Skip to main content
F1000Research logoLink to F1000Research
. 2016 Apr 28;5:F1000 Faculty Rev-770. [Version 1] doi: 10.12688/f1000research.7927.1

Recent advances in understanding psoriasis

Franziska C Eberle 1, Jürgen Brück 1, Julia Holstein 1, Kiyoshi Hirahara 2,a, Kamran Ghoreschi 1,b
PMCID: PMC4850872  PMID: 27158469

Abstract

T helper (Th) cells producing interleukin (IL)-17, IL-22, and tumor necrosis factor (TNF) form the key T cell population driving psoriasis pathogenesis. They orchestrate the inflammation in the skin that results in the proliferation of keratinocytes and endothelial cells. Besides Th17 cells, other immune cells that are capable of producing IL-17-associated cytokines participate in psoriatic inflammation. Recent advances in psoriasis research improved our understanding of the cellular and molecular players that are involved in Th17 pathology and inflammatory pathways in the skin. The inflammation-driving actions of TNF in psoriasis are already well known and antibodies against TNF are successful in the treatment of Th17-mediated psoriatic skin inflammation. A further key cytokine with potent IL-17-/IL-22-promoting properties is IL-23. Therapeutics directly neutralizing IL-23 or IL-17 itself are now extending the therapeutic spectrum of antipsoriatic agents and further developments are on the way. The enormous progress in psoriasis research allows us to control this Th17-mediated inflammatory skin disease in many patients.

Keywords: psoriasis, T helper cells, skin inflammation, keratinocyte

Introduction

Psoriasis is one of the most common chronic diseases, affecting 2–3% of the adult population and 0.5–1% of children. Due to the frequency of the disease worldwide and its clinical characteristics, psoriasis has gained the interest of many scientists in academia as well as industrial research. The easy accessibility of the skin allows scientists to study cells and mediators in inflamed skin and their relevance in disease pathogenesis in detail. Recent advances in psoriasis pathogenesis improved our understanding of disease mechanisms and resulted in the development of new immunobiologics and small molecules that help to control the chronic inflammation 1. Here we summarize the recent findings on the cellular and molecular players that presumably contribute to psoriasis development. In general, psoriasis is considered to be an autoimmune disease and most scientists agree on the central importance of T cells in disease pathogenesis 24, yet the psoriatic inflammation may originate from epidermal epithelial cells and innate immune cells. Clearly, a close interaction between mediators and cells of the innate and adaptive immune systems and keratinocytes and endothelial cells is present in psoriasis 5, 6.

Aberrant keratinocyte biology as a pathogenic driver in psoriasis

Decades ago, psoriasis was primarily thought to be caused by aberrant keratinocytes resulting in uncontrolled proliferation of the epidermal cell layers. Early studies on the cellular ‘turnover’ of epidermal cells supported this hypothesis 7. The keratinocytes in psoriasis are characterized not only by strong proliferation but also by an altered expression of certain keratins like keratin 16. The concept of altered keratinocytes as pathogenic cells causing psoriasis has gained new attention since different reports published in the beginning of this millennium showed that genetic alterations in epidermal transcription factors can cause skin disorders that resemble human psoriasis clinically and histologically. Mice with altered expression of JunB/c-Jun or phosphorylation of STAT3 in keratinocytes develop skin inflammation with histological and molecular characteristics of psoriasis 8, 9. Interestingly, in both models, the psoriatic skin inflammation seems to depend on the presence of immune cells including T cells and their cytokines. In fact, there is a close interaction between cytokines and keratinocytes. A number of cytokines present in psoriatic inflammation promotes keratinocyte proliferation. Intradermal injections of T helper 17 (Th17)-associated cytokines like interleukin (IL)-23 or IL-21 into mouse skin induce epidermal hyperplasia with morphological characteristics of human psoriasis associated with the infiltration of inflammatory T cells. On the other hand, keratinocytes themselves are a cellular source of cytokines. The most famous member is IL-8, a cytokine originally discovered in psoriatic scales 10. Another example is the cytokine IL-15, which is expressed in psoriatic epidermis and protects keratinocytes from apoptosis. Interestingly, soluble IL-15Rα has been shown to dampen the psoriatic inflammation by suppressing cytokine secretion from keratinocytes and the expansion of IL-17-producing T cells 11. Moreover, keratinocytes are a major source of IL-1 production 12. Factors such as cytosolic DNA can trigger inflammasome activation and IL-1 secretion by keratinocytes, which contribute to the psoriatic inflammation 13. Other mediators that are linked to psoriasis pathogenesis and that are produced by keratinocytes include antimicrobial peptides like S100A8/9, β-defensins, and cathelicidin (LL-37) 3. Taken together, genetic alterations in transcription factors and environmental triggering factors affecting keratinocytes presumably facilitate the manifestation of psoriasis, yet psoriasis pathogenesis seems to be dominated by the activation of immune cells rather than alterations in keratinocytes.

Central role of immune cells

Several observations support the importance of immune cells in the pathogenesis of psoriasis. One is the transfer of the disease by bone marrow cells. Case reports from individuals undergoing bone marrow transplantation for hematological disorders have linked the disappearance of psoriasis as well as the development of psoriasis in the recipient to the skin status of the donor 14, 15. The other observation is that immunosuppressive agents originally introduced for the prevention of organ transplant rejection showed unexpected benefits on the clinical course of psoriasis 16, 17. Subsequently, immunosuppressive agents like cyclosporine or methotrexate have been established in the treatment of psoriasis. Genetic data on human leukocyte antigen (HLA) associations as well as data on the presence of oligoclonal T cells in lesional skin and their reactivity towards cutaneous antigens further underline the importance of immune cells in psoriasis pathogenesis. Putative autoantigens in psoriasis include keratins, heat shock proteins, the antimicrobial peptide LL37, and the melanocytic antigen ADAMTS-like protein 5 (ADAMTSL5) 1820. The recent discovery of ADAMTSL5 as a potential autoantigen in psoriasis is a key finding. The recognition of this protein is restricted to epidermal CD8 + T cells of patients with psoriasis and a HLA-C*06:02 genotype. Stimulation of ADAMTSL5-specific CD8 + T cells results in IL-17A production 20. Of note, HLA-C*06:02 is known as the HLA locus with the strongest genetic association with psoriasis. In addition to the linkage to certain HLA genotypes, recent investigations revealed that psoriasis is also linked to polymorphisms in genes encoding certain cytokines, cytokine receptors, and transcription factors. Today, there is a widely accepted consensus that psoriasis is an immune cell-mediated disease.

A prototypic Th17 disease

Among the gene polymorphisms that have been linked to psoriasis are genes encoding IL23A, IL23R, STAT3, RUNX3, and TYK2. All of these genes are associated with the Th17 immune response 1. Th17 cells are characterized by the expression of their lineage-defining cytokine IL-17A. In addition, Th17 cells can produce other cytokines like IL-17F, IL-21, IL-22, tumor necrosis factor (TNF), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Some Th17 populations also secrete IL-9 or IL-10, depending on the signals they receive during initial activation. The differentiation and activation of the Th17 population from naïve T cells depend on cytokines like IL-6, IL-21, IL-1, TGF-β, and IL-23 21. Strikingly, IL-23, its receptor, and its downstream signaling molecule STAT3 are all linked to the genetic susceptibility for developing psoriasis. Of note, the transcription factor STAT3 is also activated by IL-6 and IL-21 and, together with the other Th17-characterizing transcription factor RORγ, STAT3 is responsible for IL-17A and IL-17F expression 22. Skin-infiltrating Th17 cells seem to be the central players orchestrating psoriasis pathogenesis ( Figure 1). They interact with tissue cells like keratinocytes and endothelial cells and with various immune cells including dendritic cells (DCs) and neutrophilic granulocytes. The reactivation of memory Th17 cells is presumably responsible for the chronic course of the disease.

Figure 1. Immune cells and T helper 17 (Th17)-associated cytokines implicated in psoriasis pathogenesis.

Figure 1.

Characteristic markers and cytokines related to the interleukin (IL)-17/IL-23 immune signature of T cells, dendritic cells (DCs), and associated immune cells in psoriatic skin inflammation.

Contribution of skin-resident immune cells

It has become obvious that there is a critical population of memory T cells that resides in the tissue and is involved in the local immune response 23. Those specific memory T cells were named “resident-memory T” (T RM) cells. T RM cells preferentially reside in epithelial barrier tissues such as the respiratory tract, reproductive tract, and skin 2426. T RM cells can respond rapidly to pathogenic invaders in the epithelial barrier site, so T RM cells are crucial for the protection of the host from harmful microorganisms. The pathogenic role of T RM cells in immune-mediated diseases including skin diseases like psoriasis is gaining more evidence. A recent study revealed the augmentation of T RM cells in the local inflamed skin of patients with psoriasis 27. Moreover, T RM cells in psoriatic skin express higher levels of both IL17A and IL22 compared to those in the skin of healthy individuals. The majority of T RM cells in the epidermis express CD103. T RM cells residing in the dermis show lower expression of this marker 27. IL-9-producing T RM cells have also been reported to be present in conditions of skin inflammation like in psoriasis 28. Besides T cells, DCs can reside in the skin. DCs are a key population of the immune system, bridging the breaks between innate and adaptive immunity. Among the heterogeneous DC population, CD1c -CD11c + DCs represent a population of inflammatory dermal DCs. Ultraviolet exposure reduces the number of inflammatory CD1c -CD11c + dermal DCs in patients with psoriasis 29, while the number of CD1c +CD11c + so-called resident DCs remains unaffected 30. A potent marker that allows the discrimination of inflammatory CD1c -CD11c + DCs from resident CD1c +CD11c + DCs in patients with psoriasis is TNF-related apoptosis-inducing ligand (TRAIL) 31. More intensive studies are needed to identify the environmental signals that induce specific features of T RM cells and resident DCs in the skin under steady state and inflammatory conditions.

Phenotype of dendritic cells in psoriasis

In general, DCs are a heterogeneous population. In the skin, different types of DCs have been described. The distinct populations are characterized by the expression of certain surface markers and mediators. In psoriasis, certain DC populations like plasmacytoid DCs (pDCs) and dermal myeloid DCs (mDCs) dominate the inflammatory skin, while the number of epidermal Langerhans cells seems to stay stable as compared to non-lesional skin. During initial inflammation, an increased number of pDCs is activated, which results in the release of type I interferon (IFN-α) 32. Interestingly, complexes formed by self-DNA or self-RNA and the antimicrobial peptide LL37 have been shown to activate pDCs through Toll-like receptor 9 (TLR9) or TLR7/8, respectively 33, 34. Recently, a novel mechanism of pDC activation has been described. As shown for antimicrobial peptides, the Th17-associated cytokine IL-26 can also form complexes with DNA from dying bacterial or host tissue cells and these complexes also promote IFN-α production by pDCs through TLR9 stimulation 35. These innate mechanisms seem to be relevant for pDC activation in psoriasis pathogenesis. The activation of pDCs is followed by an increase of CD11c + mDCs, which express TNF, inducible nitric oxide synthase (iNOS), and IL-23. As mentioned above, inflammatory CD11c + mDCs do not express CD1c in contrast to skin-resident CD1c + mDCs. Another DC population that is capable of producing IL-23 is the so-called 6-sulfo LacNAc-expressing population (slanDCs) 36, 37. Moreover, CD163 + macrophages can produce IL-23 ( Figure 1). Taken together, the major function of DCs and macrophages in psoriasis pathogenesis is to provide the signals that promote the Th17 inflammation.

Non-T cell sources of IL-17A and IL-22 in psoriasis

As we discussed before, the IL-23/IL-17A and IL-23/IL-22 axes play a pivotal role in the pathogenesis of psoriasis 38. Besides Th17 cells, IL-17A and/or IL-22 are produced by other types of immune cells including innate lymphoid cells (ILCs) 3, and gamma delta (γδ) T cells 3941. ILCs have recently been identified as a unique population of innate immune cells that lack antigen-specific receptors. They can be stimulated by cytokines and they produce a series of effector cytokines 40. ILCs are now recognized to be divided into three main groups based on the feature of producing lineage-defining cytokines and specific transcription factors 40, 42, 43. Among these groups of ILCs, ILC3 including lymphoid tissue inducer (LTi) cells are characterized by the production of IL-17A and/or IL-22 accompanied with high expression of Rorγt 40, 44, 45. In the case of humans, ILC3 can be distinguished into several subpopulations based on expression patterns of natural killer (NK) cell markers like NKp44 and NKp46 46. Among these subpopulations, NKp44 + ILC3 were reported to contribute to the pathogenesis of psoriasis, since IL-17A- and IL-22-producing NKp44 + ILC3 were increased in both the peripheral blood and the skin of patients with psoriasis 47. The crucial role of ILC3 subpopulations in psoriasis pathogenesis is supported by the finding that Rorγt +CD56 + ILC3, which are capable of producing IL-22, are highly accumulated in the skin of patients with psoriasis 48. Another cellular source of IL-17A in the skin is the γδ T cell population 49. The majority of dermal γδ T cells express a T cell receptor (TCR) containing Vγ4 together with the chemokine receptor CCR6 50. In an experimental model of psoriasis-like inflammation in mice using the TLR7-agonist imiquimod, dermal Vγ4 + γδ T cells persist in the skin and contribute to skin inflammation by producing IL-17A and IL-17F 51, 52. Consistent with these findings, the increased number of γδ T cells, which produce large amounts of IL-17A, was detected in the affected skin of patients with psoriasis 53. More recently, mast cells have also been reported as producers of IL-17A and IL-22 in psoriasis 54. Similarly, neutrophils have been suggested as a further cellular source of IL-17A and IL-22. Of note, all immune cells mentioned also produce TNF, a factor well established in psoriasis pathogenesis and treatment. Taken together, various types of immune cells produce the psoriasis-driving cytokines TNF, IL-17A, and IL-22 ( Figure 1).

Immunotherapies supporting the role of TNF and IL-17A in psoriasis

Based on the immunopathogenesis, antipsoriatic therapies target antigen-presenting cells (APCs), T cells, or their cytokines ( Table 1). Modern small molecules like dimethyl fumarate and the PDE4 inhibitor apremilast both primarily act on APCs. By interfering with intracellular signaling pathways like NRF2 or second messengers like cAMP, they impair the production of pro-inflammatory DC cytokines like IL-23 and in contrast induce the release of anti-inflammatory IL-10. Since they also inhibit IL-12 and TNF production by APCs, dimethyl fumarate and apremilast treatment both result in the suppression of Th17 and Th1 responses 55, 56. Thus, silencing IL-23 expression by DCs by small molecules or by RNA interference (RNAi) technology, as recently tested in preclinical settings of autoimmune disease, is an attractive approach 57, 58. A new class of modern immunosuppressants is the class of JAK inhibitors 59. These compounds interfere with the signaling pathways of numerous cytokines and hormones. Selective JAK inhibitors inhibit the activation and differentiation of multiple Th cell subsets, but they also inhibit the effects of cytokines on non-T cells and non-immune cells 60. Thus, the mode of action of all of these compounds is not restricted to APCs and T cells. Although they may also affect other immune cells and tissue cells, they underline the importance of cytokine signaling in psoriasis 61. To improve our understanding of psoriasis pathogenesis, it is more helpful to focus on therapeutics targeting single cytokines.

Table 1. Modern immunotherapies targeting interleukin (IL)-17/IL-23.

The table summarizes approved therapeutics and compounds that are in advanced stage development (according to www.clinicaltrials.gov) and some of their effects on the Th17 response. cAMP, cyclic adenosine monophosphate; DC, dendritic cell; HO-1, heme oxygenase-1; IL, interleukin; NRF2, nuclear factor (erythroid-derived 2)-like 2; PKC, protein kinase C; Th1, T helper type 1; Th2, T helper type 2; Th17, T helper type 17; TNF, tumor necrosis factor.

Immune-modifying
category
Immunotherapeutic Target(s) Effect on the Th17 response
Small molecules with
intracellular mode of
action
Apremilast cAMP/PKC signaling in DCs &
macrophages
Inhibits IL-23 and TNF production,
induces IL-10
Dimethyl fumarate NRF2/HO-1 signaling in DCs
& macrophages
Inhibits IL-23 production, induces
IL-10, promotes IL-4-producing
Th2 cells
Tofacitinib
Ruxolitinib
Baricitinib
JAK1/JAK3
JAK1/JAK2
JAK1/JAK2
Inhibit signaling by IL-22 and Th17-
promoting cytokines IL-6, IL-21,
and IL-23
Anti-cytokine antibodies
and fusion proteins
Etanercept
Adalimumab
Infliximab
Golimumab
Certolizumab pegol
TNF/lymphotoxin
TNF
TNF
TNF
TNF
Impair DC activation and IL-23
production
Ustekinumab IL-12/IL-23p40 Impairs Th17 and Th1 responses
Tildrakizumab
Guselkumab
BI 655066
IL-23p19
IL-23p19
IL-23p19
Impair Th17 responses
Secukinumab
Ixekizumab
IL-17A
IL-17A
Inhibit effects of IL-17A
Anti-receptor antibody Brodalumab IL-17RA Inhibits IL-17A and IL-17F signaling

The first generation of antipsoriatic biologics targeting cytokines focused on TNF. These immunotherapeutics are highly effective in the treatment of psoriasis of skin and joints since they neutralize the effects of TNF on multiple cell types. In psoriatic skin, where TNF is mainly produced by DCs and macrophages, the neutralization of this cytokine rapidly decreases the expression of the Th17-promoting IL-23p40 and some other mediators 62, 63. This initial action of TNF neutralization on IL-23 in the skin is followed by the reduction of IL-17A, IL-22, IFN-γ, and TNF. The second generation of anti-psoriatic biologics targeting cytokines focuses directly on the Th17 cytokines IL-23 and IL-17A. The neutralization of p40, a cytokine unit shared by IL-23 and IL-12, is also effective in the treatment of psoriasis and psoriatic arthritis and directly interferes with the activation of Th17 as well as Th1 cells. Importantly, selective inhibition of the IL-23 unit p19 also improves psoriasis. Currently, three antibodies targeting p19 are in phase 3 development for the treatment of psoriasis. Neutralization of IL-23 results in decreased numbers of skin-infiltrating T cells, mDCs, pDCs, and neutrophils, while epidermal Langerhans cells remain unaffected 64. Finally, IL-17A itself became a therapeutic target in psoriasis. The first monoclonal antibody directed against IL-17A is already approved for the treatment of psoriasis and psoriatic arthritis 65. A second anti-IL-17A antibody recently received a positive opinion by the European Medicines Agency 66. Systemic neutralization of IL-17A lowers the expression of IL-17A, IL-17F, IL-22, TNF, IL-6, IL-8, and p40 in the skin 65. Of note, an antibody blocking the IL-17 receptor A (IL-17RA) is also effective in psoriasis and is in phase 3 development. These new approaches emphasize the significance of the Th17 pathway in psoriasis.

Remaining questions

Recent findings have helped us to improve our understanding of psoriasis pathogenesis. We now allocate the mechanisms of previously established antipsoriatic treatments to their effects on the Th17 pathway. This is best illustrated for the use of recombinant IL-4 or the small molecule dimethyl fumarate, which both suppress Th17 cell development 55, 67, 68. The new generation of antipsoriatic biologics directly targeting IL-23 or IL-17A underlines the central role of these cytokines in psoriasis pathogenesis. Although we have a battery of systemic treatments for our patients, we do not know which patient will respond adequately to a certain drug. There is still a significant number of primary and secondary non-responders. The lack and the loss of response are mainly observed in patients treated with oral therapeutics and TNF antagonists. Possibly, this will be similar in patients receiving modern drugs directly interfering with the Th17 axis. One of the most important future developments is the establishment of testing methods to predict the clinical response to certain targeted therapies using immunobiologics or small molecules. Definition of useful biomarkers may help to identify responders in an early stage. Some markers like IL-8, IL-19, NOS2, or S100 proteins are typically expressed in psoriatic skin 10, 67, 6971, but ideal biomarkers are still not established. Furthermore, we have to understand why the same body sites can develop psoriatic plaques, even after long periods of remission. One study demonstrates that epidermal CD8 + T RM and also CD4 + T cells reside in the skin even after successful treatment and retain their capability of responding rapidly with the production of IL-17A and IL-22, respectively, upon ex vivo stimulation 27. Another issue to be studied is the exact mechanism that causes the development of paradoxical psoriasis in patients without prior history of psoriasis who receive TNF antagonists for the treatment of inflammatory colitis or rheumatoid arthritis. Unraveling these immunological mechanisms may also help us to understand the different phenotypes of psoriasis. One example is given by recent findings on pustular psoriasis. Genetic studies could link IL-36RN deficiency and CARD14 mutations to the susceptibility for generalized pustular psoriasis 72, 73. Thus, interfering with inflammasome activation or IL-1 family cytokines may be of benefit in such patients 74. Similarly, it is of interest to understand why certain environmental factors like infections and drugs but also acquired immunodeficiency result in treatment-resistant cases of psoriasis. To complete our understanding of this chronic inflammatory disease affecting a large proportion of our global population, further research in immunology, genetics, epigenetics, microbiology, and molecular biology is needed.

Abbreviations

DC, dendritic cells; HLA, human leukocyte antigen; IL, interleukin; ILC, innate lymphoid cell; mDC, myeloid dendritic cell; NK cell, natural killer cell; pDC, plasmacytoid dendritic cell; Th cell, T helper cell; TLR, Toll-like receptor; T RM cell, resident-memory T cell.

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Mario Fabri, Department of Dermatology and Venereology, University of Cologne, Cologne, Germany

  • Thomas Herzinger, Department of Dermatology and Allergy, Ludwig Maximilian University, Munich, Germany

Funding Statement

This work was supported by the Deutsche Forschungsgemeinschaft (DFG) Sonderforschungsbereich (SFB) 685 (to Kamran Ghoreschi) and SFB TR-156 (to Franziska C. Eberle and Kamran Ghoreschi).

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

[version 1; referees: 2 approved]

References

  • 1. Belge K, Brück J, Ghoreschi K: Advances in treating psoriasis. F1000Prime Rep. 2014;6:4. 10.12703/P6-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Nestle FO, Kaplan DH, Barker J: Psoriasis. N Engl J Med. 2009;361(5):496–509. 10.1056/NEJMra0804595 [DOI] [PubMed] [Google Scholar]
  • 3. Lowes MA, Suárez-Fariñas M, Krueger JG: Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–55. 10.1146/annurev-immunol-032713-120225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Boehncke WH, Schön MP: Psoriasis. Lancet. 2015;386(9997):983–94. 10.1016/S0140-6736(14)61909-7 [DOI] [PubMed] [Google Scholar]
  • 5. Heidenreich R, Röcken M, Ghoreschi K: Angiogenesis drives psoriasis pathogenesis. Int J Exp Pathol. 2009;90(3):232–48. 10.1111/j.1365-2613.2009.00669.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Yazdi AS, Röcken M, Ghoreschi K: Cutaneous immunology: basics and new concepts. Semin Immunopathol. 2016;38(1):3–10. 10.1007/s00281-015-0545-x [DOI] [PubMed] [Google Scholar]
  • 7. Weinstein GD, McCullough JL, Ross P: Cell proliferation in normal epidermis. J Invest Dermatol. 1984;82(6):623–8. 10.1111/1523-1747.ep12261462 [DOI] [PubMed] [Google Scholar]
  • 8. Zenz R, Eferl R, Kenner L, et al. : Psoriasis-like skin disease and arthritis caused by inducible epidermal deletion of Jun proteins. Nature. 2005;437(7057):369–75. 10.1038/nature03963 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 9. Sano S, Chan KS, Carbajal S, et al. : Stat3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat Med. 2005;11(1):43–9. 10.1038/nm1162 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 10. Schröder JM, Christophers E: Identification of C5a des arg and an anionic neutrophil-activating peptide (ANAP) in psoriatic scales. J Invest Dermatol. 1986;87(1):53–8. [DOI] [PubMed] [Google Scholar]
  • 11. Bouchaud G, Gehrke S, Krieg C, et al. : Epidermal IL-15Rα acts as an endogenous antagonist of psoriasiform inflammation in mouse and man. J Exp Med. 2013;210(10):2105–17. 10.1084/jem.20130291 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 12. Gross O, Yazdi AS, Thomas CJ, et al. : Inflammasome activators induce interleukin-1α secretion via distinct pathways with differential requirement for the protease function of caspase-1. Immunity. 2012;36(3):388–400. 10.1016/j.immuni.2012.01.018 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 13. Dombrowski Y, Peric M, Koglin S, et al. : Cytosolic DNA triggers inflammasome activation in keratinocytes in psoriatic lesions. Sci Transl Med. 2011;3(82):82ra38. 10.1126/scitranslmed.3002001 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 14. Eedy DJ, Burrows D, Bridges JM, et al. : Clearance of severe psoriasis after allogenic bone marrow transplantation. BMJ. 1990;300(6729):908. 10.1136/bmj.300.6729.908 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Snowden JA, Heaton DC: Development of psoriasis after syngeneic bone marrow transplant from psoriatic donor: further evidence for adoptive autoimmunity. Br J Dermatol. 1997;137(1):130–2. 10.1046/j.1365-2133.1997.17821869.x [DOI] [PubMed] [Google Scholar]
  • 16. Mueller W, Herrmann B: Cyclosporin A for psoriasis. N Engl J Med. 1979;301(10):555. 10.1056/NEJM197909063011015 [DOI] [PubMed] [Google Scholar]
  • 17. Harper JI, Keat AC, Staughton RC: Cyclosporin for psoriasis. Lancet. 1984;2(8409):981–2. 10.1016/S0140-6736(84)91194-2 [DOI] [PubMed] [Google Scholar]
  • 18. Besgen P, Trommler P, Vollmer S, et al. : Ezrin, maspin, peroxiredoxin 2, and heat shock protein 27: potential targets of a streptococcal-induced autoimmune response in psoriasis. J Immunol. 2010;184(9):5392–402. 10.4049/jimmunol.0903520 [DOI] [PubMed] [Google Scholar]
  • 19. Lande R, Botti E, Jandus C, et al. : The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat Commun. 2014;5: 5621. 10.1038/ncomms6621 [DOI] [PubMed] [Google Scholar]
  • 20. Arakawa A, Siewert K, Stöhr J, et al. : Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med. 2015;212(13):2203–12. 10.1084/jem.20151093 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 21. Ghoreschi K, Laurence A, Yang XP, et al. : Generation of pathogenic T H17 cells in the absence of TGF-β signalling. Nature. 2010;467(7318):967–71. 10.1038/nature09447 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 22. Hirahara K, Ghoreschi K, Laurence A, et al. : Signal transduction pathways and transcriptional regulation in Th17 cell differentiation. Cytokine Growth Factor Rev. 2010;21(6):425–34. 10.1016/j.cytogfr.2010.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Park CO, Kupper TS: The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med. 2015;21(7):688–97. 10.1038/nm.3883 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Iijima N, Iwasaki A: T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science. 2014;346(6205):93–8. 10.1126/science.1257530 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 25. Gaide O, Emerson RO, Jiang X, et al. : Common clonal origin of central and resident memory T cells following skin immunization. Nat Med. 2015;21(6):647–53. 10.1038/nm.3860 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Watanabe R, Gehad A, Yang C, et al. : Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci Transl Med. 2015;7(279):279ra39. 10.1126/scitranslmed.3010302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Cheuk S, Wikén M, Blomqvist L, et al. : Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. J Immunol. 2014;192(7):3111–20. 10.4049/jimmunol.1302313 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 28. Schlapbach C, Gehad A, Yang C, et al. : Human T H9 cells are skin-tropic and have autocrine and paracrine proinflammatory capacity. Sci Transl Med. 2014;6(219):219ra8. 10.1126/scitranslmed.3007828 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 29. Heier I, Søyland E, Krogstad AL, et al. : Sun exposure rapidly reduces plasmacytoid dendritic cells and inflammatory dermal dendritic cells in psoriatic skin. Br J Dermatol. 2011;165(4):792–801. 10.1111/j.1365-2133.2011.10430.x [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 30. Zaba LC, Krueger JG, Lowes MA: Resident and "inflammatory" dendritic cells in human skin. J Invest Dermatol. 2009;129(2):302–8. 10.1038/jid.2008.225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Zaba LC, Fuentes-Duculan J, Eungdamrong NJ, et al. : Identification of TNF-related apoptosis-inducing ligand and other molecules that distinguish inflammatory from resident dendritic cells in patients with psoriasis. J Allergy Clin Immunol. 2010;125(6):1261–1268.e9. 10.1016/j.jaci.2010.03.018 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 32. Nestle FO, Conrad C, Tun-Kyi A, et al. : Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med. 2005;202(1):135–43. 10.1084/jem.20050500 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 33. Lande R, Gregorio J, Facchinetti V, et al. : Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature. 2007;449(7162):564–9. 10.1038/nature06116 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 34. Ganguly D, Chamilos G, Lande R, et al. : Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. J Exp Med. 2009;206(9):1983–94. 10.1084/jem.20090480 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 35. Meller S, Di Domizio J, Voo KS, et al. : T H17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26. Nat Immunol. 2015;16(9):970–9. 10.1038/ni.3211 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 36. Schäkel K, Kannagi R, Kniep B, et al. : 6-Sulfo LacNAc, a novel carbohydrate modification of PSGL-1, defines an inflammatory type of human dendritic cells. Immunity. 2002;17(3):289–301. 10.1016/S1074-7613(02)00393-X [DOI] [PubMed] [Google Scholar]
  • 37. Günther C, Blau K, Förster U, et al. : Reduction of inflammatory slan (6-sulfo LacNAc) dendritic cells in psoriatic skin of patients treated with etanercept. Exp Dermatol. 2013;22(8):535–40. 10.1111/exd.12190 [DOI] [PubMed] [Google Scholar]
  • 38. Nograles KE, Zaba LC, Guttman-Yassky E, et al. : Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways. Br J Dermatol. 2008;159(5):1092–102. 10.1111/j.1365-2133.2008.08769.x [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 39. Chien YH, Meyer C, Bonneville M: γδ T cells: first line of defense and beyond. Annu Rev Immunol. 2014;32:121–55. 10.1146/annurev-immunol-032713-120216 [DOI] [PubMed] [Google Scholar]
  • 40. Artis D, Spits H: The biology of innate lymphoid cells. Nature. 2015;517(7534):293–301. 10.1038/nature14189 [DOI] [PubMed] [Google Scholar]
  • 41. Schön MP: The plot thickens while the scope broadens: a holistic view on IL-17 in psoriasis and other inflammatory disorders. Exp Dermatol. 2014;23(11):804–6. 10.1111/exd.12541 [DOI] [PubMed] [Google Scholar]
  • 42. Shih HY, Sciumè G, Poholek AC, et al. : Transcriptional and epigenetic networks of helper T and innate lymphoid cells. Immunol Rev. 2014;261(1):23–49. 10.1111/imr.12208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Serafini N, Vosshenrich CA, Di Santo JP: Transcriptional regulation of innate lymphoid cell fate. Nat Rev Immunol. 2015;15(7):415–28. 10.1038/nri3855 [DOI] [PubMed] [Google Scholar]
  • 44. Takatori H, Kanno Y, Watford WT, et al. : Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J Exp Med. 2009;206(1):35–41. 10.1084/jem.20072713 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Sciumé G, Hirahara K, Takahashi H, et al. : Distinct requirements for T-bet in gut innate lymphoid cells. J Exp Med. 2012;209(13):2331–8. 10.1084/jem.20122097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Cella M, Fuchs A, Vermi W, et al. : A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 2009;457(7230):722–5. 10.1038/nature07537 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 47. Villanova F, Flutter B, Tosi I, et al. : Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J Invest Dermatol. 2014;134(4):984–91. 10.1038/jid.2013.477 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 48. Dyring-Andersen B, Geisler C, Agerbeck C, et al. : Increased number and frequency of group 3 innate lymphoid cells in nonlesional psoriatic skin. Br J Dermatol. 2014;170(3):609–16. 10.1111/bjd.12658 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 49. Gray EE, Suzuki K, Cyster JG: Cutting edge: Identification of a motile IL-17-producing gammadelta T cell population in the dermis. J Immunol. 2011;186(11):6091–5. 10.4049/jimmunol.1100427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Pantelyushin S, Haak S, Ingold B, et al. : Rorγt + innate lymphocytes and γδ T cells initiate psoriasiform plaque formation in mice. J Clin Invest. 2012;122(6):2252–6. 10.1172/JCI61862 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 51. Ramírez-Valle F, Gray EE, Cyster JG: Inflammation induces dermal Vγ 4 + γδT17 memory-like cells that travel to distant skin and accelerate secondary IL-17-driven responses. Proc Natl Acad Sci U S A. 2015;112(26):8046–51. 10.1073/pnas.1508990112 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 52. Hartwig T, Pantelyushin S, Croxford AL, et al. : Dermal IL-17-producing γδ T cells establish long-lived memory in the skin. Eur J Immunol. 2015;45(11):3022–33. 10.1002/eji.201545883 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 53. Cai Y, Shen X, Ding C, et al. : Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation. Immunity. 2011;35(4):596–610. 10.1016/j.immuni.2011.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 54. Mashiko S, Bouguermouh S, Rubio M, et al. : Human mast cells are major IL-22 producers in patients with psoriasis and atopic dermatitis. J Allergy Clin Immunol. 2015;136(2):351–9.e1. 10.1016/j.jaci.2015.01.033 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 55. Ghoreschi K, Brück J, Kellerer C, et al. : Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. J Exp Med. 2011;208(11):2291–303. 10.1084/jem.20100977 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 56. Schafer PH, Parton A, Gandhi AK, et al. : Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br J Pharmacol. 2010;159(4):842–55. 10.1111/j.1476-5381.2009.00559.x [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
  • 57. Geisel J, Brück J, Glocova I, et al. : Sulforaphane protects from T cell-mediated autoimmune disease by inhibition of IL-23 and IL-12 in dendritic cells. J Immunol. 2014;192(8):3530–9. 10.4049/jimmunol.1300556 [DOI] [PubMed] [Google Scholar]
  • 58. Brück J, Pascolo S, Fuchs K, et al. : Cholesterol Modification of p40-Specific Small Interfering RNA Enables Therapeutic Targeting of Dendritic Cells. J Immunol. 2015;195(5):2216–23. 10.4049/jimmunol.1402989 [DOI] [PubMed] [Google Scholar]
  • 59. Ghoreschi K, Laurence A, O'Shea JJ: Selectivity and therapeutic inhibition of kinases: to be or not to be? Nat Immunol. 2009;10(4):356–60. 10.1038/ni.1701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Ghoreschi K, Jesson MI, Li X, et al. : Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol. 2011;186(7):4234–43. 10.4049/jimmunol.1003668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ghoreschi K, Gadina M: Jakpot! New small molecules in autoimmune and inflammatory diseases. Exp Dermatol. 2014;23(1):7–11. 10.1111/exd.12265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Brunner PM, Koszik F, Reininger B, et al. : Infliximab induces downregulation of the IL-12/IL-23 axis in 6-sulfo-LacNac (slan) + dendritic cells and macrophages. J Allergy Clin Immunol. 2013;132(5):1184–1193.e8. 10.1016/j.jaci.2013.05.036 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 63. Tsianakas A, Brunner PM, Ghoreschi K, et al. : The single-chain anti-TNF-α antibody DLX105 induces clinical and biomarker responses upon local administration in patients with chronic plaque-type psoriasis. Exp Dermatol. 2016. 10.1111/exd.12927 [DOI] [PubMed] [Google Scholar]
  • 64. Kopp T, Riedl E, Bangert C, et al. : Clinical improvement in psoriasis with specific targeting of interleukin-23. Nature. 2015;521(7551):222–6. 10.1038/nature14175 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 65. Hueber W, Patel DD, Dryja T, et al. : Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2010;2(52):52ra72. 10.1126/scitranslmed.3001107 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 66. Griffiths CE, Reich K, Lebwohl M, et al. : Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet. 2015;386(9993):541–51. 10.1016/S0140-6736(15)60125-8 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 67. Ghoreschi K, Thomas P, Breit S, et al. : Interleukin-4 therapy of psoriasis induces Th2 responses and improves human autoimmune disease. Nat Med. 2003;9(1):40–6. 10.1038/nm804 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 68. Guenova E, Skabytska Y, Hoetzenecker W, et al. : IL-4 abrogates T H17 cell-mediated inflammation by selective silencing of IL-23 in antigen-presenting cells. Proc Natl Acad Sci U S A. 2015;112(7):2163–8. 10.1073/pnas.1416922112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Witte E, Kokolakis G, Witte K, et al. : IL-19 is a component of the pathogenetic IL-23/IL-17 cascade in psoriasis. J Invest Dermatol. 2014;134(11):2757–67. 10.1038/jid.2014.308 [DOI] [PubMed] [Google Scholar]
  • 70. Quaranta M, Knapp B, Garzorz N, et al. : Intraindividual genome expression analysis reveals a specific molecular signature of psoriasis and eczema. Sci Transl Med. 2014;6(244):244ra90. 10.1126/scitranslmed.3008946 [DOI] [PubMed] [Google Scholar]
  • 71. Schonthaler HB, Guinea-Viniegra J, Wculek SK, et al. : S100A8-S100A9 protein complex mediates psoriasis by regulating the expression of complement factor C3. Immunity. 2013;39(6):1171–81. 10.1016/j.immuni.2013.11.011 [DOI] [PubMed] [Google Scholar]
  • 72. Marrakchi S, Guigue P, Renshaw BR, et al. : Interleukin-36-receptor antagonist deficiency and generalized pustular psoriasis. N Engl J Med. 2011;365(7):620–8. 10.1056/NEJMoa1013068 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 73. Berki DM, Liu L, Choon SE, et al. : Activating CARD14 Mutations Are Associated with Generalized Pustular Psoriasis but Rarely Account for Familial Recurrence in Psoriasis Vulgaris. J Invest Dermatol. 2015;135(12):2964–70. 10.1038/jid.2015.288 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
  • 74. Hüffmeier U, Wätzold M, Mohr J, et al. : Successful therapy with anakinra in a patient with generalized pustular psoriasis carrying IL36RN mutations. Br J Dermatol. 2014;170(1):202–4. 10.1111/bjd.12548 [DOI] [PubMed] [Google Scholar]

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES