSPHATE_BIOSYNTHETIC_PROCESS‒0.49‒1.680.000.03
      GO_INFLAMMATORY_RESPONSE‒0.40‒1.680.000.03
      GO_SYNAPTIC_TRANSMISSION_GLUTAMATERGIC‒0.62‒1.680.000.03
      GO_CELLULAR_RESPONSE_TO_ESTRADIOL_STIMULUS‒0.58‒1.680.010.03
      GO_PEPTIDYL_SERINE_MODIFICATION‒0.44‒1.680.000.03
      GO_MITOTIC_DNA_INTEGRITY_CHECKPOINT‒0.46‒1.670.000.03
      GO_PROTEIN_MODIFICATION_BY_SMALL_PROTEIN_CONJUGATION_OR_REMOVAL‒0.38‒1.670.000.03
      GO_RRNA_MODIFICATION‒0.60‒1.670.010.03
      GO_REGULATION_OF_INTERFERON_BETA_PRODUCTION‒0.52‒1.670.000.03
      GO_POSITIVE_REGULATION_OF_TRANSPORTER_ACTIVITY‒0.48‒1.670.000.03
      GO_LYMPHOCYTE_ACTIVATION_INVOLVED_IN_IMMUNE_RESPONSE‒0.48‒1.670.000.03
      GO_DNA_MODIFICATION‒0.48‒1.670.000.04
      GO_SOMATIC_RECOMBINATION_OF_IMMUNOGLOBULIN_GENE_SEGMENTS‒0.61‒1.670.010.04
      GO_INTERMEDIATE_FILAMENT_ORGANIZATION‒0.64‒1.670.010.04
      GO_CLEAVAGE_INVOLVED_IN_RRNA_PROCESSING‒0.62‒1.670.010.04
      GO_INNATE_IMMUNE_RESPONSE‒0.39‒1.670.000.04
      GO_INTRACELLULAR_STEROID_HORMONE_RECEPTOR_SIGNALING_PATHWAY‒0.49‒1.670.000.04
      GO_PROTEIN_LOCALIZATION_TO_NUCLEUS‒0.43‒1.670.000.04
      GO_NEURONAL_STEM_CELL_POPULATION_MAINTENANCE‒0.63‒1.660.010.04
      GO_SULFUR_COMPOUND_TRANSPORT‒0.58‒1.660.010.04
      GO_DETECTION_OF_MECHANICAL_STIMULUS_INVOLVED_IN_SENSORY_PERCEPTION‒0.59‒1.660.010.04
      GO_REGULATION_OF_GLUCOSE_TRANSPORT‒0.46‒1.660.000.04
      GO_VITAMIN_TRANSPORT‒0.57‒1.660.010.04
      GO_POSITIVE_REGULATION_OF_TELOMERASE_ACTIVITY‒0.58‒1.660.010.04
      GO_PROTEIN_LOCALIZATION_TO_ORGANELLE‒0.39‒1.660.000.04
      GO_IMMUNOGLOBULIN_PRODUCTION_INVOLVED_IN_IMMUNOGLOBULIN_MEDIATED_IMMUNE_RESPONSE‒0.61‒1.660.010.04
      GO_TRANSLESION_SYNTHESIS‒0.53‒1.660.010.04
      GO_TRANSCRIPTION_FROM_RNA_POLYMERASE_I_PROMOTER‒0.55‒1.660.000.04
      GO_CARDIAC_CELL_DEVELOPMENT‒0.52‒1.660.000.04
      GO_RESPONSE_TO_RADIATION‒0.40‒1.660.000.04
      GO_REGULATION_OF_CELL_CYCLE_G1_S_PHASE_TRANSITION‒0.43‒1.660.000.04
      GO_REGULATION_OF_PROTEIN_MODIFICATION_BY_SMALL_PROTEIN_CONJUGATION_OR_REMOVAL‒0.41‒1.660.000.04
      GO_NEGATIVE_REGULATION_OF_BLOOD_PRESSURE‒0.54‒1.660.000.04
      GO_NEGATIVE_REGULATION_OF_LYMPHOCYTE_APOPTOTIC_PROCESS‒0.58‒1.660.010.04
      GO_NUCLEAR_TRANSPORT‒0.40‒1.660.000.04
      GO_ANION_TRANSMEMBRANE_TRANSPORT‒0.41‒1.660.000.04
      GO_REACTIVE_OXYGEN_SPECIES_BIOSYNTHETIC_PROCESS‒0.59‒1.660.010.04
      GO_CYTOSKELETON_ORGANIZATION‒0.38‒1.650.000.04
      GO_REGULATION_OF_WATER_LOSS_VIA_SKIN‒0.63‒1.650.010.04
      GO_NEGATIVE_REGULATION_OF_VASCULATURE_DEVELOPMENT‒0.47‒1.650.000.04
      GO_TRNA_MODIFICATION‒0.49‒1.650.000.04
      GO_GLOMERULUS_DEVELOPMENT‒0.51‒1.650.000.04
      GO_TERMINATION_OF_RNA_POLYMERASE_II_TRANSCRIPTION‒0.48‒1.650.000.04
      GO_SPLICEOSOMAL_SNRNP_ASSEMBLY‒0.52‒1.650.000.04
      GO_REGULATION_OF_PROTEASOMAL_PROTEIN_CATABOLIC_PROCESS‒0.42‒1.650.000.04
      GO_POSITIVE_REGULATION_OF_CYCLIN_DEPENDENT_PROTEIN_KINASE_ACTIVITY‒0.54‒1.650.010.04
      GO_POSITIVE_REGULATION_OF_CHROMOSOME_SEGREGATION‒0.59‒1.650.010.04
      GO_PROTEIN_STABILIZATION‒0.44‒1.650.000.04
      GO_ACTIVATION_OF_PHOSPHOLIPASE_C_ACTIVITY‒0.60‒1.650.010.04
      GO_PROTEIN_DEPOLYMERIZATION‒0.59‒1.650.010.04
      GO_REGULATION_OF_NUCLEOTIDE_CATABOLIC_PROCESS‒0.53‒1.650.010.04
      GO_TELOMERE_MAINTENANCE_VIA_TELOMERASE‒0.65‒1.640.010.04
      GO_RESPONSE_TO_X_RAY‒0.57‒1.640.010.04
      GO_RESPONSE_TO_ALCOHOL‒0.40‒1.640.000.04
      GO_REPRODUCTIVE_SYSTEM_DEVELOPMENT‒0.39‒1.640.000.04
      GO_INFLAMMATORY_RESPONSE_TO_ANTIGENIC_STIMULUS‒0.60‒1.640.000.04
      GO_POSITIVE_REGULATION_OF_TELOMERE_MAINTENANCE‒0.51‒1.640.000.04
      GO_TUBE_MORPHOGENESIS‒0.40‒1.640.000.04
      GO_ORGANIC_ANION_TRANSPORT‒0.40‒1.640.000.04
      GO_CYTOSKELETON_DEPENDENT_INTRACELLULAR_TRANSPORT‒0.44‒1.640.000.04
      GO_UROGENITAL_SYSTEM_DEVELOPMENT‒0.40‒1.640.000.04
      GO_GLYCOSPHINGOLIPID_BIOSYNTHETIC_PROCESS‒0.59‒1.640.020.04
      GO_NEGATIVE_REGULATION_OF_EXTRINSIC_APOPTOTIC_SIGNALING_PATHWAY_VIA_DEATH_DOMAIN_RECEPTORS‒0.54‒1.640.010.04
      GO_RESPONSE_TO_ALKALOID‒0.44‒1.640.000.04
      GO_ORGANIC_ACID_TRANSPORT‒0.41‒1.640.000.04
      GO_CERAMIDE_BIOSYNTHETIC_PROCESS‒0.52‒1.630.010.04
      GO_REGULATION_OF_GENERATION_OF_PRECURSOR_METABOLITES_AND_ENERGY‒0.47‒1.630.000.04
      GO_DEFENSE_RESPONSE_TO_VIRUS‒0.43‒1.630.000.04
      GO_DEFENSE_RESPONSE‒0.37‒1.630.000.04
      GO_POSITIVE_REGULATION_OF_ACTIN_CYTOSKELETON_REORGANIZATION‒0.65‒1.630.010.04
      GO_PROTEIN_EXPORT_FROM_NUCLEUS‒0.55‒1.630.010.04
      GO_GANGLIOSIDE_BIOSYNTHETIC_PROCESS‒0.65‒1.630.010.04
      GO_TRANSCRIPTION_COUPLED_NUCLEOTIDE_EXCISION_REPAIR‒0.46‒1.630.010.05
      GO_REGULATION_OF_DEFENSE_RESPONSE‒0.38‒1.630.000.05
      GO_MACROPHAGE_ACTIVATION‒0.57‒1.630.010.04
      GO_MACROAUTOPHAGY‒0.40‒1.630.000.04
      GO_ERROR_PRONE_TRANSLESION_SYNTHESIS‒0.61‒1.630.010.05
      GO_POSITIVE_REGULATION_OF_CARBOHYDRATE_METABOLIC_PROCESS‒0.48‒1.630.000.05
      GO_SNRNA_METABOLIC_PROCESS‒0.46‒1.630.000.05
      GO_RESPONSE_TO_BIOTIC_STIMULUS‒0.37‒1.630.000.05
      GO_NEGATIVE_REGULATION_OF_POTASSIUM_ION_TRANSPORT‒0.56‒1.630.010.05
      GO_AORTA_DEVELOPMENT‒0.52‒1.630.000.05
      GO_OSSIFICATION‒0.40‒1.620.000.05
      GO_NEGATIVE_REGULATION_OF_PROTEOLYSIS‒0.39‒1.620.000.05
      GO_HISTONE_MRNA_METABOLIC_PROCESS‒0.56‒1.620.010.05
      GO_ADAPTIVE_IMMUNE_RESPONSE‒0.41‒1.620.000.05
      GO_REGULATION_OF_MEMBRANE_PERMEABILITY‒0.47‒1.620.000.05
      GO_POSITIVE_REGULATION_OF_TELOMERE_MAINTENANCE_VIA_TELOMERE_LENGTHENING‒0.54‒1.620.010.05
      GO_DNA_TEMPLATED_TRANSCRIPTION_INITIATION‒0.41‒1.620.000.05
      GO_PEPTIDYL_AMINO_ACID_MODIFICATION‒0.37‒1.620.000.05
      GO_LONG_TERM_SYNAPTIC_POTENTIATION‒0.53‒1.620.000.05
      GO_REGULATION_OF_MACROPHAGE_DERIVED_FOAM_CELL_DIFFERENTIATION‒0.56‒1.620.020.05
      GO_POSITIVE_REGULATION_OF_VASCULAR_ENDOTHELIAL_GROWTH_FACTOR_RECEPTOR_SIGNALING_PATHWAY‒0.65‒1.620.020.05
      GO_RESPONSE_TO_CYTOKINE‒0.37‒1.620.000.05
      GO_EPITHELIAL_CELL_APOPTOTIC_PROCESS‒0.57‒1.620.010.05
      GO_NEPHRON_DEVELOPMENT‒0.441.620.000.05
      Abbreviations: ES, enrichment score; FDR, false discovery rate; GO, gene ontology; NES, normalized enrichment score.
      Supplementary Table S4List of 48 Epigenetic Modifier Drugs Used for Screening
      ProbeMolecular

      Weight (g/mol)
      Used with Recommend Concentration (μM), IC90Treatment DurationDomain: Protein TargetDescription
      (+)-JQ1456.990.23 daysBRD: BET familyBET BRD inhibitor
      GSK J4417.5053 daysKDM: JMJD3/UTXHistone KDM inhibitor
      UNC 1215529.7213 daysKme: L3MBTL3L3MBTL3 Kme reader domain inhibitor
      SGC-CBP30509.0413 daysBRD: CREBBP/EP300CBP/p300 BRD inhibitor
      I-CBP 112468.5933 daysBRD: CREBBP/EP300CBP/p300 BRD inhibitor
      (R)-PFI 2 hydrochloride535.9813 daysMT: SETD7SETD7 inhibitor
      UNC 1999569.7433 daysMT: EZH2/H1EZH2/EZH1 inhibitor
      PFI 3321.3713 daysBRD: SMARCA2/4SMARCA2/4 and polybromo 1 inhibitor
      UNC 0642546.7013 daysMT: G9a/GLPG9a and GLP inhibitor
      A 366329.4413 daysMT: G9a/GLPG9a/GLP inhibitor
      BAZ2-ICR357.4113 daysBRD: BAZ2A/2BBAZ2 inhibitor
      OICR 9429555.5933 daysWD40: WDR5WDR5 antagonist
      OF 1440.3113 daysBRD: BRPF1/2/3BRPF1B and BRPF2 inhibitor
      GSK LSD 1 dihydrochloride289.2413 daysKDM: LSD1LSD1 inhibitor
      SGC 707298.3413 daysMT: PRMT3allosteric inhibitor of PRMT3
      NI 57383.4213 daysBRD: BRPF1/2/3BRPF bromodomain inhibitor
      BI 9564353.4113 daysBRD: BRD9/7BRD9 and BRD7 inhibitor; orally active
      I-BRD9497.5513 daysBRD: BRD9BRD9 inhibitor
      GSK 2801371.4533 daysBRD: BAZ2A/2BBAZ2A and BAZ2B inhibitor
      MS049 oxalate salt338.4053 daysMT: PRMT4/6PRMT4 and PRMT6 inhibitor
      MS 023 dihydrochloride360.3213 daysMT: PRMT type 1type I PRMT inhibitor
      NVS-CECR2-1495.6813 daysBRD: CECR2CECR2 inhibitor
      BAY 299429.4713 daysBRD: BRPF2/TAF1BRD1 and TAF1 inhibitor
      TP 472333.3813 daysBRD: BRD9/7BRD9/7 inhibitor
      TP 064458.6013 daysMT: PRMT4PRMT 4 inhibitor
      BAY 598525.3413 daysMT: SMYD2SMYD2 inhibitor
      A 395486.6513 daysKme: EEDEED inhibitor
      GSK 343541.6933 daysMT: EZH2SAM-competitive EZH2 inhibitor
      A 196359.2513 daysMT: SUV420H1/H2SUV420H1/H2 inhibitor
      GSK 6853409.4813 daysBRD: BRPF1BBRPF1 inhibitor
      L Moses dihydrochloride433.3853 daysBRD: PCAF/GCN5PCAF bromodomain inhibitor
      A 485536.480.83 daysHAT: p300/CBPp300/CBP HAT inhibitor; orally bioavailable
      BAY 6035396.4913 daysMT: SMYD3SMYD3 inhibitor
      GSK 864558.6033 daysDEHYDR: IDH1 mutantAllosteric inhibitor of mutant IDH1
      SGC 3027787.3733 daysMT: PRMT7PRMT7 inhibitor pro-drug
      MRK 740464.5733 daysMT: PRDM9PRDM9 inhibitor
      GSK 8814527.6113 daysBRD: BAZ2A, BAZ2BATAD2/2B inhibitor
      BAY 850654.2533 daysBRD: ATAD2ATAD2A inhibitor
      TP 238495.0413 daysBRD: CECR2, BPTF (FALZ)CECR2 and BPTF/FALZ inhibitor
      GSK 4027377.2813 daysBRD: KAT2B (PCAF), KAT2A (GCN5)KAT2B/PCAF and KAT2A/GCN5 inhibitor
      VinSpinin738.960.33 daysTudor Domain: Spin1Spin family inhibitor
      SGC 0946618.5717 daysMT: DOT1LHighly potent and selective DOT1L inhibitor
      GSK 591 dihydrochloride453.4117 daysMT: PRMT5Potent and selective PRMT5 inhibitor
      LLY 283342.350.57 daysMT: PRMT5PRMT5 inhibitor
      Tasquinimod406.3617 daysHDAC4HDAC4 negative allosteric modulator; also binds S100A9; antiangiogenic
      TMP269514.520.0257 daysHDACClass IIa HDAC inhibitor
      TSA302.370.37 daysHDACClass I HDAC inhibitor
      5 AZA228.2157 daysMT: DNMTDNMT1 inhibitor
      Abbreviations: 5-AZA, 5-azacytidine; BRD, bromodomain; IC90, the 90% maximal inhibitory concentration; TSA, trichostatin A

      References

        • Altevogt P.
        • Hubbe M.
        • Ruppert M.
        • Lohr J.
        • Hoegen P von
        • Sammar M.
        • et al.
        The alpha 4 integrin chain is a ligand for alpha 4 beta 7 and alpha 4 beta 1.
        J Exp Med. 1995; 182: 345-355
        • Andreoli F.
        • Jorge Moura Barbosa A.
        • Daniele Parenti M.
        • Del Rio A.
        Modulation of epigenetic targets for anticancer therapy: clinicopathological relevance, structural data and drug discovery perspectives.
        Curr Pharm Des. 2013; 19: 578-613
        • Arasa J.
        • Terencio M.C.
        • Andrés R.M.
        • Marín-Castejón A.
        • Valcuende-Cavero F.
        • Payá M.
        • et al.
        Defective induction of COX-2 expression by psoriatic fibroblasts promotes pro-inflammatory activation of macrophages.
        Front Immunol. 2019; 10: 536
        • Bhatla T.
        • Wang J.
        • Morrison D.J.
        • Raetz E.A.
        • Burke M.J.
        • Brown P.
        • et al.
        Epigenetic reprogramming reverses the relapse-specific gene expression signature and restores chemosensitivity in childhood B-lymphoblastic leukemia.
        Blood. 2012; 119: 5201-5210
        • Cai Y.
        • Shen X.
        • Ding C.
        • Qi C.
        • Li K.
        • Li X.
        • et al.
        Pivotal Role of Dermal IL-17-Producing γδ T cells in Skin Inflammation.
        Immunity. 2011; 35 ([published correction appears in Immunity 2011;35:649]): 596-610
        • Chan J.R.
        • Blumenschein W.
        • Murphy E.
        • Diveu C.
        • Wiekowski M.
        • Abbondanzo S.
        • et al.
        IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2–dependent mechanisms with implications for psoriasis pathogenesis.
        J Exp Med. 2006; 203: 2577-2587
        • Chan T.C.
        • Hawkes J.E.
        • Krueger J.G.
        Interleukin 23 in the skin: role in psoriasis pathogenesis and selective interleukin 23 blockade as treatment.
        Ther Adv Chronic Dis. 2018; 9: 111-119
        • Chandra A.
        • Senapati S.
        • Roy S.
        • Chatterjee G.
        • Chatterjee R.
        Epigenome-wide DNA methylation regulates cardinal pathological features of psoriasis.
        Clin Epigenet. 2018; 10: 108
        • Clark R.A.
        Gone but not forgotten: lesional memory in psoriatic skin.
        J Invest Dermatol. 2011; 131: 283-285
        • Coentro J.Q.
        • Pugliese E.
        • Hanley G.
        • Raghunath M.
        • Zeugolis D.I.
        Current and upcoming therapies to modulate skin scarring and fibrosis.
        Adv Drug Deliv Rev. 2019; 146: 37-59
        • Cribbs A.P.
        • Terlecki-Zaniewicz S.
        • Philpott M.
        • Baardman J.
        • Ahern D.
        • Lindow M.
        • et al.
        Histone H3K27me3 demethylases regulate human Th17 cell development and effector functions by impacting on metabolism.
        Proc Natl Acad Sci USA. 2020; 117: 6056-6066
        • Elices M.J.
        • Osborn L.
        • Takada Y.
        • Crouse C.
        • Luhowskyj S.
        • Hemler M.E.
        • et al.
        VCAM-1 on activated endothelium interacts with the leukocyte integrin VLA-4 at a site distinct from the VLA-4/fibronectin binding site.
        Cell. 1990; 60: 577-584
        • Ellinghaus E.
        • Ellinghaus D.
        • Stuart P.E.
        • Nair R.P.
        • Debrus S.
        • Raelson J.V.
        • et al.
        Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2.
        Nat Genet. 2010; 42: 991-995
        • Fardi M.
        • Solali S.
        • Farshdousti Hagh M.F.
        Epigenetic mechanisms as a new approach in cancer treatment: an updated review.
        Genes Dis. 2018; 5: 304-311
        • Farria A.
        • Li W.
        • Dent S.Y.R.
        KATs in cancer: functions and therapies.
        Oncogene. 2015; 34: 4901-4913
        • Filippakopoulos P.
        • Qi J.
        • Picaud S.
        • Shen Y.
        • Smith W.B.
        • Fedorov O.
        • et al.
        Selective inhibition of BET bromodomains.
        Nature. 2010; 468: 1067-1073
        • Fits van der L.
        • Mourits S.
        • Voerman J.S.A.
        • Kant M.
        • Boon L.
        • Laman J.D.
        • et al.
        Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis.
        J Immunol. 2009; 182: 5836-5845
        • Ganesan A.
        • Arimondo P.B.
        • Rots M.G.
        • Jeronimo C.
        • Berdasco M.
        The timeline of epigenetic drug discovery: from reality to dreams.
        Clin Epigenet. 2019; 11: 174
        • Gu X.
        • Nylander E.
        • Coates P.J.
        • Fahraeus R.
        • Nylander K.
        Correlation between reversal of DNA methylation and clinical symptoms in psoriatic epidermis following narrow-band UVB phototherapy.
        J Invest Dermatol. 2015; 135: 2077-2083
        • Gubán B.
        • Vas K.
        • Balog Z.
        • Manczinger M.
        • Bebes A.
        • Groma G.
        • et al.
        Abnormal regulation of fibronectin production by fibroblasts in psoriasis.
        Br J Dermatol. 2016; 174: 533-541
        • Gudjonsson J.E.
        • Krueger G.
        A role for epigenetics in psoriasis: methylated cytosine-guanine sites differentiate lesional from nonlesional skin and from normal skin.
        J Invest Dermatol. 2012; 132: 506-508
        • Gupta R.
        • Ahn R.
        • Lai K.
        • Mullins E.
        • Debbaneh M.
        • Dimon M.
        • et al.
        Landscape of long noncoding RNAs in psoriatic and healthy skin.
        J Invest Dermatol. 2016; 136: 603-609
        • Hammitzsch A.
        • Tallant C.
        • Fedorov O.
        • O’Mahony A.
        • Brennan P.E.
        • Hay D.A.
        • et al.
        CBP30, a selective CBP/p300 bromodomain inhibitor, suppresses human Th17 responses.
        Proc Natl Acad Sci USA. 2015; 112: 10768-10773
        • Hawkes J.E.
        • Yan B.Y.
        • Chan T.C.
        • Krueger J.G.
        Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis.
        J Immunol. 2018; 201: 1605-1613
        • Hedrick M.N.
        • Lonsdorf A.S.
        • Shirakawa A.K.
        • Richard Lee C.C.
        • Liao F.
        • Singh S.P.
        • et al.
        CCR6 is required for IL-23–induced psoriasis-like inflammation in mice.
        J Clin Invest. 2009; 119: 2317-2329
        • Hemmerle T.
        • Zgraggen S.
        • Matasci M.
        • Halin C.
        • Detmar M.
        • Neri D.
        Antibody-mediated delivery of interleukin 4 to the neo-vasculature reduces chronic skin inflammation.
        J Dermatol Sci. 2014; 76: 96-103
        • Huggenberger R.
        • Detmar M.
        The cutaneous vascular system in chronic skin inflammation.
        J Investig Dermatol Symp Proc. 2011; 15: 24-32
        • Jostes S.
        • Nettersheim D.
        • Fellermeyer M.
        • Schneider S.
        • Hafezi F.
        • Honecker F.
        • et al.
        The bromodomain inhibitor JQ1 triggers growth arrest and apoptosis in testicular germ cell tumours in vitro and in vivo.
        J Cell Mol Med. 2017; 21: 1300-1314
        • Kalluri R.
        • Zeisberg M.
        Fibroblasts in cancer.
        Nat Rev Cancer. 2006; 6: 392-401
        • Kelsh-Lasher R.M.
        • Ambesi A.
        • Bertram C.
        • McKeown-Longo P.J.
        Integrin α4β1 and TLR4 Cooperate to Induce fibrotic Gene Expression in Response to fibronectin’s EDA Domain.
        J Invest Dermatol. 2017; 137: 2505-2512
        • Kohan M.
        • Muro A.F.
        • White E.S.
        • Berkman N.
        EDA-containing cellular fibronectin induces fibroblast differentiation through binding to alpha4beta7 integrin receptor and MAPK/Erk 1/2-dependent signaling.
        FASEB J. 2010; 24: 4503-4512
        • Krošel M.
        • Gabathuler M.
        • Maciukiewicz M.
        • Moser L.
        • Lee G.I.
        • Marks M.
        • et al.
        Individual functions of the histone acetyl transferases CBP and p300 in regulating the inflammatory response of synovial fibroblasts.
        J Autoimmun. 2021; 123102709
        • Krueger G.G.
        • Jorgensen C.M.
        Defined system to assess the in vitro induction of a psoriasis phenotype on normal keratinocytes by fibroblasts from psoriatic subjects.
        J Cutan Med Surg. 1997; 2: 20-25
        • Kwon A.
        • Chae I.H.
        • You E.
        • Kim S.H.
        • Ahn S.Y.
        • Lee O.J.
        • et al.
        Extra domain A-containing fibronectin expression in Spin90-deficient fibroblasts mediates cancer-stroma interaction and promotes breast cancer progression.
        J Cell Physiol. 2020; 235: 4494-4507
        • Lasko L.M.
        • Jakob C.G.
        • Edalji R.P.
        • Qiu W.
        • Montgomery D.
        • Digiammarino E.L.
        • et al.
        Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours [published correction appears in Nature 2018;558:E1].
        Nature. 2017; 550: 128-132
        • Lee E.
        • Trepicchio W.L.
        • Oestreicher J.L.
        • Pittman D.
        • Wang F.
        • Chamian F.
        • et al.
        Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris.
        J Exp Med. 2004; 199: 125-130
        • Masalha M.
        • Ben-Dov I.Z.
        • Ram O.
        • Meningher T.
        • Jacob-Hirsch J.
        • Kassem R.
        • et al.
        H3K27Ac modification and gene expression in psoriasis.
        J Dermatol Sci. 2021; 103: 93-100
        • Michaelides M.R.
        • Kluge A.
        • Patane M.
        • Van Drie J.H.V.
        • Wang C.
        • Hansen T.M.
        • et al.
        Discovery of Spiro oxazolidinediones as selective, orally bioavailable inhibitors of p300/CBP histone acetyltransferases.
        ACS Med Chem Lett. 2018; 9: 28-33
        • Nadeem A.
        • Al-Harbi N.O.
        • Al-Harbi M.M.
        • El-Sherbeeny A.M.
        • Ahmad S.F.
        • Siddiqui N.
        • et al.
        Imiquimod-induced psoriasis-like skin inflammation is suppressed by BET bromodomain inhibitor in mice through RORC/IL-17A pathway modulation.
        Pharmacol Res. 2015; 99: 248-257
        • Nair R.P.
        • Duffin K.C.
        • Helms C.
        • Ding J.
        • Stuart P.E.
        • Goldgar D.
        • et al.
        Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways.
        Nat Genet. 2009; 41: 199-204
        • Nestle F.O.
        • Kaplan D.H.
        • Barker J.
        Psoriasis.
        N Engl J Med. 2009; 361: 496-509
        • Ogdie A.
        • Coates L.C.
        • Gladman D.D.
        Treatment guidelines in psoriatic arthritis.
        Rheumatology (Oxford). 2020; 59: i37-i46
        • Oliveira M.F.S.P.
        • Rocha B.O.
        • Duarte G.V.
        Psoriasis: classical and emerging comorbidities.
        An Bras Dermatol. 2015; 90: 9-20
        • Ovejero-Benito M.C.
        • Reolid A.
        • Sánchez-Jiménez P.
        • Saiz-Rodríguez M.
        • Muñoz-Aceituno E.
        • Llamas-Velasco M.
        • et al.
        Histone modifications associated with biological drug response in moderate-to-severe psoriasis.
        Exp Dermatol. 2018; 27: 1361-1371
        • Picaud S.
        • Fedorov O.
        • Thanasopoulou A.
        • Leonards K.
        • Jones K.
        • Meier J.
        • et al.
        Generation of a selective small molecule inhibitor of the CBP/p300 bromodomain for leukemia therapy.
        Cancer Res. 2015; 75: 5106-5119
        • Pinborg A.
        • Loft A.
        • Romundstad L.B.
        • Wennerholm U.B.
        • Söderström-Anttila V.
        • Bergh C.
        • et al.
        Epigenetics and assisted reproductive technologies.
        Acta Obstet Gynecol Scand. 2016; 95: 10-15
        • Pollock R.A.
        • Abji F.
        • Gladman D.D.
        Epigenetics of psoriatic disease: a systematic review and critical appraisal.
        J Autoimmun. 2017; 78: 29-38
        • Priestley G.C.
        • Adams L.W.
        Hyperactivity of fibroblasts cultured from psoriatic skin: I. Faster proliferation and effect of serum withdrawal.
        Br J Dermatol. 1983; 109: 149-156
        • Roberson E.D.O.
        • Liu Y.
        • Ryan C.
        • Joyce C.E.
        • Duan S.
        • Cao L.
        • et al.
        A subset of methylated CpG sites differentiate psoriatic from normal skin.
        J Invest Dermatol. 2012; 132: 583-592
        • Romani M.
        • Pistillo M.P.
        • Banelli B.
        Epigenetic targeting of glioblastoma.
        Front Oncol. 2018; 8: 448
        • Schwager S.
        • Renner S.
        • Hemmerle T.
        • Karaman S.
        • Proulx S.T.
        • Fetz R.
        • et al.
        Antibody-mediated delivery of VEGF-C potently reduces chronic skin inflammation.
        JCI Insight. 2018; 3e124850
        • Stylianou E.
        Epigenetics: the fine-tuner in inflammatory bowel disease?.
        Curr Opin Gastroenterol. 2013; 29: 370-377
        • Tacconi C.
        • Schwager S.
        • Cousin N.
        • Bajic D.
        • Sesartic M.
        • Sundberg J.P.
        • et al.
        Antibody-mediated delivery of VEGFC ameliorates experimental chronic colitis.
        ACS Pharmacol Transl Sci. 2019; 2: 342-352
        • Tang H.
        • Jin X.
        • Li Y.
        • Jiang H.
        • Tang X.
        • Yang X.
        • et al.
        A large-scale screen for coding variants predisposing to psoriasis.
        Nat Genet. 2014; 46: 45-50
        • Taylor A.M.
        • Côté A.
        • Hewitt M.C.
        • Pastor R.
        • Leblanc Y.
        • Nasveschuk C.G.
        • et al.
        Fragment-based discovery of a selective and cell-active benzodiazepinone CBP/EP300 bromodomain inhibitor (CPI-637).
        ACS Med Chem Lett. 2016; 7: 531-536
        • Valor L.M.
        • Viosca J.
        • Lopez-Atalaya J.P.
        • Barco A.
        Lysine acetyltransferases CBP and p300 as therapeutic targets in cognitive and neurodegenerative disorders.
        Curr Pharm Des. 2013; 19: 5051-5064
        • Van Belle A.B.V.
        • Heusch M de
        • Lemaire M.M.
        • Hendrickx E.
        • Warnier G.
        • Dunussi-Joannopoulos K.
        • et al.
        IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice.
        J Immunol. 2012; 188: 462-469
        • Van Linthout S.V.
        • Miteva K.
        • Tschöpe C.
        Crosstalk between fibroblasts and inflammatory cells.
        Cardiovasc Res. 2014; 102: 258-269
        • Verma D.
        • Ekman A.K.
        • Bivik Eding C.B.
        • Enerbäck C.
        Genome-wide DNA methylation profiling identifies differential methylation in uninvolved psoriatic epidermis.
        J Invest Dermatol. 2018; 138: 1088-1093
        • Villa A.
        • Trachsel E.
        • Kaspar M.
        • Schliemann C.
        • Sommavilla R.
        • Rybak J.N.
        • et al.
        A high-affinity human monoclonal antibody specific to the alternatively spliced EDA domain of fibronectin efficiently targets tumor neo-vasculature in vivo.
        Int J Cancer. 2008; 122: 2405-2413
        • Wang Y.
        • Edelmayer R.
        • Wetter J.
        • Salte K.
        • Gauvin D.
        • Leys L.
        • et al.
        Monocytes/Macrophages play a pathogenic role in IL-23 mediated psoriasis-like skin inflammation.
        Sci Rep. 2019; 9: 5310
        • Wayner E.A.
        • Garcia-Pardo A.
        • Humphries M.J.
        • McDonald J.A.
        • Carter W.G.
        Identification and characterization of the T lymphocyte adhesion receptor for an alternative cell attachment domain (CS-1) in plasma fibronectin.
        J Cell Biol. 1989; 109: 1321-1330
        • Wu B.
        • Pan X.
        • Chen X.
        • Chen M.
        • Shi K.
        • Xu J.
        • et al.
        Epigenetic drug library screening identified an LSD1 inhibitor to target UTX-deficient cells for differentiation therapy.
        Signal Transduct Target Ther. 2019; 4: 11
        • Wu Q.
        • Heidenreich D.
        • Zhou S.
        • Ackloo S.
        • Krämer A.
        • Nakka K.
        • et al.
        A chemical toolbox for the study of bromodomains and epigenetic signaling.
        Nat Commun. 2019; 10: 1915
        • Xue Y.
        • Wen H.
        • Shi X.
        CBP/p300: intramolecular and intermolecular regulations.
        Front Biol. 2018; 13: 168-179
        • Zhang P.
        • Su Y.
        • Zhao M.
        • Huang W.
        • Lu Q.
        Abnormal histone modifications in PBMCs from patients with psoriasis vulgaris.
        Eur J Dermatol. 2011; 21: 552-557