Preprint
Review

A Multifaced Approach on Psoriasis Pathogenesis

Altmetrics

Downloads

160

Views

59

Comments

0

Submitted:

22 March 2024

Posted:

25 March 2024

You are already at the latest version

Alerts
Abstract
Psoriasis is a chronic recurrent inflammatory autoimmune pathology, with a major genetic component and several interferences of immunological cells and their cytokines. The complex orchestration of psoriasis pathogenesis is related to the synergic effect of immune cells, polygenic alterations, the presence of autoantigens and several external factors. The major act of IL-23/IL-17 axis, strongly influencing the inflammatory pattern established during the disease activity, is visible as a continuous perpetuation of the pro-inflammatory response and keratinocyte activation and proliferation leading to the development of psoriatic lesions. Genome wide association studies (GWAS) along offered a better view of psoriasis pathogenic pathways, with approximately one third of psoriasis genetic impact on psoriasis development is associated to MHC region, with genetic loci located on chromosome 6. The most eloquent genetic factor of psoriasis, PSORS1, was identified in the MHC I site. Between the several factors involved in its complex etiology, dysbiosis, due to genetic or external stimulus, exhibiting less variety and the involvement of pathogenic bacteria, cand induce a burst of proinflammatory consequences, both cutaneous and gut microbiome being involved in psoriasis pathogenic process. A complex understanding of the major pathways that interplay in order to initiate and perpetuate psoriatic diseases and its associated comorbidities can help the current clinical practice by properly evaluating completely and individualized each patient and establish the proper therapeutic management.
Keywords: 
Subject: Medicine and Pharmacology  -   Medicine and Pharmacology

1. Introduction

Psoriasis is a chronic recurrent inflammatory autoimmune pathology, with a major genetic component and several interferences of immune cells and their cytokines [1,2]. It has a worldwide prevalence of about 2-3% of the general population, with variabilities depending on the skin phenotype, lower in Asia or Africa, higher in Scandinavian populations [3,4]. It shows an increased incidence in adults and equal distribution between men and women, with an onset in early ages for female patients, especially in case of positive family history [5]. Clinically, it is expressed by several types, most of the cases corresponding to psoriasis vulgaris or plaque-type psoriasis, pathologically characterized by an abnormal proliferation and differentiation of keratinocytes [6].
The severity and of the psoriatic disease arises not only from its progressive evolution and association in about one third of the cases with development of psoriatic arthritis, but also throughout its several comorbidities, that share a common pathogenesis [7].
During the past decades studies based on unrevealing the current knowledge of psoriasis pathogenesis have enabled the development of fundamental insights that allow a targeted therapy, highly efficient, with a major impact on quality of life of these patients [8].
The complex orchestration of psoriasis pathogenesis is related to the synergic effect of immune cells, polygenic alterations, the presence of autoantigens and several external factors [9]. Over the past years the scientific researches clearly stated that psoriasis is a T-cell mediated pathology, with an excessive production of IL-17 as a consequence of IL-23 stimulation. The major act of IL-23/IL-17 axis, strongly influencing the inflammatory pattern established during the disease activity, is visible as a continuous perpetuation of the pro-inflammatory response and keratinocyte activation and proliferation leading to the development of psoriatic lesions [9,10].
Between the several factors involved in its complex etiology, dysbiosis, due to genetic or external stimulus, exhibiting less variety and the involvement of pathogenic bacteria, cand induce a burst of proinflammatory consequences, both cutaneous and gut microbiome being involved in psoriasis pathogenic process [11]. With its role of both physical and immune barrier towards external injuries, the skin has an extensive microbiome that can constitute the target of multiple factors, leading to its dysregulation and gaps in immunoregulatory processes [12]. The relevance of gut microbiome is established by the interrelation between different components of both innate and adaptive immune systems, with the major role of T cells [13].
We aimed to describe the main components of psoriasis pathogenesis and their crosstalk mechanisms in initiation and progression of psoriasis. A complex understanding of the major pathways that interplay in order to initiate and perpetuate psoriatic diseases and its associated comorbidities can help the current clinical practice by properly evaluating completely and individualized each patient and establish the proper therapeutic management, in order to obtain not only the remission of the disease and decrease its psychosocial impact, but to prevent future organ damage and improve the quality of life for our patients.

2. Genetic Disturbances in Psoriasis

Genome wide association studies (GWAS) along with next generation sequencing (NSG) analysis offered a better view of psoriasis pathogenic pathways, strongly impacted by genomic alterations (Single nucleotide polymorphisms (SNPs), copy number variations (CNVs), and epigenetic changes) and further possible therapeutic options [14]. For the past years, a paramount amount of data related to genetic characterization of a specific pathologies was generated using GWAS approach [14,15,16,17,18]. GWAS have identified 10 loci, with the most proven association, from PSORS1 to PSORS10. The reported data show an extensive interference of several genes, from the ones associated with the functionality of skin barrier, adaptive immunity related genes, especially CD8+ T and CD4+ T lymphocytes, or genes that belong to the histocompatibility complex (MHC) region on chromosome 6 [18]. Approximately one third of psoriasis genetic impact on psoriasis development is associated to MHC region, with genetic loci located on chromosome 6. The most eloquent genetic factor of psoriasis, PSORS1, was identified in the MHC I site, where human leukocyte antigen (HLA)-Cw-6 was considered the PSORS1 risk variant. PSORS1 is bounded by a 300-kb critical region in MHC where about fifteen genes have been identified, strongly related to psoriasis. Between these, data shows HLA-C to be the most likely PSORS1 gene [29]. The risk allele HLACw0602*, present in up to 10-15% of the general population, inputs a 20 fold higher risk for developing psoriasis and in proven to be present in about 60% of psoriasis patients. It has been identified especially in young subjects, with familial history of psoriasis, where various infections, predominantly Streptococccal ones, trigger the first lesions [20]. Moreover, there has been shown the presence of a five times higher psoriasis risk in subjects with homozygous HLA-Cw0602* allele compared to heterozygous ones [22]. A recent study, published in 2021, that included 124 patients with psoriasis and aimed to establish the possible correlation between HLA-Cw0602* positivity and response to treatment, showed a lower response for the cases with this risk allele [22]. PSORS2 was identified on chromosome 17q, where CARD14 (caspase recruitment domain-containing protein 14) was described to be associated to psoriasis. CARD14 was detected in a highly expressed manner in keratinocytes, with a subsequent activating action on NF-κB and an increased release of pro-inflammatory cytokines [23,24,25]. Other two genes, SLC9A3R1 and NAT9, associated to psoriasis, where found on chromosome 17q and are responsible for RUNX1 transcription factor binding, directly correlated to T cells functionality [26,27,28,29]. PSORS4 resides on chromosome 1q21 and it has been associated to the presence of LCE genes (l ate cornified envelope genes) as the deletion of LCE3B and LCE3C is highly related to psoriasis, and the expression of this protein is significantly increased in psoriatic lesions [30,31]. A major impact on the development of both psoriasis and psoriatic arthritis is attributed to rs4349859 SNP located in HLA-B gene, linked to HLA-B27 [32]. The presence of HLA-B27 predict the development of certain future PsA characteristics, as dactylitis, axial involvement and also o more severe evolution of the disease, especially in early onset cases; in an opposite manner identifying the presence of HLA-B57 inputs a decreased risk of developing psoriasis [33]. Although the genetic impact on psoriasis is clearly demonstrated, there is no evidence of a single genetic variant to initiate the disease appearance as there is required an interference of several genetic mutations, along with other pathogenic pathways.

3. Microbiome

With its role of both physical and immune barrier towards external injuries, the skin has an extensive microbiome that can constitute the target of multiple factors, leading to its dysregulation and gaps in immunoregulatory processes [34,35,36]. The largest organ in the human body plays a host to several bacteria, viruses and fungi, all contributing to maintaining a balance for the immune system. The range and distribution of skin microbiota is highly different depending on the site and unique for each individual, dependent of external conditions, associated comorbidities, sex, age or hygiene level, with marked importance to different pathologies development [37]. Multiple studies have reported changes in skin microbiome for different pathologies, including psoriasis, and the analysis of specimens from both lesions and regular areas, along with probes from controls, have enlightened important data. In 2008, Gao et al., reported that Firmicutes was the most plentiful phylum populating psoriatic skin and Actinobacteria both in unaltered samples from psoriasis patients and healthy individuals [38]. A study published by Fahlén et al. revealed that there were three major microorganisms present both in normal and psoriatic skin, Firmicutes, Proteobacteria and Actinobacteria, as well as a significant higher level of Proteobacteria in psoriasis compared to controls. Another important observation of the report is represented by the lower level of Staphylococci and Propionibacteria in psoriasis patients compared to healthy individuals [39]. In 2013, Alekseyenko et al, analyzed the of lesional and non-lesional skin samples of 75 psoriasis patients and 124 controls and stated the relationship between Firmicutes and Actinobacteria-rich cutaneotype to psoriasis. Also, an important conclusion of the report that underlines the importance of skin microbiome analysis and establishing certain biomarkers, was that the presence of major skin genra, Corynebacterium, Proponibacterium, Staphylococcus and Streptococcus, both in lesions as well as in healthy skin samples of patients with psoriasis, along with a decrease of other taxa, Cupriavidus, Methylobacterium and Schlegella [40]. In 2018, Fyhrquist et al., performed an analysis of skin microbial population related to patterns of cutaneous gene expression in patients with atopic dermatitis or psoriasis, discovered only weak relationships between potential pathogens and the expression of host transcripts in patients with psoriasis, with an important role attributed to Corynebacterium, which can perform a regulatory role, potentially protective [41]. Later, Quan C et al., in a report published in 2020, showed that there is a certainly increased difference between samples from lesions and unaffected skin, as well as controls, with Propionibacterium and Corynebacterium dominating the probes with psoriasis. The results also showed a positive correlation between the severity of the lesions and the presence of Corynebacterium species. The severity of the lesions, quantified using PASI (Psoriasis Disease Activity Index), was confirmed to be directly related to the presence of Corynebacterium species by several other scientific reports [42]. Chang et al. performed an analysis of skin bacterial species that revealed significant inequalities between the psoriasis-associated and healthy skin microbiota, sustaining that a decrease of certain regulatory species, as Staphylococcus epidermidis and Propionibacterium acnes may prompt an increase settlement of Staphylococcus aureus, event that subsequently leads to an enhanced cutaneous inflammatory process via Th17 axis [43]. Another relevant study, published by Tett et al., using high-resolution shotgun metagenomics to characterize the microbiome of psoriatic and unaltered skin from 28 patients, demonstrated that members of the genus Staphylococcus are significantly more abundant on diseased skin compared to unaffected skin. The results also showed the presence of different other bacteria, as well as Malassezia spp., an abundant fungal type of the skin [44]. Although the part of Malassezia, a lipophilic and lipid-dependent commensal fungus, in psoriasis is not completely understood, it was first described by Rivolta et al. in 1873, from a psoriasis lesion [45], and T-cells reactive to Malassezia yeast [46] and antibodies against Malassezia [47] have been found in lesional skin but not in normal subjects; also, M. globosa was the most commonly isolated species during psoriasis exacerbation [48]. It is also of utmost importance that the increase of LL-37, a cathelicidin antimicrobial peptide with major role in IFN-α production, initiation and perpetuation of psoriatic lesions, can be subsequent to Malassezia colonization of the skin [49]. An additional supporting part when analyzing skin microbiome in patients with psoriasis is represented by smoking, a well known promoter of several inflammatory autoimmune conditions [50]. The colon is the leading site of micro-organism distribution, followed by the skin, with a diversity of microbiome settled from the first years of life, influenced by genetics, lifestyle or use of certain medication [41,51,52]. Disruptions in gut microbiome may promote an increased risk of metabolic and autoimmune conditions, with a potential on initiating or sustaining an inflammatory status, including psoriasis and its well described comorbidities [53,54]. Although there are several published studies, supported by advancements in sequencing technologies, the limited number of patients included lead to slight differences in the reported reports, with the benefit of providing important up to date data and outlining future researches directions. Gut microbiome integrates an extensive number of bacterial types, mostly represented by Actynobacteria, Bacteroides, Firmicutes, Fusobacteria, Proteobacteria and Verrucomicrobia, as well as viruses, fungi, protozoa or Archaea, maintaining a symbiotic status with the host, highly influenced by age, genetics, dietary manners or environmental external factors [13,55,56,57]. Changes in local intestinal microbiome can interact with skin homeostasis by influencing systemic immunomodulatory mechanisms [56]. Several reports have concluded that the changes seen in patients with psoriasis are relative semblable to those identified in patients with inflammatory bowel diseases, with an exuberance of Actinobacteria and Firmicutes, conjunctively to Firmicutes-to Bacteroides ratio, model of altered gut epithelial barrier [53,57,58,59,60]. The study of intestinal microbiota profiling, reported by Chen et al in 2018, observed that Ruminococcus and Megasphaera, of the phylum Firmicutes, were the top-two genera of discriminant abundance in psoriasis, and decreased abundance of phylum Bacteroidetes. Analyzing the samples from 35 patients with psoriasis and 27 controls [57], Huang et al reported that the relative abundances of Firmicutes and Bacteroidetes were inverted at the phylum level, and 16 kinds of phylotype at the genus level were detected with important distinctions. In addition to them, Proteobacteria and Actinobacteria were also found to be underrepresented in psoriasis patients [59]. Shapiro et al. documented a significant increase in the Firmicutes and Actinobacteria phyla as compared with controls. At the species level, the psoriatic patients presented significant increases of Ruminoccocus gnavus, Dorea formicigenerans and Collinsella aerofaciens, while Prevotella copri and Parabacteroides distasonis were significantly lower as compared to controls [58]. The study of Masallat et al. reported statistically significant differences in Firmicutes and Bacteroidetes ratio, directly associated to PASI score. Actinobacteria was found in a high level for controls. The results suggest that the differences in gut microbiome are the source for counteracting and inducing inflammation respectively, and therefore inducing psoriasis [57]. Another study, performed on 52 patients with psoriasis, revealed that the microbiome obtained was marked by an increased Faecalibacterium and a decrease of Bacteroides, with higher values of Akkermansia and Ruminococcus genra compared to controls [61]. Discordant to these results, Scher et al. obtained a significant reduction in Akkermansia, Ruminococcus, and Pseudobutyrivibrio in the analyzed samples [60]. Several scientific publications have reported other types of bacteria found with increased levels in patients with psoriasis, mentioning Bacillus, Subdoligranum, Slackia, Christensenella, Dorea, Coprococcus, Collinsella, Blautia, Enterococcus or Lactocococcus, as well as others were determined in relatively low concentrations, Allobaculum, Alistipes, Barnsiella, Gordonibacter or Paraprevotella [52,60,62]. Regarding individual species, there are studies that report an increased level for Escherichia coli, Clostridium citroniae, Collinsella aerofaciens, Dorea formicigenerans [58,59,63]. Besides the stated role of different bacterial species, viral infections, including human papiloma virus, or fungus, such as Candida albicans, or Malassezia, have been interrelated to psoriasis. The presence of Candida albicans activate dendritic cells, via its ligand, beta-glucan, inducing the production of IL-36α with a further development of psoriasis phenotype [64]. The relevance of gut microbiome on the pathogenesis of psoriasis is settled by the interrelation between different components of both innate and adaptive immune systems [56,65,66,67,68,69,70]. The main role of gut-skin axis in psoriasis is endorsed by T cells, by the imbalance between Treg and Th17 cells [71,72]. There are data suggesting that the absence of microbiota, or its change, decreases the pro-inflammatory T cell response and further decreases the severity of cutaneous inflammation. This is further supported by several studies describing the ability of commensal bacteria to modulate T cell development. An increased epithelial permeability, subsequent to chronic inflammation derived from gut dysbiosis, is one of the underlying mechanisms of skin impairment. Another pathway for systemic inflammatory state is represented by metabolic disturbances, with activation of several pattern recognition receptors, located on epithelial cells. As a result of the altered integrity of the mucosal cells and the increased permeability, effector T cells are activated. The bidirectional relationship between gut microbiota and mucosal epithelial cells, directly impacting the protective and functional status, is driven by the specific metabolites or immune modulating factors, between which it is worth mentioning short chain fatty acids (SCFAs), retinoic acid or polysaccharide A, as a result of the fermentation of non-digestible substrates, complex polysaccharides and indigestible oligosaccharides by colonic microorganisms [58,67,73,74]. SCFAs modulate glucose and lipid metabolism [75], maintain gut mucosal integrity [76], and regulate the immune system and inflammatory responses [77]. All these actions are mediated through different mechanisms, including specific G protein coupled receptor family (GPCR) and epigenetic effects [78,79,80,81]. Disruptions in gut microbiome balance and the further, activation of T- cells via interactions with pattern recognition receptors and Toll-like receptors triggers an inflammatory process and induces autoimmune conditions such as rheumatoid arthritis [82], inflammatory bowel disease [83], systemic lupus erythematosus [84], multiple sclerosis [85], psoriasis [56,65,66,67,68,69,70,71,72], as well as other skin alterations, such as atopic dermatitis [86] and vitiligo [87].

4. Immune Cells

4.1. Keratinocytes (KCs)

Psoriasis is a pathology characterized by hyperproliferation and disturbed differentiation of KCs, cells with both structural and immune role that interfere in early stages of the disease, as well as in maintaining chronic inflammation [88]. They constitute the main cellular population altered in psoriasis as they express receptors for all types of cytokines involved in the complex pathogenesis of the disease [89,90,91]. Consequent to dermal injury there can be observed an increased secretion of antimicrobial peptides, cathelicidin, LL-37, S100 proteins, and β-defensins from keratinocytes, promoting the activation of pDCs and release of IFN-α, IFN-γ, TNF-α, and IL-1β [92,93,94]. Besides the stated role in early stages of the disease, they continue to perpetuate the inflammatory process during disease evolution. After activation, they continuously produce chemokines, in high amounts (CXCL1/2/3, CXCL8, CXCL9/10/11, CCL2, and CCL20), recruit leucocytes or antimicrobial peptides and stimulate the inflammatory process. In addition, along with fibroblasts and endothelial cells have a major contribution to tissue restructuring, consecutive to their interaction with Th17 cells [95,96,97].

4.2. T Cells

It is well-established that psoriasis is a T-cell mediated disease, as it is assumed that effector T cells transmigrate form the lymph nodes into systemic circulation, as well as they derive from the injured skin sites [98,99]. All types of T cells, CD4+ T cells (Th), CD8+ T cells (Tc), and Treg cells, are involved in the initiation and perpetuation of psoriatic lesions [100]. Proinflammatory cytokines, IL-12, IL-23 and TNF, released by dendritic cells (DC) and activated macrophages, prompt the activity of Th cells (Th1, Th17, Th22) and promote epidermal hyperproliferation with an inhibition of apoptosis [100,101]. Concomitant, IL-17 and TNFα stimulate keratinocytes to release pro-inflammatory cytokines. Psoriasis patients in the early course of disease express high levels of IFN-γ, which shifted towards IL-10 secretion in chronic patients suggesting a possible shift from Th1 to Th2 response as an adaptation of the immune system to down regulate inflammatory Th1 response [101]. Another cytokine with contribution in psoriasis pathogenesis is represented by Th9, a majority of the memory Th9 cells being either skin-tropic or skin-resident. IL-9, released by Th9, is essential for production of IFN-γ, IL-17, and IL-13 by skin tropic T cells to maximal level [102]. Treg cells, known to conserve immune stability, are found in a low level in psoriasis, and in the context of an increased pro-inflammatory cytokine environment, psoriatic Tregs behave like Th17 cells and are unable to suppress the T effector activation [103,104].

4.3. B Cells

Known to be regulators of the pathogenesis in several autoimmune diseases, by producing autoantibodies and antigen presentation, they haven't been as extensively studied as lymphocytes T in patients with psoriasis and their input on psoriasis genesis and perpetuation is not completely clear [105]. Resident cutaneous B cells contribute not only to skin homeostasis, but also to regulating reparatory processes and local microbiome. Also, experimental studies proved that in areas of inflamed skin there is a high quantity of Il-6, IL-4, GM-CSF or IFN-γ [105,106,107,108].

4.4. Dendritic Cells (DCs)

Several types of DCs (plasmacytoid DCs-pDCs, conventional DCs-cDCs, and Langerhans cells-LCs) are involved in the pathogenesis of psoriasis, with numerous reports demonstrating the presence of pDCs and cDCs in the lesional skin samples of these patients [109,110]. Consecutive to different external stimulus DCs are activated, commuted to professional antigen-presenting cells (APCs), with subsequent interaction with naive T cells and production of proinflammatory cytokines, TNF-α, IL-23, IL-12, and IL-6, that furthermore activate inflammatory reaction, keratinocyte proliferation and recruitment of neutrophils [111,112]. A subset of inflammatory Dcs, TIP-DCs, secrete TNF and inducible nitric oxide synthase (iNOS), with a pro-inflammatory response in patients with psoriasis [113,114,115]. LCs, cells with an important contribution to local immunity of the skin, showed a debatable role in psoriasis pathogenesis. Experimental studies have shown divergent results, with reports revealing that they stimulate the secretion of multiple inflammatory cytokines and induce the proliferation of IL-17A-producing γδ T cells, or others suggesting they may exert an anti-inflammatory role in psoriasis, by increasing the production of IL-10, or IL-23 in case of LCs depletion [116,117,118,119,120]. 4.5 Neutrophils. Excessive storage of neutrophils in psoriatic lesions is a typical feature of the disease as they infiltrate the dermis in early stages and subsequently migrate to the epidermis [121,122]. Subsequently to neutrophils activation, increased levels of reactive oxygen species (ROS) are released into circulation, with DCs stimulation, T cells stimulation and an altered balance between Th1 and Th2, the final result being represented by keratinocyte proliferation and increased angiogenesis [122,123,124,125,126]. Secondary to excessive ROS production, LL-37 is released, with a production of IFN-alfa by pDCs or ILP6, IL-12 and IL-23 by mDCs [127,128]. In addition to Th17 cells, neutrophils represent a category of cells that can contribute to IL-17A production and perpetuation of psoriasis development. Moreover, neutrophils are activated by keratinocytes in order to produce pro-inflammatory cytokines and augment dermal inflammation [129,130].

4.6. Macrophages

Antigen presenting cells, derived from monocytes, are shown to be present in psoriatic lesions, with TNF secretion upon activation and regulation of angiogenesis by releasing VEGF [131,132].

4.7. Natural Killer (NK) Cells

Natural killer (NK) cells are present in the lesional skin of psoriasis patients and exhibited reduced degranulation and produced lower levels of the pro-inflammatory cytokines IFN-γ and TNF-α [73].

5. Citokines

5.1. Th17 Cytokines

One of the first studies describing cytokines in the dermal samples of psoriasis patients, published in 1994 by Schlaak et al, revealed that Th1-related cytokines prevailed in the analyzed probes, with a few Th cells releasing Th-2 relate cytokines [133], results that were confirmed by others several further reports and rised the hypothesis that psoriasis can be defined as a Th1 mediated disease. Currently, it is very well established that the pathophysiology of the disease comprises a wide network of immune cells that, along with their cytokines, initiate the inflammatory response [134,135]. The Th17 cytokine family is the major effector in the pathogenesis of psoriatic disease and strongly influences the inflammatory pattern established during the disease activity [136,137]. In addition, the vast network of cells that orchestrates the pathophysiology makes psoriasis complex to study. IL-23, part of IL-12 family, IL-23 is produced by both resident [blood dendritic cell antigen (BDCA-1)/CD1c+] and inflammatory myeloid DCs (CD11c+ BDCA-1/CD1c–), as well as macrophages (CD163+) in psoriasis. It is responsible for the activation, differentiation and proliferation of Th17 cells that further initiate the release of acting cytokines IL-17A or IL-21, with effects on neutrophils draft [138,139,140,141,142,143,144].In psoriasis patients, the IL-23 pathway is activated and characterized by high level production of IL-23 by DCs and keratinocytes and increased numbers of Th17 cells [145]. IL-17is an acknowledged cytokine produced mainly, but in exclusively, by Th17 cells [146]. Besides Th1 cells, mast cells, γδ T cells, αβ T cells, and innate lymphoid cells represent major sources for IL-17 production [147,148]. From the six subtypes of IL-17 family, IL-17A and IL-17F have been proven to be found both in psoriasis plauqes, as well as in unaltered skin, with IL-17A playing a central part in the pathogenesis of the disease [146]. The aforementioned cytokines are expressed together and act in a synergist manner on upregulation of inflammatory markers. There are several mechanisms with intricate casting in psoriasis induction by which IL-17 exerts their action on immune cells, keratinocytes, and fibroblasts, inducing the release of inflammatory mediators including cytokines, IL-6, IL-1β, TNF, and granulocyte–macrophage colony-stimulating factor (GM-CSF), chemokines (CXCL1, CXCL2, CCL20, and CXCL8), matrix metalloproteinases (MMPs), antimicrobial peptides (AMPs; LL37, S100s, β-defensin), and complement [149,150,151]. Thus, initiates recruitment and activation of neutrophils, lymphocytes, and myeloid cells, with local dermal inflammation, effect developed by the synergic effects of IL-23 and possibly IL-36. Increased IL-17, produced by CD4+ Th17 cells, can also be detected in synovial fluid of patients with psoriasis. The effect of IL-17 goes beyond epithelial cells, being identified also in endothelial cells, with a pro-coagulant activation [151,152]. IL-21 is a cytokine still under current investigations regarding its entire part in psoriasis, with high amounts of IL-21 receptor (IL-21R) revealed in keratinocytes. It represents a future therapeutic target as it has been stated as a major effect on Th17 proliferation, mediated by regulatory T cells (Treg) [153,154].

5.2. Th1 Cytokines

IL-12 is an activator for Th1 differentiation and proliferation, with consequent production of TNF-β, IFN-γ and IL-2, found to be increased in patients with psoriasis [155,156]. A major implication has been reported in psoriatic arthritis (PsA), where high levels of its soluble receptor (IL-12R) are directly correlated with the degree of cutaneous involvement [157,158]. IL-12 inhibitory agents proved their efficacy as they diminish the expression of IFN-γ and inhibits the release of IL-8 by keratinocytes; additionally experimental studies have shown a protective role of this cytokine in IL-23'Th17 pathway [143].
IL-8, a cytokine with marker role in inflammation throughout neutrophils attraction, is released by macrophages, epithelial cells, endothelial cells or respiratory smooth muscles. Keratinocytes production of IL-8 is stimulated by visfatin, an adipokine produced by leukocytes and adipose tissue, under TNF-α stimulation [159,160,161].
IL-22, a member of IL-10 family, that impairs keratinocytes proliferation, is associated with psoriasis progression when detected in high levels. It is secreted by NK cells, Th17 or Th22, with a marked input on keratinocytes cycle during regenerative processes [162]. It is notably that the study of Kagami et al. showed that IL-22 deficiency caused a significant decrease in epidermal acanthosis and dermal inflammation induced by IL-23 [163]. In addition, the study published by Van Belle et al. showed that IL-22 has not only a major part in the development of pustules and acanthosis but also in neutrophil infiltration in a mouse model triggered by the Toll-like receptor (TLR) 7/8 agonist imiquimod [164].
IL-1β is a cytokine highly expressed in psoriasis samples, and two genetic variants, rs16944 and rs2853550, were proven to be associated with late initiation of the disease and a slower progression [165].
IL-6 is expressed in high levels in the psoriatic lesion. It is a pleiotropic proinflammatory cytokine that is produced by a variety of cells such as fibroblasts, macrophages, endothelial cells, and KCs in response to a variety of stimuli, which include other cytokines such as IL-1, TNF-α, and PDGF [166]. IL-6 stimulates the proliferation of human KCs. Anti–IL-6 therapies, which are effective for rheumatoid arthritis, are either ineffective for psoriasis or can induce new-onset psoriasis-like disease [167].
TNF-α has been identified as a key cytokine mediating cutaneous inflammation in the pathogenesis of psoriasis [168]. TNF is a homotrimer cytokine mostly released by immune and epithelial cells and exerts its effects by binding to two different receptors: TNFR1/p55 (expressed ubiquitously and constitutively) and TNFR2/p75 (expressed only on immune, endothelial and neuronal cells, being inducible). Over the years, several studies have reported increased levels of TNF, TNFR1 and TNFR2 are observed in psoriatic lesions, being produced by various cellular populations involved in the pathophysiology of psoriasis, such as keratinocytes, dendritic cells (DCs), and NKT, Th1, Th17 and Th22 cells [169]. TNF induces not only immune and inflammatory responses orchestrated by keratinocytes but also tissue remodeling, cell motility, cell cycling, and apoptosis [170,171]. Additionally, activated keratinocytes also produce many chemokines responsible for recruitment of neutrophils, macrophages and skin-specific memory T cells, observations that underlines the fact that TNF might be involved in both the initial phase and the chronic phase of psoriasis [172].
T reg cells that express FOXP3 constitute major regulators of the immune system and inflammatory response. They inhibit the action of various immune cellular populations either directly or by releasing cytokines as IL-10 or TGF-β that act in a suppressing manner. The activity of T reg cells is probable to be impaired in psoriasis, as there are data sustaining that STAT3, activated by pro-inflammatory cytokines, inhibits T reg. IL-10, an anti-inflammatory cytokine, produced by regulatory T cells, has been stated as a major mediator in psoriasis and proved to be an important inhibitor for both pro-inflammatory T cell responses and keratinocyte inflammatory markers. The report of Asadullah et al. showed that the levels of mRNA along with the cytokine itself were decreased significantly in patients with psoriasis compared to other dermatologic disorders. An additional reported information was that the administration of recombinant IL-10 for 30 days diminished PASI score as well as Th-1 cytokines levels [173].

6. Intracellular Signaling Pathways

6.1. Janus Kinase-Signal Transducer and Activator of Transcription (JAK/STAT Pathway).

JAK is a member of tyrosine kinase (TYK) family, which comprises four members: JAK1, JAK2, JAK3 and TYK2, with various expression depending on the cellular type [174]. The development of various inflammatory autoimmune diseases is dependent on JAKs activation and phosphorylation of STATs, throughout several proinflammatory cytokines. Although the pathogenesis of psoriasis comprises multiple types of cytokines, the major role is attributed to IL-23 and Th17, directly linked to JAK/STAT pathway [175]. An increased expression of STAT1, directly related to JAK1/JAK2, determines an activation of IFN-α/β and IFN-γ. IFN-γ and IL-12 by a TYK2-dependent mechanism have a major input on keratinocytes stimulation and promote psoriatic inflammation [176]. An important observation was made by the study published by Nada et al. that reported a positive correlation between JAK1 and PASI score, as well as a significantly higher level compared to controls [177]. Other reports, performed on mouse models of psoriasis, concluded that TYK2 is able to slow the progression of psoriasis. STAT3, a major regulator for Th17 differentiation, via IL-23 activation, has been shown to be overexpressed in psoriasis skin samples and it constitutes a key regulator for keratinocytes [176]. In a similar manner, IL-22 stimulates keratinocytes proliferation and inhibits their differentiation, consecutive to STAT3 activation [178,179].

6.2. A3 Adenosine Signaling Pathway

Adenosine generated by ATP catabolism, acts as a supressive metabolite with immunomodulatory and anti-inflammatory functions enabled through four receptor subtypes A1, A2A, A2B and A3 [180]. A3 receptors are expressed on all types of immune cells and exert a pro-inflammatory effect in the respiratory system, while also having an anti-inflammatory action against LPS-induced cytokine release. This has enabled the development of various selective A3 receptor ligands [181]. Evidence of the role of A3Ars in modulating immune cell function and also positive results obtained in rheumatoid arthritis studies, has led to the development of a specific inhibitor CF10 (piclidenoson) for the treatment of psoriasis. It has shown positive results in vitro on cell growth and a reduction of PASI score. In addition, it lowers the expression of pro-inflammatory markers, like TNF- α, IL-17, IL-23 and phosphoinositide-3-kinase (PI3K) [182].

6.3. WNT

WNT signaling pathway plays an important role in cell growth, differentiation and migration. In psoriasis, WNT signaling generates an overexpression of WNT5A, with studies showing increased levels in affected areas, together with increased expression of its receptor proteins FZD2 and FZD5 [183]. Data suggests that the epidermis is one of the main sources of WNT5A in psoriatic lesions, with effects on inflammatory responses of human mononuclear cells and blood vessel proliferation [184,185].

6.4. NF-κB Signaling

The transcription factor NF-κB direct several processes including inflammation, proliferation, immunological actions, apoptosis and differentiation. Ccompared to normal samples, psoriatic derma presents high levels of active phosphorylated NF-κB. It influences psoriasis pathogenesis by stimulating TGF expression, VEGF induction and angiogenesis; also, binds to keratinocytes, with consequent cytokine production and chemokines activation [186].

7. Conclusions

Psoriasis etiology and extensive pathogenic pathways are not yet completely described with several hypotheses being proposed. Understanding the immunologic, genetic and autoimmune hallmarks of psoriasis has known an extensive development in the last decades and elucidating the underlying mechanism of the complex pathologic process associated to psoriatic disease enabled a tremendous progress in new and individualized therapeutic approaches, further to be extend by future characterization of the disease.

Author Contributions

Conceptualization, R.C.D., A.L.B., L.E.S., H.V.P., R.A.I., Ș.C.D., C.E.B., F.A.V.; writing—original draft preparation, R.C.D., C.E.B, A.L.B., L.E.S, Ș.C.D.; writing—review and editing, R.C.D., L.E.S., Ș.C.D, H.V.P., R.A.I, C.E.B. and F.A.V.; visualization, R.C.D., A.L.B., L.E.S., H.V.P., R.A.I., Ș.C.D., C.E.B., F.A.V.; supervision, F.A.V and A.L.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest .

References

  1. Samotij, D.; Nedoszytko, B.; Bartosińska, J.; Batycka-Baran, A.; Czajkowski, R.; Dobrucki, I.; Dobrucki, L.; Górecka-Sokołowska, M.; Janaszak-Jasienicka, A.; Krasowska, D.; et al. Pathogenesis of psoriasis in the “omic” era. Part I. Epidemiology, clinical manifestation, immunological and neuroendocrine disturbances. Adv. Dermatol. Allergol. 2020, 37, 135–153. [Google Scholar] [CrossRef] [PubMed]
  2. 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]
  3. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef] [PubMed]
  4. World Health, O. Global Report on Psoriasis. World Health Organization; Geneva, Switzerland: 2016. Available online: https://www.who.int/publications/i/item/9789241565189.
  5. Parisi, R.; Iskandar, I.Y.K.; Kontopantelis, E.; Augustin, M.; Griffiths, C.E.M.; Ashcroft, D.M. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ 2020, 369, m1590. [Google Scholar] [CrossRef] [PubMed]
  6. Raharja, A.; Mahil, S.K.; Barker, J.N. Psoriasis: a brief overview. Clin. Med. 2021, 21, 170–173. [Google Scholar] [CrossRef]
  7. Takeshita, J.; Grewal, S.; Langan, S.M.; Mehta, N.N.; Ogdie, A.; Van Voorhees, A.S.; Gelfand, J.M. Psoriasis and comorbid diseases: Epidemiology. J Am Acad Dermatol. 2017, 76, 377–390. [Google Scholar] [CrossRef] [PubMed]
  8. Lee, H.-J.; Kim, M. Challenges and Future Trends in the Treatment of Psoriasis. Int. J. Mol. Sci. 2023, 24, 13313. [Google Scholar] [CrossRef] [PubMed]
  9. Conrad, C.; Gilliet, M. Psoriasis: from Pathogenesis to Targeted Therapies. Clin. Rev. Allergy Immunol. 2018, 54, 102–113. [Google Scholar] [CrossRef] [PubMed]
  10. Ghoreschi, K.; Balato, A.; Enerbäck, C.; Sabat, R. Therapeutics targeting the IL-23 and IL-17 pathway in psoriasis. Lancet 2021, 397, 754–766. [Google Scholar] [CrossRef]
  11. Chen, L.; Li, J.; Zhu, W.; Kuang, Y.; Liu, T.; Zhang, W.; Chen, X.; Peng, C. Skin and Gut Microbiome in Psoriasis: Gaining Insight Into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front. Microbiol. 2020, 11, 589726. [Google Scholar] [CrossRef]
  12. Celoria, V.; Rosset, F.; Pala, V.; Dapavo, P.; Ribero, S.; Quaglino, P.; Mastorino, L. The Skin Microbiome and Its Role in Psoriasis: A Review. Psoriasis: Targets Ther. 13. [CrossRef]
  13. Sikora, M.; Stec, A.; Chrabaszcz, M.; Knot, A.; Waskiel-Burnat, A.; Rakowska, A.; Olszewska, M.; Rudnicka, L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens 2020, 9, 463. [Google Scholar] [CrossRef] [PubMed]
  14. Nanda, H.; Ponnusamy, N.; Odumpatta, R.; Jeyakanthan, J.; Mohanapriya, A. Exploring genetic targets of psoriasis using genome wide association studies (GWAS) for drug repurposing. 3 Biotech 2020, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  15. Dand, N.; Stuart, P.E.; Bowes, J.; Ellinghaus, D.; Nititham, J.; Saklatvala, J.R.; Teder-Laving, M.; Thomas, L.F.; Traks, T.; Uebe, S.; Assmann, G.; Baudry, D.; Behrens, F.; Billi, A.C.; Brown, M.A.; Burkhardt, H.; Capon, F.; Chung, R.; Curtis, C.J.; Duckworth, M.; Ellinghaus, E.; FitzGerald, O.; Gerdes, S.; Griffiths, C.E.M.; Gulliver, S.; Helliwell, P.; Ho, P.; Hoffmann, P.; Holmen, O.L.; Huang, Z.M.; Hveem, K.; Jadon, D.; Köhm, M.; Kraus, C.; Lamacchia, C.; Lee, S.H.; Ma, F.; Mahil, S.K.; McHugh, N.; McManus, R.; Modalsli, E.H.; Nissen, M.J.; Nöthen, M.; Oji, V.; Oksenberg, J.R.; Patrick, M.T.; Perez-White, B.E.; Ramming, A.; Rech, J.; Rosen, C.; Sarkar, M.K.; Schett, G.; Schmidt, B.; Tejasvi, T.; Traupe, H.; Voorhees, J.J.; Wacker, E.M.; Warren, R.B.; Wasikowski, R.; Weidinger, S.; Wen, X.; Zhang, Z. BSTOP study group; Estonian Biobank research team; Barton, A. ; Chandran, V.; Esko, T.; Foerster, J.; Franke, A.; Gladman, D.D.; Gudjonsson, J.E.; Gulliver, W.; Hüffmeier, U.; Kingo, K.; Kõks, S.; Liao, W.; Løset, M.; Mägi, R.; Nair, R.P.; Rahman, P.; Reis, A.; Smith, C.H.; Di Meglio, P.; Barker, J.N.; Tsoi, L.C.; Simpson, M.A.; Elder, J.T. GWAS meta-analysis of psoriasis identifies new susceptibility alleles impacting disease mechanisms and therapeutic targets. medRxiv [Preprint]. 2023, 5, 2023100423296543. [Google Scholar]
  16. Devlin, B.; Roeder, K.; Wasserman, L. Genomic Control, a New Approach to Genetic-Based Association Studies. Theor. Popul. Biol. 2001, 60, 155–166. [Google Scholar] [CrossRef] [PubMed]
  17. Stuart, P.E.; Tsoi, L.C.; Nair, R.P.; Ghosh, M.; Kabra, M.; Shaiq, P.A.; Raja, G.K.; Qamar, R.; Thelma, B.K.; Patrick, M.T.; Parihar, A.; Singh, S.; Khandpur, S.; Kumar, U.; Wittig, M.; Degenhardt, F.; Tejasvi, T.; Voorhees, J. ; Weidinger, S.; Franke, A.; Abecasis, G.R.; Sharma, V.K.; Elder, J.T. Transethnic analysis of psoriasis susceptibility in South Asians and Europeans enhances fine-mapping in the MHC and genomewide. HGG Adv. 2022, 3, 100069. [PubMed]
  18. 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.; Ruether, A.; Schreiber, S.; Weichenthal, M.; Gladman, D.; Rahman, P.; Schrodi, S.J.; Prahalad, S.; Guthery, S.L.; Fischer, J.; Liao, W.; Kwok, P.Y.; Menter, A.; Lathrop, G.M.; Wise, C.A.; Begovich, A.B.; Voorhees, J.J.; Elder, J.T.; Krueger, G.G.; Bowcock, A.M.; Abecasis, G.R. Genome-wide scan reveals association of psoriasis with IL-23 and NF-kappaB pathways. Nat. Genet. 2009, 41, 199–204. [Google Scholar] [CrossRef]
  19. Di Meglio, P.; Villanova, F.; Nestle, F.O. Psoriasis. Cold Spring Harb. Perspect. Med. 2014, 4, a015354–a015354. [Google Scholar] [CrossRef]
  20. Singh, S.; Pradhan, D.; Puri, P.; Ramesh, V.; Aggarwal, S.; Nayek, A.; Jain, A. Genomic alterations driving psoriasis pathogenesis. Gene 2018, 683, 61–71. [Google Scholar] [CrossRef]
  21. Nair, R.P.; Stuart, P.E.; Nistor, I.; Hiremagalore, R.; Chia, N.V.; Jenisch, S.; Weichenthal, M.; Abecasis, G.R.; Lim, H.W.; Christophers, E.; et al. Sequence and Haplotype Analysis Supports HLA-C as the Psoriasis Susceptibility 1 Gene. Am. J. Hum. Genet. 2006, 78, 827–851. [Google Scholar] [CrossRef]
  22. Temel, B.; Adisen, E.; Gonen, S. HLA-Cw6 status and treatment responses between psoriasis patients. Indian J. Dermatol. 2021, 66, 632–637. [Google Scholar] [CrossRef]
  23. Jordan, C.T.; Cao, L.; Roberson, E.D.; 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]
  24. Scudiero, I.; Zotti, T.; Ferravante, A.; Vessichelli, M.; Vito, P.; Stilo, R. Alternative splicing of CARMA2/CARD14 transcripts generates protein variants with differential effect on NF-κB activation and endoplasmic reticulum stress-induced cell death. J Cell Physiol. 2011, 226, 3121–31. [Google Scholar] [CrossRef] [PubMed]
  25. Fuchs-Telem, D.; Sarig, O.; van Steensel, M.A.; Isakov, O.; Israeli, S.; Nousbeck, J.; Richard, K.; Winnepenninckx, V.; Vernooij, M.; Shomron, N.; et al. Familial Pityriasis Rubra Pilaris Is Caused by Mutations in CARD14. Am. J. Hum. Genet. 2012, 91, 163–170. [Google Scholar] [CrossRef] [PubMed]
  26. Helms, C.; Cao, L.; Krueger, J.G.; Wijsman, E.M.; Chamian, F.; Gordon, D.; Heffernan, M.; Daw, J.A.W.; Robarge, J.; Ott, J.; et al. A putative RUNX1 binding site variant between SLC9A3R1 and NAT9 is associated with susceptibility to psoriasis. Nat. Genet. 2003, 35, 349–356. [Google Scholar] [CrossRef] [PubMed]
  27. Ono, M.; Yaguchi, H.; Ohkura, N.; Kitabayashi, I.; Nagamura, Y.; Nomura, T.; Miyachi, Y.; Tsukada, T.; Sakaguchi, S. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature 2007, 446, 685–9. [Google Scholar] [CrossRef] [PubMed]
  28. Kitoh, A.; Ono, M.; Naoe, Y.; Ohkura, N.; Yamaguchi, T.; Yaguchi, H.; Kitabayashi, I.; Tsukada, T.; Nomura, T.; Miyachi, Y.; Taniuchi, I.; Sakaguchi, S. Indispensable role of the Runx1-Cbfbeta transcription complex for in vivo-suppressive function of FoxP3+ regulatory T cells. Immunity 2009, 31, 609–20. [Google Scholar] [CrossRef] [PubMed]
  29. Wong, W.F.; Kohu, K.; Chiba, T.; Sato, T.; Satake, M. Interplay of transcription factors in T-cell differentiation and function: the role of Runx. Immunology 2010, 132, 157–164. [Google Scholar] [CrossRef]
  30. de Cid, R.; Riveira-Munoz, E.; Zeeuwen, P.L.J.M.; Robarge, J.; Liao, W.; Dannhauser, E.N.; Giardina, E.; Stuart, P.E.; Nair, R.; Helms, C.; et al. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nat. Genet. 2009, 41, 211–215. [Google Scholar] [CrossRef]
  31. Riveira-Munoz, E.; He, S.-M.; Escaramís, G.; Stuart, P.E.; Hüffmeier, U.; Lee, C.; Kirby, B.; Oka, A.; Giardina, E.; Liao, W.; et al. Meta-Analysis Confirms the LCE3C_LCE3B Deletion as a Risk Factor for Psoriasis in Several Ethnic Groups and Finds Interaction with HLA-Cw6. J. Investig. Dermatol. 2011, 131, 1105–1109. [Google Scholar] [CrossRef]
  32. Capon, F. The Genetic Basis of Psoriasis. Int. J. Mol. Sci. 2017, 18, 2526. [Google Scholar] [CrossRef]
  33. Queiro, R.; Morante, I.; Cabezas, I.; Acasuso, B. HLA-B27 and psoriatic disease: a modern view of an old relationship. Rheumatology 2015, 55, 221–229. [Google Scholar] [CrossRef] [PubMed]
  34. Baba, H.; Masuyama, A. Yoshimura, C. (2012). Promoter of differentiation and keratinization of epidermic cell and functional beverage/food for promotion of keratinization of epidermis. 2012, U.S. Patent NO CA2614111A1.
  35. Abhishek, S. ; M. Tech.; Krishnan, S.P.; Ph.D. Epidermal Differentiation Complex: A Review on Its Epigenetic Regulation and Potential Drug Targets.. 2016, 18, 1–6. [Google Scholar] [CrossRef]
  36. Daniel, K. ; Hsu, Maxwell, A.; Fung, Hung-Lin Chen. Role of skin and gut microbiota in the pathogenesis of psoriasis, an inflammatory skin disease. Medicine in Microecology 2020, 4.
  37. Liang, X.; Ou, C.; Zhuang, J.; Li, J.; Zhang, F.; Zhong, Y.; Chen, Y. Interplay Between Skin Microbiota Dysbiosis and the Host Immune System in Psoriasis: Potential Pathogenesis. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef] [PubMed]
  38. Gao, Z.; Tseng, C.-H.; Strober, B.E.; Pei, Z.; Blaser, M.J. Substantial Alterations of the Cutaneous Bacterial Biota in Psoriatic Lesions. PLOS ONE 2008, 3, e2719. [Google Scholar] [CrossRef] [PubMed]
  39. Fahlén, A.; Engstrand, L.; Baker, B.S.; Powles, A.; Fry, L. Comparison of bacterial microbiota in skin biopsies from normal and psoriatic skin. Arch. Dermatol. Res. 2012, 304, 15–22. [Google Scholar] [CrossRef] [PubMed]
  40. Alekseyenko, A.V.; Perez-Perez, G.I.; De Souza, A.; Strober, B.; Gao, Z.; Bihan, M.; Li, K.; Methé, B.A.; Blaser, M.J. Community differentiation of the cutaneous microbiota in psoriasis. Microbiome 2013, 1, 31. [Google Scholar] [CrossRef] [PubMed]
  41. Fyhrquist, N.; Muirhead, G.; Prast-Nielsen, S.; Jeanmougin, M.; Olah, P.; Skoog, T.; Jules-Clement, G.; Feld, M.; Barrientos-Somarribas, M.; Sinkko, H.; et al. Microbe-host interplay in atopic dermatitis and psoriasis. Nat. Commun. 2019, 10, 1–15. [Google Scholar] [CrossRef]
  42. Quan, C.; Chen, X.-Y.; Li, X.; Xue, F.; Chen, L.-H.; Liu, N.; Wang, B.; Wang, L.-Q.; Wang, X.-P.; Yang, H.; et al. Psoriatic lesions are characterized by higher bacterial load and imbalance between Cutibacterium and Corynebacterium. J. Am. Acad. Dermatol. 2019, 82, 955–961. [Google Scholar] [CrossRef] [PubMed]
  43. Chang, H.-W.; Yan, D.; Singh, R.; Liu, J.; Lu, X.; Ucmak, D.; Lee, K.; Afifi, L.; Fadrosh, D.; Leech, J.; et al. Alteration of the cutaneous microbiome in psoriasis and potential role in Th17 polarization. Microbiome 2018, 6, 1–27. [Google Scholar] [CrossRef]
  44. Tett, A.; Pasolli, E.; Farina, S.; Truong, D.T.; Asnicar, F.; Zolfo, M.; Beghini, F.; Armanini, F.; Jousson, O.; De Sanctis, V.; et al. Unexplored diversity and strain-level structure of the skin microbiome associated with psoriasis. npj Biofilms Microbiomes 2017, 3, 1–12. [Google Scholar] [CrossRef]
  45. Paulino, L.C.; Tseng, C.-H.; Blaser, M.J. Analysis of Malassezia microbiota in healthy superficial human skin and in psoriatic lesions by multiplex real-time PCR. FEMS Yeast Res. 2008, 8, 460–471. [Google Scholar] [CrossRef] [PubMed]
  46. Baker, B.S.; Powles, A.; Garioch, J.J.; Hardman, C.; Fry, L. Differential T-cell reactivity to the round and oval forms of Pityrosporum in the skin of patients with psoriasis. . 1997, 136, 319–25. [Google Scholar] [PubMed]
  47. Kanda, N.; Tani, K.; Enomoto, U.; Nakai, K.; Watanabe, S. The skin fungus-induced Th1- and Th2-related cytokine, chemokine and prostaglandin E2 production in peripheral blood mononuclear cells from patients with atopic dermatitis and psoriasis vulgaris. Clin Exp Allergy 2002, 32, 1243–50. [Google Scholar] [CrossRef] [PubMed]
  48. Gomez-Moyano, E.; Crespo-Erchiga, V.; Martínez-Pilar, L.; Godoy Diaz, D.; Martínez-García, S.; Lova Navarro, M.; Vera Casaño, A. Do Malassezia species play a role in exacerbation of scalp psoriasis? J Mycol Med. 2014, 24, 87–92. [Google Scholar] [CrossRef] [PubMed]
  49. Reinholz, M.; Ruzicka, T.; Schauber, J. Cathelicidin LL-37: An Antimicrobial Peptide with a Role in Inflammatory Skin Disease. Ann. Dermatol. 2012, 24, 126–135. [Google Scholar] [CrossRef] [PubMed]
  50. Assarsson, M.; Söderman, J.; Dienus, O.; Seifert, O. Significant Differences in the Bacterial Microbiome of the Pharynx and Skin in Patients with Psoriasis Compared with Healthy Controls. Acta Dermato-Venereologica 2020, 100, adv00273. [Google Scholar] [CrossRef]
  51. McKenzie, C.; Tan, J.; Macia, L.; Mackay, C.R. The nutrition-gut microbiome-physiology axis and allergic diseases. Immunol Rev. 2017, 278, 277–295. [Google Scholar] [CrossRef] [PubMed]
  52. Hidalgo-Cantabrana, C.; Gómez, J.; Delgado, S.; Requena-López, S.; Queiro-Silva, R.; Margolles, A.; Coto, E.; Sánchez, B.; Coto-Segura, P. Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br. J. Dermatol. 2019, 181, 1287–1295. [Google Scholar] [CrossRef]
  53. Chen, Y.; Ho, H.J.; Tseng, C.; Lai, Z.; Shieh, J.; Wu, C. Intestinal microbiota profiling and predicted metabolic dysregulation in psoriasis patients. Exp. Dermatol. 2018, 27, 1336–1343. [Google Scholar] [CrossRef]
  54. Visser, M.J.E.; Kell, D.B.; Pretorius, E. Bacterial Dysbiosis and Translocation in Psoriasis Vulgaris. Front. Cell. Infect. Microbiol. 2019, 9, 7. [Google Scholar] [CrossRef]
  55. Adarsh, M.B.; Dogra, S.; Vaiphei, K.; Vaishnavi, C.; Sinha, S.; Sharma, A. Evaluation of subclinical gut inflammation using faecal calprotectin levels and colonic mucosal biopsy in patients with psoriasis and psoriatic arthritis. Br. J. Dermatol. 2019, 181, 401–402. [Google Scholar] [CrossRef] [PubMed]
  56. Spencer, S.P.; Fragiadakis, G.K.; Sonnenburg, J.L. Pursuing Human-Relevant Gut Microbiota-Immune Interactions. Immunity 2019, 51, 225–239. [Google Scholar] [CrossRef] [PubMed]
  57. Masallat, D.; Moemen, D. ; State, Ahmed. Gut bacterial microbiota in psoriasis: A case control study. African Journal of Microbiology Research 2016, 10, 1337–1343. [Google Scholar]
  58. Shapiro, J.; Cohen, N.A.; Shalev, V.; Uzan, A.; Koren, O.; Maharshak, N. Psoriatic patients have a distinct structural and functional fecal microbiota compared with controls. J. Dermatol. 2019, 46, 595–603. [Google Scholar] [CrossRef] [PubMed]
  59. Huang, L.; Gao, R.; Yu, N.; Zhu, Y.; Ding, Y.; Qin, H. Dysbiosis of gut microbiota was closely associated with psoriasis. Sci. China Life Sci. 2018, 62, 807–815. [Google Scholar] [CrossRef] [PubMed]
  60. Scher, J.U.; Ubeda, C.; Artacho, A.; M, A.; Isaac, S.; Reddy, S.M.; Marmon, S.; Neimann, A.; Brusca, S.; Patel, T.; 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, 128–139. [Google Scholar] [CrossRef] [PubMed]
  61. Codoñer, F.M.; Ramírez-Bosca, A.; Climent, E.; Carrión-Gutierrez, M.; Guerrero, M.; Pérez-Orquín, J.M.; de la Parte, J.H.; Genovés, S.; Ramón, D.; Navarro-López, V.; et al. Gut microbial composition in patients with psoriasis. Sci. Rep. 2018, 8, 1–7. [Google Scholar] [CrossRef] [PubMed]
  62. Tan, L.; Zhao, S.; Zhu, W.; Wu, L.; Li, J.; Shen, M.; Lei, L.; Chen, X.; Peng, C. The Akkermansia muciniphila is a gut microbiota signature in psoriasis. Exp. Dermatol. 2017, 27, 144–149. [Google Scholar] [CrossRef] [PubMed]
  63. Eppinga, H.; Sperna Weiland, C.J.; Thio, H.B.; van der Woude, C.J.; Nijsten, T.E.; Peppelenbosch, M.P.; Konstantinov, S.R. Similar Depletion of Protective Faecalibacterium prausnitzii in Psoriasis and Inflammatory Bowel Disease, but not in Hidradenitis Suppurativa. J Crohns Colitis 2016, 10, 1067–75. [Google Scholar] [CrossRef]
  64. Fry, L.; Baker, B.S. Triggering psoriasis: the role of infections and medications. Clin. Dermatol. 2007, 25, 606–615. [Google Scholar] [CrossRef]
  65. Lin, S.; Wang, Z.; Lam, K.-L.; Zeng, S.; Tan, B.K.; Hu, J. Role of intestinal microecology in the regulation of energy metabolism by dietary polyphenols and their metabolites. Food Nutr. Res. 2019, 63. [Google Scholar] [CrossRef]
  66. Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef]
  67. Kosiewicz, M.M.; Dryden, G.W.; Chhabra, A.; Alard, P. Relationship between gut microbiota and development of T cell associated disease. FEBS Lett. 2014, 588, 4195–4206. [Google Scholar] [CrossRef] [PubMed]
  68. Vallianou, N.; Stratigou, T.; Christodoulatos, G.S.; Dalamaga, M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Curr. Obes. Rep. 2019, 8, 317–332. [Google Scholar] [CrossRef] [PubMed]
  69. Scher, J.U.; Littman, D.R.; Abramson, S.B. Review: Microbiome in Inflammatory Arthritis and Human Rheumatic Diseases. Arthritis Rheumatol. 2015, 68, 35–45. [Google Scholar] [CrossRef] [PubMed]
  70. Stevens, B.R.; Roesch, L.; Thiago, P.; Russell, J.T.; Pepine, C.J.; Holbert, R.C.; Raizada, M.K.; Triplett, E.W. Depression phenotype identified by using single nucleotide exact amplicon sequence variants of the human gut microbiome. Mol. Psychiatry 2020, 26, 4277–4287. [Google Scholar] [CrossRef] [PubMed]
  71. Zakostelska, Z.; Málková, J.; Klimešová, K.; Rossmann, P.; Hornová, M.; Novosádová, I.; Stehlíková, Z.; Kostovcikova, M.; Hudcovic, T.; Štepánková, R.; et al. Intestinal Microbiota Promotes Psoriasis-Like Skin Inflammation by Enhancing Th17 Response. PLoS ONE 2016, 11, e0159539. [Google Scholar] [CrossRef] [PubMed]
  72. Stehlikova, Z.; Kostovcikova, K.; Kverka, M.; Rossmann, P.; Dvorak, J.; Novosadova, I.; Kostovcik, M.; Coufal, S.; Srutkova, D.; Prochazkova, P.; et al. Crucial Role of Microbiota in Experimental Psoriasis Revealed by a Gnotobiotic Mouse Model. Front. Microbiol. 2019, 10, 236. [Google Scholar] [CrossRef] [PubMed]
  73. Forbes, J.D.; Van Domselaar, G.; Bernstein, C.N. The Gut Microbiota in Immune-Mediated Inflammatory Diseases. Front. Microbiol. 2016, 7, 1081. [Google Scholar] [CrossRef]
  74. Kim, Y.-G.; Udayanga, K.G.S.; Totsuka, N.; Weinberg, J.B.; Núñez, G.; Shibuya, A. Gut Dysbiosis Promotes M2 Macrophage Polarization and Allergic Airway Inflammation via Fungi-Induced PGE2. Cell Host Microbe 2014, 15, 95–102. [Google Scholar] [CrossRef]
  75. Shen, T.D. Diet and Gut Microbiota in Health and Disease. Nestle Nutr Inst Workshop Ser. 2017, 88, 117–126. [Google Scholar] [PubMed]
  76. Plöger, S.; Stumpff, F.; Penner, G.B.; Schulzke, J.; Gäbel, G.; Martens, H.; Shen, Z.; Günzel, D.; Aschenbach, J.R. Microbial butyrate and its role for barrier function in the gastrointestinal tract. Ann. New York Acad. Sci. 2012, 1258, 52–59. [Google Scholar] [CrossRef] [PubMed]
  77. Schwarz, A.; Philippsen, R.; Schwarz, T. Induction of Regulatory T Cells and Correction of Cytokine Disbalance by Short-Chain Fatty Acids: Implications for Psoriasis Therapy. J. Investig. Dermatol. 2020, 141, 95–104. [Google Scholar] [CrossRef] [PubMed]
  78. Krejner, A.; Bruhs, A.; Mrowietz, U.; Wehkamp, U.; Schwarz, T.; Schwarz, A. Decreased expression of G-protein-coupled receptors GPR43 and GPR109a in psoriatic skin can be restored by topical application of sodium butyrate. Arch. Dermatol. Res. 2018, 310, 751–758. [Google Scholar] [CrossRef] [PubMed]
  79. Mrowietz, U.; Morrison, P.J.; Suhrkamp, I.; Kumanova, M.; Clement, B. The Pharmacokinetics of Fumaric Acid Esters Reveal Their In Vivo Effects. Trends Pharmacol. Sci. 2017, 39, 1–12. [Google Scholar] [CrossRef] [PubMed]
  80. Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef] [PubMed]
  81. Kim, C.H. Control of lymphocyte functions by gut microbiota-derived short-chain fatty acids. Cell. Mol. Immunol. 2021, 18, 1161–1171. [Google Scholar] [CrossRef] [PubMed]
  82. Zhao, T.; Wei, Y.; Zhu, Y.; Xie, Z.; Hai, Q.; Li, Z.; Qin, D. Gut microbiota and rheumatoid arthritis: From pathogenesis to novel therapeutic opportunities. Front. Immunol. 2022, 13, 1007165. [Google Scholar] [CrossRef] [PubMed]
  83. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The Gut Microbiota in Inflammatory Bowel Disease. Front. Cell. Infect. Microbiol. 2022, 12, 733992. [Google Scholar] [CrossRef]
  84. Zhang, L.; Qing, P.; Yang, H.; Wu, Y.; Liu, Y.; Luo, Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front. Immunol. 2021, 12, 686501. [Google Scholar] [CrossRef]
  85. Altieri, C.; Speranza, B.; Corbo, M.R.; Sinigaglia, M.; Bevilacqua, A. Gut-Microbiota, and Multiple Sclerosis: Background, Evidence, and Perspectives. Nutrients 2023, 15, 942. [Google Scholar] [CrossRef]
  86. Mazur, M.; Tomczak, H.; Łodyga, M.; Plagens-Rotman, K.; Merks, P.; Czarnecka-Operacz, M. The Intestinal and Skin Microbiome in Patients with Atopic Dermatitis and Their Influence on the Course of the Disease: A Literature Review. Healthcare 2023, 11, 766. [Google Scholar] [CrossRef]
  87. Bzioueche, H.; Sjödin, K.S.; West, C.E.; Khemis, A.; Rocchi, S.; Passeron, T.; Tulic, M.K. Analysis of Matched Skin and Gut Microbiome of Patients with Vitiligo Reveals Deep Skin Dysbiosis: Link with Mitochondrial and Immune Changes. J. Investig. Dermatol. 2021, 141, 2280–2290. [Google Scholar] [CrossRef] [PubMed]
  88. Kamata, M.; Tada, Y. Crosstalk: keratinocytes and immune cells in psoriasis. Front. Immunol. 2023, 14, 1286344. [Google Scholar] [CrossRef] [PubMed]
  89. Hollox, E.J.; Huffmeier, U.; Zeeuwen, P.L.; Palla, R.; Lascorz, J.; Rodijk-Olthuis, D.; van de Kerkhof, P.C.; Traupe, H.; de Jongh, G.; den Heijer, M.; Reis, A.; Armour, J.A.; Schalkwijk, J. Psoriasis is associated with increased beta-defensin genomic copy number. Nat Genet. 2008, 40, 23–5. [Google Scholar] [CrossRef] [PubMed]
  90. Niyonsaba, F. ; Ogawa. ; H.; Nagaoka, I. Human beta-defensin-2 functions as a chemotactic agent for tumour necrosis factor-alpha-treated human neutrophils. Immunology 2004, 111, 273–81. [Google Scholar]
  91. Niyonsaba, F.; Iwabuchi, K.; Matsuda, H.; Ogawa, H.; Nagaoka, I. Epithelial cell-derived human beta-defensin-2 acts as a chemotaxin for mast cells through a pertussis toxin-sensitive and phospholipase C-dependent pathway. Int Immunol. 2002, 14, 421–6. [Google Scholar] [CrossRef]
  92. Chiricozzi, A.; Guttman-Yassky, E.; Suárez-Fariñas, M.; Nograles, K.E.; Tian, S.; Cardinale, I.; Chimenti, S.; Krueger, J.G. Integrative Responses to IL-17 and TNF-α in Human Keratinocytes Account for Key Inflammatory Pathogenic Circuits in Psoriasis. J. Investig. Dermatol. 2011, 131, 677–687. [Google Scholar] [CrossRef]
  93. Wang, C.Q.; Akalu, Y.T.; Suarez-Farinas, M.; Gonzalez, J.; Mitsui, H.; Lowes, M.A.; Orlow, S.J.; Manga, P.; Krueger, J.G. IL-17 and TNF Synergistically Modulate Cytokine Expression while Suppressing Melanogenesis: Potential Relevance to Psoriasis. J. Investig. Dermatol. 2013, 133, 2741–2752. [Google Scholar] [CrossRef]
  94. Lande, R.; Botti, E.; Jandus, C.; Dojcinovic, D.; Fanelli, G.; Conrad, C.; Chamilos, G.; Feldmeyer, L.; Marinari, B.; Chon, S.; et al. The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat. Commun. 2014, 5, 5621. [Google Scholar] [CrossRef]
  95. Cai, Y.; Xue, F.; Quan, C.; Qu, M.; Liu, N.; Zhang, Y.; Fleming, C.; Hu, X.; Zhang, H.-G.; Weichselbaum, R.; et al. A Critical Role of the IL-1β–IL-1R Signaling Pathway in Skin Inflammation and Psoriasis Pathogenesis. J. Investig. Dermatol. 2018, 139, 146–156. [Google Scholar] [CrossRef] [PubMed]
  96. Kanda, N.; Shimizu, T.; Tada, Y.; Watanabe, S. IL-18 enhances IFN-gamma-induced production of CXCL9, CXCL10, and CXCL11 in human keratinocytes. Eur J Immunol. 2007, 37, 338–50. [Google Scholar] [CrossRef] [PubMed]
  97. Harper, E.G.; Guo, C.; Rizzo, H.; Lillis, J.V.; Kurtz, S.E.; Skorcheva, I.; Purdy, D.; Fitch, E.; Iordanov, M.; Blauvelt, A. Th17 Cytokines Stimulate CCL20 Expression in Keratinocytes In Vitro and In Vivo: Implications for Psoriasis Pathogenesis. J. Investig. Dermatol. 2009, 129, 2175–2183. [Google Scholar] [CrossRef] [PubMed]
  98. Deng, Y.; Chang, C.; Lu, Q. The Inflammatory Response in Psoriasis: a Comprehensive Review. Clin. Rev. Allergy Immunol. 2016, 50, 377–389. [Google Scholar] [CrossRef] [PubMed]
  99. Kagami, S.; Rizzo, H.L.; Lee, J.J.; Koguchi, Y.; Blauvelt, A. Circulating Th17, Th22, and Th1 Cells Are Increased in Psoriasis. J. Investig. Dermatol. 2010, 130, 1373–1383. [Google Scholar] [CrossRef] [PubMed]
  100. Nakajima, K. Critical role of the interleukin-23/T-helper 17 cell axis in the pathogenesis of psoriasis. J Dermatol. 2012, 39, 219–24. [Google Scholar] [CrossRef] [PubMed]
  101. Wolk, K.; Witte, E.; Wallace, E.; Döcke, W.D.; Kunz, S.; Asadullah, K.; Volk, H.D.; Sterry, W.; Sabat, R. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol. 2006, 36, 1309–23. [Google Scholar] [CrossRef] [PubMed]
  102. Singh, T.P.; Schön, M.P.; Wallbrecht, K.; Gruber-Wackernagel, A.; Wang, X.-J.; Wolf, P. Involvement of IL-9 in Th17-Associated Inflammation and Angiogenesis of Psoriasis. PLOS ONE 2013, 8, e51752. [Google Scholar] [CrossRef]
  103. Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T Cells and Immune Tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef]
  104. Zhang, K.; Li, X.; Yin, G.; Liu, Y.; Niu, X.; Hou, R. Functional characterization of CD4+CD25+ regulatory T cells differentiated in vitro from bone marrow-derived haematopoietic cells of psoriasis patients with a family history of the disorder. Br. J. Dermatol. 2007, 158, 298–305. [Google Scholar] [CrossRef]
  105. Grän, F.; Kerstan, A.; Serfling, E.; Goebeler, M.; Muhammad, K. Current Developments in the Immunology of Psoriasis. Yale J. Biol. Med. 2020, 93, 97–110. [Google Scholar] [PubMed]
  106. Lu, J.; Ding, Y.; Yi, X.; Zheng, J. CD19+ B cell subsets in the peripheral blood and skin lesions of psoriasis patients and their correlations with disease severity. Braz. J. Med Biol. Res. 2016, 49. [Google Scholar] [CrossRef] [PubMed]
  107. Mizoguchi, A.; Mizoguchi, E.; Takedatsu, H.; Blumberg, R.S.; Bhan, A.K. Chronic Intestinal Inflammatory Condition Generates IL-10-Producing Regulatory B Cell Subset Characterized by CD1d Upregulation. Immunity 2002, 16, 219–230. [Google Scholar] [CrossRef] [PubMed]
  108. Fillatreau, S.; Sweenie, C.H.; McGeachy, M.J.; Gray, D.; Anderton, S.M. B cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 2002, 3, 944–950. [Google Scholar] [CrossRef] [PubMed]
  109. Shortman, K.; Naik, S.H. Steady-state and inflammatory dendritic-cell development. Nat. Rev. Immunol. 2006, 7, 19–30. [Google Scholar] [CrossRef]
  110. Wollenberg, A.; Günther, S.; Moderer, M.; Wetzel, S.; Wagner, M.; Towarowski, A.; Tuma, E.; Rothenfusser, S.; Endres, S.; Hartmann, G. Plasmacytoid Dendritic Cells: A New Cutaneous Dendritic Cell Subset with Distinct Role in Inflammatory Skin Diseases. J. Investig. Dermatol. 2002, 119, 1096–1102. [Google Scholar] [CrossRef]
  111. Greb, J.E.; Goldminz, A.M.; Elder, J.T.; Lebwohl, M.G.; Gladman, D.D.; Wu, J.J.; Mehta, N.N.; Finlay, A.Y.; Gottlieb, A.B. Psoriasis. Nat Rev Dis Primers. 2016, 2, 16082. [Google Scholar] [CrossRef]
  112. Liang, H.; Yan, Y.; Wu, J.; Ge, X.; Wei, L.; Liu, L.; Chen, Y. Topical nanoparticles interfering with the DNA-LL37 complex to alleviate psoriatic inflammation in mice and monkeys. Sci. Adv. 2020, 6, eabb5274. [Google Scholar] [CrossRef]
  113. Wilsmann-Theis, D.; Koch, S.; Mindnich, C.; Bonness, S.; Schnautz, S.; von Bubnoff, D.; Bieber, T. Generation and functional analysis of human TNF-α/iNOS-producing dendritic cells (Tip-DC). Allergy 2013, 68, 890–8. [Google Scholar] [CrossRef]
  114. Serbina, N.V.; Salazar-Mather, T.P.; Biron, C.A.; Kuziel, W.A.; Pamer, E.G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003, 19, 59–70. [Google Scholar] [CrossRef]
  115. Aldridge, J.R. Jr.; Moseley, C.E.; Boltz, D.A.; Negovetich, N.J.; Reynolds, C.; Franks, J.; Brown, S.A.; Doherty, P.C.; Webster, R.G.; Thomas, P.G. TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection. Proc Natl Acad Sci U S A. 2009, 106, 5306–11. [Google Scholar] [CrossRef]
  116. Brand, A.; Diener, N.; Zahner, S.P.; Tripp, C.; Backer, R.A.; Karram, K.; Jiang, A.; Mellman, I.; Stoitzner, P.; Clausen, B.E. E-Cadherin is Dispensable to Maintain Langerhans Cells in the Epidermis. J. Investig. Dermatol. 2019, 140, 132–142. [Google Scholar] [CrossRef]
  117. Yan, B.; Liu, N.; Li, J.; Li, J.; Zhu, W.; Kuang, Y.; Chen, X.; Peng, C. The role of Langerhans cells in epidermal homeostasis and pathogenesis of psoriasis. J. Cell. Mol. Med. 2020, 24, 11646–11655. [Google Scholar] [CrossRef]
  118. Glitzner, E.; Korosec, A.; Brunner, P.M.; Drobits, B.; Amberg, N.; Schonthaler, H.B.; Kopp, T.; Wagner, E.F.; Stingl, G.; Holcmann, M.; et al. Specific roles for dendritic cell subsets during initiation and progression of psoriasis. EMBO Mol. Med. 2014, 6, 1312–1327. [Google Scholar] [CrossRef] [PubMed]
  119. Nakajima, K.; Kataoka, S.; Sato, K.; Takaishi, M.; Yamamoto, M.; Nakajima, H.; Sano, S. Stat3 activation in epidermal keratinocytes induces Langerhans cell activation to form an essential circuit for psoriasis via IL-23 production. J. Dermatol. Sci. 2019, 93, 82–91. [Google Scholar] [CrossRef] [PubMed]
  120. Yoshiki, R.; Kabashima, K.; Honda, T.; Nakamizo, S.; Sawada, Y.; Sugita, K.; Yoshioka, H.; Ohmori, S.; Malissen, B.; Tokura, Y.; et al. IL-23 from Langerhans Cells Is Required for the Development of Imiquimod-Induced Psoriasis-Like Dermatitis by Induction of IL-17A-Producing γδ T Cells. J. Investig. Dermatol. 2014, 134, 1912–1921. [Google Scholar] [CrossRef]
  121. Mrowietz, U. Neutrophils' sexiness is independent of trendy fashion. Exp. Dermatol. 2016, 26, 312–313. [Google Scholar] [CrossRef]
  122. Kaneko, F.; Itoh, N.; Yoshida, H.; Suzuki, M.; Ono, I. The cell-components and cytokines in the subcorneal microabscess of psoriasis. . 1991, 37, 103–12. [Google Scholar] [PubMed]
  123. Guérard, S.; Allaeys, I.; Martin, G.; Pouliot, R.; Poubelle, P.E. Psoriatic keratinocytes prime neutrophils for an overproduction of superoxide anions. Arch. Dermatol. Res. 2013, 305, 879–889. [Google Scholar] [CrossRef]
  124. Glennon-Alty, L.; Hackett, A.P.; Chapman, E.A.; Wright, H.L. Neutrophils and redox stress in the pathogenesis of autoimmune disease. Free Radic. Biol. Med. 2018, 125, 25–35. [Google Scholar] [CrossRef]
  125. Hoffmann, M.H.; Griffiths, H.R. The dual role of Reactive Oxygen Species in autoimmune and inflammatory diseases: evidence from preclinical models. Free. Radic. Biol. Med. 2018, 125, 62–71. [Google Scholar] [CrossRef] [PubMed]
  126. Chiang, C.-C.; Cheng, W.-J.; Korinek, M.; Lin, C.-Y.; Hwang, T.-L. Neutrophils in Psoriasis. Front. Immunol. 2019, 10, 2376. [Google Scholar] [CrossRef] [PubMed]
  127. Herster, F.; Bittner, Z.; Archer, N.K.; Dickhöfer, S.; Eisel, D.; Eigenbrod, T.; Knorpp, T.; Schneiderhan-Marra, N.; Löffler, M.W.; Kalbacher, H.; et al. Neutrophil extracellular trap-associated RNA and LL37 enable self-amplifying inflammation in psoriasis. Nat. Commun. 2020, 11, 1–13. [Google Scholar] [CrossRef] [PubMed]
  128. Pinegin, B.; Vorobjeva, N.; Pinegin, V. Neutrophil extracellular traps and their role in the development of chronic inflammation and autoimmunity. Autoimmun. Rev. 2015, 14, 633–640. [Google Scholar] [CrossRef] [PubMed]
  129. Keijsers, R.R.; Joosten, I.; van Erp, P.E.; Koenen, H.J.; van de Kerkhof, P.C. Cellular sources of IL-17 in psoriasis: a paradigm shift? Exp Dermatol. 2014, 23, 799–803. [Google Scholar] [CrossRef] [PubMed]
  130. 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] [PubMed]
  131. Stratis, A.; Pasparakis, M.; Rupec, R.A.; Markur, D.; Hartmann, K.; Scharffetter-Kochanek, K.; Peters, T.; van Rooijen, N.; Krieg, T.; Haase, I. Pathogenic role for skin macrophages in a mouse model of keratinocyte-induced psoriasis-like skin inflammation. J. Clin. Investig. 2006, 116, 2094–2104. [Google Scholar] [CrossRef] [PubMed]
  132. Wang, H.; Peters, T.; Kess, D.; Sindrilaru, A.; Oreshkova, T.; Van Rooijen, N.; Stratis, A.; Renkl, A.C.; Sunderkötter, C.; Wlaschek, M.; et al. Activated macrophages are essential in a murine model for T cell-mediated chronic psoriasiform skin inflammation. J. Clin. Investig. 2006, 116, 2105–2114. [Google Scholar] [CrossRef]
  133. Schlaak, J.F.; Buslau, M.; Jochum, W.; Hermann, E.; Girndt, M.; Gallati, H.; Büschenfelde, K.-H.M.Z.; Fleischer, B. T Cells Involved in Psoriasis Vulgaris Belong to the Th1 Subset. J. Investig. Dermatol. 1994, 102, 145–149. [Google Scholar] [CrossRef]
  134. Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and Type 17 Helper T Cells. New Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef]
  135. Beringer, A.; Noack, M.; Miossec, P. IL-17 in Chronic Inflammation: From Discovery to Targeting. Trends Mol. Med. 2016, 22, 230–241. [Google Scholar] [CrossRef] [PubMed]
  136. Ariza, M.-E.; Williams, M.V.; Wong, H.K. Targeting IL-17 in psoriasis: From cutaneous immunobiology to clinical application. Clin. Immunol. 2012, 146, 131–139. [Google Scholar] [CrossRef]
  137. Benhadou, F.; Mintoff, D.; Del Marmol, V. Psoriasis: keratinocytes or immune cells - which is the trigger? Dermatology 2019, 235, 91–100. [Google Scholar] [CrossRef] [PubMed]
  138. Chiricozzi, A.; Nograles, K.E.; Johnson-Huang, L.M.; Fuentes-Duculan, J.; Cardinale, I.; Bonifacio, K.M.; Gulati, N.; Mitsui, H.; Guttman-Yassky, E.; Suárez-Fariñas, M.; et al. IL-17 Induces an Expanded Range of Downstream Genes in Reconstituted Human Epidermis Model. PLOS ONE 2014, 9, e90284. [Google Scholar] [CrossRef] [PubMed]
  139. Rizzo, H.L.; Kagami, S. ; Phillips, K,G. ; Kurtz, S.E.; Jacques, S.L.; Blauvelt, A. IL-23-mediated psoriasis-like epidermal hyperplasia is dependent on IL-17A. J Immunol. 2011, 186, 1495–1502. [Google Scholar] [PubMed]
  140. Puig, L. The role of IL 23 in the treatment of psoriasis. Expert Rev. Clin. Immunol. 2017, 13, 525–534. [Google Scholar] [CrossRef]
  141. Fitch, E.; Harper, E.; Skorcheva, I.; Kurtz, S.E.; Blauvelt, A. Pathophysiology of psoriasis: Recent advances on IL-23 and Th17 cytokines. Curr. Rheumatol. Rep. 2007, 9, 461–467. [Google Scholar] [CrossRef] [PubMed]
  142. Vignali, D.A.; Kuchroo, V.K. IL-12 family cytokines: immunological playmakers. Nat Immunol. 2012, 13, 722–8. [Google Scholar] [CrossRef] [PubMed]
  143. Kulig, P.; Musiol, S.; Freiberger, S.N.; Schreiner, B.; Gyülveszi, G.; Russo, G.; Pantelyushin, S.; Kishihara, K.; Alessandrini, F.; Kündig, T.; et al. IL-12 protects from psoriasiform skin inflammation. Nat. Commun. 2016, 7, 13466. [Google Scholar] [CrossRef]
  144. Riol-Blanco, L.; Ordovas-Montanes, J.; Perro, M.; Naval, E.; Thiriot, A.; Alvarez, D.; Paust, S.; Wood, J.N.; Von Andrian, U.H. Nociceptive sensory neurons drive interleukin-23-mediated psoriasiform skin inflammation. Nature 2014, 510, 157–161. [Google Scholar] [CrossRef]
  145. Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and Type 17 Helper T Cells. New Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef]
  146. Monin, L.; Gaffen, S.L. Interleukin 17 Family Cytokines: Signaling Mechanisms, Biological Activities, and Therapeutic Implications. Cold Spring Harb. Perspect. Biol. 2018, 10, a028522. [Google Scholar] [CrossRef]
  147. Aggarwal, S.; Ghilardi, N.; Xie, M.-H.; de Sauvage, F.J.; Gurney, A.L. Interleukin-23 Promotes a Distinct CD4 T Cell Activation State Characterized by the Production of Interleukin-17. J. Biol. Chem. 2003, 278, 1910–1914. [Google Scholar] [CrossRef]
  148. Speeckaert, R.; Lambert, J.; Grine, L.; Van Gele, M.; De Schepper, S.; van Geel, N. The many faces of interleukin-17 in inflammatory skin diseases. Br. J. Dermatol. 2016, 175, 892–901. [Google Scholar] [CrossRef]
  149. Veldhoen, M. Interleukin 17 is a chief orchestrator of immunity. Nat. Immunol. 2017, 18, 612–621. [Google Scholar] [CrossRef]
  150. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. Il-17 and th17 cells. Annu Rev Immunol. 2009, 27, 485–517. [Google Scholar] [CrossRef] [PubMed]
  151. Kolls, J.K.; McCray, P.B.; Chan, Y.R. Cytokine-mediated regulation of antimicrobial proteins. Nat. Rev. Immunol. 2008, 8, 829–835. [Google Scholar] [CrossRef] [PubMed]
  152. Fitch, E.; Harper, E.; Skorcheva, I.; Kurtz, S.E.; Blauvelt, A. Pathophysiology of psoriasis: Recent advances on IL-23 and Th17 cytokines. Curr. Rheumatol. Rep. 2007, 9, 461–467. [Google Scholar] [CrossRef] [PubMed]
  153. Long, D.; Chen, Y.; Wu, H.; Zhao, M.; Lu, Q. Clinical significance and immunobiology of IL-21 in autoimmunity. J. Autoimmun. 2019, 99, 1–14. [Google Scholar] [CrossRef] [PubMed]
  154. Sarra, M.; Caruso, R.; Cupi, M.L.; Monteleone, I.; Stolfi, C.; Campione, E.; Diluvio, L.; Mazzotta, A.; Botti, E.; Chimenti, S.; et al. IL-21 Promotes Skin Recruitment of CD4+ Cells and Drives IFN-γ–Dependent Epidermal Hyperplasia. J. Immunol. 2011, 186, 5435–5442. [Google Scholar] [CrossRef]
  155. Kobayashi, M.; Fitz, L.; Ryan, M.; Hewick, R.M.; Clark, S.C.; Chan, S.; Loudon, R.; Sherman, F.; Perussia, B.; Trinchieri, G. Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J. Exp. Med. 1989, 170, 827–845. [Google Scholar] [CrossRef] [PubMed]
  156. Cheng, J.; Tu, Y.; Li, J.; Huang, C.; Liu, Z.; Liu, D. A study on the expression of interleukin (IL)-10 and IL-12 P35, P40 mRNA in the psoriatic lesions. J Tongji Med Univ. 2001, 21, 86–8. [Google Scholar] [PubMed]
  157. Elkayam, O.; Yaron, I.; Shirazi, I.; Yaron, M.; Caspi, D. Serum levels of IL-10, IL-6, IL-1ra, and sIL-2R in patients with psoriatic arthritis. Rheumatol. Int. 2000, 19, 101–105. [Google Scholar] [CrossRef] [PubMed]
  158. Bosè, F.; Capsoni, F.; Molteni, S.; Raeli, L.; Diani, M.; Altomare, A.; Garavaglia, M.; Garutti, C.; Frigerio, E.; Banfi, G.; et al. Differential expression of interleukin-2 by anti-CD3-stimulated peripheral blood mononuclear cells in patients with psoriatic arthritis and patients with cutaneous psoriasis. Clin. Exp. Dermatol. 2013, 39, 385–390. [Google Scholar] [CrossRef]
  159. Baggiolini, M.; Clark-Lewis, I. Interleukin-8, a chemotactic and inflammatory cytokine. FEBS Lett. 1992, 307, 97–101. [Google Scholar] [CrossRef]
  160. Kanda, N.; Hau, C.S.; Tada, Y.; Tatsuta, A.; Sato, S.; Watanabe, S. Visfatin Enhances CXCL8, CXCL10, and CCL20 Production in Human Keratinocytes. Endocrinology 2011, 152, 3155–3164. [Google Scholar] [CrossRef] [PubMed]
  161. Dikbas, O.; Tosun, M.; Bes, C.; Tonuk, S.B.; Aksehirli, O.Y.; Soy, M. Serum levels of visfatin, resistin and adiponectin in patients with psoriatic arthritis and associations with disease severity. Int. J. Rheum. Dis. 2014, 19, 672–677. [Google Scholar] [CrossRef]
  162. Eyerich, S.; Eyerich, K.; Pennino, D.; Carbone, T.; Nasorri, F.; Pallotta, S.; Cianfarani, F.; Odorisio, T.; Traidl-Hoffmann, C.; Behrendt, H.; et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J. Clin. Investig. 2009, 119, 3573–3585. [Google Scholar] [CrossRef] [PubMed]
  163. Kagami, S. IL-23 and Th17 cells in infections and psoriasis. Jpn. J. Clin. Immunol. 2011, 34, 13–19. [Google Scholar] [CrossRef]
  164. Van Belle, A.B.; de Heusch, M.; Lemaire, M.M.; Hendrickx, E.; Warnier, G.; Dunussi-Joannopoulos, K.; Fouser, L.A.; Renauld, J.-C.; Dumoutier, L. IL-22 Is Required for Imiquimod-Induced Psoriasiform Skin Inflammation in Mice. J. Immunol. 2012, 188, 462–469. [Google Scholar] [CrossRef]
  165. Balato, A.; Schiattarella, M.; Lembo, S.; Mattii, M.; Prevete, N.; Balato, N.; Ayala, F. Interleukin-1 family members are enhanced in psoriasis and suppressed by vitamin D and retinoic acid. Arch. Dermatol. Res. 2013, 305, 255–262. [Google Scholar] [CrossRef]
  166. Grossman, R.M.; Krueger, J.; Yourish, D.; Granelli-Piperno, A.; Murphy, D.P.; May, L.T.; Kupper, T.S.; Sehgal, P.B.; Gottlieb, A.B. Interleukin 6 is expressed in high levels in psoriatic skin and stimulates proliferation of cultured human keratinocytes. Proc. Natl. Acad. Sci. 1989, 86, 6367–6371. [Google Scholar] [CrossRef] [PubMed]
  167. Fritz, Y.; Klenotic, P.A.; Swindell, W.R.; Yin, Z.Q.; Groft, S.G.; Zhang, L.; Baliwag, J.; Camhi, M.I.; Diaconu, D.; Young, A.B.; et al. Induction of Alternative Proinflammatory Cytokines Accounts for Sustained Psoriasiform Skin Inflammation in IL-17C+IL-6KO Mice. J. Investig. Dermatol. 2016, 137, 696–705. [Google Scholar] [CrossRef] [PubMed]
  168. Campanati, A.; Orciani, M.; Lazzarini, R.; Ganzetti, G.; Consales, V.; Sorgentoni, G.; Di Primio, R.; Offidani, A. TNF-α inhibitors reduce the pathological Th1 -Th17 /Th2 imbalance in cutaneous mesenchymal stem cells of psoriasis patients. Exp Dermatol. 2017, 26, 319–324. [Google Scholar] [CrossRef] [PubMed]
  169. Kristensen, M.; Chu, C.Q.; Eedy, D.J.; Feldmann, M.; Brennan, F.M.; Breathnach, S.M. Localization of tumour necrosis factor-alpha (TNF-α) and its receptors in normal and psoriatic skin: epidermal cells express the 55-kD but not the 75-kD TNF receptor. Clin. Exp. Immunol. 1993, 94, 354–362. [Google Scholar] [CrossRef]
  170. Dowlatshahi, E.; van der Voort, E.; Arends, L.; Nijsten, T. Markers of systemic inflammation in psoriasis: a systematic review and meta-analysis. Br. J. Dermatol. 2013, 169, 266–282. [Google Scholar] [CrossRef] [PubMed]
  171. Banno, T.; Gazel, A.; Blumenberg, M. Effects of tumor necrosis factor-alpha (TNF alpha) in epidermal keratinocytes revealed using global transcriptional profiling. J Biol Chem. 2004, 279, 32633–42. [Google Scholar] [CrossRef]
  172. Zhuang, L.; Ma, W.; Cai, D.; Zhong, H.; Sun, Q. Associations between Tumor Necrosis Factor-α Polymorphisms and Risk of Psoriasis: A Meta-Analysis. PLOS ONE 2013, 8, e68827. [Google Scholar] [CrossRef] [PubMed]
  173. Asadullah, K.; Sterry, W.; Stephanek, K.; Jasulaitis, D.; Leupold, M.; Audring, H.; Volk, H.D.; Döcke, W.D. IL-10 is a key cytokine in psoriasis. Proof of principle by IL-10 therapy: a new therapeutic approach. J. Clin. Investig. 1998, 101, 783–794. [Google Scholar] [CrossRef]
  174. Schwartz, D.M.; Kanno, Y.; Villarino, A.; Ward, M.; Gadina, M.; O'Shea, J.J. Erratum: JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat. Rev. Drug Discov. 2017, 17, 78–78. [Google Scholar] [CrossRef]
  175. Sano, S.; Chan, K.S.; Carbajal, S.; Clifford, J.; Peavey, M.; Kiguchi, K.; Itami, S.; Nickoloff, B.J.; DiGiovanni, J. Stat3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat. Med. 2004, 11, 43–49. [Google Scholar] [CrossRef] [PubMed]
  176. Ishizaki, M.; Muromoto, R.; Akimoto, T.; Sekine, Y.; Kon, S.; Diwan, M.; Maeda, H.; Togi, S.; Shimoda, K.; Oritani, K.; et al. Tyk2 is a therapeutic target for psoriasis-like skin inflammation. Int. Immunol. 2013, 26, 257–267. [Google Scholar] [CrossRef] [PubMed]
  177. Nada, H.R.; El Sharkawy, D.A.; Elmasry, M.F.; Rashed, L.A.; Mamdouh, S. Expression of Janus Kinase 1 in vitiligo & psoriasis before and after narrow band UVB: a case–control study. Arch. Dermatol. Res. 2017, 310, 39–46. [Google Scholar] [CrossRef] [PubMed]
  178. Zheng, Y.; Danilenko, D.M.; Valdez, P.; Kasman, I.; Eastham-Anderson, J.; Wu, J.; Ouyang, W. Interleukin-22, a TH17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 2006, 445, 648–651. [Google Scholar] [CrossRef] [PubMed]
  179. Szilveszter, K.P.; Németh, T.; Mócsai, A. Tyrosine Kinases in Autoimmune and Inflammatory Skin Diseases. Front. Immunol. 2019, 10, 1862. [Google Scholar] [CrossRef] [PubMed]
  180. Antonioli, L.; Csóka, B.; Fornai, M.; Colucci, R.; Kókai, E.; Blandizzi, C.; Haskó, G. Adenosine and inflammation: what's new on the horizon? Drug Discov Today 2014, 19, 1051–68. [Google Scholar] [CrossRef] [PubMed]
  181. Koscsó, B.; Csóka, B. ; Pacher. P.; Haskó, G. Investigational A₃ adenosine receptor targeting agents. Expert Opin Investig Drugs 2011, 20, 757–68. [Google Scholar]
  182. Jacobson, K.A.; Merighi, S.; Varani, K.; Borea, P.A.; Baraldi, S.; Tabrizi, M.A.; Romagnoli, R.; Baraldi, P.G.; Ciancetta, A.; Tosh, D.K.; et al. A3 Adenosine Receptors as Modulators of Inflammation: From Medicinal Chemistry to Therapy. Med. Res. Rev. 2017, 38, 1031–1072. [Google Scholar] [CrossRef] [PubMed]
  183. Gudjonsson, J.E.; Johnston, A.; Stoll, S.W.; Riblett, M.B.; Xing, X.; Kochkodan, J.J.; Ding, J.; Nair, R.P.; Aphale, A.; Voorhees, J.J.; et al. Evidence for Altered Wnt Signaling in Psoriatic Skin. J. Investig. Dermatol. 2010, 130, 1849–1859. [Google Scholar] [CrossRef]
  184. Ghosh, M.C.; Collins, G.D.; Vandanmagsar, B.; Patel, K.; Brill, M.; Carter, A.; Lustig, A.; Becker, K.G.; Wood, W.W. 3rd. ; Emeche, C.D.; French, A.D.; O'Connell, M.P.; Xu, M.; Weeraratna, A.T.; Taub, D.D. Activation of Wnt5A signaling is required for CXC chemokine ligand 12-mediated T-cell migration. Blood 2009, 114, 1366–73. [Google Scholar]
  185. Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–62. [Google Scholar] [CrossRef] [PubMed]
  186. Lizzul, P.F.; Aphale, A.; Malaviya, R.; Sun, Y.; Masud, S.; Dombrovskiy, V.; Gottlieb, A.B. Differential expression of phosphorylated NF-kappaB/RelA in normal and psoriatic epidermis and downregulation of NF-kappaB in response to treatment with etanercept. J Invest Dermatol. 2005, 124, 1275–83. [Google Scholar] [CrossRef] [PubMed]
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated