Next Article in Journal
Recurrences after Pulsed Field Ablation of Atrial Fibrillation: Incidence, Mechanisms, Predictors, and Comparison with Thermal Energy
Previous Article in Journal
Evaluation of Neuromuscular Morphometry of the Vaginal Wall Using Protein Gene Product 9.5 (Pgp 9.5) and Smooth Muscle α-Actin (Sma) in Patients with Posterior Vaginal Wall Prolapse
Previous Article in Special Issue
Analysis of the Level of Adiponectin and Selected Cytokines in Patients with Knee Osteoarthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Psoriasis and Psoriatic Arthritis—Associated Genes, Cytokines, and Human Leukocyte Antigens

1
Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia
2
National Institute of Rheumatic Diseases, Nábrežie Ivana Krasku 4, 921 12 Piestany, Slovakia
*
Author to whom correspondence should be addressed.
Medicina 2024, 60(5), 815; https://doi.org/10.3390/medicina60050815
Submission received: 17 April 2024 / Revised: 13 May 2024 / Accepted: 14 May 2024 / Published: 16 May 2024
(This article belongs to the Special Issue Exploring Novel Biomarkers of Musculoskeletal Diseases)

Abstract

:
In recent years, research has intensified in exploring the genetic basis of psoriasis (PsO) and psoriatic arthritis (PsA). Genome-wide association studies (GWASs), including tools like ImmunoChip, have significantly deepened our understanding of disease mechanisms by pinpointing risk-associated genetic loci. These efforts have elucidated biological pathways involved in PsO pathogenesis, particularly those related to the innate immune system, antigen presentation, and adaptive immune responses. Specific genetic loci, such as TRAF3IP2, REL, and FBXL19, have been identified as having a significant impact on disease development. Interestingly, different genetic variants at the same locus can predispose individuals to either PsO or PsA (e.g., IL23R and deletion of LCE3B and LCE3C), with some variants being uniquely linked to PsA (like HLA B27 on chromosome 6). This article aims to summarize known and new data on the genetics of PsO and PsA, their associated genes, and the involvement of the HLA system and cytokines.

Graphical Abstract

1. Introduction

Psoriasis (PsO) is a chronic systemic autoimmune-mediated inflammatory skin disease associated with many comorbidities [1]. The range includes cardiovascular diseases and metabolic syndrome, and psychological issues such as depression and anxiety frequently accompany it. The etiology of PsO remains elusive, which makes it a subject of extensive research. It is widely regarded as a multifactorial pathology, influenced by a complex interplay of immunological, environmental, and genetic factors. The relationship between psoriasis and psoriatic arthritis (PsA) is particularly significant; according to Alinaghi et al., 19.7% of patients suffering from PsO also experience PsA, which is characterized as an inflammatory arthritis occurring in conjunction with psoriasis [2]. The co-occurrence of these conditions often complicates diagnosis and management, underscoring the need for integrated therapeutic strategies.
In 2016, the World Health Organization (WHO) addressed the challenges of psoriasis globally in its Global Report on Psoriasis [3]. The report highlighted a considerable gap in robust data concerning the incidence and prevalence of PsO. This lack of data poses challenges in comparing studies due to varied methodologies and sampling techniques. Prevalence estimates for PsO, based on a review of 68 articles from 20 countries, range dramatically, ranging from as low as 0.09% to as high as 11.4% [4,5]. Similarly, an extensive Italian study spanning from 2001 to 2005 observed PsO incidences of between 2.3 and 3.21 cases per 1000 individuals [6]. Genetic factors play a crucial role in both PsO and PsA, substantiated by familial aggregation, twin studies, and broad population-based studies [7,8]. Notably, the concordance rates of monozygotic twins range between 35% and 70%, while for dizygotic twins, the rates are between 12% and 23% [9]. These studies suggest a strong genetic component in the pathogenesis of PsO. Recent advances in genetic research, including linkage studies, genome-wide association studies (GWASs), and genome-wide meta-analyses, have been pivotal in identifying over 80 genes and loci that contribute to PsO susceptibility [10]. These discoveries not only enhance our understanding of the genetic architecture of PsO, but also pave the way for future genetic-based therapies and personalized medicine approaches.
This article aims to summarize known and new data on the genetics of PsO and PsA, their associated genes, and the involvement of the HLA system and cytokines.

2. Genetic and Pathogenetic Aspects of Psoriasis

The pathogenesis of PsO is complex, involving not only extensive interactions between immune cells and skin structures but also a strong genetic component that underlies its development and progression [11]. In PsO, keratinocytes proliferate abnormally fast, which is thought to be a response to chronic inflammatory signals. These signals originate from a variety of immune cells that infiltrate the skin, including T-cells, macrophages, and dendritic cells, contributing to the characteristic thick, scaly skin lesions seen in psoriasis.
Research has identified T-cells, particularly those expressing Th1 and Th17 cytokines, as key players in the maintenance of psoriatic plaques. These T-cells help drive the inflammatory process through the release of cytokines such as IFN-γ, IL-17, and TNF-α, which not only promote the proliferation of keratinocytes but also enhance the infiltration of additional immune cells into the skin [12,13]. Furthermore, the role of the immune system in PsO extends beyond adaptive immunity. The innate immune system, which includes natural killer cells, macrophages, and neutrophils, also plays a crucial role in the initiation and perpetuation of the disease. For instance, research has shown that psoriatic lesions contain an increased number of Langerhans cells and other antigen-presenting cells, which may contribute to the activation and maintenance of T-cell responses in the skin [14].
At the genetic level, PsO and psoriatic arthritis (PsA) share a common genetic background, with the major genetic risk factor localized to the MHC class I region on chromosome 6p21.3. This region encodes for several immune-related genes, contributing significantly to genetic susceptibility to psoriatic disease. Key genes in this region include those related to the HLA complex, which was first linked to PsO in the early 1970s, illustrating the long-standing recognition of genetic factors in PsO pathogenesis [15]. Recent advances in genetic research, particularly genome-wide association studies (GWASs), have identified numerous single-nucleotide polymorphisms (SNPs) associated with PsO. These genetic variants frequently occur in genes involved in immune regulation, such as those encoding cytokines and their receptors. For example, variants in IL-23R, which encodes the receptor for IL-23, are associated with PsO. IL-23 is critical for the differentiation and maintenance of Th17 cells, a T-cell subset strongly implicated in psoriasis due to its production of IL-17 [16]. Environmental factors also play a significant role in triggering or exacerbating PsO, especially in individuals with a genetic predisposition [17]. These factors include infections such as streptococcal pharyngitis, physical trauma to the skin (Koebner phenomenon), smoking, obesity, and significant psychological stress, while obesity is often named the main risk factor for developing psoriatic disease and it also increases the likelihood of PsA [1]. Each of these factors can initiate or worsen the inflammatory cycle that is characteristic of PsO, highlighting the complex interplay between genetics and the environment in the pathogenesis of psoriasis.
With these facts in mind, we can say, that PsO is a multifaceted disease characterized by both immune-mediated inflammation and significant genetic contributions. While much progress has been made in understanding the genetic underpinnings and immune pathways involved in PsO, ongoing research continues to unravel the complex genetic architecture and the myriad environmental interactions that influence the disease trajectory.

3. Associated Genes

The genetics of PsO reveal a rich and intricate tapestry of gene interactions and pathways, highlighting the multifactorial nature of this disease. Understanding the genetic basis of PsO has led to targeted therapies, particularly those that inhibit the Th17/IL-23 pathway [13]. Drugs targeting IL-23, such as ustekinumab, have been effective in reducing the severity of PsO by directly influencing one of the central pathways in its pathogenesis. These genetic insights not only enhance our understanding of PsO but also guide the development of more precise and effective treatments, improving outcomes for patients with this challenging condition. Genes implicated in PsO have been categorized into different functional groups, each contributing differently to the disease mechanism and therapeutic targeting:
  • Antigen Presentation: HLA-Cw6, ERAP1, ERAP2, MICA—These genes are critical for the presentation of antigenic peptides to T-cells. HLA-Cw6, for example, is one of the most strongly associated genetic markers for PsO, influencing how the immune system recognizes and responds to pathogens and self-antigens.
  • IL-12/IL-23 Axis: IL12Bp40, IL23Ap19, IL23R, JAK2, TYK2—This group of genes regulates cytokines that are pivotal for T-cell differentiation, particularly into Th1 and Th17 cells. The IL-23 receptor pathway, through its influence on Th17 cell functioning, is a primary therapeutic target, as evidenced by the efficacy of biologics that block IL-23.
  • T-cell Development and Polarization: RUNX1, RUNX3, STAT3, TAGAP, IL-4, IL-13—These genes are involved in T-cell lineage decisions and the polarization of T-cells into specific subtypes crucial for PsO pathogenesis, such as Th2 cells, which are influenced by IL-4 and IL-13.
  • Innate Immunity: CARD14, c-REL, TRAF3IP2, DDX58, IFIH1—These genes encode proteins that play roles in the innate immune response, providing the first line of defense against pathogens and initiating inflammatory responses that can lead to psoriatic plaque formation.
  • Negative Regulators of Immune Responses—TNIP1, TNFAIP3, NFKBIA, ZC3H12C, IL36RN, SOCS1: These genes help modulate and dampen the immune response, preventing uncontrolled inflammation. Dysregulation of these genes can lead to the prolonged inflammatory responses seen in PsO.

Psoriasis Susceptibility Loci (PSORS)

Research has identified multiple chromosomal regions, known as PSORS loci, that harbor genes linked to PsO. To date, at least 12 PSORS loci have been recognized. These loci represent regions of the genome where variations can significantly increase the risk of developing PsO. Table 1 provides an overview of known loci and their associated genes.

4. Human Leukocyte Antigens

The HLA system plays a crucial role in the immune system’s ability to distinguish between self and non-self. This system’s association with PsO provides insight into the genetic basis of this complex autoimmune condition. The variations in HLA class I (A, B, C) and class II (DR, DQ) antigens have been extensively studied to understand their roles in PsO susceptibility and progression.
  • HLA Class I Associations
HLA-A: In their research findings, Singh et al. noted a significantly higher prevalence of HLA-A1, A24, and A28 subtypes in psoriatic patients compared to healthy controls. This suggests a predisposition that may enhance the presentation of pathogenic peptides to immune cells, triggering an autoimmune response [30].
HLA-B: Interestingly, in cases without arthritis, the HLA-B13 subtype was more common in healthy controls than in psoriatic patients, which might indicate a protective effect against PsO. In cases with arthritis, studies identified subtypes such as HLA-B7 and B27 as being particularly associated with PsA, pointing to a genetic link between PsO and joint inflammation. According to other authors, HLA-B13, -B16, -B17, -B38, -B39, and -Cw6 are associated with or without arthritis [31].
HLA-C: The Cw6 subtype shows a strong association with PsO, which is significantly elevated among psoriatic patients. This subtype is known to affect the immune system’s response, possibly by influencing the skin’s inflammatory environment [32,33].
  • HLA Class II Associations
HLA-DR3 and DR53 were found at higher rates in psoriatic patients. The presence of DR3 exclusively in psoriatic patients underlines its potential as a biomarker for PsO susceptibility. HLA-DQ1 co-occurrence with DR53 further supports the idea of a multifactorial genetic landscape influencing PsO pathology.

5. Cytokines

Cytokines play a pivotal role in facilitating the interactions between cells that lead to the abnormal structures and functions observed in PsO. These include the abnormal proliferation and differentiation of keratinocytes, excessive growth of blood vessels, activation of immune cells, and promotion of abnormal immune responses. Pro-inflammatory cytokines, in particular, are critical in the development and exacerbation of PsO [34,35,36]. Secukinumab, an IL-17A antibody, and Guselkumab, a selective IL-23 inhibitor, were developed and tested based on the IL-23/IL-17 axis [37,38,39]. Also, non-coding RNAs (ncRNAs) have been studied in association with connective tissue metabolism, inflammation, and cell proliferation, linking them to cytokine signaling and T-cell activation [40,41,42]. Type I Interferons (INFs) are products of plasmacytoid dendritic cells (DCs) in the early psoriatic stage. Type I IFNs modulate the production of IFN-γ and IL-17 and play a crucial role in the differentiation and activation of T-cells, especially Th1 and Th17 cells [43].
IL-12 and IL-23 are heterodimeric pleiotropic proteins that share the p40 subunit, which is encoded by IL12B, which is vital for the differentiation of Th1 and Th17 cells, respectively. IL-12 also includes a distinct p35 subunit, while IL-23 has a unique p19 subunit that is encoded by IL23A. Elevated expressions of the p19 and p40 subunits have been observed in psoriatic skin lesions, whereas the expression of the p35 subunit does not increase, highlighting the significance of IL-23 in the pathogenesis of PsO [12,44]. So, the final effect on psoriatic susceptibility depends on the IL-12/IL-23 ratio [45,46].
IL-17, the main cytokine produced by Th17 cells, plays a crucial role in the pathogenesis of psoriasis. Additionally, neutrophils, mast cells, and natural killer (NK) cells also produce IL-17, which is implicated in the pathogenesis of inflammatory bowel diseases [47]. IL-17 is considered the primary regulator of psoriatic cutaneous inflammation and is key in linking the innate and adaptive immune responses [48,49,50]. IL-17, particularly IL-17A, emerges as a significant cytokine in the pathogenesis of psoriatic disease. Mast cells, γδ T-cells, αβ T-cells, and innate lymphoid cells are the primary sources of IL-17A in the lesioned skin and synovial fluid of patients. IL-17A targets various cells, including keratinocytes, neutrophils, endothelial cells, fibroblasts, osteoclasts, chondrocytes, and osteoblasts in the skin and joints, stimulating them to produce antimicrobial peptides, chemokines, and pro-inflammatory cytokines. This activity promotes tissue inflammation and bone remodeling, highlighting the critical role of the IL-23/IL-17A axis in the disease’s pathogenesis, leading to new biologic treatments targeting these cytokines [51,52].
IL-22 stimulates keratinocytes in human skin in various ways. In conjunction with IL-17, it can induce keratinocyte proliferation and suppress their differentiation during the tissue-remodeling phase seen in PsO [53]. IL-9, a pro-inflammatory cytokine, enhances the production of IL-17, IL-13, IFN-γ, and TNF-α in psoriasis, contributing to the inflammatory response [35]. IL-33 is a recently discovered mediator of the IL-1 family [54]. Certain cytokines, like HLA antigens, are linked to risks for both psoriasis and psoriatic arthritis. Genetic associations with both conditions have been identified for cytokines such as IL-12B, IL-23A, IL-23R, IL-2/IL-21, and TNF-α [14].
Tumor necrosis factor α (TNFα) plays a significant role in various inflammatory skin conditions, including psoriasis. TNF-α influences the antigen-presenting capabilities of DCs and stimulates T-cell infiltration [55]. It also activates the expression of C-reactive protein, a component of the acute phase response, and various cytokines, such as IL-6 and IL-23, in addition to inducing chemokines like CXCL8/IL-8 and CCL20, which are crucial for neutrophil and myeloid DC and Th17 cell recruitment, respectively. TNF-α is a vital regulator of the IL-23/Th17 axis in PsO [56].

6. Genetic and Pathogenetic Aspects of Psoriatic Arthritis

Psoriatic arthritis (PsA) is an inflammatory arthritis that occurs in 20–30% of patients diagnosed with psoriasis [57]. PsA is influenced by genetic, immunologic, and environmental factors, with epidemiological studies indicating a strong genetic component [58,59]. The genetic predisposition to PsA is significant, as evidenced by a recurrence rate of 30–55% among siblings and first-degree relatives [60,61]. The adoption of GWASs, including the use of the immunochip, has profoundly shifted our understanding of disease pathogenesis in psoriasis PsO [62]. These studies, involving over 15,000 PsO cases and 27,000 healthy controls, have identified more than 60 risk loci and have also elucidated pathways involved in the pathogenesis of PsO, specifically those related to the innate immune system, antigen presentation, and the acquired or adaptive immune response [63,64]. Genetic research has uncovered significant dominant effects of the major histocompatibility complex (MHC) region, including both HLA and non-HLA alleles. Genome-wide association studies have played a pivotal role in pinpointing key genes within critical signaling pathways, such as IL-23/IL-17, RANK, and NFκB [65].
Given that PsA frequently occurs alongside PsO, with an estimated prevalence of 30%, it is not unexpected that many of the genetic variants identified are common to both conditions. However, GWAS scans and meta-analyses focused on PsA, involving over 3000 PsA cases and 13,000 controls, have identified fewer variants reaching genome-wide significance. Over 20 variants have achieved this threshold, including genes like HLA-A, HLA-B, HLA-C, IL-12B, IL-23R, IL-23A, and others, highlighting the genetic underpinnings specific to PsA [64,66,67,68,69]. Similar to PsO, GWAS scans and meta-analyses in PsA have revealed pathways involved in its pathogenesis, particularly those related to the innate immune system, antigen presentation, and the acquired or adaptive immune response [70,71]. These findings have identified key signaling pathways and genetic markers, such as those affecting epidermal differentiation, innate immunity, antigen processing, and adaptive immunity, that are crucial in understanding the mechanisms of PsA. HLA genetic markers specifically associated with PsA are detailed further in Table 2.
To discern PsA-specific or PsA-weighted genetic variants, researchers compared GWAS results from PsA patients to those from patients with cutaneous PsO without joint involvement. Identifying risk loci unique to the development of PsA in PsO patients has proven challenging, though emerging evidence highlights loci reaching genome-wide significance that are uniquely associated with PsA and not PsO, such as CSF2, PTPN22, TNFAIP3, HLA-B, and IL23R [64,66,76,84]. The majority of genetic findings in these studies show only modest odds ratios, except for in the HLA region. Specific alleles such as HLA-B08, HLA-B27, HLA-B38, and HLA-B39 are linked with a significantly increased risk of developing PsA. Conversely, HLA-C*06 is associated with a reduced risk of PsA, acting as a protective factor in comparison to patients with PsO. However, a recent study suggests that the genetic variance for PsA may not be as extensive as previously thought [85].
In vitro models observed the impact of genistein (a soy-derived isoflavone known for its anti-inflammatory properties) on psoriatic cells [86]. Different studies on the dietary effects of PsA focused on omega-3 and omega-6 fatty acids, reporting that certain genetic profiles might reduce the risk of PsA in the presence of a high dietary intake of omega-3 [87]. Lysosomal dysfunction in psoriasis might also contribute to chronic inflammation [88]. Lin et al. identified an abnormality in the apoptosis of CD14+ monocytes, which can lead to prolonged inflammatory responses, in patients with PsA [89]. Table 3 presents pooled data based on the latest research and data from two recent systematic reviews, showing non-HLA genetic markers categorized into three levels of evidence: strong, moderate, and conflicting [90,91]. No genetic markers reached a strong level of evidence for a definitive positive, negative, or neutral association with the presence of PsA.

7. Conclusions

Reviews in the field of PsO and PsA focus on evaluating key genetic markers related to the most significant HLA markers for PsO and PsA, and the IL-12–IL-23–IL-17 axis [81,91,99]. The genetic marker HLA-C06, also known as PSOR1, is particularly crucial, accounting for up to 50% of the heritability of PsO in the general population. Within PsO populations, studies show that patients with PsO who also have the HLA-C*06 marker are less likely to develop PsA [100]. Despite numerous studies on this association, further high-quality research is needed to confirm the largely negative relationship between HLA-C*06 and the onset of PsA. A recent case-control study found that the polymorphisms studied in the IL-12B and IL-23R genes did not show a significant association with psoriasis susceptibility in a southern European cohort [101].
This review found moderate evidence supporting the association of HLA-B*27 with concurrent PsA in patients with PsO. This marker is notably prevalent (90%) in ankylosing spondylitis (AS), and while it is also more common in other spondylarthritis conditions than in the healthy population, it is less frequent than in AS [102,103]. HLA-B27 levels were found to be higher in PsO patients who developed arthritis compared to those who did not. This suggests that HLA-B27 might help differentiate between PsO patients who will and will not develop PsA, considering that both conditions are part of the spondylarthritis spectrum.
From a genetic perspective of the IL-17/IL-23 axis, there was moderate evidence indicating no significant differences in SNPs in the IL23 gene between PsA and PsO patients. However, the review highlighted limited evidence showing a higher occurrence of the SNP rs79877597 in the IL17 gene in PsA patients compared to PsO patients. While the IL-17/IL-23 axis is important for the development of psoriatic disease in general, these findings suggest its limited relevance in the development of PsA among PsO patients.
Studies on the incidence of PsO and PsA in first- and second-degree relatives have indicated stronger heritability of PsA compared to psoriasis alone. This suggests the existence of PsA-specific risk loci. Identifying such loci could aid in developing therapies that are more effective for PsA, especially as a considerable portion of patients are non-responsive to current treatments. Notably, evidence of a PsA-specific locus has been found at HLA-B27 within the MHC region. Recent studies have also identified non-HLA risk loci specific to PsA at IL23R, PTPN22, and on chromosome 5q31. Further functional characterization of these loci will enhance understanding of the pathways underlying PsA and facilitate the application of genetic findings in patient therapy [34]. Diagnosing PsA presents unique challenges due to its greater clinical heterogeneity compared to other autoimmune diseases, like PsO or Rheumatoid Arthritis (RA). This diversity in symptoms and disease manifestations, coupled with a lower rate of accurate diagnoses, complicates the ability to conduct consistent and reproducible genetic research. As fewer patients are correctly diagnosed, gathering reliable data for genetic studies is hindered, impacting the development of targeted therapies and advancements in understanding the genetic foundation of PsA [104,105].
Achievements with TNF inhibitors have led to significant improvements across multiple aspects of psoriatic arthritis. These improvements encompass not only the primary signs and symptoms of arthritis but also extend to dactylitis and enthesitis, as well as skin manifestations. Furthermore, there have been enhancements in functional status and quality of life, along with a notable reduction in the progression of radiographic damage [106]. While combination therapy has been shown to be very effective for plaque psoriasis, with promising combinations of pioglitazone, the overall approach remains complicated and complex, especially for patients with serious comorbidities [107,108]. FDA-approved JAK-STAT inhibitors are also showing promise, not only in PsA but also in different inflammatory conditions [109]. Etanercept is viewed as effective for juvenile idiopathic arthritis, which may have implications for similar strategies for psoriatic arthritis [110]. Clinical trials show sustained improvements in disease activity being achieved with Guselkumab treatment for PsA patients, indicating its potential as an effective long-term therapy option [111]. Understanding the polymorphic nature of PsA is crucial for creating individualized treatment plans [112].
In conclusion, advances in next-generation sequencing (e.g., single-cell analysis) have led to the identification of more accurate and reliable genetic markers for PsA [113,114]. There has been substantial progress in understanding the genetic underpinnings of PsA, revealing that some loci, such as TRAF3IP2, REL, and FBXL19, have a strong effect, while others, like IL23R and deletions of LCE3B and LCE3C, predispose individuals to both PsO and PsA, with certain markers, like HLA B27 at 5q31, being uniquely associated with PsA [115,116]. This enhances the potential for targeted screening within the psoriasis population to identify those at higher risk for PsA and to apply the long-needed precision medicine approach [117]. Additionally, combining genetic markers with laboratory (e.g., inflammatory markers of bone metabolism) and clinical markers (such as comorbidities and lifestyle factors) is crucial to providing targeted therapies [118]. This ongoing research continues to inform the EULAR recommendations for managing PsA with pharmacological therapies, which was last updated in 2019 [119]. Future gene function studies could provide deeper insights into disease pathogenesis, improving early diagnosis and treatment options.

Author Contributions

M.Z.: investigation, writing—original draft, writing—review and editing; L.D.: writing—review and editing, funding acquisition; S.H.: conceptualization, writing—original draft, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study is the result of the project implementation of the Center for Advanced Therapies of Chronic Inflammatory Diseases of the Musculoskeletal System (CPT-ZOPA) grant, ITMS2014+: 313011W410, which was supported by the Operational Programme Integrated infrastructure funded by the European Regional Development Fund.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mrowietz, U.; Lauffer, F.; Sondermann, W.; Gerdes, S.; Sewerin, P. Psoriasis as a Systemic Disease. Dtsch. Arztebl. Int. 2024; online ahead of print. [Google Scholar] [CrossRef]
  2. Alinaghi, F.; Calov, M.; Kristensen, L.E.; Gladman, D.D.; Coates, L.C.; Jullien, D.; Gottlieb, A.B.; Gisondi, P.; Wu, J.J.; Thyssen, J.P.; et al. Prevalence of Psoriatic Arthritis in Patients with Psoriasis: A Systematic Review and Meta-Analysis of Observational and Clinical Studies. J. Am. Acad. Dermatol. 2019, 80, 251–265.e19. [Google Scholar] [CrossRef] [PubMed]
  3. World Health Organization. Global Report on Psoriasis; World Health Organization: Geneva, Switzerland, 2016; ISBN 978-92-4-156518-9. [Google Scholar]
  4. Gibbs, S. Skin Disease and Socioeconomic Conditions in Rural Africa: Tanzania. Int. J. Dermatol. 1996, 35, 633–639. [Google Scholar] [CrossRef] [PubMed]
  5. Pariser, D.; Schenkel, B.; Carter, C.; Farahi, K.; Brown, T.M.; Ellis, C.N. Psoriasis Patient Interview Study Group A Multicenter, Non-Interventional Study to Evaluate Patient-Reported Experiences of Living with Psoriasis. J. Dermatolog. Treat. 2016, 27, 19–26. [Google Scholar] [CrossRef] [PubMed]
  6. Vena, G.A.; Altomare, G.; Ayala, F.; Berardesca, E.; Calzavara-Pinton, P.; Chimenti, S.; Giannetti, A.; Girolomoni, G.; Lotti, T.; Martini, P.; et al. Incidence of Psoriasis and Association with Comorbidities in Italy: A 5-Year Observational Study from a National Primary Care Database. Eur. J. Dermatol. 2010, 20, 593–598. [Google Scholar] [CrossRef] [PubMed]
  7. Chandran, V. The Genetics of Psoriasis and Psoriatic Arthritis. Clin. Rev. Allergy Immunol. 2013, 44, 149–156. [Google Scholar] [CrossRef]
  8. Karmacharya, P.; Chakradhar, R.; Ogdie, A. The Epidemiology of Psoriatic Arthritis: A Literature Review. Best. Pract. Res. Clin. Rheumatol. 2021, 35, 101692. [Google Scholar] [CrossRef]
  9. Bowcock, A.M. The Genetics of Psoriasis and Autoimmunity. Annu. Rev. Genom. Hum. Genet. 2005, 6, 93–122. [Google Scholar] [CrossRef]
  10. Ran, D.; Cai, M.; Zhang, X. Genetics of Psoriasis: A Basis for Precision Medicine. Precis. Clin. Med. 2019, 2, 120–130. [Google Scholar] [CrossRef]
  11. Dand, N.; Mahil, S.K.; Capon, F.; Smith, C.H.; Simpson, M.A.; Barker, J.N. Psoriasis and Genetics. Acta Derm. Venereol. 2020, 100, 5647. [Google Scholar] [CrossRef]
  12. Chamian, F.; Lowes, M.A.; Lin, S.-L.; Lee, E.; Kikuchi, T.; Gilleaudeau, P.; Sullivan-Whalen, M.; Cardinale, I.; Khatcherian, A.; Novitskaya, I.; et al. Alefacept Reduces Infiltrating T Cells, Activated Dendritic Cells, and Inflammatory Genes in Psoriasis Vulgaris. Proc. Natl. Acad. Sci. USA 2005, 102, 2075–2080. [Google Scholar] [CrossRef] [PubMed]
  13. Harden, J.L.; Krueger, J.G.; Bowcock, A.M. The Immunogenetics of Psoriasis: A Comprehensive Review. J. Autoimmun. 2015, 64, 66–73. [Google Scholar] [CrossRef] [PubMed]
  14. O’Rielly, D.D.; Rahman, P. Genetics of Psoriatic Arthritis. Best. Pract. Res. Clin. Rheumatol. 2014, 28, 673–685. [Google Scholar] [CrossRef] [PubMed]
  15. Valentova, V.; Galajda, P.; Péč, M.; Mokan, M.; Pec, J. Genetics of Psoriasis-Short Resume. Acta Medica Martiniana 2011, 11, 7–15. [Google Scholar] [CrossRef]
  16. Chandran, V. Genetics of Psoriasis and Psoriatic Arthritis. Indian J. Dermatol. 2010, 55, 151–156. [Google Scholar] [CrossRef] [PubMed]
  17. Yan, D.; Gudjonsson, J.E.; Le, S.; Maverakis, E.; Plazyo, O.; Ritchlin, C.; Scher, J.U.; Singh, R.; Ward, N.L.; Bell, S.; et al. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J. Investig. Dermatol. 2021, 141, 2112–2122.e3. [Google Scholar] [CrossRef] [PubMed]
  18. Jordan, C.T.; Cao, L.; Roberson, E.D.O.; Pierson, K.C.; Yang, C.-F.; Joyce, C.E.; Ryan, C.; Duan, S.; Helms, C.A.; Liu, Y.; et al. PSORS2 Is due to Mutations in CARD14. Am. J. Hum. Genet. 2012, 90, 784–795. [Google Scholar] [CrossRef] [PubMed]
  19. Samuelsson, L.; Stiller, C.; Friberg, C.; Nilsson, C.; Inerot, A.; Wahlström, J. Association Analysis of Cystatin A and Zinc Finger Protein 148, Two Genes Located at the Psoriasis Susceptibility Locus PSORS5. J. Investig. Dermatol. 2004, 122, 1399–1400. [Google Scholar] [CrossRef] [PubMed]
  20. Duffin, K.C.; Chandran, V.; Gladman, D.D.; Krueger, G.G.; Elder, J.T.; Rahman, P. Genetics of Psoriasis and Psoriatic Arthritis: Update and Future Direction. J. Rheumatol. 2008, 35, 1449–1453. [Google Scholar]
  21. Sun, L.-D.; Li, W.; Yang, S.; Fan, X.; Yan, K.-L.; Liang, Y.-H.; Gao, M.; Cui, Y.; Xiao, F.-L.; Du, W.-H.; et al. Evidence for a Novel Psoriasis Susceptibility Locus at 9q33-9q34 in Chinese Hans. J. Investig. Dermatol. 2007, 127, 1140–1144. [Google Scholar] [CrossRef]
  22. Vasku, V.; Bienertova Vasku, J.; Slonková, V.; Kanková, K.; Vasku, A. Matrix Metalloproteinase-2 Promoter Variability in Psoriasis. Arch. Dermatol. Res. 2009, 301, 467–473. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, Z.; Yuan, J.; Tian, Z.; Xu, J.; Lu, Z. Investigation of 36 Non-HLA (Human Leucocyte Antigen) Psoriasis Susceptibility Loci in a Psoriatic Arthritis Cohort. Arch. Dermatol. Res. 2017, 309, 71–77. [Google Scholar] [CrossRef] [PubMed]
  24. Occella, C.; Bleidl, D.; Nozza, P.; Mascelli, S.; Raso, A.; Gimelli, G.; Gimelli, S.; Tassano, E. Identification of an Interstitial 18p11.32-P11.31 Duplication Including the EMILIN2 Gene in a Family with Porokeratosis of Mibelli. PLoS ONE 2013, 8, e61311. [Google Scholar] [CrossRef] [PubMed]
  25. Tsunemi, Y.; Saeki, H.; Nakamura, K.; Sekiya, T.; Hirai, K.; Fujita, H.; Asano, N.; Kishimoto, M.; Tanida, Y.; Kakinuma, T.; et al. Interleukin-12 P40 Gene (IL12B) 3′-Untranslated Region Polymorphism Is Associated with Susceptibility to Atopic Dermatitis and Psoriasis Vulgaris. J. Dermatol. Sci. 2002, 30, 161–166. [Google Scholar] [CrossRef] [PubMed]
  26. Friberg, C.; Björck, K.; Nilsson, S.; Inerot, A.; Wahlström, J.; Samuelsson, L. Analysis of Chromosome 5q31-32 and Psoriasis: Confirmation of a Susceptibility Locus but No Association with SNPs within SLC22A4 and SLC22A5. J. Investig. Dermatol. 2006, 126, 998–1002. [Google Scholar] [CrossRef] [PubMed]
  27. Duffin, K.C.; Freeny, I.C.; Schrodi, S.J.; Wong, B.; Feng, B.-J.; Soltani-Arabshahi, R.; Rakkhit, T.; Goldgar, D.E.; Krueger, G.G. Association between IL13 Polymorphisms and Psoriatic Arthritis Is Modified by Smoking. J. Investig. Dermatol. 2009, 129, 2777–2783. [Google Scholar] [CrossRef]
  28. Nagai, N.; Kashiwamura, S.; Nakanishi, K.; Shinka, S. [Interleukin-4 (IL-4)]. Gan Kagaku Ryoho 1994, 21, 1099–1108. [Google Scholar]
  29. Yang, P.; Lu, Y.; Li, M.; Zhang, K.; Li, C.; Chen, H.; Tao, D.; Zhang, S.; Ma, Y. Identification of RNF114 as a Novel Positive Regulatory Protein for T Cell Activation. Immunobiology 2014, 219, 432–439. [Google Scholar] [CrossRef]
  30. Singh, S.; Singh, U.; Singh, S. Human Leukocyte Antigen in Patients with Psoriasis. Indian J. Dermatol. Venereol. Leprol. 2011, 77, 535. [Google Scholar] [CrossRef]
  31. Gladman, D.D.; Anhorn, K.A.; Schachter, R.K.; Mervart, H. HLA Antigens in Psoriatic Arthritis. J. Rheumatol. 1986, 13, 586–592. [Google Scholar]
  32. Dand, N.; Duckworth, M.; Baudry, D.; Russell, A.; Curtis, C.J.; Lee, S.H.; Evans, I.; Mason, K.J.; Alsharqi, A.; Becher, G.; et al. HLA-C*06:02 Genotype Is a Predictive Biomarker of Biologic Treatment Response in Psoriasis. J. Allergy Clin. Immunol. 2019, 143, 2120–2130. [Google Scholar] [CrossRef] [PubMed]
  33. Ho, P.Y.P.C.; Barton, A.; Worthington, J.; Thomson, W.; Silman, A.J.; Bruce, I.N. HLA-Cw6 and HLA-DRB1*07 Together Are Associated with Less Severe Joint Disease in Psoriatic Arthritis. Ann. Rheum. Dis. 2007, 66, 807–811. [Google Scholar] [CrossRef] [PubMed]
  34. Budu-Aggrey, A.; Bowes, J.; Barton, A. Identifying a Novel Locus for Psoriatic Arthritis. Rheumatology 2016, 55, 25–32. [Google Scholar] [CrossRef] [PubMed]
  35. Meephansan, J.; Subpayasarn, U.; Ohtsuki, M.K. Pathogenic Role of Cytokines and Effect of Their Inhibition in Psoriasis; IntechOpen: Rijeka, Croatia, 2017; ISBN 978-953-51-3252-3. [Google Scholar]
  36. Suzuki, E.; Mellins, E.D.; Gershwin, M.E.; Nestle, F.O.; Adamopoulos, I.E. The IL-23/IL-17 Axis in Psoriatic Arthritis. Autoimmun. Rev. 2014, 13, 496–502. [Google Scholar] [CrossRef] [PubMed]
  37. Gladman, D.D. Recent Advances in Understanding and Managing Psoriatic Arthritis. F1000Research 2016, 5, 2670. [Google Scholar] [CrossRef] [PubMed]
  38. Mease, P.J.; McInnes, I.B.; Kirkham, B.; Kavanaugh, A.; Rahman, P.; van der Heijde, D.; Landewé, R.; Nash, P.; Pricop, L.; Yuan, J.; et al. Secukinumab Inhibition of Interleukin-17A in Patients with Psoriatic Arthritis. N. Engl. J. Med. 2015, 373, 1329–1339. [Google Scholar] [CrossRef] [PubMed]
  39. Boehncke, W.-H.; Brembilla, N.C.; Nissen, M.J. Guselkumab: The First Selective IL-23 Inhibitor for Active Psoriatic Arthritis in Adults. Expert. Rev. Clin. Immunol. 2021, 17, 5–13. [Google Scholar] [CrossRef] [PubMed]
  40. Lin, S.-H.; Ho, J.-C.; Li, S.-C.; Chen, J.-F.; Hsiao, C.-C.; Lee, C.-H. MiR-146a-5p Expression in Peripheral CD14+ Monocytes from Patients with Psoriatic Arthritis Induces Osteoclast Activation, Bone Resorption, and Correlates with Clinical Response. J. Clin. Med. 2019, 8, 110. [Google Scholar] [CrossRef] [PubMed]
  41. Wade, S.M.; McGarry, T.; Wade, S.C.; Fearon, U.; Veale, D.J. Serum MicroRNA Signature as a Diagnostic and Therapeutic Marker in Patients with Psoriatic Arthritis. J. Rheumatol. 2020, 47, 1760–1767. [Google Scholar] [CrossRef]
  42. Van Raemdonck, K.; Umar, S.; Palasiewicz, K.; Romay, B.; Volkov, S.; Arami, S.; Sweiss, N.; Shahrara, S. TLR7 Endogenous Ligands Remodel Glycolytic Macrophages and Trigger Skin-to-Joint Crosstalk in Psoriatic Arthritis. Eur. J. Immunol. 2021, 51, 714–720. [Google Scholar] [CrossRef]
  43. Gregorio, J.; Meller, S.; Conrad, C.; Di Nardo, A.; Homey, B.; Lauerma, A.; Arai, N.; Gallo, R.L.; Digiovanni, J.; Gilliet, M. Plasmacytoid Dendritic Cells Sense Skin Injury and Promote Wound Healing through Type I Interferons. J. Exp. Med. 2010, 207, 2921–2930. [Google Scholar] [CrossRef]
  44. Lee, E.; Trepicchio, W.L.; Oestreicher, J.L.; Pittman, D.; Wang, F.; Chamian, F.; Dhodapkar, M.; Krueger, J.G. Increased Expression of Interleukin 23 P19 and P40 in Lesional Skin of Patients with Psoriasis Vulgaris. J. Exp. Med. 2004, 199, 125–130. [Google Scholar] [CrossRef]
  45. Gottlieb, A.B.; Chamian, F.; Masud, S.; Cardinale, I.; Abello, M.V.; Lowes, M.A.; Chen, F.; Magliocco, M.; Krueger, J.G. TNF Inhibition Rapidly Down-Regulates Multiple Proinflammatory Pathways in Psoriasis Plaques. J. Immunol. 2005, 175, 2721–2729. [Google Scholar] [CrossRef]
  46. Piskin, G.; Tursen, U.; Sylva-Steenland, R.M.R.; Bos, J.D.; Teunissen, M.B.M. Clinical Improvement in Chronic Plaque-Type Psoriasis Lesions after Narrow-Band UVB Therapy Is Accompanied by a Decrease in the Expression of IFN-Gamma Inducers-IL-12, IL-18 and IL-23. Exp. Dermatol. 2004, 13, 764–772. [Google Scholar] [CrossRef]
  47. Hohenberger, M.; Cardwell, L.A.; Oussedik, E.; Feldman, S.R. Interleukin-17 Inhibition: Role in Psoriasis and Inflammatory Bowel Disease. J. Dermatolog Treat. 2018, 29, 13–18. [Google Scholar] [CrossRef]
  48. Fletcher, J.M.; Moran, B.; Petrasca, A.; Smith, C.M. IL-17 in Inflammatory Skin Diseases Psoriasis and Hidradenitis Suppurativa. Clin. Exp. Immunol. 2020, 201, 121–134. [Google Scholar] [CrossRef]
  49. Lai, Y.; Li, D.; Li, C.; Muehleisen, B.; Radek, K.A.; Park, H.J.; Jiang, Z.; Li, Z.; Lei, H.; Quan, Y.; et al. The Antimicrobial Protein REG3A Regulates Keratinocyte Proliferation and Differentiation after Skin Injury. Immunity 2012, 37, 74–84. [Google Scholar] [CrossRef]
  50. Paroli, M.; Spadea, L.; Caccavale, R.; Spadea, L.; Paroli, M.P.; Nante, N. The Role of Interleukin-17 in Juvenile Idiopathic Arthritis: From Pathogenesis to Treatment. Medicina 2022, 58, 1552. [Google Scholar] [CrossRef]
  51. Blauvelt, A.; Chiricozzi, A. The Immunologic Role of IL-17 in Psoriasis and Psoriatic Arthritis Pathogenesis. Clin. Rev. Allergy Immunol. 2018, 55, 379–390. [Google Scholar] [CrossRef]
  52. Raychaudhuri, S.P.; Raychaudhuri, S.K. Mechanistic Rationales for Targeting Interleukin-17A in Spondyloarthritis. Arthritis Res. Ther. 2017, 19, 51. [Google Scholar] [CrossRef] [PubMed]
  53. Boniface, K.; Bernard, F.-X.; Garcia, M.; Gurney, A.L.; Lecron, J.-C.; Morel, F. IL-22 Inhibits Epidermal Differentiation and Induces Proinflammatory Gene Expression and Migration of Human Keratinocytes. J. Immunol. 2005, 174, 3695–3702. [Google Scholar] [CrossRef]
  54. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an Interleukin-1-like Cytokine That Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-Associated Cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef]
  55. Goldminz, A.M.; Au, S.C.; Kim, N.; Gottlieb, A.B.; Lizzul, P.F. NF-κB: An Essential Transcription Factor in Psoriasis. J. Dermatol. Sci. 2013, 69, 89–94. [Google Scholar] [CrossRef]
  56. Zaba, L.C.; Cardinale, I.; Gilleaudeau, P.; Sullivan-Whalen, M.; Suárez-Fariñas, M.; Fuentes-Duculan, J.; Novitskaya, I.; Khatcherian, A.; Bluth, M.J.; Lowes, M.A.; et al. Amelioration of Epidermal Hyperplasia by TNF Inhibition Is Associated with Reduced Th17 Responses. J. Exp. Med. 2007, 204, 3183–3194. [Google Scholar] [CrossRef]
  57. Gladman, D.D.; Antoni, C.; Mease, P.; Clegg, D.O.; Nash, P. Psoriatic Arthritis: Epidemiology, Clinical Features, Course, and Outcome. Ann. Rheum. Dis. 2005, 64 (Suppl. 2), ii14–ii17. [Google Scholar] [CrossRef]
  58. Lowes, M.A.; Suárez-Fariñas, M.; Krueger, J.G. Immunology of Psoriasis. Annu. Rev. Immunol. 2014, 32, 227–255. [Google Scholar] [CrossRef]
  59. O’Rielly, D.D.; Rahman, P. Genetic, Epigenetic and Pharmacogenetic Aspects of Psoriasis and Psoriatic Arthritis. Rheum. Dis. Clin. N. Am. 2015, 41, 623–642. [Google Scholar] [CrossRef]
  60. Chandran, V.; Schentag, C.T.; Brockbank, J.E.; Pellett, F.J.; Shanmugarajah, S.; Toloza, S.M.A.; Rahman, P.; Gladman, D.D. Familial Aggregation of Psoriatic Arthritis. Ann. Rheum. Dis. 2009, 68, 664–667. [Google Scholar] [CrossRef]
  61. Myers, A.; Kay, L.J.; Lynch, S.A.; Walker, D.J. Recurrence Risk for Psoriasis and Psoriatic Arthritis within Sibships. Rheumatology 2005, 44, 773–776. [Google Scholar] [CrossRef] [PubMed]
  62. Cortes, A.; Brown, M.A. Promise and Pitfalls of the Immunochip. Arthritis Res. Ther. 2011, 13, 101. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Helms, C.; Liao, W.; Zaba, L.C.; Duan, S.; Gardner, J.; Wise, C.; Miner, A.; Malloy, M.J.; Pullinger, C.R.; et al. A Genome-Wide Association Study of Psoriasis and Psoriatic Arthritis Identifies New Disease Loci. PLoS Genet. 2008, 4, e1000041. [Google Scholar] [CrossRef]
  64. Stuart, P.E.; Nair, R.P.; Tsoi, L.C.; Tejasvi, T.; Das, S.; Kang, H.M.; Ellinghaus, E.; Chandran, V.; Callis-Duffin, K.; Ike, R.; et al. Genome-Wide Association Analysis of Psoriatic Arthritis and Cutaneous Psoriasis Reveals Differences in Their Genetic Architecture. Am. J. Hum. Genet. 2015, 97, 816–836. [Google Scholar] [CrossRef]
  65. Hile, G.; Kahlenberg, J.M.; Gudjonsson, J.E. Recent Genetic Advances in Innate Immunity of Psoriatic Arthritis. Clin. Immunol. 2020, 214, 108405. [Google Scholar] [CrossRef]
  66. Bowes, J.; Budu-Aggrey, A.; Huffmeier, U.; Uebe, S.; Steel, K.; Hebert, H.L.; Wallace, C.; Massey, J.; Bruce, I.N.; Bluett, J.; et al. Dense Genotyping of Immune-Related Susceptibility Loci Reveals New Insights into the Genetics of Psoriatic Arthritis. Nat. Commun. 2015, 6, 6046. [Google Scholar] [CrossRef]
  67. Nair, R.P.; Duffin, K.C.; Helms, C.; Ding, J.; Stuart, P.E.; Goldgar, D.; Gudjonsson, J.E.; Li, Y.; Tejasvi, T.; Feng, B.-J.; et al. Genome-Wide Scan Reveals Association of Psoriasis with IL-23 and NF-kappaB Pathways. Nat. Genet. 2009, 41, 199–204. [Google Scholar] [CrossRef]
  68. Stuart, P.E.; Nair, R.P.; Ellinghaus, E.; Ding, J.; Tejasvi, T.; Gudjonsson, J.E.; Li, Y.; Weidinger, S.; Eberlein, B.; Gieger, C.; et al. Genome-Wide Association Analysis Identifies Three Psoriasis Susceptibility Loci. Nat. Genet. 2010, 42, 1000–1004. [Google Scholar] [CrossRef] [PubMed]
  69. Winchester, R.; FitzGerald, O. The Many Faces of Psoriatic Arthritis: Their Genetic Determinism. Rheumatology 2020, 59, i4–i9. [Google Scholar] [CrossRef]
  70. Ellinghaus, E.; Stuart, P.E.; Ellinghaus, D.; Nair, R.P.; Debrus, S.; Raelson, J.V.; Belouchi, M.; Tejasvi, T.; Li, Y.; Tsoi, L.C.; et al. Genome-Wide Meta-Analysis of Psoriatic Arthritis Identifies Susceptibility Locus at REL. J. Investig. Dermatol. 2012, 132, 1133–1140. [Google Scholar] [CrossRef]
  71. Hüffmeier, U.; Mössner, R. Complex Role of TNF Variants in Psoriatic Arthritis and Treatment Response to Anti-TNF Therapy: Evidence and Concepts. J. Investig. Dermatol. 2014, 134, 2483–2485. [Google Scholar] [CrossRef]
  72. Aterido, A.; Cañete, J.D.; Tornero, J.; Ferrándiz, C.; Pinto, J.A.; Gratacós, J.; Queiró, R.; Montilla, C.; Torre-Alonso, J.C.; Pérez-Venegas, J.J.; et al. Genetic Variation at the Glycosaminoglycan Metabolism Pathway Contributes to the Risk of Psoriatic Arthritis but Not Psoriasis. Ann. Rheum. Dis. 2019, 78, e214158. [Google Scholar] [CrossRef]
  73. Bowes, J.; Ashcroft, J.; Dand, N.; Jalali-Najafabadi, F.; Bellou, E.; Ho, P.; Marzo-Ortega, H.; Helliwell, P.S.; Feletar, M.; Ryan, A.W.; et al. Cross-Phenotype Association Mapping of the MHC Identifies Genetic Variants That Differentiate Psoriatic Arthritis from Psoriasis. Ann. Rheum. Dis. 2017, 76, 1774–1779. [Google Scholar] [CrossRef]
  74. Eder, L.; Chandran, V.; Pellett, F.; Shanmugarajah, S.; Rosen, C.F.; Bull, S.B.; Gladman, D.D. Differential Human Leucocyte Allele Association between Psoriasis and Psoriatic Arthritis: A Family-Based Association Study. Ann. Rheum. Dis. 2012, 71, 1361–1365. [Google Scholar] [CrossRef]
  75. Eder, L.; Chandran, V.; Pellet, F.; Shanmugarajah, S.; Rosen, C.F.; Bull, S.B.; Gladman, D.D. Human Leucocyte Antigen Risk Alleles for Psoriatic Arthritis among Patients with Psoriasis. Ann. Rheum. Dis. 2012, 71, 50–55. [Google Scholar] [CrossRef]
  76. Winchester, R.; Minevich, G.; Steshenko, V.; Kirby, B.; Kane, D.; Greenberg, D.A.; FitzGerald, O. HLA Associations Reveal Genetic Heterogeneity in Psoriatic Arthritis and in the Psoriasis Phenotype. Arthritis Rheum. 2012, 64, 1134–1144. [Google Scholar] [CrossRef]
  77. Elkayam, O.; Segal, R.; Caspi, D. Human Leukocyte Antigen Distribution in Israeli Patients with Psoriatic Arthritis. Rheumatol. Int. 2004, 24, 93–97. [Google Scholar] [CrossRef]
  78. Pollock, R.A.; Chandran, V.; Pellett, F.J.; Thavaneswaran, A.; Eder, L.; Barrett, J.; Rahman, P.; Farewell, V.; Gladman, D.D. The Functional MICA-129 Polymorphism Is Associated with Skin but Not Joint Manifestations of Psoriatic Disease Independently of HLA-B and HLA-C. Tissue Antigens 2013, 82, 43–47. [Google Scholar] [CrossRef]
  79. Liao, H.-T.; Lin, K.-C.; Chang, Y.-T.; Chen, C.-H.; Liang, T.-H.; Chen, W.-S.; Su, K.-Y.; Tsai, C.-Y.; Chou, C.-T. Human Leukocyte Antigen and Clinical and Demographic Characteristics in Psoriatic Arthritis and Psoriasis in Chinese Patients. J. Rheumatol. 2008, 35, 891–895. [Google Scholar]
  80. Cabaleiro, T.; Román, M.; Gallo, E.; Ochoa, D.; Tudelilla, F.; Talegón, M.; Prieto-Pérez, R.; García-Díez, A.; Daudén, E.; Abad-Santos, F. Association between Psoriasis and Polymorphisms in the TNF, IL12B, and IL23R Genes in Spanish Patients. Eur. J. Dermatol. 2013, 23, 640–645. [Google Scholar] [CrossRef]
  81. Chen, L.; Tsai, T.-F. HLA-Cw6 and Psoriasis. Br. J. Dermatol. 2018, 178, 854–862. [Google Scholar] [CrossRef] [PubMed]
  82. Coto-Segura, P.; Coto, E.; González-Lara, L.; Alonso, B.; Gómez, J.; Cuesta-Llavona, E.; Queiro, R. Gene Variant in the NF-κB Pathway Inhibitor NFKBIA Distinguishes Patients with Psoriatic Arthritis within the Spectrum of Psoriatic Disease. Biomed. Res. Int. 2019, 2019, 1030256. [Google Scholar] [CrossRef] [PubMed]
  83. Okada, Y.; Han, B.; Tsoi, L.C.; Stuart, P.E.; Ellinghaus, E.; Tejasvi, T.; Chandran, V.; Pellett, F.; Pollock, R.; Bowcock, A.M.; et al. Fine Mapping Major Histocompatibility Complex Associations in Psoriasis and Its Clinical Subtypes. Am. J. Hum. Genet. 2014, 95, 162–172. [Google Scholar] [CrossRef]
  84. Soomro, M.; Hum, R.; Barton, A.; Bowes, J. Genetic Studies Investigating Susceptibility to Psoriatic Arthritis: A Narrative Review. Clin. Ther. 2023, 45, 810–815. [Google Scholar] [CrossRef]
  85. O’Rielly, D.D.; Jani, M.; Rahman, P.; Elder, J.T. The Genetics of Psoriasis and Psoriatic Arthritis. J. Rheumatol. Suppl. 2019, 95, 46–50. [Google Scholar] [CrossRef]
  86. Bocheńska, K.; Moskot, M.; Smolińska-Fijołek, E.; Jakóbkiewicz-Banecka, J.; Szczerkowska-Dobosz, A.; Słomiński, B.; Gabig-Cimińska, M. Impact of Isoflavone Genistein on Psoriasis in in Vivo and in Vitro Investigations. Sci. Rep. 2021, 11, 18297. [Google Scholar] [CrossRef]
  87. Xu, X.; Xu, X.; Zakeri, M.A.; Wang, S.-Y.; Yan, M.; Wang, Y.-H.; Li, L.; Sun, Z.-L.; Wang, R.-Y.; Miao, L.-Z. Assessment of Causal Relationships between Omega-3 and Omega-6 Polyunsaturated Fatty Acids in Autoimmune Rheumatic Diseases: A Brief Research Report from a Mendelian Randomization Study. Front. Nutr. 2024, 11, 1356207. [Google Scholar] [CrossRef]
  88. Klapan, K.; Frangež, Ž.; Markov, N.; Yousefi, S.; Simon, D.; Simon, H.-U. Evidence for Lysosomal Dysfunction within the Epidermis in Psoriasis and Atopic Dermatitis. J. Investig. Dermatol. 2021, 141, 2838–2848.e4. [Google Scholar] [CrossRef]
  89. Lin, S.-H.; Hsu, C.-Y.; Li, S.-C. Increased Circulating CD14+ Monocytes in Patients with Psoriatic Arthritis Presenting Impaired Apoptosis Activity. Biomedicines 2024, 12, 775. [Google Scholar] [CrossRef]
  90. Chen, J.; Yuan, F.; Fan, X.; Wang, Y. Psoriatic Arthritis: A Systematic Review of Non-HLA Genetic Studies and Important Signaling Pathways. Int. J. Rheum. Dis. 2020, 23, 1288–1296. [Google Scholar] [CrossRef]
  91. Mulder, M.L.M.; van Hal, T.W.; Wenink, M.H.; Koenen, H.J.P.M.; van den Hoogen, F.H.J.; de Jong, E.M.G.J.; van den Reek, J.M.P.A.; Vriezekolk, J.E. Clinical, Laboratory, and Genetic Markers for the Development or Presence of Psoriatic Arthritis in Psoriasis Patients: A Systematic Review. Arthritis Res. Ther. 2021, 23, 168. [Google Scholar] [CrossRef] [PubMed]
  92. Yang, Q.; Liu, H.; Qu, L.; Fu, X.; Yu, Y.; Yu, G.; Tian, H.; Yu, Y.; Sun, D.; Peng, J.; et al. Investigation of 20 Non-HLA (Human Leucocyte Antigen) Psoriasis Susceptibility Loci in Chinese Patients with Psoriatic Arthritis and Psoriasis Vulgaris. Br. J. Dermatol. 2013, 168, 1060–1065. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, Y.-W.; Kang, J.-H.; Lin, H.-C. Increased Risk of Psoriasis Following Obstructive Sleep Apnea: A Longitudinal Population-Based Study. Sleep. Med. 2012, 13, 285–289. [Google Scholar] [CrossRef] [PubMed]
  94. Loft, N.D.; Skov, L.; Rasmussen, M.K.; Gniadecki, R.; Dam, T.N.; Brandslund, I.; Hoffmann, H.J.; Andersen, M.R.; Dessau, R.B.; Bergmann, A.C.; et al. Genetic Polymorphisms Associated with Psoriasis and Development of Psoriatic Arthritis in Patients with Psoriasis. PLoS ONE 2018, 13, e0192010. [Google Scholar] [CrossRef] [PubMed]
  95. Bowes, J.; Eyre, S.; Flynn, E.; Ho, P.; Salah, S.; Warren, R.B.; Marzo-Ortega, H.; Coates, L.; McManus, R.; Ryan, A.W.; et al. Evidence to Support IL-13 as a Risk Locus for Psoriatic Arthritis but Not Psoriasis Vulgaris. Ann. Rheum. Dis. 2011, 70, 1016–1019. [Google Scholar] [CrossRef] [PubMed]
  96. Eder, L.; Chandran, V.; Pellett, F.; Pollock, R.; Shanmugarajah, S.; Rosen, C.F.; Rahman, P.; Gladman, D.D. IL13 Gene Polymorphism Is a Marker for Psoriatic Arthritis among Psoriasis Patients. Ann. Rheum. Dis. 2011, 70, 1594–1598. [Google Scholar] [CrossRef] [PubMed]
  97. Işık, S.; Sılan, F.; Kılıç, S.; Hız, M.M.; Ögretmen, Z.; Özdemir, Ö. 308G/A and 238G/A Polymorphisms in the TNF-α Gene May Not Contribute to the Risk of Arthritis among Turkish Psoriatic Patients. Egypt. Rheumatol. 2016, 38, 313–317. [Google Scholar] [CrossRef]
  98. Tejasvi, T.; Stuart, P.E.; Chandran, V.; Voorhees, J.J.; Gladman, D.D.; Rahman, P.; Elder, J.T.; Nair, R.P. TNFAIP3 Gene Polymorphisms Are Associated with Response to TNF Blockade in Psoriasis. J. Investig. Dermatol. 2012, 132, 593–600. [Google Scholar] [CrossRef] [PubMed]
  99. Vecellio, M.; Hake, V.X.; Davidson, C.; Carena, M.C.; Wordsworth, B.P.; Selmi, C. The IL-17/IL-23 Axis and Its Genetic Contribution to Psoriatic Arthritis. Front. Immunol. 2020, 11, 596086. [Google Scholar] [CrossRef] [PubMed]
  100. Boehncke, W.-H.; Schön, M.P. Psoriasis. Lancet 2015, 386, 983–994. [Google Scholar] [CrossRef] [PubMed]
  101. Zervou, M.; Goulielmos, G.; Castro-Giner, F.; Chiotaki, R.; Sidiropoulos, P.; Krueger-Krasagakis, S. Interleukin-12B (IL-12B) and Interleukin-23R (IL-23R) Gene Polymorphisms Do Not Confer Susceptibility to Psoriasis in a Southern European Population: A Case-Control Study. Int. J. New Technol. Res. (IJNTR) 2016, 2, 67–70. [Google Scholar]
  102. Brown, M.A.; Xu, H.; Li, Z. Genetics and the Axial Spondyloarthritis Spectrum. Rheumatology 2020, 59, iv58–iv66. [Google Scholar] [CrossRef]
  103. Feld, J.; Chandran, V.; Haroon, N.; Inman, R.; Gladman, D. Axial Disease in Psoriatic Arthritis and Ankylosing Spondylitis: A Critical Comparison. Nat. Rev. Rheumatol. 2018, 14, 363–371. [Google Scholar] [CrossRef]
  104. Carvalho, A.L.; Hedrich, C.M. The Molecular Pathophysiology of Psoriatic Arthritis-The Complex Interplay between Genetic Predisposition, Epigenetics Factors, and the Microbiome. Front. Mol. Biosci. 2021, 8, 662047. [Google Scholar] [CrossRef] [PubMed]
  105. Laborde, C.M.; Larzabal, L.; González-Cantero, Á.; Castro-Santos, P.; Díaz-Peña, R. Advances of Genomic Medicine in Psoriatic Arthritis. J. Pers. Med. 2022, 12, 35. [Google Scholar] [CrossRef]
  106. Cassell, S.; Kavanaugh, A. Psoriatic Arthritis: Pathogenesis and Novel Immunomodulatory Approaches to Treatment. J. Immune Based Ther. Vaccines 2005, 3, 6. [Google Scholar] [CrossRef]
  107. Kulakova, E.; Baлeнтинoвнa, K.Э.; Rybalkin, S.B.; Бopиcoвич, P.C. Experience of using genetic engineering biological therapy in a patient with psoriasis and chronic renal failure under hemodialysis. Vestn. Dermatol. Venerol. 2024. [Google Scholar] [CrossRef]
  108. Snehasis, N.; Zafar, S.; Herve, N.; Siri, P.; Ali, K.H. Efficacy and Safety of Various Drug Combinations in Treating Plaque Psoriasis: A Meta-Analysis. F1000Research 2024, 13, 453. [Google Scholar] [CrossRef]
  109. Potlabathini, T.; Pothacamuri, M.A.; Bandi, V.V.; Anjum, M.; Shah, P.; Molina, M.; Dutta, N.; Adzhymuratov, O.; Mathew, M.; Sadu, V.; et al. FDA-Approved Janus Kinase-Signal Transducer and Activator of Transcription (JAK-STAT) Inhibitors for Managing Rheumatoid Arthritis: A Narrative Review of the Literature. Cureus 2024, 16, e59978. [Google Scholar] [CrossRef]
  110. van Dijk, B.T.; Bergstra, S.A.; van den Berg, J.M.; Schonenberg-Meinema, D.; van Suijlekom-Smit, L.W.A.; van Rossum, M.A.J.; Koopman-Keemink, Y.; ten Cate, R.; Allaart, C.F.; Brinkman, D.M.C.; et al. Increasing the Etanercept Dose in a Treat-to-Target Approach in Juvenile Idiopathic Arthritis: Does It Help to Reach the Target? A Post-Hoc Analysis of the BeSt for Kids Randomised Clinical Trial. Pediatr. Rheumatol. 2024, 22, 53. [Google Scholar] [CrossRef]
  111. Coates, L.C.; Gossec, L.; Zimmermann, M.; Shawi, M.; Rampakakis, E.; Shiff, N.J.; Kollmeier, A.P.; Xu, X.L.; Nash, P.; Mease, P.J.; et al. Guselkumab Provides Durable Improvement across Psoriatic Arthritis Disease Domains: Post Hoc Analysis of a Phase 3, Randomised, Double-Blind, Placebo-Controlled Study. RMD Open 2024, 10, e003977. [Google Scholar] [CrossRef] [PubMed]
  112. Agrawal, A.; Bhagwati, J. The Spectrum of Psoriatic Arthritis: A Polymorphic Puzzle. Int. J. Toxicol. Pharmacol. Res. 2024, 14, 197–202. [Google Scholar]
  113. Jung, S.; Lee, J.S. Single-Cell Genomics for Investigating Pathogenesis of Inflammatory Diseases. Mol. Cells 2023, 46, 120–129. [Google Scholar] [CrossRef] [PubMed]
  114. Penkava, F.; Velasco-Herrera, M.D.C.; Young, M.D.; Yager, N.; Nwosu, L.N.; Pratt, A.G.; Lara, A.L.; Guzzo, C.; Maroof, A.; Mamanova, L.; 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, 4767. [Google Scholar] [CrossRef] [PubMed]
  115. Chang, M.; Li, Y.; Yan, C.; Callis-Duffin, K.P.; Matsunami, N.; Garcia, V.E.; Cargill, M.; Civello, D.; Bui, N.; Catanese, J.J.; et al. Variants in the 5q31 Cytokine Gene Cluster Are Associated with Psoriasis. Genes. Immun. 2008, 9, 176–181. [Google Scholar] [CrossRef] [PubMed]
  116. Shen, C.; Gao, J.; Yin, X.; Sheng, Y.; Sun, L.; Cui, Y.; Zhang, X. Association of the Late Cornified Envelope-3 Genes with Psoriasis and Psoriatic Arthritis: A Systematic Review. J. Genet. Genom. 2015, 42, 49–56. [Google Scholar] [CrossRef]
  117. Krishnan, V.S.; Kõks, S. Transcriptional Basis of Psoriasis from Large Scale Gene Expression Studies: The Importance of Moving towards a Precision Medicine Approach. Int. J. Mol. Sci. 2022, 23, 6130. [Google Scholar] [CrossRef]
  118. Yan, D.; Blauvelt, A.; Dey, A.K.; Golpanian, R.S.; Hwang, S.T.; Mehta, N.N.; Myers, B.; Shi, Z.; Yosipovitch, G.; Bell, S.; et al. New Frontiers in Psoriatic Disease Research, Part II: Comorbidities and Targeted Therapies. J. Investig. Dermatol. 2021, 141, 2328–2337. [Google Scholar] [CrossRef]
  119. Gossec, L.; Baraliakos, X.; Kerschbaumer, A.; de Wit, M.; McInnes, I.; Dougados, M.; Primdahl, J.; McGonagle, D.G.; Aletaha, D.; Balanescu, A.; et al. EULAR Recommendations for the Management of Psoriatic Arthritis with Pharmacological Therapies: 2019 Update. Ann. Rheum. Dis. 2020, 79, 700–712. [Google Scholar] [CrossRef] [PubMed]
Table 1. PSORS with cytogenetic localizations and associated genes.
Table 1. PSORS with cytogenetic localizations and associated genes.
Locus NameCytogenetic LocalizationGenes, Their Products, and Their Roles
PSORS16p21.3Human leucocyte antigens (HLAs), especially HLACw6 [15].
Antigen presentation, strong association with PsO.
PSORS217q24-25Missense mutation of CARD14 in keratinocytes leads to overexpression of NFκB, IL8, chemokine ligand 20, IL36, and ILγ [18].
Skin barrier functions and cellular signaling pathways.
PSORS34q34Immunologically important proteins, including IL15 [15].
Influencing the inflammatory response and skin cell proliferation.
PSORS41q21S100 calcium-binding proteins are overexpressed in keratinocytes of psoriatic patients and are responsible for chemotaxis of leucocytes [15].
Epidermal differentiation and barrier formation.
PSORS53q21Cystatin A and zinc finger protein 148 [19].
Regulation of the immune system and inflammatory processes.
PSORS619p13-q13JUNB gene produces one member of AP-1-family transcription factors controlling differentiation of keratinocytes, the KIR gene product associated with HLA antigens [20].
T-cell activation and immune response modulation.
PSORS71p35-p34Gene EPS 15 codes intracellular substrate for EGF receptor, which is highly expressed in psoriatic skin [21].
Immune system and skin integrity.
PSORS816qThe NOD2/CARD15 gene is associated with psoriasis and Crohn’s disease [22].
Cellular proliferation, apoptosis, and the body’s inflammatory responses.
PSORS94q31Polymorphism of the IL-15 gene is connected with interleukin production and inflammation [23].
Inflammation and the immune system.
PSORS1018p11.23EMILIN2 gene regulates apoptosis and survival of epidermal keratinocytes [24].
Cytokine production and immune regulation.
PSORS115q31.1-q33.1IL-12B affects the balance of Th1/Th2 cells [25], SLC22A4,5 organic cation transporters [26], IL-13-regulating T-cell-mediated immunity [27], and IL-4,5 as Th2 cell products [28].
Immune system signaling.
PSORS1220q13RNF114 ring finger protein is a positive regulator of T-cell activation [29].
Inflammation and immune system responses.
Table 2. HLA genetic markers with supporting evidence of their association with PsA.
Table 2. HLA genetic markers with supporting evidence of their association with PsA.
MarkerEvidenceAssociationStudy
HLA-B*08Conflicting-[72,73,74,75,76]
HLA-B*13Conflicting-[72,77,78]
HLA-B*18Conflicting-[74,75,76]
HLA-B*27
(EH27.1 and 2)
ModeratePositive
HLA-B*27ModeratePositive[72,73,74,75,76,78,79]
HLA-B*37Conflicting-[72,76]
HLA-B*38Conflicting-[74,75,76,77,78]
HLA-B*38
(EHB38.1)
ModerateNegative[74,75,76]
HLA-B*39Conflicting-
HLA-B*57
(EH57.1)
ModerateNegative
HLA-B*57ModerateNo association[72,74,75,78]
HLA-C*01ModerateNo association[74,75,76]
HLA-C*02Conflicting-[72,74,75,76]
HLA-C*06ModerateNegative[72,73,78,79,80,81,82]
HLA-C*07Conflicting-[72,74,75]
HLA-C*12Conflicting-[74,75]
HLA-DRB1*03ModerateNo association[72,77]
HLA-DQB1*02Conflicting-
HLA-B Glu45Conflicting-[73,83]
Table 3. Non-HLA genetic markers with supporting evidence of their association with PsA.
Table 3. Non-HLA genetic markers with supporting evidence of their association with PsA.
MarkerEvidenceAssociationStudy
GJB2 SNP rs3751385ModeratePositive[92]
ERAP1 SNP rs27524ModeratePositive
IL1RNModerateNo association[67,93]
IL12B SNP rs2082412ModeratePositive[92]
IL12B rs3212227ModerateNo association[80,94]
IL12B rs6887695ModerateNo association
IL12B SNP rs7709212ModeratePositive[23]
IL13rs1800925ModeratePositive[95,96]
IL13 rs20541Conflicting-[67,95]
IL23A SNP rs2066807ModerateNo association[67,93]
IL23R SNP rs2201841Conflicting-
LCE SNP rs1886734ModeratePositive[92]
PTTI SNP rs2451697ModeratePositive
RUNX3 SNP rs7536201ModeratePositive[23]
TNFa-238ModerateNo association[80,97]
TNFa-308ModerateNo association
TNF alpha-induced protein 3 rs610604ModerateNo association[98]
TNIP1 SNP rs17728338ModeratePositive[92]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zalesak, M.; Danisovic, L.; Harsanyi, S. Psoriasis and Psoriatic Arthritis—Associated Genes, Cytokines, and Human Leukocyte Antigens. Medicina 2024, 60, 815. https://doi.org/10.3390/medicina60050815

AMA Style

Zalesak M, Danisovic L, Harsanyi S. Psoriasis and Psoriatic Arthritis—Associated Genes, Cytokines, and Human Leukocyte Antigens. Medicina. 2024; 60(5):815. https://doi.org/10.3390/medicina60050815

Chicago/Turabian Style

Zalesak, Marek, Lubos Danisovic, and Stefan Harsanyi. 2024. "Psoriasis and Psoriatic Arthritis—Associated Genes, Cytokines, and Human Leukocyte Antigens" Medicina 60, no. 5: 815. https://doi.org/10.3390/medicina60050815

Article Metrics

Back to TopTop