eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3-4/2006
vol. 31
 
Share:
Share:

Review paper
Cytokine network in psoriasis. Cross-talk between keratinocytes and cells

Anna Wojas-Pelc
,
Marta Ciszek
,
Maria Kurnyta
,
Janusz Marcinkiewicz

Centr Eur J Immunol 2006; 31 (3-4): 111-116
Online publish date: 2007/01/16
Article file
- Cytokine.pdf  [0.15 MB]
Get citation
 
 

Introduction


Etiology and pathogenesis of psoriasis.


Genetic basis

Psoriasis is a common chronic inflammatory skin disease characterized by localized hyperproliferation of keratinocytes. The disease has certain distinct but overlapping clinical phenotypes including chronic plaque lesions (psoriasis vulgaris), acute and usually self-limiting guttate type eruptions, seborrhoeic psoriasis, pustular lesions, and at least 10% of these patients develop arthritis [1]. Although the etiology of psoriasis remains unknown, it is generally assumed that it is a T-cell mediated autoimmune disease.
In recent years, genetic analyses of multiply affected families have identified some susceptibility variants for psoriasis. One of the most compelling susceptibility factors for psoriasis is the presence of HLA-Cw*0602 allele, which was found in about 50% of psoriasis patients [2]. Psoriasis is not genetically homogenous diseases, but interestingly, it is the only chronic inflammatory disease that has a strong association with HLA-C. Interestingly, in recent years, genetic connections between psoriasis and other chronic inflammatory diseases such as atopic dermatitis, rheumatoid arthritis and Crohn’s disease have been demonstrated [3]. Type I psoriasis, defined by the onset of psoriasis before age of 40 years, had a stronger genetic basis as a greater proportion of patients had a family history of psoriasis, stronger HLA associations (HLA-Cw*0602) and more severe symptoms. Patients with type II psoriasis were characterized by a later age of onset, after 40 years of age, and were found to have lower familial tendency [4, 5].
Triggering factors, both external (which directly interact with the skin) and systemic, can elicit psoriasis in genetically predisposed individuals. Psoriatic lesions can be induced by various forms of cutaneous injury (trauma, sunburn), drug eruptions or viral exanthems. For example, in 90% of psoriatic skin samples, polymerase chain reaction has revealed DNA of human papillomaviruses (mainly EV HPV5) [6]. In addition to that, considerable clinical evidence exists for the role of stress in onset and exacerbation of psoriasis [7].
Several drugs have been incriminated as inducers of psoriasis (lithium, b-blockers, interferons, angiotensin-
-converting enzyme inhibitors) as well as increased alcohol consumption and increased incidence of smoking [8, 9]. Although all these factors have been implicated, infection with b-hemolytic streptococci is the only well defined external trigger that has convincingly been associated with induction and/or aggravation of psoriasis [1]. It has been reported that patients with chronic plaque psoriasis can experience an exacerbation after streptococcal throat infection [10]. It is assumed that streptococcal toxins can act as superantigens, resulting in a complex cascade of T-cell, Langerhans cell and keratinocyte activation and interactions [11]. Moreover, it has been postulated that psoriasis is mediated by T cells that cross-react with epitopes which are common to streptococcal M proteins and type I keratins that are up-regulated in psoriatic lesions [12].

Histology of psoriatic lesions

Psoriasis is a papulosquamous disease with variable morphology, distribution, severity and course. The morphology of psoriatic lesion can range from small tear shaped papules (guttate psoriasis) to pustules (pustular psoriasis) and generalized erythema and scale (erythrodermic psoriasis). These different forms of psoriasis may be localized or widespread and disabling [13, 14]. Chronic plaque psoriasis, the most common variant of psoriasis vulgaris, is characterized by sharply demarcated and erythematous papulosquamous lesions. The classic findings of erythema, thickening and scaling are reflections of elongated dilated vertical dermal capillaries that are close to the skin surface, epidermal acanthosis plus cellular infiltrates, and abnormal keratinization. Increased keratinocyte proliferation observed in psoriasis is a consequence of an increase in the proliferating cell compartment in the basal and suprabasal levels of the epidermis; the number of cycling cells is increased approximately sevenfold. Multiple growth factors, in particular transforming growth factor-a (TGF-a), epidermal growth factor (EGF), keratinocyte growth factor (KGF), insulin-like growth factor I (IGF-I) appear to be an important autocrine mediators of these events. Increased level of these factors and increased expression of their receptors were found in psoriatic lesions and cultured psoriatic keratinocytes [15-17].
The histopathological findings in an active lesion are diagnostic for psoriasis. In the dermis the capillaries are increased in number and length. Marked edema is seen especially at the tops of the papillae, in these place there is
a mixed perivascular infiltrate of lymphocytes, macrophages and neutrophils. The epidermis is acanthotic with focal accumulations of neutrophils and lymphocytes which have migrated from the underlaying dermis. Above these foci, the granular layer is absent and the stratum corneum contains flattened nuclei. Accumulations of neutrophils in the stratum corneum (microabscess of Munro) and in the epidermis (spongiform pustule of Kogoj) are pathognomonic for psoriasis. Therefore psoriatic keratinocytes proliferate and mature rapidly so that terminal differentiation, normally occurring in granular keratinocytes, is incomplete. Hence, squamous keratinocytes aberrantly contain intact nuclei
(so-called parakeratosis) (figure 1).
Cytokine network: Cross-talk between keratinocytes and the immune cells in psoriasis
The pathogenesis of psoriasis remains primarily unclear. Nevertheless, the presence of T-lymphocyte subsets in an early phase of the disease and the response to T-cell targeting therapies strongly suggest that these cells are the driving force in the pathogenesis of psoriasis [18, 19]. The clinical features of psoriasis, such as the hyperproliferation of keratinocytes, inflammation and increased neovascularization, reflect the pathological interplay between keratinocytes and immune cells [20]. Psoriasis is considered to be a primary chronic inflammatory disorder mediated by type-1 T memory cells [3]. Although CD4+ T cells seem to be essential for initiating psoriatic lesions, CD8+ T cells may also play an important role in the pathogenesis of psoriasis as dermal T lymphocytes are a mixture of CD4+ and CD8+ T cells. Increased numbers of T cells are a highly consistent finding, while neutrophils are quite variably expressed in psoriatic lesions from different patients [21, 22]. Apart from T cells, dendritic cells (DC) form another major class of leukocytes that are found in increased numbers in psoriatic skin lesions along with mastocytes and natural killer cells (NK-T cells) [23]. Skin DCs, such as Langerhans cells (resident immature dendritic cells) and IDEC – inflammatory epidermal dendritic cells (CD83+, DC-LAMP+) play a crucial role in the development of optimal cutaneous immune responses and are known as strong polarisers of T cell responses (table 1) [3, 24].
In the pathogenic models of psoriasis all these immune cells along with keratinocytes contribute to the development of chronic skin inflammation through the production of cytokines [20]. They produce a number of effector and regulatory cytokines, predominantly of Th1-type, creating very complex cytokine network (figure 2). Psoriatic keratinocytes continuously produce enormously wide spectrum of cytokines showing distinct biological functions (TNF-α, IL-1, IL-6,
IL-7, IL-8, IL-15, IL-18, IL-19, IL-20, IL-23) [1].
TNF-α, IL-1, IL-6, the major proinflammatory cytokines, activate keratinocytes in an autocrine manner for the production of other inflammatory mediators, such as ROS, NO and various cytokines [25-27]. Some of these cytokines are responsible for accumulation of inflammatory immune cells in psoriatic plaques. For example, IL-8 plays a role in the migration of neutrophils and mast cells to lesion sites, while IL-7 and IL-15 activate CD8+ T cells [28, 29]. The exact role of TNF-α in pathogenesis of psoriatic lesions is still unclear, however, anti-TNF-α therapy demonstrated significant antipsoriatic effects indicating that this cytokine plays a crucial role in this disease [25, 26].
It is well documented that Th1 cytokines, such as IL-23, IFN-g and TNF-α predominate in psoriatic lesions [1, 30]. IFN-g is produced by both CD4+ Th1 and CD8+ Tc1 cells, and may be a central effector cytokine in psoriasis. However, this is IL-23, the cytokine produced by keratinocytes and/or skin DC activated with IL-18, that plays a principal role in activating T cells in psoriasis [31, 32]. IL-23 is a heterodimer, sharing a p40 subunit with
IL-12 but having a distinct p19 subunit. IL-23 stimulates T cells for IFN-g production polarizing the immune response into type-1. It has been recently postulated that IL-23, but not IL-12, plays a major role in the perpetuation of the inflammation process in psoriasis [33, 34].
Other cytokines contribute to hyperplasia of keratinocytes, the major hallmark of psoriatic lesions. IL-19, IL-20 and IL-22, play a special role in psoriasis; they all belong to
a family of cytokines structurally related to IL-10, but in contrast to IL-10, they have proinflammatory activities
[35, 36]. The increased IL-19, IL-20, IL-22 mRNA expression and detection of their receptors in lesional psoriatic skin suggests that these cytokines are involved in the pathogenesis of psoriasis [37, 38]. A number of separate studies confirmed recently this hypothesis. It has been shown that IL-19 stimulates dermal macrophages for production of IL-6 and TNF-α [39]. In addition, it has been demonstrated that IL-19, as well as IL-20, stimulates CD8+ T cells to produce KGF (keratinocyte growth factor), which contributes to sustaining the hyperproliferative status of keratinocytes [37, 40]. On the other hand IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of keratinocytes [41].
The pattern of cytokines involved in pathogenesis of psoriasis may be defined as proinflammatory and type-1 (Th1 inducers), and is characteristic for many inflammatory autoimmune diseases. However, unique for psoriasis is the effect of cross–talk between hyperproliferating keratinocytes and immune cells. At inflammatory psoriatic lesions both keratinocytes and inflammatory cells produce high amounts of ROS and NO which leads to the expression of heme oxygenase-1 (HO-1), a stress inducible enzyme with anti-oxidant, cytoprotective and anti-inflammatory properties (figure 2) [42].
In recent years our knowledge about the role of cytokines in pathogensis of psoriasis has improved dramatically. The question is how to use this knowledge to treat this disease.

Major strategies of psoriasis treatment.
Cytokine therapies

The traditional strategy of treatment of psoriasis is to minimize the severity of psoriatic lesions. First–line therapy of psoriasis includes topical application of agents which affect keratinocyte proliferation and production of inflammatory mediators involved in pathogenesis of psoriatic skin inflammation [43]. However, the role of topical therapeutic agents in improving the balance between oxidants and antioxidants is unclear and not consistent with the suggested, pathological role of oxidative stress in psoriasis For example, phototherapy (UVB- ultraviolet B, PUVA – photosensitizing medicaments + ultraviolet A), is one of the forms of traditional topical therapy to treat psoriasis via generation of ROS [44].
As all of the above proinflammatory mediators are under control of anti-inflammatory agents, the anti-inflammatory therapy (e.g. systemic cytokine therapy) seems to be a rational and effective strategy in the treatment of psoriasis [18].
New anti-inflammatory drugs, biologically based agents (e.g anti-TNF agents), are devoid of the side-effects characteristic for traditional systemic anti-psoriatic agents such as methotrexate and cyclosporine, and, most importantly, they can precisely target steps in the pathogenesis of psoriasis [18, 26]. All anti-TNF agents currently in clinical use, namely the monoclonal antibodies infliximab and adalimumab, as well as the soluble TNF receptor etanercept, markedly decrease not only joint inflammation but also skin inflammation in patients with psoriatic arthritis. In addition, they are effective in decreasing the clinical activity of skin lesions in patients with the severe form of psoriasis vulgaris [45].
In our opinion, future cytokine therapies in the treatment of psoriasis should include IL-10 therapy (for its ability to inhibit Th1 response) and anti–IL-23 therapy (for its ability to stimulate Th1 response).

Conclusions

In the present paper we have summarized the current data confirming the role of cytokines in physiological and pathological status of the skin. It is commonly accepted that cytokines of Th1-type play an essential role in the development of autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease and psoriasis. In the last decade, an enhanced understanding of the role of cytokines in pathogenesis of psoriasis has led to the development and utilization of new drugs. However, further complex studies are necessary to determine the role of cytokines, ROS and HO-1 system in the development and resolution of psoriatic lesions [46].


Acknowledgements

We wish to thank Maria Ciszek and Dr Piotr Kochan for their assistance in preparation of this manuscript. This work was supported by a grant No 501/P/22/L.

References

1. Gudjonsson JE, Johnston A, Sigmundsdottir H, et al. (2004): Immunopathogenic mechanisms in psoriasis. Clin Exp Immunol 135: 1-8.
2. Krueger JG, Bowcock A (2005): Psoriasis pathophysiology: current concepts of pathogenesis. Ann Rheum Dis 64 Suppl 2: 30-36.
3. Krueger G, Ellis CN (2005): Psoriasis-recent advances in understanding its pathogenesis and treatment. J Am Acad Dermatol 53 (1 Suppl 1): S94-S100.
4. Henseler T, Christophers E (1985): Psoriasis of early and late onset: characterization of two types of psoriasis vulgaris. J Am Acad Dermatol 13: 450-456.
5. Rahman P, Elder JT (2005): Genetic epidemiology of psoriasis and psoriatic arthritis. Ann Rheum Dis 64: Suppl 2: ii37-ii39.
6. Majewski S, Jabłonska S (2003): Epidermodysplasia verruciformis human papillomaviruses in skin cancer and psoriasis. Przegl Dermatol 5: 327-333.
7. Fortune DG, Richards HL, Main CJ, et al. (1998): J Am Acad Dermatol 39: 196-201.
8. Tsankov N, Angelova I, Kazandjieva J (2000): Drug-induced psoriasis. Recognition and management. Am J Clin Dermatol 1: 159-165.
9. Higgins E (2000): Alcohol, smoking and psoriasis. Clin Exp Dermatol 25: 107-110.
10. Wardrop P, Weller R, Marais J, et al. (1998): Tonsillitis and chronic psoriasis. Clin Otolaryngol 23: 67-68.
11. Kormeili T, Lowe NJ, Yamauchi PS (2004): Psoriasis: immunopathogenesis and evolving immunomodulators and systemic therapies; U.S. experiences. Br J Dermatol 151: 3-15.
12. McFadden J, Valdimarsson H, Fry L (1991): Cross-reactivity between streptococcal M surface antigen and human skin. Br J Dermatol 125: 443-447.
13. Christophers E (2001): Psoriasis-epidemiology and clinical spectrum. Clin Exp Dermatol 26: 314-320.
14. Langley RG, Krueger GG, Griffiths CE (2005): Psoriasis: epidemiology, clinical features, and quality of life. Ann Rheum Dis 64: ii18-ii23.
15. Elder LT, Klein SB, Tavakkol A, et al. (1990): Growth factor and proto-oncogene expression in psoriasis. J Invest Dermatol 95 (5 Suppl): 7S-9S.
16. Mascia F, Mariani V, Girolomoni G, et al. (2003): Blockade of the EGF receptor induces a deranged chemokine expression in keratinocytes leading to enhanced skin inflammation. Am J Pathol 163: 303-312.
17. Peus D, Beyerle A, Vasa M, et al. (2004): Antipsoriatic drug anthralin induces EGF receptor phosphorylation in keratinocytes: requirement for H(2)O(2) generation. Exp Dermatol 13: 78-85.
18. Koo J, Khera P (2005): Update on the mechanisms and efficacy of biological therapies for psoriasis. J Dermatol Sci 38: 75-87.
19. Ellis CN, Krueger GG, Alefacept Clinical Study Group (2001): Treatment of chronic plaque psoriasis by selective targeting of memory effector T lymphocytes. N Engl J Med 345: 248-255.
20. Albanesi C, Scarponi C, Giustizieri ML, et al. (2005): Keratinocytes in inflammatory skin diseases. Curr Drug Targets Inflamm Allergy 4: 329-334.
21. Schaerli P, Britschgi M, Keller M, et al. (2004): Characterization of human T cells that regulate neutrophilic skin inflammation. J Immunol 173: 2151-2158.
22. Wetzel A, Wetzig T, Haustein UF, et al. (2006): Increased neutrophil adherence in psoriasis: role of the human endothelial cell receptor Thy-1 (CD90). J Invest Dermatol 126: 441-452.
23. McGregor JM, Barker JN, Ross EL, et al. (1992): Epidermal dendritic cells in psoriasis possess a phenotype associated with antigen presentation: in situ expression of beta 2-integrins.
J Am Acad Dermatol 27: 383-388.
24. Yu Y, Tang L, Wang J, et al. (2002): Psoriatic lesional keratinocytes promote the maturation of human monocyte-derived Langerhans cells. Dermatology 204: 94-99.
25. Lowes MA, Chamian F, Abello MV, et al. (2005): Increase in TNF-αlpha and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proc Natl Acad Sci 102: 19057-19062.
26. Gottlieb AB, Chamian F, Masud S, et al. (2005): TNF inhibition rapidly down-regulates multiple proinflammatory pathways in psoriasis plaques. J Immunol 175: 2721-2729.
27. Pelle E, Mammone T, Maes D, et al. (2005): Keratinocytes act as a source of reactive oxygen species by transferring hydrogen peroxide to melanocytes. J Invest Dermatol 124: 793-797.
28. Jiang WY, Chattedee AD, Raychaudhuri SP, et al. (2001): Mast cell density and IL-18 expression in nonlesional and lesional psoriatic skin. In. J Dermatol 40: 699-703.
29. McInnes IB, Gracie JA (2004): Interleukin-15: a new cytokine target for the treatment of inflammatory diseases. Curr Opin Pharmacol 4: 392-397.
30. Schottelius AJ, Moldawer LL, Dinarello CA, et al. (2004): Biology of tumor necrosis factor-alpha- implications for psoriasis. Exp Dermatol 13: 193-222.
31. Wittmann M, Purwar R, Hartmann C, et al. (2005): Human keratinocytes respond to interleukin-18: implication for the course of chronic inflammatory skin diseases. J Invest Dermatol 124: 1225-1233.
32. Langrish CL, Chen Y, Blumenschein WM, et al. (2005): IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.
33. Piskin G, Sylva-Steenland RM, Bos JD, et al. (2006): In vitro and in situ expression of IL-23 by keratinocytes in healthy skin and psoriasis lesions: enhanced expression in psoriatic skin. J Immunol 176: 1908-1915.
34. Lee E, Trepicchio WL, Oestreicher JL, et al. (2004): Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med 199: 125-130.
35. Fickenscher H, Hor S, Kupers H, et al. (2002): The interleukin-10 family of cytokines. Trends Immunol 23: 89-96.
36. Jung M, Sabat R, Kratzschmar J, et al. (2004): Expression profiling of IL-10-regulated genes in human monocytes and peripheral blood mononuclear cells from psoriatic patients during IL-10 therapy. Eur J Immunol 34: 481-493.
37. Wei CC, Chen WY, Wang YC, et al. (2005): Detection of IL-20 and its receptors on psoriatic skin. Clin Immunol 117: 65-72.
38. Rich BE (2003): IL-20: a new target for the treatment of inflammatory skin disease. Expert Opin Ther Targets 7: 165-174.
39. Liao YC, Liang WG, Chen FW, et al. (2002): IL-19 induces production of IL-6 and TNF-αlpha and results in cell apoptosis through TNF-αlpha. J Immunol 169: 4288-4297.
40. Li HH, Lin YC, Chen PJ, et al. (2005): Interleukin-19 upregulates keratinocyte growth factor and is associated with psoriasis. Br J Dermatol 153: 591-595.
41. Boniface K, Bernard FX, Garcia M, et al. (2005): IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. J Immunol 174: 3695-3702.
42. Hanselmann C, Mauch C, Werner S (2001): Haem oxygenase-1: a novel player in cutaneous wound repair and psoriasis? Biochem J 353: 459-466.
43. Lange RW, Hayden PJ, Chignell CF, et al. (1998): Anthralin stimulates keratinocyte-derived proinflammatory cytokines via generation of reactive oxygen species. Inflamm Res 47: 174-181.
44. Lebwohl M, Ting PT, Koo JY (2005): Psoriasis treatment: traditional therapy. Ann Rheum Dis 64 Suppl 2: ii83-ii86.
45. Sfikakis PP, Iliopoulos A, Elezoglou A, et al. (2005): Psoriasis induced by anti-tumor necrosis factor therapy: a paradoxical adverse reaction. Arthritis Rheum 52: 2513-2518.
46. Wojas-Pelc A, Marcinkiewicz J (2006): What is a role of heme oxygenase-1 in psoriasis? Current concepts of pathogenesis. Int J Exp Pathol [in press].
Copyright: © 2007 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.