Review Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Dermatol. May 2, 2014; 3(2): 28-35
Published online May 2, 2014. doi: 10.5314/wjd.v3.i2.28
Looking within the lesion: Large scale transcriptional profiling of psoriatic plaques
Claudia Mimoso, Miroslav Blumenberg, RO Perelman Department of Dermatology, Department of Biochemistry and Molecular Pharmacology, NYU Cancer Institute, NYU Langone Medical Center, New York, NY 10016, United States
Author contributions: Mimoso C, Blumenberg M contributed to the manuscript.
Correspondence to: Miroslav Blumenberg, PhD, RO Perelman Department of Dermatology, Department of Biochemistry and Molecular Pharmacology, NYU Cancer Institute, NYU Langone Medical Center, 455 First Avenue, New York, NY 10016, United States. miroslav.blumenberg@nyumc.org
Telephone: +1-212-2635924   Fax: +1-212-2638752
Received: November 22, 2013
Revised: January 23, 2014
Accepted: March 13, 2014
Published online: May 2, 2014

Abstract

Psoriasis is a lifelong, chronic, recurring and highly variable skin disease. Psoriatic plaques are formed through induction of inflammation in the epidermis and deregulation of keratinocyte proliferation and differentiation. This results in red or silvery scaly patches on the surface of the epidermis. To look within the lesions and define the changes in gene expression in psoriasis, investigators compared the transcriptomes of psoriatic plaques, of uninvolved skin of patients and of skin from healthy individuals. In several large studies with many patients, the genes expressed at much higher level in psoriatic plaques included those responsible for the cell cycle, keratinocyte differentiation, and response to wounding; conversely, lipid and fatty acid metabolism enzymes were expressed at reduced levels. The nonlesional and healthy skin appeared fairly similar. The largest study included paired biopsies from 85 individual patients. The same group used transcription profiling to follow the course of treatment in a set of patients, and correlated changes in the transcriptome of blood samples of psoriatic patients. Importantly, a noninvasive technique involving tape-stripping of skin, has been shown effective in transcriptional studies of psoriasis. Current efforts are focused on deconvoluting the contributions of various cell types in psoriasis, keratinocytes, lymphocytes, fibroblasts etc. Taken as a whole, these efforts will lead to personalized medicine, i.e., to specific, individualized treatments of patients with psoriasis.

Key Words: Cytokines, Inflammation, Metaa nalysis, Microarrays, Skinomics

Core tip: Dermatology was among the first medical specialties to adopt bioinformatics methodology, and Psoriasis, with its high prevalence, among the first diseases. Genome-wide association studies identified close to 50 genetic predisposition loci, to date. Recently, large-scale transcriptome analysis using DNA microarrays identified the important signaling pathways and regulators of gene expression in psoriasis. These efforts, and the fundamental knowledge they provide will lead to personalized medicine, i.e., to specific, individualized treatments of psoriatic patients in the near future.



INTRODUCTION

According to the National Psoriasis Foundation, psoriasis affects 7.5 million Americans and 125 million people worldwide. It is a chronic autoimmune disease with a multifactorial (genetic and environmental) etiology[1]. Past research suggests that external, internal and/or environmental triggers, such as stress, systemic illnesses and environmental allergens, combined with the genetic predisposition, may result in an altered immunity and an increased risk for the development of psoriasis[2]. However, the initial trigger for psoriasis and development of psoriatic lesions remains unknown[3-5]. While the exact causes are unknown, in psoriasis the immune system sends out incorrect signals that speed up the proliferation of epidermal keratinocytes. While normal keratinocytes mature and desquamate in about a month, psoriatic skin ones reach the surface in only 3-4 d and, instead of sloughing off individually, they accumulate to form large flaking scraps of skin[2].

Psoriasis is typically a lifelong, chronic recurring condition. It can vary in severity from small localized areas to covering the entire body. The diagnosis of psoriasis is based on the appearance of skin, not on blood tests or specialized diagnostic procedures. Occasionally a skin biopsy may be needed to rule out other proliferative skin disorders. Psoriatic plaques are formed through an increase in inflammation in the epidermis, deregulation of cell cycle processes, increase in keratinocyte proliferation and epidermal differentiation changes. Together this results in the formation of raised, red or silvery scaly patches on the surface of the stratum corneum.

The genetic predisposition for psoriasis was known through family-based and population-based epidemiological studies, which suggested that genetic factors play a key role in the development of psoriasis[6-7]. Perhaps a third of psoriatic patients report a family history of psoriasis; reports on monozygotic twins find a 70% chance of a twin developing psoriasis if the other twin has psoriasis while this number is around 20% for paternal twins. More recently, genome-wide association scans have fine-mapped the nine susceptibility loci (PSORS1-PSORS9) and located many previously unsuspected genomic markers on human chromosomes[6,8-11]. A current list of psoriasis susceptibility loci is given in Table 1. However, the known genetic factors for psoriasis do not account for all observed genetic susceptibility to psoriasis; additional genetic factors remain to be discovered[6]. Thus, the genetic contribution to psoriasis is not fully understood[3].

Table 1 Psoriasis susceptibility loci identified using genome-wide association studies.
PsoriasisChromosomeGeneLocusRef.
11p31.3IL-23RNair et al[11]
11p31.3IL-23RLiu et al[48]
11p31.3IL-23Rrs9988642Tsoi et al[8]
21p36RUNX3rs7536201Tsoi et al[8]
31p36.11IL28RArs7552167Tsoi et al[8]
41p36.23SLC45A1, TNFRSF9rs11121129Tsoi et al[8]
51q21PSORS4Julià et al[49]
61q21LCE3Drs6701216Liu et al[48]
71q21.3LCE3Brs4112788de Cid et al[1]
820q12-q13.12NCOA5rs2903908Zervou et al[50]
920q12-q13.2CD40rs4810485Zervou et al[50]
1020q13.13RNF114rs1056198Tsoi et al[8]
1122q11.21UBE2L3rs4821124Tsoi et al[8]
122p15B3GNT2rs10865331Tsoi et al[8]
132p16.1FLJ16341, RELrs62149416Tsoi et al[8]
142q14.2IL1RNJulià et al[49]
152q24.2KCNH7, IFIH1rs17716942Tsoi et al[8]
163p12.1CADM2Hiruma et al[51]
174q27Liu et al[48]
185q15ERAP1rs27432Tsoi et al[8]
195q31IL-13, IL-4rs1295685Tsoi et al[8]
205q31.1-q33.1IL-12Brs12188300Tsoi et al[8]
215q32-q33.1TNIP1rs17728338Bowes et al[52]
226p21.3HLA-CKnight et al[53]
236p25.3EXOC2, IRF4rs9504361Tsoi et al[8]
246q21TRAF3IP2rs33980500Ellinghaus et al[10]
256q23TNFAIP3rs582757Tsoi et al[8]
266q25.3TAGAPrs2451258Tsoi et al[8]
277p14.1ELMO1rs2700987Tsoi et al[8]
289p12DDX58rs11795343Tsoi et al[8]
299q31KLF4rs10979182Tsoi et al[8]
309q34TSC1rs1076160Bowes et al[52]
3110q22.3ZMIZ1rs1250546Tsoi et al[8]
3211q11-q13RPS6KA4, PRDX5rs645078Tsoi et al[8]
3311q22.3ZC3H12Crs4561177Tsoi et al[8]
3411q23.3ETS1rs3802826Tsoi et al[8]
3512q13.3STAT2, IL23Ars2066819Tsoi et al[8]
3613q12LHFPLiu et al[48]
3713q14.11COG6rs7993214Liu et al[48]
3814q13NFKBIArs8016947Tsoi et al[8]
3915q21rs3803369Liu et al[48]
4016p11.2PRSS53, FBXL19rs12445568Tsoi et al[8]
4116p13.13PRM3, SOCS1rs367569Tsoi et al[8]
4217q11.2-q12NOS2rs28998802Tsoi et al[8]
4317q21.31PTRF, STAT3, STAT5A/Brs963986Tsoi et al[8]
4417q25CARD14rs11652075Tsoi et al[8]
4518q21.2POL1, STARD6, MBD2rs545979Tsoi et al[8]
4618q22.1SERPINB8Julià et al[49]
4719p13.2TYK2rs34536443Tsoi et al[8]
4819p13.2ILF3, CARM1rs892085Tsoi et al[8]

Of the five types of psoriasis (plaque, guttate, inverse, pustular, and erythrodermic), the most common is the plaque psoriasis. Plaque psoriasis is seen as red and white silvery hues of scaly patches appearing on the top of the epidermis. Plaques frequently occur on the extensor aspects of the knees and elbows, but can affect any area, including the scalp, genitals, palms and soles. Fingernails and toenails are often affected, which can be an independent symptom. Additionally, psoriasis can be associated with inflammation of the joints, which is known as psoriatic arthritis. Guttate psoriasis presents as numerous small, scaly, pink or red lesions over large areas of the body, the trunk, limbs and scalp. Inverse or flexural psoriasis occurs in skin folds, e.g., around the genitals, the armpits or under the breasts. Pustular psoriasis presents as raised pus-filled bumps, commonly on the hands and feet (i.e., palmoplantar pustulosis), or generalized, occurring randomly widespread on any part of the body. Erythrodermic psoriasis involves the widespread inflammation and exfoliation over most of the body skin. While the shared symptoms, i.e., the underlying inflammation and epidermal hyperproliferation, characterize all types of psoriasis, the distinct clinical presentations, the extensive and dispersed genetic underpinnings and inconsistent, variable clinical responses argue that psoriasis comprises a cluster of related but distinct disorders.

The superb international success in GWAS mapping the psoriasis susceptibility loci has been joined recently by equally outstanding transcriptional profiling studies from several laboratories that recruited very impressive numbers of patients and samples (Table 2). These studies provide deep and comprehensive insights into the molecular mechanisms of the pathology of psoriasis. Also an international effort, the transcriptional profiling is lead by two teams in the United States, that of Drs. G. Gudjonsson and J.T. Elder at the University of Michigan, and the team of Dr. J.G. Krueger at the Rockefeller University. The researchers compared the genes expressed in psoriatic plaques with those expressed in the nonlesional skin of patients, and both of these with the skin of healthy control subjects. Investigators also searched for diagnostic markers of psoriasis in the blood of patients. The current status and insights from these efforts is the subject of this review.

Table 2 Transcriptional profiling studies targeting psoriasis in the GEO database.
MicroarraysPlatformSamplesRef.
Comparison studies
GSE3424814 + 14HG-U133_Plus_2Lesional + NonLes[21]
GSE4166224 + 24HG-U133_Plus_2Lesional + NonLes[21]
GSE4166315 + 15 (+ 51 treated)HG-U133_Plus_2Lesional + NonLes[21]
GSE3099985 + 85HG-U133_Plus_2Lesional + NonLes[19]
GSE1190315 + 15 (+ 59 treated)HG-U133A_2Lesional + NonLes[18]
GSE671013 + 13HG-U133ALesional + NonLes[24]
GSE1490521 + 33 + 30HG-U133_Plus_2Healthy + Les + NonLes[16]
GSE1335564 + 58 + 58HG-U133_Plus_2Healthy + Les + NonLes[12]
GSE3240720 + 20 (+ 20 IFNg treated)HG-U133A_2Healthy + NonLes[17]
Related studies
GSE42305Monocytes[54]
GSE41905Kcytes transfected wt Antimir31[55]
GSE31652All lesional, treated placebo[56]
GSE26952Nonlesional only Psor AD[57]
GSE18948PBMCs[58]
GSE11307PCR study1
GSE6601Psor vs AD[59]
GSE417453 + 3RNA Sequencing[60]
GSE2686611 + 11 (different regions)HG-U133A_2Single vs double amplification[61]
GSE307682 +4 (+ 8 flare and relapse)HG-U133A_2Small number of samples[62]
GSE27373 + 4 + 4HG_U95Av2Small array[63]
DISCUSSION

In a very influential study Gudjonsson et al[12] analyzed a large cohort of psoriatic patients and healthy controls using transcriptional profiling. Importantly, their analysis included 58 paired samples of lesional and nonlesional skin, allowing comparisons of matched samples from the same patients, and 64 control biopsies, allowing large-scale comparisons of lesional and nonlesional skin with healthy skin[11-15]. The sheer size of this study allowed the authors to identify close to a thousand differentially expressed genes in the lesional skin. The genes overrepresented in the psoriasis lesions included Serpins, β-defensin-2, S100A genes and IL-8. Suppressed genes included β-cellulin, IL1F7 and CCL27. The ontological categories induced in the lesions incorporated cell cycle, expected in this hyperproliferative disease, keratinocyte differentiation markers and three categories that contained cytokines, chemokines and their receptors, namely immune response, defense response and response to wounding[14]. The suppressed ontological categories incorporated lipid and fatty acid metabolism. The nonlesional and healthy skin, however, appeared rather similar, confirming the results of Yao et al[16] (see below).

In an important follow-up a team in United Kingdom, collaborating with Dr Gudjonsson, used sophisticated bioinformatics methodologies to classify psoriatic patients and identify distinct molecular subtypes[15]. Again, the nonlesional and healthy skin appeared quite similar. Among the psoriatic plaque samples, two subtypes were identified using multidimensional scaling, one a tightly clustered group of patients at the apex of the less congruent and more dispersed subtype. The authors proposed that TGF and the ErbB pathways may be involved in distinguishing the two subtypes.

The ground-breaking large-scale transcriptional profiling of psoriatic samples was reported by the team of Dr Krueger in 2009[16]. They analyzed 33 lesional, 30 non-lesional and 21 healthy control samples. The nonlesional skin was more similar to healthy skin of other donors that to the lesional skins from the same patient. The transcriptional signatures of the plaque biopsies pointed to the infiltration of T cells and dendritic cells in the lesions. Yao et al[16] recognized the signatures of several cytokines implicated in psoriasis. Specifically, they compared genes differentially expressed in the lesions with the gene sets regulated by IFN-α, IFN-γ and by TNF-α in keratinocytes. The significant overlaps substantiated the proposed roles of these cytokines in psoriasis. Several members of IFN-α family, IFN-α1, IFN-α2, IFN-α6, IFN-α7, IFN-α8, IFN-α14 and IFN-α21, were overexpressed in the lesions. The results validated the TNF-α-targeting and the T cell targeting therapies currently in wide use to treat psoriasis, as well as suggested IFN-α as a potential target.

Interestingly, in a separate study, Dr. Krueger’s team found that a single injection of IFN-γ into the dermis of nonlesional sites of psoriatics can recapitulate the transcription profile changes seen in the psoriatics plaques[17]. Apparently, IFN-γ can initiate the psoriasiform immune responses by promoting influx of T cells and dendritic cells. A similar influx was seen even in the IFN-γ-injected sites of healthy, non-psoriatic individuals.

The same team followed transcriptional changes in psoriatic patients treated with Etanercept[18]. Baseline transcriptional profiles were compared with those in treatment for up to 12 wk. The patients were divided into responders (11 patients) and non-responders (4 patients). Interestingly, the TNF-α-regulated genes (e.g., IL-1β and IL-8) were silenced in both groups; however the responders specifically inactivated the genes associated with the Th17 immune responses. The study highlighted the distinguishing and important role of the Th17 pathway in the pathology of psoriasis.

The largest transcriptional profiling study of psoriasis patients, to date, was reported by Suárez-Fariñas et al[19] in 2012. The Rockefeller University team compared 85 matched pairs of lesional and nonlesional biopsies from patients. The impressively large study identified 2725 individual genes differentially expressed 2-fold or more in the plaques. Serpins and S100A proteins were among the most overexpressed genes, but also many proteases/peptidases, including Kallikrein-related peptidase-6, -13, etc. Conversely, β-cellulin, CCL27 and lipid and fatty acid metabolism enzymes were found suppressed in the plaques, as seen by others[12,19]. The authors confirmed the results of transcriptional profiling using extensive RT-PCR and immunohistological experiments.

In this study by Suárez-Fariñas et al[19], the sets of regulated genes were compared with the sets identified in two previous studies[12,16]. Very high correlation was seen (scores ranging from 0.83 to 0.94) demonstrating very high concordance of the gene expression changes in psoriasis across the three large studies in two different centers. The concordance among different studies received extensive scrutiny[20-21], and it was found that, provided appropriate statistical methodologies are used, the studies are very highly concordant. The concordance allowed a meta analysis of psoriasis transcriptomics studies[22]. The meta analysis identified over 1000 genes that were consistently differentially expressed over 5 different studies. Moreover, this study provided a link between changes in the psoriasis transcriptome and atherosclerosis signaling, lipid and fatty acid metabolism and cardiovascular disease, thus providing a crucial link between the psoriatic skin conditions and these systemic diseases. Tian et al[22] in 2012, defined a “core” 20-gene set that distinguishes the psoriatic lesions. Interestingly, this core contained genes overexpressed even in psoriatic skin after successful treatment, as well as distinct genes epigenetically labeled by differential methylation in plaques.

Suárez-Fariñas et al[19] also compared serum protein levels of 12 important secreted proteins detected as overrepresented in psoriatic plaques[19]. In large cohort of approximately 150 patients and as many controls, all 12 proteins were found at increased levels in the sera of patients. The proteins included CCL2, CCL22, CXCL5 and TNF-α, which are all markers of psoriasis.

Using transient unresponsiveness to the stimulation of dendritic cells as a model of chronic inflammation, such as in psoriasis, Filkor et al[23] found the expression of feedback regulators of innate immunity to be suppressed, such as TNFAIP3 and TNFAIP8; these are also suppressed in the dermis of psoriatic patients.

In a study of matched lesional and nonlesional samples from 13 patients, in 2007, Reischl et al. identified 179 genes differentially expressed 2-fold or more[24]. Interestingly, 16 statistically significant genes were associated with the Wnt/β-catenin pathway, leading the authors to propose an important role for this pathway in psoriasis. Attempts to distinguish differences in the transcriptomes of plaques from different body sites, and between patients with symmetric and asymmetric plaques have not been successful[25]. In a more limited study of just 44 genes, Aubert et al[26] found that in psoriasis of the scalp, treatment with topical steroids restores expression of the 10 inflammation-related genes to the more basal, healthy levels. Similar results were found in a study of 5 lesional and 5 nonlesional samples, compared with similar number of blood samples[27]. Using a completely different approach, involving meta analysis of data in public repositories, specifically the BodyMap gene expression database[28], and RNA sequencing, Itoh et al[29] found very similar sets of differentially expressed genes. Others have noted the overlaps between genes differentially expressed in psoriasis and regulated by cytokines in epidermal keratinocytes[30-36].

In a study of matched lesional and nonlesional samples from 15 patients and 6 healthy controls, the team of Bowcock et al[37] a collaboration of Washington U. and Baylor U., also found overexpression of serpins and S100A proteins, but also of keratins KRT6, KRT16 and KRT17, known markers of epidermal hyperproliferation[38]. These studies also addressed the transcription factors responsible for the expression of differentially expressed genes and found NF-κB and AP1 sites evident, as expected. In addition, sites for nuclear receptors, RORa1, VDR and PPAR are found in the regulated genes, as are the motifs bound by Ikaros proteins, zinc finger transcription factors characteristic for lymphoid cell lineages. A similar set of transcription factors associated with psoriasis, additionally including E2F1 was proposed in another study[39]. Using a completely different approach, involving proteomics, NF-κB, AP1, STAT1 and STAT3 proteins were identified as important in psoriasis transcriptional deregulation[40].

In an exciting and sophisticated skinomics approach, Swindell et al[41] were able to assign most of the differentially expressed genes in the psoriatic plaques to the different cell types that contribute to the disease[41]. Specifically, they found that the genes induced in the plaques derive mainly from the activated keratinocytes, 56%, infiltrating T-cells, 14%, and macrophages, 11%. The suppressed genes were derived from the adipose, epidermis and dermis 4%. Swindell et al[41] also distinguished the patients who responded to Etanercept from the non-responders by their respective transcriptional profiles. Moreover, they confirmed the induction of genes responding to several cytokines, including IFN-γ, IL-1, IL-17A and TNF-α.

Importantly, skin samples can be obtained using noninvasive and (almost) painless technique of tape-stripping. This method provides RNA samples of quality and quantity adequate for microarray analysis[42]. Using tape-stripping followed by RT-PCR, Benson et al[43] have detected increased levels of mRNAs for TNFα, IFγ and KRT16, among others, in psoriatic plaques.

CONCLUSIONS AND FUTURE PROSPECTS

Whereas the future is inherently unpredictable, currently several trends seem to guide the research in transcriptional changes in psoriasis. First, stratification of patients into categories (e.g., etanercept responders) will allow personalized medicine approaches to be developed and used in the treatment of psoriasis. Second, the exact roles of the immune cell types, the cytokines and chemokines they produce and the signaling pathways consequently activated in the responding keratinocytes will provide scores of additional targets, which will further advance patient-specific treatments. And third, an exciting new area of research, that of the effects of the cutaneous microbiome on psoriasis initiation, progression and resolution[44-47] has the potential to revolutionize our conceptual and practical approach to this intractable and difficult problem.

ACKNOWLEDGMENTS

The authors thank the Ronald O Perelman Department of Dermatology, NYU School of Medicine for support, the NYU Genomics Facility for technical assistance with our microarray analyses. In addition, the authors would like to thank Mr. Angelo Piccirillo and Ms. Valerie Holmes from the Ossining High School Science Research Program, for all of their guidance and unwavering support.

Footnotes

P- Reviewers: Guarneri F, Hermida MD, Vasconcellos C S- Editor: Qi Y L- Editor: A E- Editor: Wu HL

References
1.  de Cid R, Riveira-Munoz E, Zeeuwen PL, Robarge J, Liao W, Dannhauser EN, Giardina E, Stuart PE, Nair R, Helms C. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nat Genet. 2009;41:211-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 399]  [Cited by in F6Publishing: 383]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
2.  Halprin KM. Epidermal “turnover time”--a re-examination. Br J Dermatol. 1972;86:14-19.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Duffin KC, Chandran V, Gladman DD, Krueger GG, Elder JT, Rahman P. Genetics of psoriasis and psoriatic arthritis: update and future direction. J Rheumatol. 2008;35:1449-1453.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263-271.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Bowcock AM, Krueger JG. Getting under the skin: the immunogenetics of psoriasis. Nat Rev Immunol. 2005;5:699-711.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Stuart PE, Nair RP, Ellinghaus E, Ding J, Tejasvi T, Gudjonsson JE, Li Y, Weidinger S, Eberlein B, Gieger C. Genome-wide association analysis identifies three psoriasis susceptibility loci. Nat Genet. 2010;42:1000-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 253]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
7.  Sun LD, Cheng H, Wang ZX, Zhang AP, Wang PG, Xu JH, Zhu QX, Zhou HS, Ellinghaus E, Zhang FR. Association analyses identify six new psoriasis susceptibility loci in the Chinese population. Nat Genet. 2010;42:1005-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in F6Publishing: 255]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
8.  Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, Ding J, Li Y, Tejasvi T, Gudjonsson JE. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat Genet. 2012;44:1341-1348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 745]  [Cited by in F6Publishing: 706]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
9.  Strange A, Capon F, Spencer CC, Knight J, Weale ME, Allen MH, Barton A, Band G, Bellenguez C, Bergboer JG. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42:985-990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 766]  [Cited by in F6Publishing: 777]  [Article Influence: 55.5]  [Reference Citation Analysis (0)]
10.  Ellinghaus E, Ellinghaus D, Stuart PE, Nair RP, Debrus S, Raelson JV, Belouchi M, Fournier H, Reinhard C, Ding J. Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet. 2010;42:991-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 283]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
11.  Nair RP, Duffin KC, Helms C, Ding J, Stuart PE, Goldgar D, Gudjonsson JE, Li Y, Tejasvi T, Feng BJ. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat Genet. 2009;41:199-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 997]  [Cited by in F6Publishing: 1039]  [Article Influence: 69.3]  [Reference Citation Analysis (0)]
12.  Gudjonsson JE, Ding J, Johnston A, Tejasvi T, Guzman AM, Nair RP, Voorhees JJ, Abecasis GR, Elder JT. Assessment of the psoriatic transcriptome in a large sample: additional regulated genes and comparisons with in vitro models. J Invest Dermatol. 2010;130:1829-1840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 173]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
13.  Ding J, Gudjonsson JE, Liang L, Stuart PE, Li Y, Chen W, Weichenthal M, Ellinghaus E, Franke A, Cookson W. Gene expression in skin and lymphoblastoid cells: Refined statistical method reveals extensive overlap in cis-eQTL signals. Am J Hum Genet. 2010;87:779-789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 150]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
14.  Gudjonsson JE, Ding J, Li X, Nair RP, Tejasvi T, Qin ZS, Ghosh D, Aphale A, Gumucio DL, Voorhees JJ. Global gene expression analysis reveals evidence for decreased lipid biosynthesis and increased innate immunity in uninvolved psoriatic skin. J Invest Dermatol. 2009;129:2795-2804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
15.  Ainali C, Valeyev N, Perera G, Williams A, Gudjonsson JE, Ouzounis CA, Nestle FO, Tsoka S. Transcriptome classification reveals molecular subtypes in psoriasis. BMC Genomics. 2012;13:472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
16.  Yao Y, Richman L, Morehouse C, de los Reyes M, Higgs BW, Boutrin A, White B, Coyle A, Krueger J, Kiener PA. Type I interferon: potential therapeutic target for psoriasis? PLoS One. 2008;3:e2737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 218]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
17.  Johnson-Huang LM, Suárez-Fariñas M, Pierson KC, Fuentes-Duculan J, Cueto I, Lentini T, Sullivan-Whalen M, Gilleaudeau P, Krueger JG, Haider AS. A single intradermal injection of IFN-γ induces an inflammatory state in both non-lesional psoriatic and healthy skin. J Invest Dermatol. 2012;132:1177-1187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
18.  Zaba LC, Suárez-Fariñas M, Fuentes-Duculan J, Nograles KE, Guttman-Yassky E, Cardinale I, Lowes MA, Krueger JG. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 239]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
19.  Suárez-Fariñas 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;132:2552-2564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
20.  Suárez-Fariñas M, Lowes MA, Zaba LC, Krueger JG. Evaluation of the psoriasis transcriptome across different studies by gene set enrichment analysis (GSEA). PLoS One. 2010;5:e10247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 139]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
21.  Bigler J, Rand HA, Kerkof K, Timour M, Russell CB. Cross-study homogeneity of psoriasis gene expression in skin across a large expression range. PLoS One. 2013;8:e52242.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Tian S, Krueger JG, Li K, Jabbari A, Brodmerkel C, Lowes MA, Suárez-Fariñas M. Meta-analysis derived (MAD) transcriptome of psoriasis defines the “core” pathogenesis of disease. PLoS One. 2012;7:e44274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 131]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
23.  Filkor K, Hegedűs Z, Szász A, Tubak V, Kemény L, Kondorosi É, Nagy I. Genome wide transcriptome analysis of dendritic cells identifies genes with altered expression in psoriasis. PLoS One. 2013;8:e73435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
24.  Reischl J, Schwenke S, Beekman JM, Mrowietz U, Stürzebecher S, Heubach JF. Increased expression of Wnt5a in psoriatic plaques. J Invest Dermatol. 2007;127:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Quekenborn-Trinquet V, Fogel P, Aldana-Jammayrac O, Ancian P, Demarchez M, Rossio P, Richards HL, Kirby B, Nguyen C, Voegel JJ. Gene expression profiles in psoriasis: analysis of impact of body site location and clinical severity. Br J Dermatol. 2005;152:489-504.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Aubert J, Reiniche P, Fogel P, Poulin Y, Lui H, Lynde C, Shapiro J, Villemagne H, Soto P, Voegel JJ. Gene expression profiling in psoriatic scalp hair follicles: clobetasol propionate shampoo 0.05% normalizes psoriasis disease markers. J Eur Acad Dermatol Venereol. 2010;24:1304-1311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
27.  Coda AB, Icen M, Smith JR, Sinha AA. Global transcriptional analysis of psoriatic skin and blood confirms known disease-associated pathways and highlights novel genomic “hot spots” for differentially expressed genes. Genomics. 2012;100:18-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
28.  Hishiki T, Kawamoto S, Morishita S, Okubo K. BodyMap: a human and mouse gene expression database. Nucleic Acids Res. 2000;28:136-138.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Itoh K, Kawasaki S, Kawamoto S, Seishima M, Chiba H, Michibata H, Wakimoto K, Imai Y, Minesaki Y, Otsuji M. Identification of differentially expressed genes in psoriasis using expression profiling approaches. Exp Dermatol. 2005;14:667-674.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Mee JB, Johnson CM, Morar N, Burslem F, Groves RW. The psoriatic transcriptome closely resembles that induced by interleukin-1 in cultured keratinocytes: dominance of innate immune responses in psoriasis. Am J Pathol. 2007;171:32-42.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Yano S, Banno T, Walsh R, Blumenberg M. Transcriptional responses of human epidermal keratinocytes to cytokine interleukin-1. J Cell Physiol. 2008;214:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Yano S, Banno T, Walsh R, Blumenberg M. Transcriptional responses of human epidermal keratinocytes to cytokine interleukin-1. J Cell Physiol. 2008;214:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Yano S, Strober BE, Haider SA, Blumenberg M. Transcriptional profiling analysis applied to psoriasis. Giornale Italiano di Dermatologia e Venerologia. 2007;142:519-531.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Gazel A, Rosdy M, Bertino B, Tornier C, Sahuc F, Blumenberg M. A characteristic subset of psoriasis-associated genes is induced by oncostatin-M in reconstituted epidermis. J Invest Dermatol. 2006;126:2647-2657.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Banno T, Gazel A, Blumenberg M. Effects of tumor necrosis factor-alpha (TNF alpha) in epidermal keratinocytes revealed using global transcriptional profiling. J Biol Chem. 2004;279:32633-32642.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Banno T, Adachi M, Mukkamala L, Blumenberg M. Unique keratinocyte-specific effects of interferon-gamma that protect skin from viruses, identified using transcriptional profiling. Antivir Ther. 2003;8:541-554.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Bowcock AM, Shannon W, Du F, Duncan J, Cao K, Aftergut K, Catier J, Fernandez-Vina MA, Menter A. Insights into psoriasis and other inflammatory diseases from large-scale gene expression studies. Hum Mol Genet. 2001;10:1793-1805.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Zhou X, Krueger JG, Kao MC, Lee E, Du F, Menter A, Wong WH, Bowcock AM. Novel mechanisms of T-cell and dendritic cell activation revealed by profiling of psoriasis on the 63,100-element oligonucleotide array. Physiol Genomics. 2003;13:69-78.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Lu X, Du J, Liang J, Zhu X, Yang Y, Xu J. Transcriptional regulatory network for psoriasis. J Dermatol. 2013;40:48-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
40.  Piruzian E, Bruskin S, Ishkin A, Abdeev R, Moshkovskii S, Melnik S, Nikolsky Y, Nikolskaya T. Integrated network analysis of transcriptomic and proteomic data in psoriasis. BMC Syst Biol. 2010;4:41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Swindell WR, Johnston A, Voorhees JJ, Elder JT, Gudjonsson JE. Dissecting the psoriasis transcriptome: inflammatory- and cytokine-driven gene expression in lesions from 163 patients. BMC Genomics. 2013;14:527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 95]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
42.  Wong R, Tran V, Morhenn V, Hung SP, Andersen B, Ito E, Wesley Hatfield G, Benson NR. Use of RT-PCR and DNA microarrays to characterize RNA recovered by non-invasive tape harvesting of normal and inflamed skin. J Invest Dermatol. 2004;123:159-167.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Benson NR, Papenfuss J, Wong R, Motaal A, Tran V, Panko J, Krueger GG. An analysis of select pathogenic messages in lesional and non-lesional psoriatic skin using non-invasive tape harvesting. J Invest Dermatol. 2006;126:2234-2241.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Blaser MJ. Harnessing the power of the human microbiome. Proc Natl Acad Sci U S A. 2010;107:6125-6126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
45.  Gao Z, Tseng CH, Strober BE, Pei Z, Blaser MJ. Substantial alterations of the cutaneous bacterial biota in psoriatic lesions. PLoS One. 2008;3:e2719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 285]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
46.  Paulino LC, Tseng CH, Blaser MJ. Analysis of Malassezia microbiota in healthy superficial human skin and in psoriatic lesions by multiplex real-time PCR. FEMS Yeast Res. 2008;8:460-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
47.  Paulino LC, Tseng CH, Strober BE, Blaser MJ. Molecular analysis of fungal microbiota in samples from healthy human skin and psoriatic lesions. J Clin Microbiol. 2006;44:2933-2941.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Liu Y, Helms C, Liao W, Zaba LC, Duan S, Gardner J, Wise C, Miner A, Malloy MJ, Pullinger CR. A genome-wide association study of psoriasis and psoriatic arthritis identifies new disease loci. PLoS Genet. 2008;4:e1000041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 528]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
49.  Julià A, Tortosa R, Hernanz JM, Cañete JD, Fonseca E, Ferrándiz C, Unamuno P, Puig L, Fernández-Sueiro JL, Sanmartí R. Risk variants for psoriasis vulgaris in a large case-control collection and association with clinical subphenotypes. Hum Mol Genet. 2012;21:4549-4557.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Zervou MI, Goulielmos GN, Castro-Giner F, Boumpas DT, Tosca AD, Krueger-Krasagakis S. A CD40 and an NCOA5 gene polymorphism confer susceptibility to psoriasis in a Southern European population: a case-control study. Hum Immunol. 2011;72:761-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
51.  Hiruma A, Ikeda S, Terui T, Ozawa M, Hashimoto T, Yasumoto S, Nakayama J, Kubota Y, Iijima M, Sueki H. A novel splicing variant of CADM2 as a protective transcript of psoriasis. Biochem Biophys Res Commun. 2011;412:626-632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
52.  Bowes J, Orozco G, Flynn E, Ho P, Brier R, Marzo-Ortega H, Coates L, McManus R, Ryan AW, Kane D. Confirmation of TNIP1 and IL23A as susceptibility loci for psoriatic arthritis. Ann Rheum Dis. 2011;70:1641-1644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 97]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
53.  Knight J, Spain SL, Capon F, Hayday A, Nestle FO, Clop A, Barker JN, Weale ME, Trembath RC. Conditional analysis identifies three novel major histocompatibility complex loci associated with psoriasis. Hum Mol Genet. 2012;21:5185-5192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
54.  Hyder LA, Gonzalez J, Harden JL, Johnson-Huang LM, Zaba LC, Pierson KC, Eungdamrong NJ, Lentini T, Gulati N, Fuentes-Duculan J. TREM-1 as a potential therapeutic target in psoriasis. J Invest Dermatol. 2013;133:1742-1751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
55.  Xu N, Meisgen F, Butler LM, Han G, Wang XJ, Söderberg-Nauclér C, Ståhle M, Pivarcsi A, Sonkoly E. MicroRNA-31 is overexpressed in psoriasis and modulates inflammatory cytokine and chemokine production in keratinocytes via targeting serine/threonine kinase 40. J Immunol. 2013;190:678-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 147]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
56.  Krueger JG, Fretzin S, Suárez-Fariñas M, Haslett PA, Phipps KM, Cameron GS, McColm J, Katcherian A, Cueto I, White T. IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis. J Allergy Clin Immunol. 2012;130:145-54.e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
57.  De Benedetto A, Rafaels NM, McGirt LY, Ivanov AI, Georas SN, Cheadle C, Berger AE, Zhang K, Vidyasagar S, Yoshida T. Tight junction defects in patients with atopic dermatitis. J Allergy Clin Immunol. 2011;127:773-786.e1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
58.  Suárez-Fariñas M, Shah KR, Haider AS, Krueger JG, Lowes MA. Personalized medicine in psoriasis: developing a genomic classifier to predict histological response to Alefacept. BMC Dermatol. 2010;10:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
59.  de Jongh GJ, Zeeuwen PL, Kucharekova M, Pfundt R, van der Valk PG, Blokx W, Dogan A, Hiemstra PS, van de Kerkhof PC, Schalkwijk J. High expression levels of keratinocyte antimicrobial proteins in psoriasis compared with atopic dermatitis. J Invest Dermatol. 2005;125:1163-1173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 216]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
60.  Jabbari A, Suárez-Fariñas M, Dewell S, Krueger JG. Transcriptional profiling of psoriasis using RNA-seq reveals previously unidentified differentially expressed genes. J Invest Dermatol. 2012;132:246-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
61.  Mitsui H, Suárez-Fariñas M, Belkin DA, Levenkova N, Fuentes-Duculan J, Coats I, Fujita H, Krueger JG. Combined use of laser capture microdissection and cDNA microarray analysis identifies locally expressed disease-related genes in focal regions of psoriasis vulgaris skin lesions. J Invest Dermatol. 2012;132:1615-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
62.  Johnson-Huang LM, Pensabene CA, Shah KR, Pierson KC, Kikuchi T, Lentini T, Gilleaudeau P, Sullivan-Whalen M, Cueto I, Khatcherian A. Post-therapeutic relapse of psoriasis after CD11a blockade is associated with T cells and inflammatory myeloid DCs. PLoS One. 2012;7:e30308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
63.  Kulski JK, Kenworthy W, Bellgard M, Taplin R, Okamoto K, Oka A, Mabuchi T, Ozawa A, Tamiya G, Inoko H. Gene expression profiling of Japanese psoriatic skin reveals an increased activity in molecular stress and immune response signals. J Mol Med (Berl). 2005;83:964-975.  [PubMed]  [DOI]  [Cited in This Article: ]