Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 2.
Published in final edited form as: J Allergy Clin Immunol. 2018 May 24;143(2):604–618. doi: 10.1016/j.jaci.2018.03.021

Ichthyosis molecular fingerprinting shows profound TH17 skewing and a unique barrier genomic signature

Kunal Malik a,*, Helen He a,*, Thy Nhat Huynh c, Gary Tran c, Kelly Mueller c, Kristina Doytcheva c, Yael Renert-Yuval d, Tali Czarnowicki a,b, Shai Magidi a, Margaret Chou a, Yeriel D Estrada a, Huei-Chi Wen a, Xiangyu Peng a, Hui Xu a, Xiuzhong Zheng b, James G Krueger b, Amy S Paller c,*, Emma Guttman-Yassky a,b,*
PMCID: PMC7195861  NIHMSID: NIHMS1057361  PMID: 29803800

Abstract

Background

Ichthyoses are a group of rare skin disorders lacking effective treatments. Although genetic mutations are progressively delineated, comprehensive molecular phenotyping of ichthyotic skin could suggest much-needed pathogenesis-based therapy.

Objective

We sought to profile the molecular fingerprint of the most common orphan ichthyoses.

Methods

Gene, protein, and serum studies were performed on skin and blood samples from 29 patients (congenital ichthyosiform erythroderma, n = 9; lamellar ichthyosis, n = 8; epidermolytic ichthyosis, n = 8; and Netherton syndrome, n = 4), as well as age-matched healthy control subjects (n = 14), patients with psoriasis (n = 30), and patients with atopic dermatitis (AD; n = 16).

Results

Using criteria of a fold change of greater than 2 and a false discovery rate of less than 0.05, 132 differentially expressed genes were shared commonly among all ichthyoses, including many IL-17 and TNF-α–coregulated genes, which are considered hallmarks of psoriasis (defensin beta 4A, kynureninase, and vanin 3). Although striking upregulation of TH17 pathway genes (IL17F and IL36B/G) resembling that seen in patients with psoriasis was common to all patients with ichthyoses in a severity-related manner, patients with Netherton syndrome showed the greatest T-cell activation(inducible costimulator [ICOS]) and a broader immune phenotype with TH1/IFN-γ, OASL, and TH2/IL-4 receptor/IL-5 skewing, although less than seen in patients with AD (all P < .05). Ichthyoses lacked the epidermal differentiation and tight junction alterations of patients with AD (loricrin, filaggrin, and claudin 1) but showed characteristic alterations in lipid metabolism genes (ELOVL fatty acid elongase 3 and galanin), with parallel reductions in extracellular lipids and corneocyte compaction in all ichthyoses except epidermolytic ichthyosis, suggesting phenotypic variations. Transepidermal water loss, a functional barrier measure, significantly correlated with IL-17–regulated gene expression (IL17F and IL36A/IL36B/IL36G).

Conclusion

Similar to patients with AD and psoriasis, in whom cytokinedysregulation and barrier impairment orchestrate disease phenotypes, psoriasis-like immune dysregulation and lipid alterations characterize the ichthyoses. These data support the testing of IL-17/IL-36–targeted therapeutics for patients with ichthyosis similar to those used in patients with psoriasis.

Keywords: Psoriasis, atopic dermatitis, immune, inflammation, epidermal barrier, Netherton syndrome, congenital ichthyosiform erythroderma, lamellar ichthyosis, epidermolytic, ichthyosis, IL-17, TNF-α, IL-36, targeted therapy, precision medicine


The ichthyoses are a heterogeneous group of rare genetic, debilitating cutaneous disorders14 that share defective keratinization, a compromised epidermal barrier, and increased transepidermal water loss (TEWL).2, 3, 5 Patients present with xerosis, hyperkeratosis, often severe erythema, and reduced quality of life.610

Current treatments lack specificity and efficacy and are largely limited to topical emollients, keratolytics, and oral retinoids,811 all with potential toxicity.3, 1215 Although corrective gene therapy is evolving, it is still experimental and limited to model systems.16, 17 Thus an unmet need exists for pathogenesis-based therapeutic approaches.

Data derived from animal or human models suggest abnormalities in cornification, lipid homeostasis, and keratinocyte adhesion/desquamation among various ichthyoses, implying shared defects in barrier architecture.1826 Human skin and blood studies have been limited to a few patients or select ichthyosis subtypes and have shown abnormalities in lipid, cornified envelope (CE), and/or other differentiation measures.18, 2740

Several observations link ichthyoses, particularly Netherton syndrome (NS), to atopic dermatitis (AD), which is marked by epidermal barrier defects and immune dysregulation.41 NS shares the eosinophilia and increased TH2/atopy-related markers (thymic stromal lymphopoietin [TSLP], chemokine like 17 [CCL17]/thymus and activation-regulated chemokine, and IgE) of AD,18, 20, 4249 and polymorphisms of serine protease inhibitor, Kazal-type 5 (mutated in patients with NS) are associated with AD.5052 Moreover, clinical improvement in association with reduced TH2 marker expression has been noted in single cases of NS treated with omalizumab/anti-IgE44, 49 and infliximab/anti–TNF-α,49 implying potential reversibility of ichthyosis through immune-targeted treatments.

Recently, using a limited RT-PCR panel, our group showed enhanced TH17/IL-23 responses in patients with ichthyoses with minimal TH2 inflammation and preserved expression of the terminal differentiation markers filaggrin (FLG), loricrin(LOR), periplakin (PPL).53 These data linked the ichthyoses with psoriasis, a disease with a proved TH17/IL-23–centered pathogenesis.5456 Variable increases in levels of inflammatory cytokines (IL-17, IL-23, IL-8, IL-1β, TNF-α, CCL20, S100As, CCL17/thymus and activation-regulated chemokine, and TSLP) have been reported in patients with ichthyoses21, 46, 48, 49, 5761 but only in limited animal model studies,46, 6062 keratinocyte cultures,21, 25 or specific markers of ichthyosis subtypes in human subjects.48, 49, 58, 59 Thus a global comprehensive molecular phenotyping across the ichthyoses is warranted.

The present study is the first detailed genomic and cellular profiling of the most common orphan forms of ichthyosis(NS, autosomal recessive congenital ichthyosis [ARCI] subtypes of lamellar ichthyosis [LI] and congenital ichthyosiform erythroderma [CIE], and epidermolytic ichthyosis [EI]) compared with control subjects. To identify whether ichthyosis bears similarity to either AD (TH2 skewed) or psoriasis(TH17/IL-23 polarized), we included these common diseases as comparators. Our fingerprinting showed ichthyotic skin to harbor robust TH17/IL-23 skewing and activation of genes considered to be molecular classifiers of psoriasis (kynureninase [KYNU], vanin 1 [VNN1], and vanin 3 [VNN3]). Patients with ichthyoses showed less TH2 expression than patients with AD (and in many cases less than control subjects) and lacked its characteristic differentiation abnormalities. Significant correlation between disease severity and IL-17activation in skin and blood suggests systemic TH17 activation and further supports the use of systemically administered IL-17 antagonists as therapy.

Methods

Patients’ characteristics and samples

Institutional review board–approved consent was provided by subjects (≥12 years) and parents (<18 years), and demographics, medical history, physical examination, clinical severity scores (Ichthyosis Area and Severity Index [IASI], including Ichthyosis Area and Severity Index–erythema [IASI-E], and Ichthyosis Area and Severity Index–scaling [IASI-S]), pruritus scores (5-D itch and Numeric Rating Scale), and TEWL were obtained, as reported.53 The IASI score consists of erythema (IASI-E) and scaling (IASI-S) subscores and quantifies score intensity within body regions.53 Four-millimeter biopsy specimens and sera were obtained (see details in the Methods section in this article’s Online Repository at www.jacionline.org). Because patients with ichthyosis were significantly younger than those in the psoriasis and AD groups, a sensitivity analysis was performed, confirming no significant differences in results when adjusting for age (data not shown; Table I and see the Methods section in this article’s Online Repository).

Table I.

Patients’ demographics

Parameter Control subjects (n = 21) Patients with ichthyosis (n = 29) P value, control subjects vs patients with ichthyosis Patients with NS (n = 4) Patients with LI (n = 8) Patients with CIE (n = 9) Patients with EI (n = 8) P value, subtypes
Age (y) Mean ± SD 26.1 ± 20.4 21.2 ± 15.4 .362 15.2 ± 7.4 27.7 ± 15.6 19.5 ± 15.6 19.7 ± 18.3 .551
(range) (6.4-57) (2.2-56.9) (6.3-23) (9.2-56.9) (4.0-44) (2.2-55)

Sex F/M 10/11 16/13 .810 2/2 6/2 3/6 5/3 .358

Race EA/AA/As/H 16/1/0/4 20/3/3/3 .443 2/1/0/1 6/1/0/1 6/1/2/0 6/0/1/1 .590

IASI score Mean (±SD) NA 27.9 ± 8.4 NA 31.0 ± 10.8 29.2 ± 7.5 23.5 ± 9.2 30.4 ± 6.8 .284

AA, African American; As, Asian; EA, European American; F, female; H, Hispanic; M, male; NA, not applicable.

Gene-expression analyses

HGU133Plus2.0 microarrays (GSE#108640; Affymetrix, Santa Clara, Calif) were used for gene arrays, as previously described.63 RNA was extracted for RT-PCR with the Qiagen miRNeasy Mini Kit (Qiagen, Valencia, Calif), as previously described.64, 65 Expression values were normalized to human acidic ribosomal protein. Primers and probes are listed in Table E1 in this article’s Online Repository at www.jacionline.org. See the Methods section in this article’s Online Repository for details.

Serum immunoassays

Electrochemiluminescence immunoassays (Singulex, Alameda, Calif) were used to quantify IL-17A in sera from patients with ichthyosis, as previously described.66

Immunostaining and immunofluorescence

Immunohistochemistry was performed on frozen sections by using the listed antibodies (see Table E2 in this article’s Online Repository at www.jacionline.org), as previously described.67Immunofluorescence staining for neutral lipids with Nile red was performed, as previously eported.68, 69 See the Methods section in this article’s Online Repository for details.

Statistical analysis

Statistical analysis was performed with R software (www.R-project.org) and packages available through Bioconductor(www.bioconductor.org). Quality control was performed, as previously described.63 Expression values were obtained by using the GC Robust Multiarray Average algorithm. Batch effects were removed by identifying and adjusting for surrogate variables with the sva package.70 Probe sets with 15 or more samples and expression values of greater than 3 were kept for analysis. Data were log2-transformed and fitted with a linear model. Fold changes (FCHs) between comparisons of interest were estimated, and hypothesis testing was conducted by using contrasts under the general framework for linear models in the R software limma package. P values from the moderated (paired) t test were adjusted for multiple hypotheses by using the Benjamini-Hochberg procedure. Genes with FCHs of greater than 2 and a false discovery rate (FDR) of less than 0.05 were considered differentially expressed. Hierarchical clustering of samples/conditions used a McQuitty agglomeration algorithm. Gene-set variance analysis was performed by using unsupervised sample-wise enrichment.71 Gene-set overrepresentation analysis was performed with XGR software72 and crowd-extracted expression of differential signature disease signatures.73

All 29 samples from patients with ichthyoses, 21 control subjects, and 10 patients with psoriasis and AD were available for RT-PCR analyses. mRNA expression RT-PCR data were log2-transformed and fitted to a linear model as above. Values of less than the limit of detection were imputed as 20% of the minimal value over the limit of detection, as previously eported.74 See the Methods section in this article’s Online Repository for details.

Results

Twenty-nine patients (age, 2.2-56.9 years; mean age, 21.2 years; 16 female and 13 male patients) with ichthyosis and known mutations, including 4 with NS, 8 with ARCI-LI, 9 with ARCI-CIE, and 8 with EI, were enrolled (Table I and see Table E3 in this article’s Online Repository at www.jacionline.org). Healthy control subjects (n = 21) were between 6.4 and 57 years old (mean age, 26.1 years). Data from previous studies on 16 patients with moderate-to-severe AD (age, 33-69 years; mean age, 49.8 years; mean SCORAD score, 61.2)63 and 30 patients with psoriasis (age, 33–67 years; mean age, 48.5 years; mean Psoriasis Area and Severity Index score, 23.6)75 served as comparators.

Genomic profiling identifies the common and individual ichthyosis phenotypes

Gene arrays were performed on 21 of the 29 patients with ichthyoses (6 patients with ARCI-CIE, 7 patients with ARCI-LI, 5 patients with EI, and 3 patients with NS) and 14 healthy age-matched control subjects.53 Although a principal component analysis of microarray data demonstrated considerable overlap among the ichthyosis subtypes, it also distinguished between individual subtypes and healthy control subjects, which clustered separately (see Fig E1 in this article’s Online Repository at www.jacionline.org). Using criteria of an FCH of greater than 2 and an FDR of less than 0.05, we identified the common ichthyosis molecular phenotype, consisting of a core set of 132 differentially expressed genes (DEGs; 113 upregulated and 19 downregulated genes), as well as unique genomic signatures of each subtype (Fig 1, A). Among upregulated genes in the common ichthyosis transcriptome were many psoriasis hallmark genes, including Defensin beta4A (DEFB4A), kynureninase (KYNU), VNN3, kallikrein 8 (KLK8), late cornified envelope 3D (LCE3D), inducible nitric oxide synthase regulator gene arginase 1, the NOD-like receptor gene caspase recruitment domain family member 6 (CARD6), and the novel psoriasis autoantigen phospholipase A2, group 4D (PLA2G4D) (Fig 1, A, and see Table E4 in this article’s Online Repository at www.jacionline.org).76, 77

Fig 1.

Fig 1.

A, Venn diagram showing DEGs comparing ichthyosis subtypes: NS, LI, CIE, and EI. The box represents the common ichthyosis molecular phenotype. B, Heat map representing DEGs upregulated and downregulated in the common ichthyosis phenotype, with a table of fold changes of these genes in patients with psoriasis (PSO), AD, and ichthyosis subtypes. C, Venn diagram depicting how gene alterations of the common ichthyosis phenotype shown to be shared in Fig 1, A, overlap with the molecular signatures of AD and psoriasis. Red, Upregulated; blue, downregulated. +P < .1, *P < .05, **P < .01, and ***P < .001.

However, many DEGs differed among the unique subtypes. To assess these differences, we evaluated uniquely expressed DEGs (Fig 1, A). Of all ichthyoses, patients with EI expressed the greatest number of unique DEGs (223 upregulated and 150 downregulated genes), with upregulation in KLKs (KLK7 and KLK10), keratins (KRT4, KRT72, and KRT9), cell cycle/proliferation (cell division cycle 6/20/25A), and cell transport molecules (dynein axonemal assembly factor 3, clathrin interactor 1, and cilia and flagella associated protein 97). Two hundred thirty-four DEGs were unique to patients with LI, 61 of which were upregulated, including the psoriasis-associated cytokine IL-20 and IL-17–related genes (IL36A and lipocalin 2 [LCN2]). Downregulated DEGs in patients with LI consisted of lipid metabolism (arachidonate 15-lipoxygenase, type B, Acyl-CoA wax alcohol acyltransferase 1, and fatty acid binding protein 7 [FABP7]), TH1 (CXCL9/CXCL11), and TH2 (CCL8/CCL17 and IL31RA) markers, as well keratins (KRT35/KRT40/KRT75/KRT81/KRT83/KRT85/KRT86) and keratin-associated protein 1. One hundred fifty-eight DEGs were unique to patients with CIE, including upregulation of the junctional protein desmocollin 1 and downregulation of metalloproteases (ADAM metallopeptidase domain 33 and matrix metallopeptidase [MMP] 9), aquaporin 1(AQP1), collagens (COL14A1), and integrins (ITGA11 and ITGA5). Patients with NS expressed the fewest unique DEGs (63 upregulated and 33 downregulated DEGs), including upregulation of microtubule-associated proteins (MAP4/7), the interferon-induced molecule 2′-5′-Oligoadenylate synthetase 3, and the TNF receptor superfamily member 11a (see Table E4).

We further assessed how different subtypes vary in their inflammatory profiles through analysis of a previously published immune gene subset63, 67, 78 represented in a heat map (see Fig E2 in this article’s Online Repository at www.jacionline.org). Relative to other ichthyoses, patients with NS demonstrated the greatest upregulation of T-cell activation markers (MMP12, inducible costimulator [ICOS], and C-C motif chemokine receptor 7), interferon/TH1 products (IFNG and 2′−5′-oligoadenylate synthetase-like protein [OASL]), and TH2 (IL4R, IL5, IL13, and CCL18/CCL26) markers, although increases in the latter group were much smaller than in patients with AD. TH17 (PI3, S100A9/12, and IL17A/F) measures were upregulated in all patients with ichthyoses, with the greatest increases in patients with NS; CCL20 and LCN2 were exceptions and greatest in patients with LI (see Fig E2).

The common upregulated ichthyosis profile overlaps with psoriasis

We compared the global ichthyosis profile with that of psoriasis and AD, which can clinically resemble ichthyoses.42, 47, 48, 49, 51, 52 As shown in an unsupervised hierarchical clustering heat map of the top 100 upregulated ichthyosis DEGs (Fig 1, B, and by individual samples in Fig E3 and Table E4 in this article’s Online Repository at www.jacionline.org), there is a striking similarity to psoriasis and little similarity to AD. Upregulated genes in patients with ichthyosis included IL-17/TNF-α–associated molecules (IL36B/IL36G, peptidase inhibitor 3 [PI3], and S100A7A/S100A9/S100A12), with many showing psoriasis-level expression (Fig 1, B). The intersection of the common ichthyosis lesional phenotype with AD and psoriasis signatures is represented in Fig 1, C, as a Venn diagram. When compared with normal skin, 37 DEGs were shared among all diseases, including upregulation of CARD6, DEFB4A, IL36G, LCE3D, PI3, S100A9/12, and VNN3. Notably, the common ichthyosis molecular phenotype demonstrated larger overlap with psoriasis than AD, with psoriasis sharing 92 (69%) of 132 DEGs and AD sharing only 38 (28%) of 132 DEGs. Only ichthyosis and psoriasis showed unique upregulation in KLK8, KYNU, and PLA2G4D (see Table E5 in this article’s Online Repository at www.jacionline.org).

An IL-17/TNF-α gene signature is shared among ichthyosis subtypes

Because an IL-17–skewed pattern was shared across ichthyoses and many IL-17–related genes are synergistically/additively induced by IL-17/TNF-α, we evaluated individual ichthyoses for a subset of IL-17– and TNF-α–regulated genes (Fig 2). All ichthyosis subtypes demonstrated highly significant increases in IL-17/TNF-α hallmark psoriasis genes (KYNU, VNN3, Wiskott-Aldrich syndrome like, dedicator of cytokinesis 9, and kinesin family member 1B; Fig 2); these were not as highly upregulated or were even downregulated in patients with AD.

FIG 2.

FIG 2.

Heat map of additive and synergistic IL-17/TNF-a–induced genes across ichthyosis subtypes (NS, LI, CIE, and EI), with fold changes also depicted for psoriasis (PSO) and AD lesions versus normal skin. Red, Upregulated; blue, downregulated. 1P < .1, *P < .05, **P < .01, and ***P < .001

To validate the array data, we also measured mRNA expression of representative IL-17/TNF-α–regulated genes using quantitative PCR/RT-PCR in the 29 patients with ichthyoses. As seen in the box plots in Fig 3, A, all subtypes showed similar highly significant increases in expression of IL-17/TNF-α–induced gene mRNA (IL17F, IL36A/IL36B/IL36G, KYNU, epiregulin [EREG], and VNN1/3) to psoriasis (P < .05 for all). A heat map depicting FCHs of mRNA expressions in skin of patients with lesional ichthyosis versus normal skin (see Fig E4in this article’s Online Repository at www.jacionline.org) also shows overall greater similarity to psoriasis but not AD.

Fig 3.

Fig 3.

A, Real-time/RT-PCR–derived mRNA expression of TH17/TNF-α–induced cytokines and markers in normal (N), psoriasis (PSO), AD, NS, LI, CIE, and EI lesions versus normal skin represented as box plots. Red dots indicate means, black dots indicate individual samples, horizontal black lines indicate medians, and vertical black lines indicate values within 1.5 times the interquartile range of the median; the top and bottom of the boxes indicate 75th and 25th percentiles, respectively. P values above horizontal lines denote significance among groups. B-H,Immunohistochemistry staining of IL-36R in normal (NRML), AD, and psoriasis (PSO) lesions compared with CIE, LI, EI, and NS tissues (×10 magnification). Scale bars = 100 μm.

IL-36α, IL-36β, and IL-36γ subunits exert their proinflammatory effects through binding

IL-36 receptor (IL-36R), which activates intracellular signaling pathways.79 To extend our findings, we analyzed IL-36R protein expression through immunohistochemistry. IL-36R is known to form feedback loops driving IL-17 cytokine production in patients with psoriasis.80, 81 Although in normal (Fig 3, B) and AD lesions (Fig 3, C) IL-36R showed minimal epidermal expression, widespread expression of IL-36R was noted in psoriatic epidermis (Fig 3, D) and patients with all types of ichthyoses (Fig 3, EH).

Different ichthyosis subtypes show unique epidermal differentiation and lipid genomic alterations

Ichthyoses were historically considered disorders of lipid transport and epidermal barrier alterations. Hence we assessed genes relating to the epidermal differentiation complex, CE, and lipids across each subtype, as reported in a heat map (Fig 4, A). Although patients with AD showed significant downregulation of differentiation products versus control subjects (LOR, FLG, PPL, envoplakin, corneodesmin, late cornified envelope 1B/E, and annexin A9 [ANXA9]),64 ichthyosis showed minimal downregulation of differentiation products, with some measures even showing upregulation (EREG, ANXA9, psoriasis susceptibility 1 candidate 2, LOR, and FLG) versus normal skin (Fig 4, A, yellow box). Significant decreases in claudins (CLDNs; CLDN1/8/23) were seen in patients with AD and those with psoriasis, with variable and less significant reductions across the ichthyoses (Fig 4, A). Lipids were most prominently decreased in patients with AD but were also downregulated in those with psoriasis. Among ichthyoses, patients with LI demonstrated the greatest and most significant lipid downregulation (ELOVL fatty acid elongase 3, galanin [GAL], hydroxyacid oxidase 2, and FABP7), whereas patients with EI generally showed preserved expression of lipid-associated genes (Fig 4, A, green box).

Fig 4.

Fig 4.

A, Heat map of epidermal differentiation complex (EDC), CE, and lipid genes expressed across normal skin (N)and skin of patients with psoriasis (PSO), AD, and ichthyoses(NS, LI, CIE, and EI), showing FCHs versus normal skin. Greenand yellow boxes indicate gene sets displaying differences in patients with ichthyoses from those with the other conditions. Red, Upregulated; blue, downregulated. +P < .1, *P < .05, **P < .01, ***P < .001. B-H, Lipid staining using Nile red immunofluorescence in normal (NRML), AD, and psoriasis (PSO) lesions compared with tissue from patients with CIE, LI, EI, and NS, with insets showing decreased corneocyte compaction and reduced extracellular lipids (white arrows; ×20 magnification).

To better visualize stratum corneum (SC) intercellular lipids and validate array findings, we performed Nile red immunofluorescence staining in skin from patients with ichthyoses, patients with AD, patients with psoriasis, and healthy subjects (Fig 4, BH). Although control skin (Fig 4, B) was characterized by extracellular lipids organized in lamellar sheets between compacted corneocytes, patients with ichthyoses (Fig 4, EH) showed variable patterns in extracellular lipid layers. Patients with CIE and those with NS showed similar lipid staining as control subjects with compact corneocytes and prominent lipid lamellae, whereas patients with EI demonstrated less distinct lamellar layering and foci of greater intensity (“lacunae-like” defects). Consistent with its greatest downregulation of lipid genes among ichthyoses (Fig 4, A), patients with LI showed diminished corneocyte compaction, visibly reduced lipid lamellae, and intensely staining intracellular, globular lipid-like structures. Qualitatively, patients with AD demonstrated the most profound decreases in lipid layers and SC compaction (Fig 4, C), whereas patients with psoriasis (Fig 4, D) revealed more compact SC and less profound lipid defects.

Pathway analysis highlights similarities to psoriasis

We also performed gene-set variation analysis using previously defined gene lists for IL-17–regulated immune axes.67 This is represented as bar plots depicting increases in IL-17–upregulated and synergistic/additive IL-17/TNF-α genes in patients with all types of ichthyoses, most profoundly in patients with NS and psoriasis compared with patients with AD and control subjects, which showed smaller activation of these gene sets (P < .05 for all; Fig 5, A and B). Expression of CE genes was decreased significantly in patients with AD but relatively unchanged or even increased in patients with psoriasis and ichthyoses (Fig 5, C). Lipid genes (Fig 5, D) were reduced significantly in patients with AD (P < .001) and, to a lesser extent, in patients with ichthyoses, except EI, which resembled a lipid profile of normal skin.

Fig 5.

Fig 5.

A-D, Gene-set variance analyses (GSVA) for subsets of genes upregulated with IL-17 (Fig 5, A), genes synergistically and additively induced by IL-17/TNF-α (Fig 5, B), CE genes (Fig 5, C), and lipid metabolism genes (Fig 5, D). Expression is represented as a GSVA score for normal (N) skin and skin from patients with psoriasis (PSO), AD, NS, LI, CIE, and EI. Means ± SEMs are shown. P values above horizontal lines denote significance among groups. +P < .1, *P < .05, **P < .01, and ***P < .001. E, Disease signature enrichment analyses of upregulated genes in ichthyosis lesions versus control lesions performed against curated disease signature sets from the Broad Institute. Top representative hits of polarized inflammatory/immune diseases are represented as a bar plot, and all outputs of the enrichment analyses are presented in Table E9. The green line indicates a false discovery rate cutoff of 0.05.

Pathway enrichment analyses using function-based pathway databases (KEGG, REACTOME, Biocarta, and the Pathway Interaction Database; see Tables E6E10 in this article’s Online Repository at www.jacionline.org) revealed significantly upregulated pathways in the common and individual ichthyosis signatures (FDR < 0.05). These included focal adhesion and mitogen-activated protein kinase signaling for patients with CIE; cyclins, cell-cycle regulation, and lipoprotein metabolism for patients with EI; chemokine signaling and extracellular matrix for patients with LI; and extracellular matrix for patients with NS. We further evaluated gene enrichment of the ichthyosis phenotype with available signatures of other polar inflammatory diseases (see Table E11 in this article’s Online Repository at www.jacionline.org).73 Ichthyosis showed the most significant overlap with the psoriasis vulgaris fingerprint but also shared its profile with other TH17-mediated dermatoses (acne) and TH2 conditions (AD; Fig 5, E).82

IL-17– and TNF-α–regulated genes correlate with ichthyosis severity and epidermal barrier disruption

To determine how molecular and cellular biomarkers in the skin of patients with ichthyosis correlated with clinical severity scores (IASI and its subscores) and epidermal barrier disruption (TEWL), we performed Spearman correlation coefficient analysis presented as correlation plots in Fig 6, A and B, and Fig E4. The highest correlations with the inflammatory component of IASI (IASI-E) included IL-17/TNF-α–regulated genes, such as IL17F (r = 0.71, P = 9.54 × 10−6), IL36G (r = 0.62, P = .0002), KYNU (r = 0.57, P = .001), and KLK13 (r = 0.57, P = .001; Fig 6, A, and see Fig E5 and Table E11 in this article’s Online Repository at www.jacionline.org). Highly significant correlations were also noted between select markers and total IASI scores (IL17F and KLK13, P < .05) and TEWL levels (IL17F, EREG, IL36A, IL36B, IL36G, KLK13, and VNN3; all P < .01; Fig 6, A, and see Fig E5).

Fig 6.

Fig 6.

A, Representative Spearman correlations between real-time/RT-PCR–derived mRNA expression of skin markers with IASI-E and TEWL. Dots represent ichthyosis subtypes: orange dots, CIE; green dots, EI; blue dots, LI; and purple dots, NS. B,Serum IL-17A levels in normal (N) and ichthyosis (ICH) serum represented as box plots. The mean of each group is shown as a large red dot, black dots are individual samples, horizontal black lines indicate medians, vertical black lines indicate values within 1.5 times the interquartile range of the median, and the top and bottom boundaries of the boxes indicate 75th and 25th percentiles, respectively. P values above horizontal lines denote significance among groups. C, Spearman correlation heat map of clinical (IASI-E, IASI total, IASI-S, pruritus, and TEWL), transcriptional, and serum immune markers. The black boxdemonstrates groups of tightly clustered markers/parameters. +P < .1, *P < .05, **P < .01, and ***P < .001.

We also assessed IL-17A levels in serum. IL-17A activation extended to the blood compartment, with highly significant increases in serum from patients with ichthyosis versus control subjects (P < .001; Fig 6, B, and see Fig E6 in this article’s Online Repository at www.jacionline.org) and the highest increases in patients with NS (P < .01), highlighting its high-grade immune activation.

A Spearman rank correlation coefficient heat map of all clinical parameters (IASI, IASI-E, IASI-S, pruritus, and TEWL) in patients with ichthyosis compared with skin mRNA and serum expression is presented in Fig 6, C(red, positive correlations; blue, negative correlations; color intensity reflects correlative strength). A black box demonstrates IL-17−associated markers in ichthyotic skin (KYNU, VNN1/3, IL36A/G, and IL17F) and serum IL-17A clustering tightly with disease severity (IASI-E), whereas epidermal dysfunction (TEWL) correlated most with EREG and IL36B mRNA. Individual correlations are shown in Table E12 in this article’s Online Repository at www.jacionline.org.

Discussion

Disrupted terminal differentiation, lipid homeostasis, increased TEWL, and associations with atopy have historically linked ichthyoses to AD.2, 3, 18, 2741, 43, 5052, 8386 However, our recent profiling of ichthyotic skin using a limited gene-expression approach demonstrated enhanced TH17/IL-23 response with marginal TH2 skewing and lack of abnormalities in 3 differentiation proteins (FLG, LOR, and PPL), aligning it more closely to the psoriasis profile.53

The present study is the first comprehensive genomic skin fingerprinting of the most common orphanichthyosis subtypes to date, with comparisons to tissue from healthy control subjects, patients with AD, and patients with psoriasis. Our data further establish a predominant TH17/IL-23 and IL-36 signature underlying the common and individual ichthyosis phenotypes, expanding on our prior report53 and profiling of single patients with erythrokeratodermia-cardiomyopathy (mutation in desmoplakin [DSP])87 and ichthyosis Curth-Macklin (keratin 1 [KRT1]).88 TNF-αantagonism is beneficial in patients with psoriasis, likely because of its effects on IL-17/TNF-α synergistically/additively induced genes rather than on TNF-α–regulated genes alone.89, 90 Here we show a striking increase in expression of IL-17/TNF-α synergistic genes, including those representing molecular hallmarks of psoriasis (KYNU, VNN3, IL36B/G, and EREG) across ichthyoses and particularly NS. Furthermore, the expression of these IL-17/TNF-α–regulated genes significantly correlated with sores on IASI-E, the inflammatory component of ichthyosis severity. Among these psoriasis classifiers are (1) KYNU, which is involved in tryptophan metabolism91; (2) VNN3, expression of which is increased by TH17/TH1 but not by TH2 cytokines,92, 93 as suggested by its increased expression in patients with ichthyosis and psoriasis but not patients with AD; and (3) IL36A/IL36B/IL36G, proinflammatory cytokinescoregulated in keratinocytes by IL-17/TNF-α94 that amplify IL-17 cytokine production80, 81 and are overexpressed in psoriatic skin,95, 96 discriminating it from AD skin.53, 80, 81, 97 Our study shows increased mRNA and protein expression of IL-36 family members and IL-36R and highly significant correlations with ichthyosis severity and TEWL, further suggesting IL-36/IL-36R involvement in the TH17 axis in the ichthyoses, similar to psoriasis.44, 48, 49, 58, 59, 96 Our data also extend the prominent severity-associated TH17 profile in ichthyosis to the blood compartment, supporting an accentuated systemic inflammatory phenotype, particularly in patients with NS.

Beyond the common ichthyosis phenotype, the present study is the first to highlight the unique genomic signatures of each ichthyosis variant. NS showed the greatest increases in inflammatory pathways and particularly those related to TH17 versus other subtypes. In mouse NS models a few TH17 markers showed increases,20, 60 and in 1 patient with NS, TH17 markers were reduced with TNF-α antagonism and clinical improvement.49 Patients with NS carry serine protease inhibitor, Kazal-type 5 mutations, leading to unopposed KLK5–protease-activated receptor 2–TSLP activation and induction of TH2 and TNF-α, an identified gene target of KLK5–protease-activated receptor 2 signaling.46, 47, 98 Contrary to patients with NS, which has been linked classically to the atopic diathesis18, 46, 48, 5052, 60, 86 and harbors TH2 and pronounced TH17 activation, TH2 skewing was largely absent in other ichthyoses. This might be attributable to reciprocal regulation of the TH17 and TH2 axes99101 or might be secondary to KYNU metabolites, which were implicated in TH2 downregulation.91 Hence TH17 upregulation in patients with ichthyoses might be the outcome of complex interactions between different arms of adaptive immunity that extend to the systemic level, as evidenced in our report.

Ichthyoses have barrier defects,2, 5, 53, 102 as reflected by increased TEWL,2, 3, 5, 18, 28, 30, 31, 53, 102 and many forms have defective lamellar body transport and lipid homeostasis. Hence we comprehensively analyzed epidermal barrier genes, including those contributing to terminal differentiation, tight junctions (TJs), and lipid metabolism to dissect contributions of each component to impaired barrier function. A preserved differentiation profile was seen across ichthyoses, supporting our limited prior data and other past studies and possibly reflecting the relative lack of TH2-mediated suppression of epidermal differentiation.36, 60, 103105 CLDN1/CLDN8/CLDN23, which are crucial for TJ formation and highly suppressed in skin of patients with AD,64, 106 were variably downregulated in patients with ichthyoses, although less significantly than in those with AD and even psoriasis. IL-17 has been suggested to suppress TJs and adhesion molecules (including CLDN1/CLDN4 and occludin), perhaps explaining their reduced expression in patients with ichthyosis.107

Our gene profiling, corroborated by Nile red staining, also demonstrates unique abnormalities in SC lipid structure among the ichthyoses. Although overall lipid measures (FA2H, FABP7, and ELOVL3) were suppressed in all patients with ichthyoses (except those with EI) compared with control subjects, the greatest downregulation was found in patients with LI, which was even more pronounced than in patients with AD and psoriasis. Our patients with LI all harbored mutations in both alleles of TGM1, which encodes transglutaminase 1.108, 109 Transglutaminase 1 cross-links cutaneous ω-hydroxyceramides to terminal differentiation proteins, playing an important role in CE assembly.110, 111 Thus TGM1 inactivity in patients with LI might drive the LI barrier abnormalities in addition to the underlying inflammation/immune activation, particularly of TH17. Notably, decreased lipid levels were shown to upregulate psoriasis-like inflammation in mice (IL-17, IL-22, IL-23, S100As, and β-defensins),112 and preadipocytes treated with IL-17 have reduced lipid-related gene expression,113, 114 highlighting the need to further investigate the role of IL-17 in epidermal lipid defects in patients with ichthyosis.

Although ichthyosis and psoriasis have different primary causes, our data highlight the similarities between these 2 diseases. In patients with psoriasis, TH17 T cells become self-reactive to endogenous proteins,54, 55, 115126 and the IL-17 produced by T cells, along with IL-26 and IL-29, trigger major genomic changes in epidermal keratinocytes, including induction of cytokines (ie, IL-36) that drive the typical epidermal hyperplasia.93, 95, 117, 118, 125, 127129 Many other IL-17–induced products create feed-forward inflammatory responses, leading to chronic T-cell activation in the skin and sustained reactive responses of keratinocytes and other cells creating the psoriasis phenotype. IL-17 blockade with therapeutic antibodies resolves most of the clinical or molecular psoriasis phenotype.55, 120, 130, 131

In contrast, the ichthyoses are caused by genetic defects primarily in keratinocyte structural genes or enzymes, leading to barrier abnormalities that are most often present at birth. In patients with ichthyosis, initial triggering of IL-17–dominated inflammatory responses stems presumably from exposure to microbes or other antigens through a disrupted barrier, which one predicts sustains immune activation by constant antigenic exposure. However, skin responses to IL-17 have the same potential to alter keratinocyte transcriptional/differentiation programs, and these can create feed-forward inflammatory loops, as in patients with psoriasis. We propose that the shared phenotypic features among ichthyosis subtypes result from primary barrier abnormalities, which in turn lead to cutaneous molecular and structural responses to IL-17–centered inflammation with hyperplasia and TJ/lipid changes. Given that so many patients with known genetic defects in the proteins/lipids that are critical to normal barrier function show shared IL-17/IL-23 skewing, it would be unlikely that the explanation is an additional primary underlying genetic abnormality of innate immunity. The extent to which IL-17–centered inflammation contributes to overall disease pathogenesis and clinical disease can only be determined by means of direct antagonism of IL-17 or upstream regulators. Recently, we reported 2 cases of syndromic ichthyoses from a primary defect in DSP(erythrokeratodermia-cardiomyopathy and severe dermatitis, multiple allergies, and metabolic wasting syndromes) treated successfully with IL-12/23p40 antagonism87 with improved ichthyosis severity (both erythroderma and scaling) and TEWL, suggesting that TH17/IL-23 activation could contribute to disease pathogenesis. Our data form the basis for an ongoing clinical trial ( NCT03041038) to test the therapeutic efficacy of IL-17A antagonism in patients with ichthyosis, which can better dissect the complex relationship of IL-17 and the underlying barrier defects.

Our study has several limitations. First, although this is the largest characterization of ichthyosis subtypes, we acknowledge the limited sample size because of disease rarity. Second, we could only screen for IL-17A in sera from patients with ichthyosis, suggesting the need for a more comprehensive investigation of systemic TH17 activation in patients with ichthyosis.

In summary, ichthyotic skin and serum have strong TH17 skewing, with an increase in markers coregulated by IL-17/TNF-α, preserved epidermal differentiation, and downregulation of lipid metabolism and TJ genes best resembling psoriasis. Our study offers new insights into the possible role of the IL-17 pathway in perpetuating the barrier defects of ichthyosis and advocates for testing of IL-17, IL-36,96 and IL-23 antagonism in patients with these diseases. Such studies will not only help dissect disease pathogenesis but might also provide hope for better treatment for these devastating disorders.

Supplementary Material

Malik Suppl FigE1
Malik Suppl TabE4
Malik Suppl TabE5
Malik Suppl TabE6
Malik Suppl TabE7
Malik Suppl TabE8
Malik Suppl TabE9
Malik Suppl txt and STab E1-3
Malik Suppl FigE2
Malik Suppl FigE3
Malik Suppl FigE4
Malik Suppl FigE5
Malik Suppl FigE6
Malik Suppl TabE10
Malik Suppl TabE11
Malik Suppl TabE12

Clinical implications.

Our report offers novel insights into the role of IL-17/IL-36 responses and unique barrier alterations in ichthyosis pathogenesis, advocating for directed immune antagonism in these patients. This comprehensive molecular phenotyping could direct much-needed targeted therapeutics for these diseases.

Acknowledgments

Supported by the Foglia Family Foundation Endowment. We acknowledge Core resources provided by the Northwestern University Skin Disease Research Center (NIAMS P30AR057216). Genetic testing of the majority of patients was performed in the laboratory of Dr. Keith Choate, Yale University.

Abbreviations used

AD

Atopic dermatitis

ARCI

Autosomal recessive congenital ichthyosis

CCL17

Chemokine like 17

CE

Cornified envelope

CIE

Congenital ichthyosiform erythroderma

CLDN

Claudin

DEG

Differentially expressed gene

EI

Epidermolytic ichthyosis

EREG

Epiregulin

FCH

Fold change

FDR

False discovery rate

FLG

Filaggrin

IASI

Ichthyosis Area and Severity Index

IASI-E

Ichthyosis Area and Severity Index–erythema

IASI-S

Ichthyosis Area and Severity Index–scaling

IL-36R

IL-36 receptor

KLK

Kallikrein

KYNU

Kynureninase

LI

Lamellar ichthyosis

LOR

Loricrin

NS

Netherton syndrome

OASL

29-59-oligoadenylate synthetase-like protein

PPL

Periplakin

SC

Stratum corneum

TEWL

Transepidermal water loss

TJ

Tight junction

TSLP

Thymic stromal lymphopoietin

VNN3

Vanin 3

Footnotes

Disclosure of potential conflict of interest: J. G. Krueger has received grants and or consultancy fees from Pfizer, Amgen, Janssen, Lilly, Merck, Novartis, Kadmon, Dermira, Boehringer, Innovaderm, Kyowa, BMS, Serono, BiogenIdec, Delenex, AbbVie, Sanofi, Baxter, Paraxel, Xenoport, and Kineta. A. S. Paller has received grants and/or consultancy fees from Amgen, Celgene, Dermira, Eli Lilly, GlaxoSmithKline-Stiefel, Novartis, Pfizer, Regeneron/Sanofi, Valeant, Galderma, Janssen, Anacor, and Vitae and grants from Astellas and LEO Pharma. E. Guttman-Yassky has received grants and/or consultancy fees from Celgene, Dermira, Janssen Biotech, LEO Pharma, Merck, Novartis, Regeneron/Sanofi, BMS, AbbVie, Amgen, Anacor, Celsus Therapeutics, Drais, Galderma, Genentech, Glenmark, Pfizer, Stiefel/GlaxoSmithKline, Vitae, Mitsubishi Tanabe, Eli Lilly, Allergan, and Escalier. The rest of the authors declare that they have no relevant conflicts of interest.

References

  • 1.Chamcheu JC, Siddiqui IA, Syed DN, Adhami VM, Liovic M, Mukhtar H. Keratin gene mutations in disorders of human skin and its appendages. Arch Biochem Biophys 2011;508:123–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Marukian NV, Choate KA. Recent advances in understanding ichthyosis pathogenesis. F1000Res 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.DiGiovanna JJ, Robinson-Bostom L. Ichthyosis: etiology, diagnosis, and management. Am J Clin Dermatol 2003;4:81–95. [DOI] [PubMed] [Google Scholar]
  • 4.Oji V, Tadini G, Akiyama M, Blanchet Bardon C, Bodemer C, Bourrat E, et al. Revised nomenclature and classification of inherited ichthyoses: results of the First Ichthyosis Consensus Conference in Soreze 2009. J Am Acad Dermatol 2010;63:607–41. [DOI] [PubMed] [Google Scholar]
  • 5.Elias PM, Williams ML, Holleran WM, Jiang YJ, Schmuth M. Pathogenesis of permeability barrier abnormalities in the ichthyoses: inherited disorders of lipid metabolism. J Lipid Res 2008;49:697–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Diaz LZ, Browning JC, Smidt AC, Rizzo WB, Levy ML. Complications of ichthyosis beyond the skin. Dermatol Ther 2013;26:39–45. [DOI] [PubMed] [Google Scholar]
  • 7.Ganemo A, Sjoden PO, Johansson E, Vahlquist A, Lindberg M. Health-related quality of life among patients with ichthyosis. Eur J Dermatol 2004;14:61–6. [PubMed] [Google Scholar]
  • 8.Styperek AR, Rice ZP, Kamalpour L, Pavlis M, Kuo J, Culler S, et al. Annual direct and indirect health costs of the congenital ichthyoses. Pediatr Dermatol 2010;27:325–36. [DOI] [PubMed] [Google Scholar]
  • 9.Mazereeuw-Hautier J, Dreyfus I, Barbarot S, Serrentino L, Bourdon-Lanoy E, Ezzedine K, et al. Factors influencing quality of life in patients with inherited ichthyosis: a qualitative study in adults using focus groups. Br J Dermatol 2012;166:646–8. [DOI] [PubMed] [Google Scholar]
  • 10.Ganemo A, Lindholm C, Lindberg M, Sjoden PO, Vahlquist A. Quality of life in adults with congenital ichthyosis. J Adv Nurs 2003;44:412–9. [DOI] [PubMed] [Google Scholar]
  • 11.Oji V, Traupe H. Ichthyosis: clinical manifestations and practical treatment options. Am J Clin Dermatol 2009;10:351–64. [DOI] [PubMed] [Google Scholar]
  • 12.DiGiovanna JJ. Isotretinoin effects on bone. J Am Acad Dermatol 2001; 45(suppl):S176–82. [DOI] [PubMed] [Google Scholar]
  • 13.Digiovanna JJ, Mauro T, Milstone LM, Schmuth M, Toro JR. Systemic retinoids in the management of ichthyoses and related skin types. Dermatol Ther 2013;26:26–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Saurat JH. Side effects of systemic retinoids and their clinical management. J Am Acad Dermatol 1992;27(suppl):S23–8. [DOI] [PubMed] [Google Scholar]
  • 15.Vahlquist A, Ganemo A, Virtanen M. Congenital ichthyosis: an overview of current and emerging therapies. Acta Derm Venereol 2008;88:4–14. [DOI] [PubMed] [Google Scholar]
  • 16.Akiyama M, Sugiyama-Nakagiri Y, Sakai K, McMillan JR, Goto M, Arita K, et al. Mutations in lipid transporter ABCA12 in harlequin ichthyosis and functional recovery by corrective gene transfer. J Clin Invest 2005;115: 1777–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Choate KA, Medalie DA, Morgan JR, Khavari PA. Corrective gene transfer in the human skin disorder lamellar ichthyosis. Nat Med 1996;2:1263–7. [DOI] [PubMed] [Google Scholar]
  • 18.Oji V, Traupe H. Ichthyoses: differential diagnosis and molecular genetics. Eur J Dermatol 2006;16:349–59. [PubMed] [Google Scholar]
  • 19.Descargues P, Deraison C, Bonnart C, Kreft M, Kishibe M, Ishida-Yamamoto A, et al. Spink5-deficient mice mimic Netherton syndrome through degradation of desmoglein 1 by epidermal protease hyperactivity. Nat Genet 2005;37:56–65. [DOI] [PubMed] [Google Scholar]
  • 20.Furio L, Pampalakis G, Michael IP, Nagy A, Sotiropoulou G, Hovnanian A. KLK5 inactivation reverses cutaneous hallmarks of Netherton syndrome. PLoS Genet 2015;11:e1005389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Blunder S, Ruhl R, Moosbrugger-Martinz V, Krimmel C, Geisler A, Zhu H, et al. Alterations in epidermal eicosanoid metabolism contribute to inflammation and impaired late differentiation in FLG-mutated atopic dermatitis. J Invest Dermatol 2017;137:706–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hirabayashi T, Anjo T, Kaneko A, Senoo Y, Shibata A, Takama H, et al. PNPLA1 has a crucial role in skin barrier function by directing acylceramide biosynthesis. Nat Commun 2017;8:14609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Casal ML, Wang P, Mauldin EA, Lin G, Henthorn PS. A defect in NIPAL4 is associated with autosomal recessive congenital ichthyosis in American bulldogs. PLoS One 2017;12:e0170708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Numata S, Teye K, Karashima T, Matsuda M, Hamada T, Hashimoto T. Functional study of TGM1 missense mutations in autosomal recessive congenital ichthyosis. Exp Dermatol 2016;25:657–9. [DOI] [PubMed] [Google Scholar]
  • 25.O’Shaughnessy RF, Choudhary I, Harper JI. Interleukin-1 alpha blockade prevents hyperkeratosis in an in vitro model of lamellar ichthyosis. Hum Mol Genet 2010;19:2594–605. [DOI] [PubMed] [Google Scholar]
  • 26.Bauer A, Waluk DP, Galichet A, Timm K, Jagannathan V, Sayar BS, et al. A de novo variant in the ASPRV1 gene in a dog with ichthyosis. PLoS Genet 2017;13: e1006651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Descargues P, Deraison C, Prost C, Fraitag S, Mazereeuw-Hautier J, D’Alessio M, et al. Corneodesmosomal cadherins are preferential targets of stratum corneum trypsin- and chymotrypsin-like hyperactivity in Netherton syndrome. J Invest Dermatol 2006;126:1622–32. [DOI] [PubMed] [Google Scholar]
  • 28.Vahidnezhad H, Youssefian L, Saeidian AH, Zeinali S, Mansouri P, Sotoudeh S, et al. Gene-targeted next generation sequencing identifies PNPLA1 mutations in patients with a phenotypic spectrum of autosomal recessive congenital ichthyosis: the impact of consanguinity. J Invest Dermatol 2017;137:678–85. [DOI] [PubMed] [Google Scholar]
  • 29.Saito R, Boyce A, Hsu CK, Rashidghamat E, Hide M, Wedgeworth EK, et al. Predictive phenotyping of inherited ichthyosis by next-generation DNA sequencing. Br J Dermatol 2017;176:249–51. [DOI] [PubMed] [Google Scholar]
  • 30.Pigg MH, Bygum A, Ganemo A, Virtanen M, Brandrup F, Zimmer AD, et al. Spectrum of autosomal recessive congenital ichthyosis in Scandinavia: clinical characteristics and novel and recurrent mutations in 132 patients. Acta Derm Venereol 2016;96:932–7. [DOI] [PubMed] [Google Scholar]
  • 31.Ullah R, Ansar M, Durrani ZU, Lee K, Santos-Cortez RL, Muhammad D, et al. Novel mutations in the genes TGM1 and ALOXE3 underlying autosomal recessive congenital ichthyosis. Int J Dermatol 2016;55:524–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Takeichi T, Nomura T, Takama H, Kono M, Sugiura K, Watanabe D, et al. Deficient stratum corneum intercellular lipid in a Japanese patient with lamellar ichthyosis by a homozygous deletion mutation in SDR9C7. Br J Dermatol 2017; 177:e62–4. [DOI] [PubMed] [Google Scholar]
  • 33.Kusakabe M, Nagai M, Nakano E, Jitsukawa O, Nishigori C, Yamanishi KA. Japanese case of ichthyosiform erythroderma with a novel mutation in NIPAL4/ichthyin. Acta Derm Venereol 2017;97:397–8. [DOI] [PubMed] [Google Scholar]
  • 34.Smith FJ, Irvine AD, Terron-Kwiatkowski A, Sandilands A, Campbell LE, Zhao Y, et al. Loss-of-function mutations in the gene encoding filaggrin cause ichthyosis vulgaris. Nat Genet 2006;38:337–42. [DOI] [PubMed] [Google Scholar]
  • 35.Li H, Lorie EP, Fischer J, Vahlquist A, Torma H. The expression of epidermal lipoxygenases and transglutaminase-1 is perturbed by NIPAL4 mutations: indications of a common metabolic pathway essential for skin barrier homeostasis. J Invest Dermatol 2012;132:2368–75. [DOI] [PubMed] [Google Scholar]
  • 36.Kawashima J, Akiyama M, Takizawa Y, Takahashi S, Matsuo I, Shimizu H. Structural, enzymatic and molecular studies in a series of nonbullous congenital ichthyosiform erythroderma patients. Clin Exp Dermatol 2005;30:429–31. [DOI] [PubMed] [Google Scholar]
  • 37.Thomas AC, Tattersall D, Norgett EE, O’Toole EA, Kelsell DP. Premature terminal differentiation and a reduction in specific proteases associated with loss of ABCA12 in Harlequin ichthyosis. Am J Pathol 2009;174:970–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ishida-Yamamoto A, Eady RA, Underwood RA, Dale BA, Holbrook KA. Filaggrin expression in epidermolytic ichthyosis (epidermolytic hyperkeratosis). Br J Dermatol 1994;131:767–79. [DOI] [PubMed] [Google Scholar]
  • 39.Dale BA, Holbrook KA, Fleckman P, Kimball JR, Brumbaugh S, Sybert VP. Heterogeneity in harlequin ichthyosis, an inborn error of epidermal keratinization: variable morphology and structural protein expression and a defect in lamellar granules. J Invest Dermatol 1990;94:6–18. [DOI] [PubMed] [Google Scholar]
  • 40.Choate KA, Williams ML, Elias PM, Khavari PA. Transglutaminase 1 expression in a patient with features of harlequin ichthyosis: case report. J Am Acad Dermatol 1998;38:325–9. [DOI] [PubMed] [Google Scholar]
  • 41.Malajian D, Guttman-Yassky E. New pathogenic and therapeutic paradigms in atopic dermatitis. Cytokine 2015;73:311–8. [DOI] [PubMed] [Google Scholar]
  • 42.Nomura T, Akiyama M, Sandilands A, Nemoto-Hasebe I, Sakai K, Nagasaki A, et al. Specific filaggrin mutations cause ichthyosis vulgaris and are significantly associated with atopic dermatitis in Japan. J Invest Dermatol 2008;128:1436–41. [DOI] [PubMed] [Google Scholar]
  • 43.Wells RS, Kerr CB. Clinical features of autosomal dominant and sex-linked ichthyosis in an English population. Br Med J 1966;1:947–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Yalcin AD. A case of netherton syndrome: successful treatment with omalizumab and pulse prednisolone and its effects on cytokines and immunoglobulin levels. Immunopharmacol Immunotoxicol 2016;38:162–6. [DOI] [PubMed] [Google Scholar]
  • 45.Hannula-Jouppi K, Laasanen SL, Ilander M, Furio L, Tuomiranta M, Marttila R, et al. Intrafamily and interfamilial phenotype variation and immature immunity in patients with Netherton syndrome and Finnish SPINK5 founder mutation. JAMA Dermatol 2016;152:435–42. [DOI] [PubMed] [Google Scholar]
  • 46.Briot A, Deraison C, Lacroix M, Bonnart C, Robin A, Besson C, et al. Kalli-krein 5 induces atopic dermatitis-like lesions through PAR2-mediated thymic stromal lymphopoietin expression in Netherton syndrome. J Exp Med 2009; 206:1135–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Briot A, Lacroix M, Robin A, Steinhoff M, Deraison C, Hovnanian A. Par2 inactivation inhibits early production of TSLP, but not cutaneous inflammation, in Netherton syndrome adult mouse model. J Invest Dermatol 2010;130:2736–42. [DOI] [PubMed] [Google Scholar]
  • 48.Akagi A, Kitoh A, Moniaga CS, Fujimoto A, Fujikawa H, Shimomura Y, et al. Case of Netherton syndrome with an elevated serum thymus and activation-regulated chemokine level. J Dermatol 2013;40:752–3. [DOI] [PubMed] [Google Scholar]
  • 49.Fontao L, Laffitte E, Briot A, Kaya G, Roux-Lombard P, Fraitag S, et al. Infliximab infusions for Netherton syndrome: sustained clinical improvement correlates with a reduction of thymic stromal lymphopoietin levels in the skin. J Invest Dermatol 2011;131:1947–50. [DOI] [PubMed] [Google Scholar]
  • 50.Kabesch M, Carr D, Weiland SK, von Mutius E. Association between polymorphisms in serine protease inhibitor, kazal type 5 and asthma phenotypes in a large German population sample. Clin Exp Allergy 2004;34:340–5. [DOI] [PubMed] [Google Scholar]
  • 51.Kato A, Fukai K, Oiso N, Hosomi N, Murakami T, Ishii M. Association of SPINK5 gene polymorphisms with atopic dermatitis in the Japanese population. Br J Dermatol 2003;148:665–9. [DOI] [PubMed] [Google Scholar]
  • 52.Walley AJ, Chavanas S, Moffatt MF, Esnouf RM, Ubhi B, Lawrence R, et al. Gene polymorphism in Netherton and common atopic disease. Nat Genet 2001; 29:175–8. [DOI] [PubMed] [Google Scholar]
  • 53.Paller AS, Renert-Yuval Y, Suprun M, Esaki H, Oliva M, Huynh TN, et al. An IL-17-dominant immune profile is shared across the major orphan forms of ichthyosis. J Allergy Clin Immunol 2017;139:152–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Papp KA, Leonardi C, Menter A, Ortonne JP, Krueger JG, Kricorian G, et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N Engl J Med 2012;366:1181–9. [DOI] [PubMed] [Google Scholar]
  • 55.Gordon KB, Blauvelt A, Papp KA, Langley RG, Luger T, Ohtsuki M, et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque psoriasis. N Engl J Med 2016;375:345–56. [DOI] [PubMed] [Google Scholar]
  • 56.Kim J, Bissonnette R, Lee J, Correa da Rosa J, Suarez-Farinas M, Lowes MA, et al. The spectrum of mild to severe psoriasis vulgaris is defined by a common activation of IL-17 pathway genes, but with key differences in immune regulatory genes. J Invest Dermatol 2016;136:2173–82. [DOI] [PubMed] [Google Scholar]
  • 57.Van Gysel D, Koning H, Baert MR, Savelkoul HF, Neijens HJ, Oranje AP. Clinico-immunological heterogeneity in Comel-Netherton syndrome. Derma- tology 2001;202:99–107. [DOI] [PubMed] [Google Scholar]
  • 58.Renner ED, Hartl D, Rylaarsdam S, Young ML, Monaco-Shawver L, Kleiner G, et al. Comel-Netherton syndrome defined as primary immunodeficiency. J Allergy Clin Immunol 2009;124:536–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hosomi N, Fukai K, Nakanishi T, Funaki S, Ishii M. Caspase-1 activity of stratum corneum and serum interleukin-18 level are increased in patients with Netherton syndrome. Br J Dermatol 2008;159:744–6. [DOI] [PubMed] [Google Scholar]
  • 60.Furio L, de Veer S, Jaillet M, Briot A, Robin A, Deraison C, et al. Transgenic kallikrein 5 mice reproduce major cutaneous and systemic hallmarks of Netherton syndrome. J Exp Med 2014;211:499–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Haneda T, Imai Y, Uchiyama R, Jitsukawa O, Yamanishi K. Activation of molecular signatures for antimicrobial and innate defense responses in skin with transglutaminase 1 deficiency. PLoS One 2016;11:e0159673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Roth W, Kumar V, Beer HD, Richter M, Wohlenberg C, Reuter U, et al. Keratin 1 maintains skin integrity and participates in an inflammatory network in skin through interleukin-18. J Cell Sci 2012;125:5269–79. [DOI] [PubMed] [Google Scholar]
  • 63.Khattri S, Shemer A, Rozenblit M, Dhingra N, Czarnowicki T, Finney R, et al. Cyclosporine in patients with atopic dermatitis modulates activated inflammatory pathways and reverses epidermal pathology. J Allergy Clin Immunol 2014;133: 1626–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Esaki H, Ewald DA, Ungar B, Rozenblit M, Zheng X, Xu H, et al. Identification of novel immune and barrier genes in atopic dermatitis by means of laser capture microdissection. J Allergy Clin Immunol 2015;135:153–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ewald DA, Malajian D, Krueger JG, Workman CT, Wang T, Tian S, et al. Meta-analysis derived atopic dermatitis (MADAD) transcriptome defines a robust AD signature highlighting the involvement of atherosclerosis and lipid metabolism pathways. BMC Med Genomics 2015;8:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ungar B, Garcet S, Gonzalez J, Dhingra N, Correa da Rosa J, Shemer A, et al. An integrated model of atopic dermatitis biomarkers highlights the systemic nature of the disease. J Invest Dermatol 2017;137:603–13. [DOI] [PubMed] [Google Scholar]
  • 67.Dhingra N, Shemer A, Correa da Rosa J, Rozenblit M, Fuentes-Duculan J, Gittler JK, et al. Molecular profiling of contact dermatitis skin identifies allergen-dependent differences in immune response. J Allergy Clin Immunol 2014;134:362–72. [DOI] [PubMed] [Google Scholar]
  • 68.Guttman-Yassky E, Suarez-Farinas M, Chiricozzi A, Nograles KE, Shemer A, Fuentes-Duculan J, et al. Broad defects in epidermal cornification in atopic dermatitis identified through genomic analysis. J Allergy Clin Immunol 2009; 124:1235–44.e58. [DOI] [PubMed] [Google Scholar]
  • 69.Czarnowicki T, Malajian D, Khattri S, Correa da Rosa J, Dutt R, Finney R, et al. Petrolatum: Barrier repair and antimicrobial responses underlying this “inert” moisturizer. J Allergy Clin Immunol 2016;137:1091–102, e1–7. [DOI] [PubMed] [Google Scholar]
  • 70.Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 2007;8:118–27. [DOI] [PubMed] [Google Scholar]
  • 71.Hanzelmann S, Castelo R, Guinney JGSVA. gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 2013;14:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fang H, Knezevic B, Burnham KL, Knight JC. XGR software for enhanced interpretation of genomic summary data, illustrated by application to immunological traits. Genome Med 2016;8:129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang Z, Monteiro CD, Jagodnik KM, Fernandez NF, Gundersen GW, Rouillard AD, et al. Extraction and analysis of signatures from the Gene Expression Omnibus by the crowd. Nat Commun 2016;7:12846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Esaki H, Brunner PM, Renert-Yuval Y, Czarnowicki T, Huynh T, Tran G, et al. Early-onset pediatric atopic dermatitis is TH2 but also TH17 polarized in skin. J Allergy Clin Immunol 2016;138:1639–51. [DOI] [PubMed] [Google Scholar]
  • 75.Suarez-Farinas M, Li K, Fuentes-Duculan J, Hayden K, Brodmerkel C, Krueger JG. Expanding the psoriasis disease profile: interrogation of the skin and serum of patients with moderate-to-severe psoriasis. J Invest Dermatol 2012;132:2552–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Fuentes-Duculan J, Bonifacio KM, Hawkes JE, Kunjravia N, Cueto I, Li X, et al. Autoantigens ADAMTSL5 and LL37 are significantly upregulated in active psoriasis and localized with keratinocytes, dendritic cells and other leukocytes. Exp Dermatol 2017;26:1075–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cheung KL, Jarrett R, Subramaniam S, Salimi M, Gutowska-Owsiak D, Chen YL, et al. Psoriatic T cells recognize neolipid antigens generated by mast cell phospholipase delivered by exosomes and presented by CD1a. J Exp Med 2016;213:2399–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Malik K, Ungar B, Garcet S, Dutt R, Dickstein D, Zheng X, et al. Dust mite induces multiple polar T cell axes in human skin. Clin Exp Allergy 2017;47: 1648–60. [DOI] [PubMed] [Google Scholar]
  • 79.Walsh PT, Fallon PG. The emergence of the IL-36 cytokine family as novel targets for inflammatory diseases. Ann N Y Acad Sci 2018;1417:23–34. [DOI] [PubMed] [Google Scholar]
  • 80.Tortola L, Rosenwald E, Abel B, Blumberg H, Schafer M, Coyle AJ, et al. Psoriasiform dermatitis is driven by IL-36-mediated DC-keratinocyte crosstalk. J Clin Invest 2012;122:3965–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Carrier Y, Ma HL, Ramon HE, Napierata L, Small C, O’Toole M, et al. Inter-regulation of Th17 cytokines and the IL-36 cytokines in vitro and in vivo: implications in psoriasis pathogenesis. J Invest Dermatol 2011;131:2428–37. [DOI] [PubMed] [Google Scholar]
  • 82.Li J, Ueno A, Fort Gasia M, Luider J, Wang T, Hirota C, et al. Profiles of lamina propria T helper cell subsets discriminate between ulcerative colitis and Crohn’s disease. Inflamm Bowel Dis 2016;22:1779–92. [DOI] [PubMed] [Google Scholar]
  • 83.Samuelov L, Sprecher E. Peeling off the genetics of atopic dermatitis-like congenital disorders. J Allergy Clin Immunol 2014;134:808–15. [DOI] [PubMed] [Google Scholar]
  • 84.Sandilands A, Terron-Kwiatkowski A, Hull PR, O’Regan GM, Clayton TH, Watson RM, et al. Comprehensive analysis of the gene encoding filaggrin uncovers prevalent and rare mutations in ichthyosis vulgaris and atopic eczema. Nat Genet 2007;39:650–4. [DOI] [PubMed] [Google Scholar]
  • 85.Tay YK, Khoo BP, Goh CL. The epidemiology of atopic dermatitis at a tertiary referral skin center in Singapore. Asian Pac J Allergy Immunol 1999;17:137–41. [PubMed] [Google Scholar]
  • 86.Hannula-Jouppi K, Laasanen SL, Heikkila H, Tuomiranta M, Tuomi ML, Hilvo S, et al. IgE allergen component-based profiling and atopic manifestations in patients with Netherton syndrome. J Allergy Clin Immunol 2014;134:985–8. [DOI] [PubMed] [Google Scholar]
  • 87.Paller AS, Czarnowicki T, Renert-Yuval Y, Holland K, Huynh T, Sadlier M, et al. The spectrum of manifestations in DSP (desmoplakin) SR6 domain mutations: immunophenotyping and response to ustekinumab. J Am Acad Dermatol 2018; 78:498–505.e2. [DOI] [PubMed] [Google Scholar]
  • 88.Ortega-Recalde O, Silgado D, Fetiva C, Fonseca DJ, Laissue P. Transcriptomic analysis of skin in a case of ichthyosis Curth-Macklin caused by a KRT1 mutation. Br J Dermatol 2016;175:1372–5. [DOI] [PubMed] [Google Scholar]
  • 89.Chiricozzi A, Guttman-Yassky E, Suarez-Farinas M, Nograles KE, Tian S, Cardinale I, et al. Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key infl ry pathogenic circuits in psoriasis. J Invest Dermatol 2011;131:677–87. [DOI] [PubMed] [Google Scholar]
  • 90.Zaba LC, Suarez-Farinas M, Fuentes-Duculan J, Nograles KE, Guttman-Yassky E, Cardinale I, et al. Effective treatment of psoriasis with etanercept is linked to suppression of IL-17 signaling, not immediate response TNF genes. J Allergy Clin Immunol 2009;124:1022-10.e1-395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Harden JL, Lewis SM, Lish SR, Suarez-Farinas M, Gareau D, Lentini T, et al. The tryptophan metabolism enzyme L-kynureninase is a novel inflammatory factor in psoriasis and other inflammatory diseases. J Allergy Clin Immunol 2016;137: 1830–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jansen PA, Kamsteeg M, Rodijk-Olthuis D, van Vlijmen-Willems IM, de Jongh GJ, Bergers M, et al. Expression of the vanin gene family in normal and inflamed human skin: induction by proinflammatory cytokines. J Invest Dermatol 2009; 129:2167–74. [DOI] [PubMed] [Google Scholar]
  • 93.Kamsteeg M, Jansen PA, van Vlijmen-Willems IM, van Erp PE, Rodijk-Olthuis D, van der Valk PG, et al. Molecular diagnostics of psoriasis, atopic dermatitis, allergic contact dermatitis and irritant contact dermatitis. Br J Dermatol 2010; 162:568–78. [DOI] [PubMed] [Google Scholar]
  • 94.Dietrich D, Martin P, Flacher V, Sun Y, Jarrossay D, Brembilla N, et al. Interleukin- 36 potently stimulates human M2 macrophages, Langerhans cells and keratinocytes to produce pro-inflammatory cytokines. Cytokine 2016;84:88–98. [DOI] [PubMed] [Google Scholar]
  • 95.Johnston A, Xing X, Wolterink L, Barnes DH, Yin Z, Reingold L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol 2017;140:109–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mahil SK, Catapano M, Di Meglio P, Dand N, Ahlfors H, Carr IM, et al. An analysis of IL-36 signature genes and individuals with IL1RL2 knockout mutations validates IL-36 as a psoriasis therapeutic target. Sci Transl Med 2017;9. [DOI] [PubMed] [Google Scholar]
  • 97.D’Erme AM, Wilsmann-Theis D, Wagenpfeil J, Holzel M, Ferring-Schmitt S, Sternberg S, et al. IL-36gamma (IL-1F9) is a biomarker for psoriasis skin lesions. J Invest Dermatol 2015;135:1025–32. [DOI] [PubMed] [Google Scholar]
  • 98.Malik K, Heitmiller KD, Czarnowicki T. An update on the pathophysiology of atopic dermatitis. Dermatol Clin 2017;35:317–26. [DOI] [PubMed] [Google Scholar]
  • 99.Choy DF, Hart KM, Borthwick LA, Shikotra A, Nagarkar DR, Siddiqui S, et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci Transl Med 2015;7:301ra129. [DOI] [PubMed] [Google Scholar]
  • 100.Newcomb DC, Boswell MG, Huckabee MM, Goleniewska K, Dulek DE, Reiss S, et al. IL-13 regulates Th17 secretion of IL-17A in an IL-10-dependent manner. J Immunol 2012;188:1027–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Newcomb DC, Boswell MG, Zhou W, Huckabee MM, Goleniewska K, Sevin CM, et al. Human TH17 cells express a functional IL-13 receptor and IL-13 attenuates IL-17A production. J Allergy Clin Immunol 2011; 127:1006–13, e1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kirchmeier P, Zimmer A, Bouadjar B, Rosler B, Fischer J. Whole-exome-sequencing reveals small deletions in CASP14 in patients with autosomal recessive inherited ichthyosis. Acta Derm Venereol 2017;97:102–4. [DOI] [PubMed] [Google Scholar]
  • 103.Sehra S, Yao Y, Howell MD, Nguyen ET, Kansas GS, Leung DY, et al. IL-4 regulates skin homeostasis and the predisposition toward allergic skin inflammation. J Immunol 2010;184:3186–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Howell MD, Gallo RL, Boguniewicz M, Jones JF, Wong C, Streib JE, et al. Cytokine milieu of atopic dermatitis skin subverts the innate immune response to vaccinia virus. Immunity 2006;24:341–8. [DOI] [PubMed] [Google Scholar]
  • 105.Kim BE, Leung DY, Boguniewicz M, Howell MD. Loricrin and involucrin expression is down-regulated by Th2 cytokines through STAT-6. Clin Immunol 2008;126:332–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.De Benedetto A, Rafaels NM, McGirt LY, Ivanov AI, Georas SN, Cheadle C, et al. Tight junction defects in patients with atopic dermatitis. J Allergy Clin Immunol 2011;127:773–86, e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Yuki T, Tobiishi M, Kusaka-Kikushima A, Ota Y, Tokura Y. Impaired tight junctions in atopic dermatitis skin and in a skin-equivalent model treated with interleukin-17. PLoS One 2016;11:e0161759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Akiyama M, Sakai K, Yanagi T, Fukushima S, Ihn H, Hitomi K, et al. Transglutaminase1 preferred substrate peptide K5 is an efficient tool in diagnosis of lamellar ichthyosis. Am J Pathol 2010;176:1592–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Pietrusinski M, Stanczyk-Przyluska A, Chlebna-Sokol D, Borkowska E, Kaluzewski B, Borowiec M. Identification and clinical consequences of a novel mutation in the gene for transglutaminase 1 in a patient with lamellar ichthyosis. Clin Exp Dermatol 2015;40:921–3. [DOI] [PubMed] [Google Scholar]
  • 110.Nemes Z, Marekov LN, Fesus L, Steinert PM. A novel function for transglutaminase 1: attachment of long-chain omega-hydroxyceramides to involucrin by ester bond formation. Proc Natl Acad Sci U S A 1999;96:8402–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Nemes Z, Marekov LN, Steinert PM. Involucrin cross-linking by transglutaminase 1. Binding to membranes directs residue specificity. J Biol Chem 1999;274:11013–21. [DOI] [PubMed] [Google Scholar]
  • 112.Nakajima K, Terao M, Takaishi M, Kataoka S, Goto-Inoue N, Setou M, et al. Barrier abnormality due to ceramide deficiency leads to psoriasiform inflammation in a mouse model. J Invest Dermatol 2013;133:2555–65. [DOI] [PubMed] [Google Scholar]
  • 113.Lee SH, Jhun J, Byun JK, Kim EK, Jung K, Lee JE, et al. IL-17 axis accelerates the inflammatory progression of obese in mice via TBK1 and IKBKE pathway. Immunol Lett 2017;184:67–75. [DOI] [PubMed] [Google Scholar]
  • 114.Zuniga LA, Shen WJ, Joyce-Shaikh B, Pyatnova EA, Richards AG, Thom C, et al. IL-17 regulates adipogenesis, glucose homeostasis, and obesity. J Immunol 2010; 185:6947–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Guttman-Yassky E, Krueger JG, Lebwohl MG. Systemic immune mechanisms in atopic dermatitis and psoriasis with implications for treatment. Exp Dermatol 2018;27:409–17. [DOI] [PubMed] [Google Scholar]
  • 116.Lopez-Ferrer A, Vilarrasa E, Puig L. Secukinumab (AIN457) for the treatment of psoriasis. Expert Rev Clin Immunol 2015;11:1177–88. [DOI] [PubMed] [Google Scholar]
  • 117.Krueger JG, Ferris LK, Menter A, Wagner F, White A, Visvanathan S, et al. Anti-IL-23A mAb BI 655066 for treatment of moderate-to-severe psoriasis: safety, efficacy, pharmacokinetics, and biomarker results of a single-rising-dose, randomized, double-blind, placebo-controlled trial. J Allergy Clin Immunol 2015;136:116–24.e7. [DOI] [PubMed] [Google Scholar]
  • 118.Harden JL, Krueger JG, Bowcock AM. The immunogenetics of Psoriasis: a comprehensive review. J Autoimmun 2015;64:66–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Griffiths CE, Reich K, Lebwohl M, van de Kerkhof P, Paul C, Menter A, et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet 2015;386:541–51. [DOI] [PubMed] [Google Scholar]
  • 120.Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, et al. Secukinumab in plaque psoriasis—results of two phase 3 trials. N Engl J Med 2014;371:326–38. [DOI] [PubMed] [Google Scholar]
  • 121.Chiricozzi A, Saraceno R, Chimenti MS, Guttman-Yassky E, Krueger JG. Role of IL-23 in the pathogenesis of psoriasis: a novel potential therapeutic target? Expert Opin Ther Targets 2014;18:513–25. [DOI] [PubMed] [Google Scholar]
  • 122.Martin DA, Towne JE, Kricorian G, Klekotka P, Gudjonsson JE, Krueger JG, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol 2013;133:17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lowes MA, Russell CB, Martin DA, Towne JE, Krueger JG. The IL-23/T17 pathogenic axis in psoriasis is amplified by keratinocyte responses. Trends Immunol 2013;34:174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Chiricozzi A, Krueger JG. IL-17 targeted therapies for psoriasis. Expert Opin Investig Drugs 2013;22:993–1005. [DOI] [PubMed] [Google Scholar]
  • 125.Krueger JG, Fretzin S, Suarez-Farinas M, Haslett PA, Phipps KM, Cameron GS, et al. IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis. J Allergy Clin Immunol 2012;130:145–54.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sticherling M. Psoriasis and autoimmunity. Autoimmun Rev 2016;15:1167–70. [DOI] [PubMed] [Google Scholar]
  • 127.Sofen H, Smith S, Matheson RT, Leonardi CL, Calderon C, Brodmerkel C, et al. Guselkumab (an IL-23-specific mAb) demonstrates clinical and molecular response in patients with moderate-to-severe psoriasis. J Allergy Clin Immunol 2014;133:1032–40. [DOI] [PubMed] [Google Scholar]
  • 128.Lew W, Bowcock AM, Krueger JG. Psoriasis vulgaris: cutaneous lymphoid tissue supports T-cell activation and ‘‘type 1’’ inflammatory gene expression. Trends Immunol 2004;25:295–305. [DOI] [PubMed] [Google Scholar]
  • 129.Lebwohl M. Psoriasis. Lancet 2003;361:1197–204. [DOI] [PubMed] [Google Scholar]
  • 130.Gordon KB, Langley RG, Gottlieb AB, Papp KA, Krueger GG, Strober BE, et al. A phase III, randomized, controlled trial of the fully human IL-12/23 mAb briakinumab in moderate-to-severe psoriasis. J Invest Dermatol 2012; 132:304–14. [DOI] [PubMed] [Google Scholar]
  • 131.Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008;371:1665–74. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Malik Suppl FigE1
Malik Suppl TabE4
Malik Suppl TabE5
Malik Suppl TabE6
Malik Suppl TabE7
Malik Suppl TabE8
Malik Suppl TabE9
Malik Suppl txt and STab E1-3
Malik Suppl FigE2
Malik Suppl FigE3
Malik Suppl FigE4
Malik Suppl FigE5
Malik Suppl FigE6
Malik Suppl TabE10
Malik Suppl TabE11
Malik Suppl TabE12

RESOURCES