Abstract
Psoriasis is a chronic inflammatory condition characterized by systemic immune dysregulation. Over the past several years, advances in genetics, microbiology, immunology, and mouse models have revealed the complex interplay between the heritable and microenvironmental factors that drive the development of psoriatic inflammation. In the first of this two-part review series, the authors will discuss the newest insights into the pathogenesis of psoriatic disease and highlight how the evolution of these scientific fields has paved the way for a more personalized approach to psoriatic disease treatment.
Introduction
Psoriasis is a chronic, inflammatory disease affecting an estimated 3 percent of the U.S. population (Rachakonda et al., 2014). In its skin manifestation, psoriasis most commonly presents with plaques as psoriasis vulgaris (PsV), but also includes rarer subtypes such as guttate, pustular, erythrodermic, and inverse psoriasis. Approximately 30 percent of patients with psoriasis have psoriatic arthritis (PsA) (Mease et al., 2013), which can manifest as synovitis, enthesitis, dactylitis, and spondylitis. Due to the overlap between the skin manifestation of psoriasis, PsA, and other comorbidities associated with the systemic inflammation from this immune-mediated disease state, some researchers, providers, and patients, as well as the National Institutes of Health, consider these conditions to be part of a spectrum of psoriatic disease. It has only been in the last few decades that researchers have truly begun to unravel the pathogenesis of psoriasis and PsA. Since then, this research has evolved rapidly.
While the mechanism of psoriasis is now understood to involve IL-23/IL-17 signaling, there is increasing recognition that psoriasis is driven by a complex interplay between numerous other intrinsic and extrinsic factors. Recent innovations in next generation sequencing and single cell profiling coupled with increases in computing power have enabled a multi-omics approach to understand the genetic landscape of psoriasis, tissue-specific immune micro-environments, and host-microbiome interactions. These data provide a more nuanced understanding of psoriasis and PsA, revealing novel pathways and mechanisms that contribute to the development of PsA, particular psoriatic disease subtypes, and characteristic patterns of inflammation.
In the first of this two-part review series, the authors highlight the latest developments in genetics, environmental triggers, and immunology of psoriasis and PsA, novel insights from mouse models, and how these new developments have enabled more personalized treatment for psoriatic disease.
The genetics of psoriasis and psoriatic arthritis
Genetic architecture of psoriasis
Genetic susceptibility to PsV is polygenic, according to the sum of multiple genetic risk factors. To date, genome-wide association studies (GWAS) have identified over 63 genetic risk loci, which account for 28% of PsV heritability (Tsoi et al., 2017) while broader genome-wide heritability for PsV is estimated at 50% (Li et al., 2020). There is significant genetic concordance between ethnic groups, but eleven risk loci differ in Han Chinese and Caucasian populations (Yin et al., 2015). GWAS have revealed valuable insights into disease pathogenesis (Table 1). Of particular importance are genes associated with antigen presentation, such as MHC class I: HLA-C*06:02, HLA-C*12:03, HLA-B*57:01, HLA-B*38:01, and HLA-A*02:01. Coding mutations in ERAP1 and ERAP2, involved in peptide processing, have also been linked to PsV. Genetic risk loci have been identified in the NFκB pathway, which mediates keratinocyte (KC) proliferation and differentiation, production of pro-inflammatory cytokines in Th17 cells, clonal expansion of T cells, and Wnt signaling in osteoblasts (Goldminz et al., 2013, Ma and Hottiger, 2016, Takao et al., 2003). These include genetic variants in TRAF3IP2, CARD14, TNFSF15, TNIP1, IKBKE, CHUK, REL, NFKBIA, and non-coding mutations in TNFAIP3. Other genetic risk loci, such as IFIH1, DDX58, KLF4, ZC3H12C, CARD14 and CARM1 are involved in type I interferon signaling and the innate immune response, while variants in IL23R, IL12B, IL23A, and TYK2 play a role in adaptive (IL-23/IL-17) immunity. PsV genes also include those involved in host defense and maintenance of the skin epithelial barrier, such as beta-defensin and LCE3B/LCE3C.
Table 1.
Major identified genetic risk loci for psoriasis and their associated pathways.
These genes are associated with pustular psoriasis
Associated with psoriasis vulgaris and guttate psoriasis
Genetics of psoriatic arthritis and psoriasis subtypes
GWAS of PsA have found significant overlap with PsV (Bowes et al., 2015, Stuart et al., 2015). Surprisingly, only a few genetic variants so far have been identified as being discrepant between PsA and PsV, one of the main ones being HLA-C*06:02, which is a risk factor for PsV but protective for PsA, with other discrepant variants observed near IL23R, TNFAIP3, LCE3A, and TNFRSF9 (Stuart et al., 2015). This may indicate a more prominent role for environmental factors in the development of PsA. Notably, mice expressing three distinct targeted mutations of ZF7 ubiquitin-binding motif of the TNFAIP3 gene, were found to develop distal arthritis through IL-17-mediated NFκB activation (Razani et al., 2020). The genetic differences between psoriasis and PsA are also enriched in regulatory elements for lymphocytes, including CD8+ T cells (Patrick et al., 2018), which are present in the synovial fluid of PsA patients and correlate with disease severity (Menon et al., 2014, Steel et al., 2020). Interestingly, different HLA alleles have been found to be preferentially associated with PsA features such as synovitis, sacroiliitis, dactylitis, and enthesitis (FitzGerald et al., 2015).
Regarding psoriasis subtypes, HLA-C*06:02 has been found to be highly associated with guttate psoriasis (Gudjonsson et al., 2006). Coding mutations in IL36RN, CARD14, AP1S3, SERPINA3, and MPO that affect IL-1 and IL-36 signaling have been associated with the clinical spectrum of pustular psoriasis (Supplementary Table 1). Psoriasis genetic variants overlap with Crohn’s disease (Ellinghaus et al., 2012) and several cardiovascular disease phenotypes (Lu et al., 2013) but share no common genetic features with atopic dermatitis (Baurecht et al., 2015).
Recent discoveries in genetics of psoriasis and psoriatic arthritis
Sequencing data from a large Chinese cohort has revealed a potentially important, unexpected role for small genomic insertions and deletions in psoriasis susceptibility (Zhen et al., 2019). As these variants are not adequately captured in GWAS, additional studies are warranted. Mendelian randomization (Emdin et al., 2017) has allowed researchers to demonstrate that obesity is not only associated with psoriasis, but that obesity is a causal risk factor for psoriasis (Budu-Aggrey et al., 2019, Ogawa et al., 2019). Future studies employing Mendelian randomization are likely to help define the directionality between psoriasis and its comorbidities. Finally, several studies have highlighted the potential powerful role of psoriatic genetics in precision medicine. A meta-analysis demonstrated that HLA-C*06:02 is associated with favorable response of PsV patients to ustekinumab (van Vugt et al., 2019), while another study demonstrated that lack of HLA-C*06:02 is associated with better response to adalimumab (Dand et al., 2019). Using a machine learning approach involving ~200 genetic loci, researchers have developed a model to predict PsA with area under the receiver operator curve of 0.82 (Patrick et al., 2018).
Environmental triggers of psoriasis and psoriatic arthritis
The increased risk of PsV with smoking (Armstrong et al., 2014) may be modulated by interactions with genetic factors such as HLA-C*06:02 and CYP1A1 (Jin et al., 2009, Krämer and Esser, 2006). Although smoking is associated with a higher risk of PsA in the general population, amongst patients with psoriasis, there is either a decreased incidence of PsA or no significant relationship (Pezzolo and Naldi, 2019). Unlike smoking, the effects of alcohol consumption on psoriasis incidence is unclear (Brenaut et al., 2013, Dai et al., 2019).
Medications such as beta blockers, lithium, anti-malarials, imiquimod, nonsteroidal anti-inflammatory drugs, interferon-α, and terbinafine have been linked to the induction and exacerbation psoriasis (Balak and Hajdarbegovic, 2017, Kim and Del Rosso, 2010). More recently, paradoxical psoriasis induced by TNF-α inhibitors (Mazloom et al., 2018) has been shown to be associated with increased production of type I interferons by plasmacytoid dendritic cells in a T cell independent fashion (Conrad et al., 2018).
Obesity and diet are modifiable risk factors for psoriasis. Psoriasis patients have a higher prevalence and incidence of obesity (Armstrong et al., 2012). Animal studies have shown that diet-induced obese mice have more severe psoriasiform dermatitis, suggesting that the chronic inflammatory state caused by adipokines in excessive fat tissue could be a pathogenic link between obesity and psoriasis (Kanemaru et al., 2015). A meta-analysis demonstrated that weight loss through dietary intervention in obese individuals improves pre-existing psoriasis and prevents de novo psoriasis (Mahil et al., 2019).
However, more recent studies suggest that mechanisms other than obesity may mediate the impact of diet on psoriasis. For example, an increase of saturated fatty acids (SFAs) in healthy, lean mice alone was sufficient to induce an exacerbation of psoriasiform inflammation, and reduction of nutritional saturated fatty acids diminished the psoriatic phenotype in non-obese mice (Herbert et al., 2018). Another study showed that exposure to a high-sugar and moderate-fat Western diet (WD), even in the absence of obesity, was enough to induce skin inflammation in mice in as little as 4 weeks through the recruitment of IL-17A-producing γδ-type T cells (Shi et al., 2020). Interestingly, WD-induced skin inflammation was blocked by systemic antibiotic treatment, suggesting a critical role of gut dysbiosis in diet-induced inflammation. Further studies are warranted to determine if specific diets, in the absence of weight reduction, would bring meaningful clinical improvement in psoriasis and PsA.
Biomechanical stress is another major microenvironmental factor in psoriatic inflammation (Belasco and Wei, 2019). For example, animal models show that hind limb unloading reduced Achilles tendon inflammation (Jacques et al., 2014). Furthermore, mechanical stress induced expression of IL-17A by entheseal T cells at anatomic locations commonly affected by arthritis in mice (Reinhardt et al., 2016). Local biomechanical stress is also thought to promote differentiation of murine osteoclasts in the joint (Gracey et al., 2020). This is supported by a recent study demonstrating that erosive disease in mice is confined to mechano-sensitive regions and driven by mesenchymal cells that recruit monocytes via CXCL1 and CCL2 prior to differentiation into mature osteoclasts (Cambre et al., 2018). The pathogenic importance of exogenous mechanical loading suggests that rather than inherent genetic predisposition, dynamic interplay between the immune system and microenvironmental factors plays a major role in the development of PsA.
Various infections have been known to trigger or worsen psoriasis. HIV has been linked to a higher prevalence of psoriasis (Ceccarelli et al., 2019), worsening of pre-existing psoriasis, eruptive onset of de novo psoriasis (Duvic et al., 1987), and more recalcitrant disease (Menon et al., 2010), which may be due to an increased CD8+ to CD4+ T cell ratio, HIV viral protein superantigens (Ceccarelli et al., 2019), and a shared genetic architecture of the MHC I region (Chen et al., 2012). Preceding streptococcal pharyngitis (Telfer et al., 1992), possibly through similarities between streptococcal antigens and keratinocyte proteins, may induce cross-reactivity in streptococcal specific T cells leading to activation of psoriasis (Prinz, 2001). Periodontitis has also been associated with psoriasis, with psoriasis patients demonstrating more gingival inflammation, alveolar bone loss, and missing teeth (Qiao et al., 2019), and showing a significant reduction in psoriasis severity following non-surgical periodontitis treatment (Ucan Yarkac et al., 2020).
Emerging research now suggests that interactions between the skin and gut microbiome and the host immune system may play a key pathogenic role in the development of psoriasis and PsA (Supplementary Table 1). Bacteria play crucial roles in host IL-17 mediated mucosal barrier immunity, Th17 cell development, and RORγt+ Treg immune homeostasis (Zheng et al., 2020). PsV skin is enriched in pro-inflammatory Streptococcus and Staphylococcus, though not all studies observed this enrichment. Both Streptococcus and Staphylococcus aureus elicit a strong Th17-type response in skin T effector cells resulting in induction of IL-17A, IL-17F, and IL-22 cytokines in murine models (Okada et al., 2020). Interestingly, murine models of PsA and PsV do not develop inflammatory disease when treated with antibiotics or raised in a germ free environment (Rath et al., 1996, Zákostelská et al., 2016). In addition to increased pro-inflammatory bacteria, there is a reduction in immunomodulatory Propionibacterium, a major producer of propionate (Yan et al., 2017), a short chain fatty acid (SCFA) that promotes regulatory T cell homeostasis (Smith et al., 2013).
Similarly, the gut microbiome in PsV has demonstrated a decrease in Bacteroides and F. prausnitzii, a gut commensal that produces the immunomodulatory SCFA, butyrate (Furusawa et al., 2013). The only study of the gut microbiome in PsA showed decreases in Akkermansia, Ruminococcus, and Pseudobutyrivibrio, which were associated with decreased levels of immunomodulatory medium-chain fatty acids (MCFA) (Scher et al., 2015).
In addition to its role in disease pathogenesis, the microbiome may also be a modulator of therapeutic response. Gut bacteria are not only involved in drug metabolism, but also indirectly influence drug efficacy through effects on host gene expression and interactions between drugs and bacterial metabolites (Scher et al., 2020). For example, PsV and PsA patients who responded to secukinumab, had a higher abundance of intestinal Citrobacter, Staphylococcus, and Hafnia/Obesumbacterium (Yeh et al., 2019). However, there was no difference in the gut microbiome between responders and non-responders to ustekinumab (Yeh et al., 2019).
Not surprisingly, the microbiome is now a prime target for novel therapeutics. Already, clinical trials are investigating the efficacy of probiotic supplements for the treatment of psoriasis. One double-blinded, placebo-controlled clinical trial did not find any significant clinical improvement in PsV with a probiotic containing Lactobacillus (Kaur et al., 2007). However, a different study found that PsV patients who received an oral probiotic cocktail were more likely to achieve PASI-75 at 12 weeks (Navarro-López et al., 2019). Oral supplements of Bifidobacteria infantis have also been found to reduce serum levels of TNF-α and C-reactive protein in PsV patients (Groeger et al., 2013). Intriguingly, a recent case report described a patient who developed sustained remission of her PsA after undergoing fecal microbiota transplantation (FMT) for a Clostridium difficile infection (Selvanderan et al., 2019). The therapeutic efficacy of FMT in patients with PsA is now under investigation through a double-blind, randomized, placebo-controlled trial (Kragsnaes et al., 2018).
The immunology of psoriasis and psoriatic arthritis
Data from GWAS and other preclinical studies suggest that while certain immunopathogenic mechanisms may be shared, there are likely also tissue specific microenvironmental factors that contribute to the phenotypic heterogeneity of psoriasis and PsA. The IL-23/IL-17 axis is common to the pathogenesis of both cutaneous psoriasis, PsA, and enthesitis (Iwakura and Ishigame, 2006, Sherlock et al., 2012). Binding of IL-23 to the heterodimeric IL-23R/IL-12Rβ1 receptor activates Tyk2 and Jak2 dependent STAT3 signaling, which promotes the expansion of Th/Tc17 cells that secrete IL-17A, IL-17F, and TNF (Hile et al., 2020). Components of innate immunity also facilitate the inflammatory cascade in cutaneous psoriasis and PsA in which plasmacytoid and conventional dendritic cells are activated in the skin (Nestle et al., 2005) and synovial fluid (Penkava et al., 2020), respectively, and produce inflammatory cytokines, including IL-23, to facilitate priming and proliferation of Th/Tc17 cells. Antimicrobial peptides, most notably cathelicidin (LL37), bind to self-DNA and stimulate plasmacytoid dendritic cells via TLR9 or serve as autoantigen presented to T cells by HLA-C*06:02. IL-17A can be also produced by group 3 innate lymphoid cells (ILC3) that are abundant in PsV and PsA (Polese et al., 2020, Soare et al., 2018).
In addition to heightened pro-inflammatory signaling, PsO involves suppression of anti-inflammatory pathways. Both psoriasis and PsA demonstrate decreased numbers of CD73+ T regulatory cells (Tregs) (Han et al., 2018), expansion of dysfunctional RORγt+ Tregs with Th17 potential (Liu Y. et al., 2020, Scher et al., 2019), and decreased levels of the anti-inflammatory cytokine IL-38 (Mercurio et al., 2018), and reduction in ILC2s (Soare et al., 2018).
Differences in tissue-specific cues result in the activation of unique pathogenic pathways in the skin and synovium. In the skin, IL-17 and IL-22 stimulate keratinocyte hyperplasia as well as CXCL8-dependent neutrophil recruitment and microabscess formation (Liang et al., 2017). In the joints, local inflammatory cytokines and growth factors promote osteoclast-osteoblast decoupling and RANKL-mediated destructive bone remodeling in PsA (Paine and Ritchlin, 2018). Chronic psoriatic inflammation also upregulates Wnt signaling (RUNX1, FUT8, and CTNNAL1) (Patrick et al., 2019) in osteoblasts of PsA patients. A recent study utilizing an in vitro cell culture system to mimic local inflammatory microenvironment of bone-forming sites validated the link between Wnt signaling and inflammation, establishing that constitutive low-intensity stimulation with TNF induces persistent expression of Wnt proteins, causing increased bone formation through NFκB signaling (Li et al., 2018). Furthermore, binding of osteoclast derived RANK to osteoblastic RANKL leads to activation of RUNX2, which mediates bone remodeling and bone formation in RANKL mutant mice (Ikebuchi et al., 2018).
Locally presented antigens are another element of the immune microenvironment that may play a key role in psoriatic inflammation. In PsV, a large proportion of cutaneous CD8+ T cells is oligoclonal, with oligoclonality usually confined to lesional skin. To date, putative autoantigens in psoriasis including LL-37, a disintegrin and metalloprotease domain containing thrombospondin type 1 motif-like 5 (ADAMTSL5)(Fuentes-Duculan et al., 2017, Yuan et al., 2019), keratin 17 (KRT17)(Yunusbaeva et al., 2018), and neolipids generated by phospholipase A2 group IVD (PLA2G4D)(Cheung et al., 2016).
Mouse models of psoriasis and psoriatic arthritis
Preclinical mouse models are a valuable tool for investigating the pathogenic role of pathways, genes, or transcripts identified in genetic analyses and other studies. These approaches model one or more components of human psoriasis, but none to date replicate the disease in its full complexity. Mouse models of psoriasis can be broadly classified as spontaneous, xenotransplants, induced/acute, and genetically engineered (reviewed in (Gudjonsson et al., 2007, Schon et al., 2020). The first models of psoriasis involved spontaneous gene mutations in Scd, Sharpin ,or Ttc resulting in a “flakey skin” phenotype (Gates and Karasek, 1965, HogenEsch et al., 1993, Sundberg et al., 1994). However, the use of these models was limited as they lacked T cell infiltration.
Other early approaches used grafts of uninvolved skin from psoriasis patients onto immunodeficient mice (athymic nude, AGR), which triggered conversion into a psoriatic plaque (Boyman et al., 2004, Fraki et al., 1982, Krueger et al., 1981). Xenografts and ex vivo psoriasis skin explants (Billi et al., 2020, Ward et al., 2015) are the most translatable approach for studying treatment efficacy and the role of skin resident versus circulating immune cells. However, they are challenging to develop, require full-thickness patient skin, and some require autologous activated T cells (Nickoloff and Wrone-Smith, 1999).
The most widely used model involves topical application of imiquimod (IMQ) a TLR7 agonist. Since 2009 (van der Fits et al., 2009), over 600 publications have used this approach and interpreted their findings, often incorrectly, as critical for psoriasis pathogenesis (Hawkes et al., 2017). However, non-lesional skin from PsV patients treated with IMQ has different transcriptomic signatures compared to lesional skin, suggesting that IMQ models recapitulate only limited aspects of psoriasis (Vinter et al., 2015).
Investigators have also studied the effects of cytokines through intradermal injections of IL-17C (Ramirez-Carrozzi et al., 2011), IL-17A (Vasseur et al., 2016), IL-36 (Foster et al., 2014), IL-21 (Caruso et al., 2009), IL-23 (Chan et al., 2006) either alone or in combination (Guilloteau et al., 2010). These models reproduce some features of psoriasis and provide insight into the pathogenic role of individual cytokines and their synergistic effects. However, the need for repeated injections limits their utility for long-term study. The development of novel DNA minicircles allowing for sustained transgenic expression of IL-23 (Sherlock et al., 2012) reproduces chronic psoriasiform skin inflammation, enthesitis, arthritis, and vascular inflammation in mice, providing an innovative model for studying psoriatic comorbidities.
Recently, advances in genetic engineering have led to the development of transgenic and knockout animal models, which have provided novel insights into psoriasis. For example, the CD18 hypomorphic model (CD18hypo PL/J background; CD18(Itgb2tm1Bay)) helped define the role of skin-infiltrating T cells (Bullard et al., 1996) and TNF-α-producing macrophages (Wang et al., 2009).
The growing interest in keratinocytes and their derived factors resulted in an influx of KC-specific transgenic models: VEGF-A (Kunstfeld et al., 2004, Xia et al., 2003), TGF-β1 (Li et al., 2004), IL-17C (Johnston et al., 2013), ectopic Tie2 (Wolfram et al., 2009) and IL-17A (Karbach et al., 2014) all resulted in flakey skin phenotypes. The Tie2, IL-17A and IL-17C models also developed cardiovascular disease (CVD), including vascular inflammation (Karbach et al., 2014, Schuler et al., 2019, Wang et al., 2009) and thrombosis (Golden et al., 2015, Wang et al., 2016), which are also seen in psoriasis patients (Gelfand et al., 2006). These models will be helpful in identifying the cellular mechanisms underlying CVD comorbidities.
The newest murine model of psoriasis overexpresses a serine protease, called kallikrein 6 (KLK6). Klk6 mice (Klk6+) develop severe psoriasiform dermatitis and arthritis that are Klk6- and protease-activated receptor 1- (PAR1; F2R) dependent (Billi et al., 2020). Other models develop PsA-like disease, including KC-overexpression of constitutively active Stat3c (Yamamoto et al., 2015), RAC1 (Winge and Marinkovich, 2019), and Il17a (Croxford et al., 2014) and KC-deletion of JunB/c-Jun (Uluckan et al., 2016, Zenz et al., 2005). These models demonstrate how epidermal factors can cause arthritis.
Precision medicine
While major advances in the treatment of psoriasis have been made over the last few decades, many patients are treatment refractory, especially those with PsA (Ritchlin and Scher, 2019), and achieving long term remission remains a significant challenge. Precision medicine can be used to address these gaps in care, by tailoring treatment based on individual patient characteristics.
Clinical features, serologic factors, and “omics” datasets can help us classify patients into subpopulations based on their likelihood to respond to a specific therapy or their probability of developing systemic comorbidities (van Vugt et al., 2018). For example, patients with nail and scalp involvement have a higher likelihood of developing PsA (Scher et al., 2019, Wilson et al., 2009). Factors such as therapeutic levels and neutralizing anti-drug antibodies levels can also help guide clinicians when assessing the initial response and sustained efficacy of a drug (Loeff et al., 2020, Pan et al., 2020, Tsakok et al., 2019). However, in the near future clinicians will be able to utilize more sophisticated approaches that integrate clinical phenotypic data with predictive or diagnostic classifiers constructed from “omic” datasets, such as for the transcriptome (Le et al., 2019, Li et al., 2014, Merleev et al., 2018, Tsoi et al., 2019), genome (Supplementary Table 1), microbiome (Supplementary Table 1), lipidome (Sorokin et al., 2018, Zeng et al., 2017), glycome (Li et al., 2019, Maverakis et al., 2015, Park et al., 2018), proteome (Broome et al., 2003, Carlen et al., 2005), and metabolome (Alonso et al., 2016, Kamleh et al., 2015, Zeng et al., 2017). Recent technologies such as single cell RNA-seq have begun to identify novel cell subsets in the skin and synovium (Cheng et al., 2018, Croft et al., 2019, Liu J. et al., 2020, Orange et al., 2018, Stephenson et al., 2018). Already, some promising biomarkers and predictive models are starting to emerge (Supplementary Table 2).
Further advancements in “omic” datasets will give rise to more precise clinically relevant psoriasis models in the future. Complementing these technologies are the advances driven by big data analysis and machine learning, which has been applied to improve the diagnostic accuracy of tissue pathology (Correa da Rosa et al., 2017, Emam et al., 2020, Foulkes et al., 2019, Orange et al., 2018, Tomalin et al., 2020, Zhang et al., 2019). The last critical element is to improve high throughput capture of multimodal clinical data from the EMR, a major requirement in the development of precision medicine (Haendel et al., 2018).
Conclusion
The latest developments in psoriatic research suggest that a multitude of interconnected factors is responsible for the heterogeneous presentation of psoriatic disease (Figure 1). Numerous genetic variants and the interactions occurring in the microenvironment between immune cells, keratinocytes, osteoclasts, and the microbiome determine the development of psoriatic disease, disease severity, therapeutic response, and the development of comorbidities such as inflammatory arthritis and cardiometabolic conditions. These advances have not only revealed novel therapeutic targets, but also provide data to optimize treatment for the individual patient. The next frontier lies in harnessing this information in the clinical setting to deliver more timely, effective, and personalized care.
Figure 1. The multifactorial pathogenesis of psoriatic disease.
The development of psoriasis and psoriatic arthritis is influenced by a combination of genetic risk loci (red), many of which are involved in regulation of IL-23 receptor signaling and the NF-kB pathway, imbalances in Treg/T17 immunity (blue), as well as changes in the gut and cutaneous microbiome (yellow). Illustration assistance provided by Heather McDonald, BioSerendipity, LLC, Elkridge, MD.
Supplementary Material
Acknowledgements:
The authors would like to thank Jerry Bagel, Jackie Domire, Joel Gelfand, George Gondo, Alice Gottlieb, Samantha Koons, and Gil Yosipovitch for review of the manuscript. Dr. Yan is supported by a grant from the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Dr. Gudjonsson is supported by P30-AR075043 and R01-AR060802. Dr. Plazyo is supported by (T32 AR007197). Dr. Scher is supported by grants from NIH/NIAMS R01AR074500, the Snyder Family Foundation, the Riley Family Foundation, and the National Psoriasis Foundation. Dr. Ward is supported by the following awards from the National Institutes of Health: R01-AR073196, P50-AR070590, R01-AR069071. Dr. Ritchlin is supported by a grant from the NIH (R01 AR0609000). Dr. Liao is supported by grants from the National Institutes of Health U01-AI119125 and National Psoriasis Foundation.
Conflicts of interest:
Dr. Gudjonsson has received grant support from AbbVie, Almirall, BMS/Celgene Novartis, Eli Lilly, and Kiowa-Kirin, and served as an advisor to Almirall, Novartis, Eli Lilly, and AnaptysBio. Dr. Scher has consulted for Abbvie, Janssen, Novartis, Sanofi, Eli Lilly, UCB, Amgen, Pfizer and received research grant support from Pfizer, Amgen.
Dr. Liao has received research grant support from Abbvie, Amgen, Janssen, Novartis, Pfizer, Regeneron, and TRex Bio. Dr. Bell is an employee of the National Psoriasis Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Alonso A, Julia A, Vinaixa M, Domenech E, Fernandez-Nebro A, Canete JD, et al. Urine metabolome profiling of immune-mediated inflammatory diseases. BMC Med 2016;14(1):133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and obesity: a systematic review and meta-analysis of observational studies. Nutr Diabetes 2012;2:e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Armstrong AW, Harskamp CT, Dhillon JS, Armstrong EJ. Psoriasis and smoking: a systematic review and meta-analysis. Br J Dermatol 2014;170(2):304–14. [DOI] [PubMed] [Google Scholar]
- Balak DM, Hajdarbegovic E. Drug-induced psoriasis: clinical perspectives. Psoriasis (Auckl) 2017;7:87–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baurecht H, Hotze M, Brand S, Buning C, Cormican P, Corvin A, et al. Genome-wide comparative analysis of atopic dermatitis and psoriasis gives insight into opposing genetic mechanisms. Am J Hum Genet 2015;96(1):104–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belasco J, Wei N. Psoriatic Arthritis: What is Happening at the Joint? Rheumatol Ther 2019;6(3):305–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billi AC, Ludwig JE, Fritz Y, Rozic R, Swindell WR, Tsoi LC, et al. KLK6 expression in skin induces PAR1-mediated psoriasiform dermatitis and inflammatory joint disease. J Clin Invest 2020;130(6):3151–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowes J, Budu-Aggrey A, Huffmeier U, Uebe S, Steel K, Hebert HL, et al. Dense genotyping of immune-related susceptibility loci reveals new insights into the genetics of psoriatic arthritis. Nat Commun 2015;6:6046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyman O, Hefti HP, Conrad C, Nickoloff BJ, Suter M, Nestle FO. Spontaneous development of psoriasis in a new animal model shows an essential role for resident T cells and tumor necrosis factor-alpha. J Exp Med 2004;199(5):731–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brenaut E, Horreau C, Pouplard C, Barnetche T, Paul C, Richard MA, et al. Alcohol consumption and psoriasis: a systematic literature review. J Eur Acad Dermatol Venereol 2013;27 Suppl 3:30–5. [DOI] [PubMed] [Google Scholar]
- Broome AM, Ryan D, Eckert RL. S100 protein subcellular localization during epidermal differentiation and psoriasis. J Histochem Cytochem 2003;51(5):675–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Budu-Aggrey A, Brumpton B, Tyrrell J, Watkins S, Modalsli EH, Celis-Morales C, et al. Evidence of a causal relationship between body mass index and psoriasis: A mendelian randomization study. PLoS Med 2019;16(1):e1002739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bullard DC, Scharffetter-Kochanek K, McArthur MJ, Chosay JG, McBride ME, Montgomery CA, et al. A polygenic mouse model of psoriasiform skin disease in CD18-deficient mice. Proc Natl Acad Sci USA 1996;93(5):2116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cambre I, Gaublomme D, Burssens A, Jacques P, Schryvers N, De Muynck A, et al. Mechanical strain determines the site-specific localization of inflammation and tissue damage in arthritis. Nat Commun 2018;9(1):4613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlen LM, Sanchez F, Bergman AC, Becker S, Hirschberg D, Franzen B, et al. Proteome analysis of skin distinguishes acute guttate from chronic plaque psoriasis. J Invest Dermatol 2005;124(1):63–9. [DOI] [PubMed] [Google Scholar]
- Caruso R, Botti E, Sarra M, Esposito M, Stolfi C, Diluvio L, et al. Involvement of interleukin-21 in the epidermal hyperplasia of psoriasis. Nat Med 2009;15(9):1013–5. [DOI] [PubMed] [Google Scholar]
- Ceccarelli M, Venanzi Rullo E, Vaccaro M, Facciolà A, d’Aleo F, Paolucci IA, et al. HIV-associated psoriasis: Epidemiology, pathogenesis, and management. Dermatol Ther 2019;32(2):e12806. [DOI] [PubMed] [Google Scholar]
- Chan JR, Blumenschein W, Murphy E, Diveu C, Wiekowski M, Abbondanzo S, et al. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med 2006;203(12):2577–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen H, Hayashi G, Lai OY, Dilthey A, Kuebler PJ, Wong TV, et al. Psoriasis patients are enriched for genetic variants that protect against HIV-1 disease. PLoS Genet 2012;8(2):e1002514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng H, Li Y, Zuo XB, Tang HY, Tang XF, Gao JP, et al. Identification of a missense variant in LNPEP that confers psoriasis risk. J Invest Dermatol 2014;134(2):359–65. [DOI] [PubMed] [Google Scholar]
- Cheng JB, Sedgewick AJ, Finnegan AI, Harirchian P, Lee J, Kwon S, et al. Transcriptional Programming of Normal and Inflamed Human Epidermis at Single-Cell Resolution. Cell Rep 2018;25(4):871–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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(11):2399–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conrad C, Di Domizio J, Mylonas A, Belkhodja C, Demaria O, Navarini AA, et al. TNF blockade induces a dysregulated type I interferon response without autoimmunity in paradoxical psoriasis. Nat Commun 2018;9(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Correa da Rosa J, Kim J, Tian S, Tomalin LE, Krueger JG, Suarez-Farinas M. Shrinking the Psoriasis Assessment Gap: Early Gene-Expression Profiling Accurately Predicts Response to Long-Term Treatment. J Invest Dermatol 2017;137(2):305–12. [DOI] [PubMed] [Google Scholar]
- Croft AP, Campos J, Jansen K, Turner JD, Marshall J, Attar M, et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 2019;570(7760):246–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Croxford AL, Karbach S, Kurschus FC, Wortge S, Nikolaev A, Yogev N, et al. IL-6 regulates neutrophil microabscess formation in IL-17A-driven psoriasiform lesions. J Invest Dermatol 2014;134(3):728–35. [DOI] [PubMed] [Google Scholar]
- Dai YX, Wang SC, Chou YJ, Chang YT, Chen TJ, Li CP, et al. Smoking, but not alcohol, is associated with risk of psoriasis in a Taiwanese population-based cohort study. J Am Acad Dermatol 2019;80(3):727–34. [DOI] [PubMed] [Google Scholar]
- Dand N, Duckworth M, Baudry D, Russell A, Curtis CJ, Lee SH, et al. HLA-C*06:02 genotype is a predictive biomarker of biologic treatment response in psoriasis. J Allergy Clin Immunol 2019;143(6):2120–30. [DOI] [PubMed] [Google Scholar]
- Dand N, Mucha S, Tsoi LC, Mahil SK, Stuart PE, Arnold A, et al. Exome-wide association study reveals novel psoriasis susceptibility locus at TNFSF15 and rare protective alleles in genes contributing to type I IFN signalling. Hum Mol Genet 2017;26(21):4301–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Cid R, Riveira-Munoz E, Zeeuwen PL, Robarge J, Liao W, Dannhauser EN, et al. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nat Genet 2009;41(2):211–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duvic M, Johnson TM, Rapini RP, Freese T, Brewton G, Rios A. Acquired immunodeficiency syndrome-associated psoriasis and Reiter’s syndrome. Arch Dermatol 1987;123(12):1622–32. [PubMed] [Google Scholar]
- Ellinghaus D, Ellinghaus E, Nair RP, Stuart PE, Esko T, Metspalu A, et al. Combined analysis of genome-wide association studies for Crohn disease and psoriasis identifies seven shared susceptibility loci. Am J Hum Genet 2012;90(4):636–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ellinghaus E, Ellinghaus D, Stuart PE, Nair RP, Debrus S, Raelson JV, et al. Genome-wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet 2010;42(11):991–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emam S, Du AX, Surmanowicz P, Thomsen SF, Greiner R, Gniadecki R. Predicting the long-term outcomes of biologics in patients with psoriasis using machine learning. Br J Dermatol 2020;182(5):1305–7. [DOI] [PubMed] [Google Scholar]
- Emdin CA, Khera AV, Kathiresan S. Mendelian Randomization. Jama 2017;318(19):1925–6. [DOI] [PubMed] [Google Scholar]
- Feng BJ, Sun LD, Soltani-Arabshahi R, Bowcock AM, Nair RP, Stuart P, et al. Multiple Loci within the major histocompatibility complex confer risk of psoriasis. PLoS Genet 2009;5(8):e1000606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FitzGerald O, Haroon M, Giles JT, Winchester R. Concepts of pathogenesis in psoriatic arthritis: genotype determines clinical phenotype. Arthritis Res Ther 2015;17:115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foster AM, Baliwag J, Chen CS, Guzman AM, Stoll SW, Gudjonsson JE, et al. IL-36 promotes myeloid cell infiltration, activation, and inflammatory activity in skin. Journal of immunology 2014;192(12):6053–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Foulkes AC, Watson DS, Carr DF, Kenny JG, Slidel T, Parslew R, et al. A Framework for Multi-Omic Prediction of Treatment Response to Biologic Therapy for Psoriasis. J Invest Dermatol 2019;139(1):100–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fraki JE, Briggaman RA, Lazarus GS. Uninvolved skin from psoriatic patients develops signs of involved psoriatic skin after being grafted onto nude mice. Science 1982;215(4533):685–7. [DOI] [PubMed] [Google Scholar]
- Frey S, Sticht H, Wilsmann-Theis D, Gerschutz A, Wolf K, Lohr S, et al. Rare Loss-of-Function Mutation in SERPINA3 in Generalized Pustular Psoriasis. J Invest Dermatol 2020. [DOI] [PubMed] [Google Scholar]
- 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(11):1075–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013;504(7480):446–50. [DOI] [PubMed] [Google Scholar]
- Gates AH, Karasek M. Hereditary Absence of Sebaceous Glands in the Mouse. Science 1965;148(3676):1471–3. [DOI] [PubMed] [Google Scholar]
- Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB. Risk of myocardial infarction in patients with psoriasis. Jama 2006;296(14):1735–41. [DOI] [PubMed] [Google Scholar]
- Genetic Analysis of Psoriasis C, the Wellcome Trust Case Control C, Strange A, Capon F, Spencer CC, Knight J, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet 2010;42(11):985–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golden JB, Groft SG, Squeri MV, Debanne SM, Ward NL, McCormick TS, et al. Chronic Psoriatic Skin Inflammation Leads to Increased Monocyte Adhesion and Aggregation. J Immunol 2015;195(5):2006–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldminz AM, Au SC, Kim N, Gottlieb AB, Lizzul PF. NF-κB: an essential transcription factor in psoriasis. J Dermatol Sci 2013;69(2):89–94. [DOI] [PubMed] [Google Scholar]
- Gracey E, Burssens A, Cambre I, Schett G, Lories R, McInnes IB, et al. Tendon and ligament mechanical loading in the pathogenesis of inflammatory arthritis. Nat Rev Rheumatol 2020;16(4):193–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groeger D, O’Mahony L, Murphy EF, Bourke JF, Dinan TG, Kiely B, et al. Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes 2013;4(4):325–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gudjonsson JE, Johnston A, Dyson M, Valdimarsson H, Elder JT. Mouse models of psoriasis. J Invest Dermatol 2007;127(6):1292–308. [DOI] [PubMed] [Google Scholar]
- Gudjonsson JE, Karason A, Runarsdottir EH, Antonsdottir AA, Hauksson VB, Jonsson HH, et al. Distinct clinical differences between HLA-Cw*0602 positive and negative psoriasis patients--an analysis of 1019 HLA-C- and HLA-B-typed patients. J Invest Dermatol 2006;126(4):740–5. [DOI] [PubMed] [Google Scholar]
- Guilloteau K, Paris I, Pedretti N, Boniface K, Juchaux F, Huguier V, et al. Skin Inflammation Induced by the Synergistic Action of IL-17A, IL-22, Oncostatin M, IL-1{alpha}, and TNF-{alpha} Recapitulates Some Features of Psoriasis. J Immunol 2010;184(9):5263–70. [DOI] [PubMed] [Google Scholar]
- Gupta R, Ahn R, Lai K, Mullins E, Debbaneh M, Dimon M, et al. Landscape of Long Noncoding RNAs in Psoriatic and Healthy Skin. J Invest Dermatol 2016;136(3):603–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haendel MA, Chute CG, Robinson PN. Classification, Ontology, and Precision Medicine. N Engl J Med 2018;379(15):1452–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han L, Sugiyama H, Zhang Q, Yan K, Fang X, McCormick TS, et al. Phenotypical analysis of ectoenzymes CD39/CD73 and adenosine receptor 2A in CD4(+) CD25(high) Foxp3(+) regulatory T-cells in psoriasis. Australas J Dermatol 2018;59(1):e31–e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haskamp S, Bruns H, Hahn M, Hoffmann M, Gregor A, Lohr S, et al. Myeloperoxidase Modulates Inflammation in Generalized Pustular Psoriasis and Additional Rare Pustular Skin Diseases. Am J Hum Genet 2020;107(3):527–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawkes JE, Gudjonsson JE, Ward NL. The Snowballing Literature on Imiquimod-Induced Skin Inflammation in Mice: A Critical Appraisal. J Invest Dermatol 2017;137(3):546–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herbert D, Franz S, Popkova Y, Anderegg U, Schiller J, Schwede K, et al. High-Fat Diet Exacerbates Early Psoriatic Skin Inflammation Independent of Obesity: Saturated Fatty Acids as Key Players. J Invest Dermatol 2018;138(9):1999–2009. [DOI] [PubMed] [Google Scholar]
- Hile G, Kahlenberg JM, Gudjonsson JE. Recent genetic advances in innate immunity of psoriatic arthritis. Clin Immunol 2020;214:108405. [DOI] [PubMed] [Google Scholar]
- HogenEsch H, Gijbels MJ, Offerman E, van Hooft J, van Bekkum DW, Zurcher C. A spontaneous mutation characterized by chronic proliferative dermatitis in C57BL mice. Am J Pathol 1993;143(3):972–82. [PMC free article] [PubMed] [Google Scholar]
- Hollox EJ, Huffmeier U, Zeeuwen PL, Palla R, Lascorz J, Rodijk-Olthuis D, et al. Psoriasis is associated with increased beta-defensin genomic copy number. Nat Genet 2008;40(1):23–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ikebuchi Y, Aoki S, Honma M, Hayashi M, Sugamori Y, Khan M, et al. Coupling of bone resorption and formation by RANKL reverse signalling. Nature 2018;561(7722):195–200. [DOI] [PubMed] [Google Scholar]
- Iwakura Y, Ishigame H. The IL-23/IL-17 axis in inflammation. J Clin Invest 2006;116(5):1218–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jacques P, Lambrecht S, Verheugen E, Pauwels E, Kollias G, Armaka M, et al. Proof of concept: enthesitis and new bone formation in spondyloarthritis are driven by mechanical strain and stromal cells. Ann Rheum Dis 2014;73(2):437–45. [DOI] [PubMed] [Google Scholar]
- Jin Y, Yang S, Zhang F, Kong Y, Xiao F, Hou Y, et al. Combined effects of HLA-Cw6 and cigarette smoking in psoriasis vulgaris: a hospital-based case-control study in China. J Eur Acad Dermatol Venereol 2009;23(2):132–7. [DOI] [PubMed] [Google Scholar]
- Johnston A, Fritz Y, Dawes SM, Diaconu D, Al-Attar PM, Guzman AM, et al. Keratinocyte overexpression of IL-17C promotes psoriasiform skin inflammation. Journal of immunology 2013;190(5):2252–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jordan CT, Cao L, Roberson ED, Duan S, Helms CA, Nair RP, et al. Rare and common variants in CARD14, encoding an epidermal regulator of NF-kappaB, in psoriasis. Am J Hum Genet 2012;90(5):796–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamleh MA, Snowden SG, Grapov D, Blackburn GJ, Watson DG, Xu N, et al. LC-MS metabolomics of psoriasis patients reveals disease severity-dependent increases in circulating amino acids that are ameliorated by anti-TNFalpha treatment. J Proteome Res 2015;14(1):557–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kanemaru K, Matsuyuki A, Nakamura Y, Fukami K. Obesity exacerbates imiquimod-induced psoriasis-like epidermal hyperplasia and interleukin-17 and interleukin-22 production in mice. Exp Dermatol 2015;24(6):436–42. [DOI] [PubMed] [Google Scholar]
- Karbach S, Croxford AL, Oelze M, Schuler R, Minwegen D, Wegner J, et al. Interleukin 17 drives vascular inflammation, endothelial dysfunction, and arterial hypertension in psoriasis-like skin disease. Arterioscler Thromb Vasc Biol 2014;34(12):2658–68. [DOI] [PubMed] [Google Scholar]
- Kaur M, Conde J, Willard JD, Taylor S, Camacho F, Fleischer AB, et al. A Randomized, Double-Blind Clinical Trial of a Probiotic Nutritional Intervention in the Treatment of Mild to Moderate Non-Scalp Psoriasis. Psoriasis Forum 2007;13a(1):12–5. [Google Scholar]
- Kim GK, Del Rosso JQ. Drug-provoked psoriasis: is it drug induced or drug aggravated?: understanding pathophysiology and clinical relevance. J Clin Aesthet Dermatol 2010;3(1):32–8. [PMC free article] [PubMed] [Google Scholar]
- Kragsnaes MS, Kjeldsen J, Horn HC, Munk HL, Pedersen FM, Holt HM, et al. Efficacy and safety of faecal microbiota transplantation in patients with psoriatic arthritis: protocol for a 6-month, double-blind, randomised, placebo-controlled trial. BMJ Open 2018;8(4):e019231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krämer U, Esser C. Cigarette smoking, metabolic gene polymorphism, and psoriasis. J Invest Dermatol 2006;126(3):693–4; author reply 5. [DOI] [PubMed] [Google Scholar]
- Krueger GG, Chambers DA, Shelby J. Involved and uninvolved skin from psoriatic subjects: are they equally diseased? Assessment by skin transplanted to congenitally athymic (nude) mice. J Clin Invest 1981;68(6):1548–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kunstfeld R, Hirakawa S, Hong YK, Schacht V, Lange-Asschenfeldt B, Velasco P, et al. Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood 2004;104(4):1048–57. [DOI] [PubMed] [Google Scholar]
- Le ST, Merleev AA, Luxardi G, Shimoda M, Adamopoulos IE, Tsoi LC, et al. 2D visualization of the psoriasis transcriptome fails to support the existence of dual-secreting IL-17A/IL-22 Th17 T cells. Front Immunol 2019:In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li AG, Wang D, Feng XH, Wang XJ. Latent TGFbeta1 overexpression in keratinocytes results in a severe psoriasis-like skin disorder. EMBO J 2004;23(8):1770–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B, Tsoi LC, Swindell WR, Gudjonsson JE, Tejasvi T, Johnston A, et al. Transcriptome analysis of psoriasis in a large case-control sample: RNA-seq provides insights into disease mechanisms. J Invest Dermatol 2014;134(7):1828–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Wu Y, Chen G, Yang Y, Zhou D, Zhang Z, et al. Deletion of the late cornified envelope genes LCE3C and LCE3B is associated with psoriasis in a Chinese population. J Invest Dermatol 2011;131(8):1639–43. [DOI] [PubMed] [Google Scholar]
- Li Q, Chandran V, Tsoi L, O’Rielly D, Nair RP, Gladman D, et al. Quantifying Differences in Heritability among Psoriatic Arthritis (PsA), Cutaneous Psoriasis (PsC) and Psoriasis vulgaris (PsV). Sci Rep 2020;10(1):4925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Q, Kailemia MJ, Merleev AA, Xu G, Serie D, Danan LM, et al. Site-Specific Glycosylation Quantitation of 50 Serum Glycoproteins Enhanced by Predictive Glycopeptidomics for Improved Disease Biomarker Discovery. Anal Chem 2019;91(8):5433–45. [DOI] [PubMed] [Google Scholar]
- Li X, Wang J, Zhan Z, Li S, Zheng Z, Wang T, et al. Inflammation Intensity-Dependent Expression of Osteoinductive Wnt Proteins Is Critical for Ectopic New Bone Formation in Ankylosing Spondylitis. Arthritis Rheumatol 2018;70(7):1056–70. [DOI] [PubMed] [Google Scholar]
- Liang Y, Sarkar MK, Tsoi LC, Gudjonsson JE. Psoriasis: a mixed autoimmune and autoinflammatory disease. Curr Opin Immunol 2017;49:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Chang HW, Huang ZM, Nakamura M, Sekhon S, Ahn R, et al. Single-cell RNA sequencing of psoriatic skin identifies pathogenic Tc17 cell subsets and reveals distinctions between CD8(+) T cells in autoimmunity and cancer. J Allergy Clin Immunol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Jarjour W, Olsen N, Zheng SG. Traitor or warrior-Treg cells sneaking into the lesions of psoriatic arthritis. Clin Immunol 2020;215:108425. [DOI] [PubMed] [Google Scholar]
- Loeff FC, Tsakok T, Dijk L, Hart MH, Duckworth M, Baudry D, et al. Clinical impact of antibodies against ustekinumab in psoriasis: an observational, cross-sectional, multicenter study. J Invest Dermatol 2020. [DOI] [PubMed] [Google Scholar]
- Lu Y, Chen H, Nikamo P, Qi Low H, Helms C, Seielstad M, et al. Association of cardiovascular and metabolic disease genes with psoriasis. J Invest Dermatol 2013;133(3):836–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma B, Hottiger MO. Crosstalk between Wnt/β-Catenin and NF-κB Signaling Pathway during Inflammation. Front Immunol 2016;7:378-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mahil SK, McSweeney SM, Kloczko E, McGowan B, Barker JN, Smith CH. Does weight loss reduce the severity and incidence of psoriasis or psoriatic arthritis? A Critically Appraised Topic. Br J Dermatol 2019;181(5):946–53. [DOI] [PubMed] [Google Scholar]
- Marrakchi S, Guigue P, Renshaw BR, Puel A, Pei XY, Fraitag S, et al. Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N Engl J Med 2011;365(7):620–8. [DOI] [PubMed] [Google Scholar]
- Maverakis E, Kim K, Shimoda M, Gershwin ME, Patel F, Wilken R, et al. Glycans in the immune system and The Altered Glycan Theory of Autoimmunity: a critical review. J Autoimmun 2015;57:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mazloom SE, Yan D, Hu JZ, Ya J, Husni ME, Warren CB, et al. TNF-α Inhibitor-Induced psoriasis: A decade of experience at the Cleveland Clinic. J Am Acad Dermatol 2018. [DOI] [PubMed] [Google Scholar]
- Mease PJ, Gladman DD, Papp KA, Khraishi MM, Thaçi D, Behrens F, et al. Prevalence of rheumatologist-diagnosed psoriatic arthritis in patients with psoriasis in European/North American dermatology clinics. J Am Acad Dermatol 2013;69(5):729–35. [DOI] [PubMed] [Google Scholar]
- Menon B, Gullick NJ, Walter GJ, Rajasekhar M, Garrood T, Evans HG, et al. Interleukin-17+CD8+ T cells are enriched in the joints of patients with psoriatic arthritis and correlate with disease activity and joint damage progression. Arthritis Rheumatol 2014;66(5):1272–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menon K, Van Voorhees AS, Bebo BF Jr., Gladman DD, Hsu S, Kalb RE, et al. Psoriasis in patients with HIV infection: from the medical board of the National Psoriasis Foundation. J Am Acad Dermatol 2010;62(2):291–9. [DOI] [PubMed] [Google Scholar]
- Mercurio L, Morelli M, Scarponi C, Eisenmesser EZ, Doti N, Pagnanelli G, et al. IL-38 has an anti-inflammatory action in psoriasis and its expression correlates with disease severity and therapeutic response to anti-IL-17A treatment. Cell Death Dis 2018;9(11):1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Merleev AA, Marusina AI, Ma C, Elder JT, Tsoi LC, Raychaudhuri SP, et al. Meta-analysis of RNA sequencing datasets reveals an association between TRAJ23, psoriasis, and IL-17A. JCI Insight 2018;3(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mossner R, Wilsmann-Theis D, Oji V, Gkogkolou P, Lohr S, Schulz P, et al. The genetic basis for most patients with pustular skin disease remains elusive. Br J Dermatol 2018;178(3):740–8. [DOI] [PubMed] [Google Scholar]
- Nair RP, Duffin KC, Helms C, Ding J, Stuart PE, Goldgar D, et al. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat Genet 2009;41(2):199–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nair RP, Stuart PE, Nistor I, Hiremagalore R, Chia NVC, Jenisch S, et al. Sequence and haplotype analysis supports HLA-C as the psoriasis susceptibility 1 gene. Am J Hum Genet 2006;78(5):827–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Navarro-López V, Martínez-Andrés A, Ramírez-Boscá A, Ruzafa-Costas B, Núñez-Delegido E, Carrión-Gutiérrez MA, et al. Efficacy and Safety of Oral Administration of a Mixture of Probiotic Strains in Patients with Psoriasis: A Randomized Controlled Clinical Trial. Acta Derm Venereol 2019;99(12):1078–84. [DOI] [PubMed] [Google Scholar]
- Nestle FO, Conrad C, Tun-Kyi A, Homey B, Gombert M, Boyman O, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 2005;202(1):135–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nickoloff BJ, Wrone-Smith T. Injection of pre-psoriatic skin with CD4+ T cells induces psoriasis. Am J Pathol 1999;155(1):145–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ogawa K, Stuart PE, Tsoi LC, Suzuki K, Nair RP, Mochizuki H, et al. A Transethnic Mendelian Randomization Study Identifies Causality of Obesity on Risk of Psoriasis. J Invest Dermatol 2019;139(6):1397–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada K, Matsushima Y, Mizutani K, Yamanaka K. The Role of Gut Microbiome in Psoriasis: Oral Administration of Staphylococcus aureus and Streptococcus danieliae Exacerbates Skin Inflammation of Imiquimod-Induced Psoriasis-Like Dermatitis. Int J Mol Sci 2020;21(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada Y, Han B, Tsoi LC, Stuart PE, Ellinghaus E, Tejasvi T, et al. Fine mapping major histocompatibility complex associations in psoriasis and its clinical subtypes. Am J Hum Genet 2014;95(2):162–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onoufriadis A, Simpson MA, Pink AE, Di Meglio P, Smith CH, Pullabhatla V, et al. Mutations in IL36RN/IL1F5 are associated with the severe episodic inflammatory skin disease known as generalized pustular psoriasis. Am J Hum Genet 2011;89(3):432–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orange DE, Agius P, DiCarlo EF, Robine N, Geiger H, Szymonifka J, et al. Identification of Three Rheumatoid Arthritis Disease Subtypes by Machine Learning Integration of Synovial Histologic Features and RNA Sequencing Data. Arthritis Rheumatol 2018;70(5):690–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paine A, Ritchlin C. Altered Bone Remodeling in Psoriatic Disease: New Insights and Future Directions. Calcif Tissue Int 2018;102(5):559–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pan S, Tsakok T, Dand N, Lonsdale DO, Loeff FC, Bloem K, et al. Using Real-World Data to Guide Ustekinumab Dosing Strategies for Psoriasis: A Prospective Pharmacokinetic-Pharmacodynamic Study. Clin Transl Sci 2020;13(2):400–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park DD, Xu G, Wong M, Phoomak C, Liu M, Haigh NE, et al. Membrane glycomics reveal heterogeneity and quantitative distribution of cell surface sialylation. Chem Sci 2018;9(29):6271–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patrick MT, Stuart PE, Raja K, Chi S, He Z, Voorhees JJ, et al. Integrative Approach to Reveal Cell Type Specificity and Gene Candidates for Psoriatic Arthritis Outside the MHC. Front Genet 2019;10:304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patrick MT, Stuart PE, Raja K, Gudjonsson JE, Tejasvi T, Yang J, et al. Genetic signature to provide robust risk assessment of psoriatic arthritis development in psoriasis patients. Nat Commun 2018;9(1):4178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penkava F, Velasco-Herrera MDC, Young MD, Yager N, Nwosu LN, Pratt AG, et al. Single-cell sequencing reveals clonal expansions of pro-inflammatory synovial CD8 T cells expressing tissue-homing receptors in psoriatic arthritis. Nat Commun 2020;11(1):4767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pezzolo E, Naldi L. The relationship between smoking, psoriasis and psoriatic arthritis. Expert Rev Clin Immunol 2019;15(1):41–8. [DOI] [PubMed] [Google Scholar]
- Polese B, Zhang H, Thurairajah B, King IL. Innate Lymphocytes in Psoriasis. Front Immunol 2020;11:242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Prinz JC. Psoriasis vulgaris--a sterile antibacterial skin reaction mediated by cross-reactive T cells? An immunological view of the pathophysiology of psoriasis. Clin Exp Dermatol 2001;26(4):326–32. [DOI] [PubMed] [Google Scholar]
- Qiao P, Shi Q, Zhang R, E L, Wang P, Wang J, et al. Psoriasis Patients Suffer From Worse Periodontal Status-A Meta-Analysis. Front Med (Lausanne) 2019;6:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol 2014;70(3):512–6. [DOI] [PubMed] [Google Scholar]
- Ramirez-Carrozzi V, Sambandam A, Luis E, Lin Z, Jeet S, Lesch J, et al. IL-17C regulates the innate immune function of epithelial cells in an autocrine manner. Nature immunology 2011;12(12):1159–66. [DOI] [PubMed] [Google Scholar]
- Rath HC, Herfarth HH, Ikeda JS, Grenther WB, Hamm TE Jr., Balish E, et al. Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. J Clin Invest 1996;98(4):945–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Razani B, Whang MI, Kim FS, Nakamura MC, Sun X, Advincula R, et al. Non-catalytic ubiquitin binding by A20 prevents psoriatic arthritis-like disease and inflammation. Nature immunology 2020;21(4):422–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reinhardt A, Yevsa T, Worbs T, Lienenklaus S, Sandrock I, Oberdorfer L, et al. Interleukin-23-Dependent gamma/delta T Cells Produce Interleukin-17 and Accumulate in the Enthesis, Aortic Valve, and Ciliary Body in Mice. Arthritis Rheumatol 2016;68(10):2476–86. [DOI] [PubMed] [Google Scholar]
- Ritchlin C, Scher JU. Strategies to Improve Outcomes in Psoriatic Arthritis. Curr Rheumatol Rep 2019;21(12):72. [DOI] [PubMed] [Google Scholar]
- Scher JU, Nayak RR, Ubeda C, Turnbaugh PJ, Abramson SB. Pharmacomicrobiomics in inflammatory arthritis: gut microbiome as modulator of therapeutic response. Nat Rev Rheumatol 2020;16(5):282–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scher JU, Ogdie A, Merola JF, Ritchlin C. Preventing psoriatic arthritis: focusing on patients with psoriasis at increased risk of transition. Nat Rev Rheumatol 2019;15(3):153–66. [DOI] [PubMed] [Google Scholar]
- Scher JU, Ubeda C, Artacho A, Attur M, Isaac S, Reddy SM, et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol 2015;67(1):128–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schon MP, Manzke V, Erpenbeck L. Animal models of psoriasis-highlights and drawbacks. J Allergy Clin Immunol 2020. [DOI] [PubMed] [Google Scholar]
- Schuler R, Brand A, Klebow S, Wild J, Veras FP, Ullmann E, et al. Antagonization of IL-17A Attenuates Skin Inflammation and Vascular Dysfunction in Mouse Models of Psoriasis. J Invest Dermatol 2019;139(3):638–47. [DOI] [PubMed] [Google Scholar]
- Selvanderan SP, Goldblatt F, Nguyen NQ, Costello SP. Faecal microbiota transplantation for Clostridium difficile infection resulting in a decrease in psoriatic arthritis disease activity. Clin Exp Rheumatol 2019;37(3):514–5. [PubMed] [Google Scholar]
- Setta-Kaffetzi N, Navarini AA, Patel VM, Pullabhatla V, Pink AE, Choon SE, et al. Rare pathogenic variants in IL36RN underlie a spectrum of psoriasis-associated pustular phenotypes. J Invest Dermatol 2013;133(5):1366–9. [DOI] [PubMed] [Google Scholar]
- Setta-Kaffetzi N, Simpson MA, Navarini AA, Patel VM, Lu HC, Allen MH, et al. AP1S3 mutations are associated with pustular psoriasis and impaired Toll-like receptor 3 trafficking. Am J Hum Genet 2014;94(5):790–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sherlock JP, Joyce-Shaikh B, Turner SP, Chao CC, Sathe M, Grein J, et al. IL-23 induces spondyloarthropathy by acting on ROR-gammat+ CD3+CD4-CD8- entheseal resident T cells. Nat Med 2012;18(7):1069–76. [DOI] [PubMed] [Google Scholar]
- Shi Z, Wu X, Yu S, Huynh M, Jena PK, Nguyen M, et al. Short-Term Exposure to a Western Diet Induces Psoriasiform Dermatitis by Promoting Accumulation of IL-17A-Producing gammadelta T Cells. J Invest Dermatol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341(6145):569–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soare A, Weber S, Maul L, Rauber S, Gheorghiu AM, Luber M, et al. Cutting Edge: Homeostasis of Innate Lymphoid Cells Is Imbalanced in Psoriatic Arthritis. J Immunol 2018;200(4):1249–54. [DOI] [PubMed] [Google Scholar]
- Sorokin AV, Domenichiello AF, Dey AK, Yuan ZX, Goyal A, Rose SM, et al. Bioactive Lipid Mediator Profiles in Human Psoriasis Skin and Blood. J Invest Dermatol 2018;138(7):1518–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steel KJA, Srenathan U, Ridley M, Durham LE, Wu SY, Ryan SE, et al. Polyfunctional, Proinflammatory, Tissue-Resident Memory Phenotype and Function of Synovial Interleukin-17A+CD8+ T Cells in Psoriatic Arthritis. Arthritis Rheumatol 2020;72(3):435–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stephenson W, Donlin LT, Butler A, Rozo C, Bracken B, Rashidfarrokhi A, et al. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat Commun 2018;9(1):791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stuart PE, Nair RP, Ellinghaus E, Ding J, Tejasvi T, Gudjonsson JE, et al. Genome-wide association analysis identifies three psoriasis susceptibility loci. Nat Genet 2010;42(11):1000–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stuart PE, Nair RP, Tsoi LC, Tejasvi T, Das S, Kang HM, et al. Genome-wide Association Analysis of Psoriatic Arthritis and Cutaneous Psoriasis Reveals Differences in Their Genetic Architecture. Am J Hum Genet 2015;97(6):816–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun LD, Cheng H, Wang ZX, Zhang AP, Wang PG, Xu JH, et al. Association analyses identify six new psoriasis susceptibility loci in the Chinese population. Nat Genet 2010;42(11):1005–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sundberg JP, Boggess D, Shultz LD, Beamer WG. The Flaky Skin (fsn) mutation chromosome? In: Sundberg JP, editor. Handbook of Mouse Mutations with Skin and Hair Abnormalities. Boca Raton: CRC Press; 1994. p. 253–68. [Google Scholar]
- Takao J, Yudate T, Das A, Shikano S, Bonkobara M, Ariizumi K, et al. Expression of NF-kappaB in epidermis and the relationship between NF-kappaB activation and inhibition of keratinocyte growth. Br J Dermatol 2003;148(4):680–8. [DOI] [PubMed] [Google Scholar]
- Tang H, Jin X, Li Y, Jiang H, Tang X, Yang X, et al. A large-scale screen for coding variants predisposing to psoriasis. Nat Genet 2014;46(1):45–50. [DOI] [PubMed] [Google Scholar]
- Telfer NR, Chalmers RJ, Whale K, Colman G. The role of streptococcal infection in the initiation of guttate psoriasis. Arch Dermatol 1992;128(1):39–42. [PubMed] [Google Scholar]
- Tiilikainen A, Lassus A, Karvonen J, Vartiainen P, Julin M. Psoriasis and HLA-Cw6. Br J Dermatol 1980;102(2):179–84. [DOI] [PubMed] [Google Scholar]
- Tomalin LE, Kim J, Correa da Rosa J, Lee J, Fitz LJ, Berstein G, et al. Early Quantification of Systemic Inflammatory Proteins Predicts Long-Term Treatment Response to Tofacitinib and Etanercept. J Invest Dermatol 2020;140(5):1026–34. [DOI] [PubMed] [Google Scholar]
- Tsakok T, Wilson N, Dand N, Loeff FC, Bloem K, Baudry D, et al. Association of Serum Ustekinumab Levels With Clinical Response in Psoriasis. JAMA Dermatol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoi LC, Rodriguez E, Degenhardt F, Baurecht H, Wehkamp U, Volks N, et al. Atopic dermatitis is an IL-13 dominant disease with greater molecular heterogeneity compared to psoriasis. J Invest Dermatol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoi LC, Spain SL, Ellinghaus E, Stuart PE, Capon F, Knight J, et al. Enhanced meta-analysis and replication studies identify five new psoriasis susceptibility loci. Nat Commun 2015;6:7001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoi LC, Spain SL, Knight J, Ellinghaus E, Stuart PE, Capon F, et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat Genet 2012;44(12):1341–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsoi LC, Stuart PE, Tian C, Gudjonsson JE, Das S, Zawistowski M, et al. Large scale meta-analysis characterizes genetic architecture for common psoriasis associated variants. Nat Commun 2017;8:15382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ucan Yarkac F, Ogrum A, Gokturk O. Effects of non-surgical periodontal therapy on inflammatory markers of psoriasis: A randomized controlled trial. J Clin Periodontol 2020;47(2):193–201. [DOI] [PubMed] [Google Scholar]
- Uluckan O, Jimenez M, Karbach S, Jeschke A, Grana O, Keller J, et al. Chronic skin inflammation leads to bone loss by IL-17-mediated inhibition of Wnt signaling in osteoblasts. Sci Transl Med 2016;8(330):330ra37. [DOI] [PubMed] [Google Scholar]
- van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. Journal of immunology 2009;182(9):5836–45. [DOI] [PubMed] [Google Scholar]
- van Vugt LJ, van den Reek J, Coenen MJH, de Jong E. A systematic review of pharmacogenetic studies on the response to biologics in patients with psoriasis. Br J Dermatol 2018;178(1):86–94. [DOI] [PubMed] [Google Scholar]
- van Vugt LJ, van den Reek J, Hannink G, Coenen MJH, de Jong E. Association of HLA-C*06:02 Status With Differential Response to Ustekinumab in Patients With Psoriasis: A Systematic Review and Meta-analysis. JAMA Dermatol 2019;155(6):708–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vasseur P, Serres L, Jegou JF, Pohin M, Delwail A, Petit-Paris I, et al. High-Fat Diet-Induced IL-17A Exacerbates Psoriasiform Dermatitis in a Mouse Model of Steatohepatitis. Am J Pathol 2016;186(9):2292–301. [DOI] [PubMed] [Google Scholar]
- Vergnano M, Mockenhaupt M, Benzian-Olsson N, Paulmann M, Grys K, Mahil SK, et al. Loss-of-Function Myeloperoxidase Mutations Are Associated with Increased Neutrophil Counts and Pustular Skin Disease. Am J Hum Genet 2020;107(3):539–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vinter H, Iversen L, Steiniche T, Kragballe K, Johansen C. Aldara(R)-induced skin inflammation: studies of patients with psoriasis. Br J Dermatol 2015;172(2):345–53. [DOI] [PubMed] [Google Scholar]
- Wang H, Peters T, Sindrilaru A, Scharffetter-Kochanek K. Key role of macrophages in the pathogenesis of CD18 hypomorphic murine model of psoriasis. J Invest Dermatol 2009;129(5):1100–14. [DOI] [PubMed] [Google Scholar]
- Wang Y, Golden JB, Fritz Y, Zhang X, Diaconu D, Camhi MI, et al. Interleukin 6 regulates psoriasiform inflammation-associated thrombosis. JCI Insight 2016;1(20):e89384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ward NL, Bhagathavula N, Johnston A, Dawes SM, Fu W, Lambert S, et al. Erlotinib-induced skin inflammation is IL-1 mediated in KC-Tie2 mice and human skin organ culture. J Invest Dermatol 2015;135(3):910–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson FC, Icen M, Crowson CS, McEvoy MT, Gabriel SE, Kremers HM. Incidence and clinical predictors of psoriatic arthritis in patients with psoriasis: a population-based study. Arthritis Rheum 2009;61(2):233–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Winge MCG, Marinkovich MP. Epidermal activation of the small GTPase Rac1 in psoriasis pathogenesis. Small GTPases 2019;10(3):163–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolfram JA, Diaconu D, Hatala DA, Rastegar J, Knutsen DA, Lowther A, et al. Keratinocyte but not endothelial cell-specific overexpression of Tie2 leads to the development of psoriasis. Am J Pathol 2009;174(4):1443–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia YP, Li B, Hylton D, Detmar M, Yancopoulos GD, Rudge JS. Transgenic delivery of VEGF to mouse skin leads to an inflammatory condition resembling human psoriasis. Blood 2003;102(1):161–8. [DOI] [PubMed] [Google Scholar]
- Yamamoto M, Nakajima K, Takaishi M, Kitaba S, Magata Y, Kataoka S, et al. Psoriatic inflammation facilitates the onset of arthritis in a mouse model. J Invest Dermatol 2015;135(2):445–53. [DOI] [PubMed] [Google Scholar]
- Yan D, Issa N, Afifi L, Jeon C, Chang HW, Liao W. The Role of the Skin and Gut Microbiome in Psoriatic Disease. Curr Dermatol Rep 2017;6(2):94–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang C, Chen M, Huang H, Li X, Qian D, Hong X, et al. Exome-Wide Rare Loss-of-Function Variant Enrichment Study of 21,347 Han Chinese Individuals Identifies Four Susceptibility Genes for Psoriasis. J Invest Dermatol 2020;140(4):799–805 e1. [DOI] [PubMed] [Google Scholar]
- Yeh NL, Hsu CY, Tsai TF, Chiu HY. Gut Microbiome in Psoriasis is Perturbed Differently During Secukinumab and Ustekinumab Therapy and Associated with Response to Treatment. Clin Drug Investig 2019;39(12):1195–203. [DOI] [PubMed] [Google Scholar]
- Yin X, Low HQ, Wang L, Li Y, Ellinghaus E, Han J, et al. Genome-wide meta-analysis identifies multiple novel associations and ethnic heterogeneity of psoriasis susceptibility. Nat Commun 2015;6:6916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan Y, Qiu J, Lin ZT, Li W, Haley C, Mui UN, et al. Identification of Novel Autoantibodies Associated With Psoriatic Arthritis. Arthritis Rheumatol 2019;71(6):941–51. [DOI] [PubMed] [Google Scholar]
- Yunusbaeva M, Valiev R, Bilalov F, Sultanova Z, Sharipova L, Yunusbayev B. Psoriasis patients demonstrate HLA-Cw*06:02 allele dosage-dependent T cell proliferation when treated with hair follicle-derived keratin 17 protein. Sci Rep 2018;8(1):6098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zákostelská Z, Málková J, Klimešová K, Rossmann P, Hornová M, Novosádová I, et al. Intestinal Microbiota Promotes Psoriasis-Like Skin Inflammation by Enhancing Th17 Response. PLoS One 2016;11(7):e0159539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng C, Wen B, Hou G, Lei L, Mei Z, Jia X, et al. Lipidomics profiling reveals the role of glycerophospholipid metabolism in psoriasis. Gigascience 2017;6(10):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zenz R, Eferl R, Kenner L, Florin L, Hummerich L, Mehic D, et al. Psoriasis-like skin disease and arthritis caused by inducible epidermal deletion of Jun proteins. Nature 2005;437(7057):369–75. [DOI] [PubMed] [Google Scholar]
- Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S, et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nature immunology 2019;20(7):928–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhen Q, Yang Z, Wang W, Li B, Bai M, Wu J, et al. Genetic Study on Small Insertions and Deletions in Psoriasis Reveals a Role in Complex Human Diseases. J Invest Dermatol 2019;139(11):2302–12 e14. [DOI] [PubMed] [Google Scholar]
- Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Research 2020;30(6):492–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zuo X, Sun L, Yin X, Gao J, Sheng Y, Xu J, et al. Whole-exome SNP array identifies 15 new susceptibility loci for psoriasis. Nat Commun 2015;6:6793. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.