Open access peer-reviewed chapter

Immunomodulatory Effect of Methotrexate Abruptly Controls Keratinocyte Activation in Psoriasis

Written By

Tamilselvi Elango, Anburaj Jeyaraj, Haripriya Dayalan, Pushpa Gnanaraj, Xinghui Li and Xuejun Zhang

Submitted: 19 December 2021 Reviewed: 21 January 2022 Published: 08 April 2022

DOI: 10.5772/intechopen.102811

From the Edited Volume

Psoriasis - New Research

Edited by Shahin Aghaei

Chapter metrics overview

413 Chapter Downloads

View Full Metrics

Abstract

In psoriatic skin, epidermal keratinocytes (KCs) undergo deregulated inflammatory response that leads to prolonged expression of inflammatory mediators as well as abnormal keratins. Due to immune and genetic factors, KCs get activated and cell balance gets disturbed. This activation is mainly due to deregulated inflammatory response. A vicious cycle of KC-immune response called KC activation cycle leads to psoriasis. In psoriatic skin, epidermal KCs undergo deregulated inflammatory response that leads to prolonged expression of inflammatory mediators as well as abnormal keratins. Methotrexate (MTX) an immunosuppressive agent has been used as a standard drug to treat severe psoriasis. Acanthosis and abnormal terminal differentiation was mainly due to the mutation in epidermal keratins. In turn, disease severity and relapsing of psoriasis are mainly due to the mutation of hyperproliferative keratins. These novel keratin mutations in psoriatic epidermis might be one of the causative factors for psoriasis. MTX strongly regulates the KC activation cycle by deregulated inflammatory markers and maintains normal keratin phenotype on hyperproliferating KC, thereby controlling acanthosis in psoriasis patients.

Keywords

  • psoriasis
  • methotrexate
  • keratins
  • inflammatory markers
  • keratinocyte

1. Introduction

Psoriasis is a chronic inflammatory skin disease that mainly characterized by acanthosis, abnormal differentiation and infiltration of leukocytes from the dermis. The factors that causing this disease are genetic, environmental and inflammatory mediators [1]. In normal skin, the transformation of basal keratinocytes (KCs) to anucleate corneocytes process will takes within 50 days. Whereas, in psoriatic skin the epidermal cell cycle is rapid and the transformation occurs within 5 days. Thereby, the stratum corneum contains fully unmatured keratinized cells which build up abnormally and forms scales like structure. Due to this, epidermis of psoriatic lesions will become thicker and also blood vessels in the papillary layer of the dermis get dilated along with effusion of inflammatory cells, such as neutrophils, infiltrate the epidermis [2]. About 80% of the epidermal skin constitutes KCs. KC play a major role in this chronic inflammatory disease. It play a special role in sensing epidermal barrier and regulating immune homeostasis [1].

1.1 Hyperproliferation of epidermis in psoriasis

Until the late 1970s, the hypothesis of psoriasis arising from abnormalities in KCs was favored, and the abnormal proliferation was treated with antiproliferative agents. Since then, the participation of KC in the pathogenesis of psoriasis has certainly been overlooked. A publication by Zenz et al. [3] has highlighted again the role of KC in the pathomechanisms leading to psoriatic lesions. Their findings favor the view that psoriasis could also be regarded as a primary KC disorder amplified by the immune system [4]. While the debate will continue among skin biologists on the different theories, it is unquestionable that KC are potential initiators of inflammation, producing a number of cytokines, adhesion molecules and growth factors.

The growth of KC is regulated by a delicate balance between molecules that control cell survival and cell death. Thus, the thickness of human epidermis remains relatively constant throughout life. This regulation is disturbed in psoriasis that leads to KC hyperproliferation with the net result of an increase in the volume of cell mass [5]. The epidermal cell cycle of hyperproliferating psoriatic KC occurs within 5 days. Effusion of growth factors and inflammatory mediators from different skin cells, are believed to regulate the epidermal hyperproliferation in psoriasis (Figure 1) [6].

Figure 1.

Illustration of the epidermal layers of the normal skin and psoriatic skin including scaliness, hyperkeratosis, and neutrophil accumulation in the stratum corneum. A indicates immunohistology of normal skin, B indicates immunohistology of psoriatic skin.

When skin cells exposed to any external factors such as environmental, chemical and internal factor like genetic, psychological and physical stress will probably activate immune cells within the KCs, which create KCs to hyperproliferate and also altered differentiation. Thus, the establishment of mutual KC-immunocyte stimulation (KC activation cycle) will leads to psoriasis [7, 8]. The hyperproliferation in psoriasis seemed to result from an increase in the number of transit amplifying cells, following depletion of the stem cell compartment [9]. As a whole, these changes suggest that intermediate filament (IF) keratin pair provides specific functional requirements to maintain epidermal KCs. KCs stability and integrity are mainly depend on keratin proteins [10, 11]. Keratins are the main structural cytoskeletal protein, an IF in all epithelia [12]. In normal skin, basal KCs express K5 and Keratin 14 (K14) keratins which helps in proliferation, whereas suprabasal cells express K1 and Keratin 10 (K10) keratins which supports differentiation process [13, 14]. Subsequently, any defects in these keratins can lead to cell fragility and are linked to a wide array of genodermatoses and cancers [15, 16].

Since genome-wide association studies (GWAS), connecting the psoriasis to the late cornified envelope gene cluster has specified that epidermal abnormalities along with hyperproliferative keratin pattern plays a major role in the pathogenesis of psoriasis [17, 18]. Studies have shown that dysfunction or mutations of keratin proteins are associated with a remarkable variety of skin disorders, such as skin blistering, inflammatory disorders and skin tumors [19]. The main aim of psoriasis treatments is to stop skin cells from growing so quickly and to remove scales. Methotrexate (MTX) is considered as the gold standard therapy for moderate to severe psoriasis [20, 21, 22, 23]. Mostly, MTX exerts various immunomodulatory effects on T cell and also control KC growth [24]. In psoriasis, MTX was found to decreases the markers involved in hyperproliferation [25].

In this chapter we will first describe KC hyperproliferation then how keratins are expressed and regulated in psoriasis, then we will describe how MTX exert its action on controlling psoriasis through its immunomodulatory effect on Keratins and KC activation.

Advertisement

2. Keratins involved in psoriasis

The hyperproliferation seemed to result from an increase in the number of transit amplifying cells, following depletion of the stem cell compartment [9, 26]. The differentiation state of epidermal KCs is reflected by the intricate expression pattern of keratins [27, 28]. Keratins are members of the large IF gene family [29] in all epithelia including the epidermis. Basal KCs express keratins (K) K5 and K14, which helps to maintain epidermal shape are replaced by differentiation keratin K1 and K10 [27], whereas in activated KCs, keratins K6, K16, and Keratin 17 (K17), which are distinct from the keratins in the healthy epidermis, are expressed [30]. Therefore, keratins K6, K16, and K17 are usually referred to as activation- and hyperproliferation-associated keratins. There are predicted deleterious mutations were highly located in α-helical rod domain of keratins which forms coiled structure to these keratins, are important to maintain the structural integrity of the skin. Thus the genetic defects of basal keratin K14 and differentiation keratin K10 in skin might leads to express abnormal keratin pair which causes thickening of epidermis (acanthosis) in psoriasis (Figure 2).

Figure 2.

Protein structure of epidermal and hyperproliferative keratins protein. (a) Schematic representation of secondary structure of all four keratins with their domain and sub domain. Whereas, S = peptide epitope; L = linker. (b) Region specific of 33 deleterious mutation localization. The left sides of the figures are the 3D structure of all four keratins CDS region. The right sides of the figures are the 3D structures of domains with mutated residues. The mutated residues are numbered according to their position on regions. The position of mutated amino acids (aa) on all four keratins and on CDS regions are provided [31].

2.1 Keratin 14

Keratin 5 and K14 pair are considered as a biochemical marker of mitotically active basal layers. This pairs are supports to maintain epidermal integrity as well as protects skin from mechanical stress. Interestingly, the K5/K14 pair is expressed in the basal layer of the epidermis, which contains epidermal stem cells and transient amplifying (TA) cells [32]. Many studies have showed that the level of K14 was considerably higher in lesional skin than in normal epidermis [33, 34, 35]. p53 activation promotes the expression of p21 and the repression of K14 during epidermal differentiation [36], whereas in psoriasis, there is downregulation of p53 [37] and also decreased Notch 1 expression [38] causes increase in K14 expression in psoriasis [39]. p75NTR is nearly absent in psoriatic KCs that are reportedly resistant to apoptosis, leads to increase TA cell turnover in psoriasis which leads to increase in K14 expression in psoriasis (Figure 3) [40].

Figure 3.

Immunofluorescence analysis of keratins in control and lesional psoriatic skin. Immunofluorescence analysis of epidermal and hyperproliferative type I keratins in frozen skin sections from patients with psoriatic skin (×20 magnification, respectively), and nuclei were visualized with 4′-6-diamidino-2-phenylindole (DAPI). Bar = 100 μm. Ab = antibody [31].

2.2 Keratin 10

The epidermis is a stratified epithelium that regenerates permanently from the basal layer. K10 pair with K1 is considered as a major differentiation keratin [29]. Normally,the basal layer contains K14, K15, and K5, whereas suprabasal, postmitotic cells switch to the expression of K1 and K10 [41, 42] is also involved in the control of cell proliferation [43]. Many studies have shown a downregulation of K1 and K10 [33, 34, 35] in psoriatic epidermis. This may be due to downregulation in E-cadherin expression [43], and increase in IL-22 [45, 46, 47, 48] levels in psoriasis. Also c-fos protein in AP-1 transcription factor highly regulates a number of genes that are involved in KC differentiation were found to be reduced in psoriasis [49, 50, 51, 52].

In psoriasis, defects in K10 expression leads to hyperproliferation of KCs by decreasing its inhibitory action on Rb phosphorylation, which leads to increased Cyclin D and E expression and also increased Phosho-Akt levels in psoriasis (Figure 3) [52].

2.3 Keratin 16

Keratin 16 expressions, reduces the fraction of cells in G1 while increasing that in S phase [52]. Many authors also shown an upregulation of K6 and K16 [33, 35, 54, 55] in psoriatic epidermis. Decrease expression of keratin 10 gene leads to hyperproliferation of basal cells, alterations in epidermal cell cycle by inducing cmyc, cyclin D1, 14-3-3σ, keratin 6 and keratin 16 [56]. Multiple transcription factors activated by the extracellular signals revealed the specific signals transduction mechanisms that respond to the corresponding growth factors and cytokines. For example, interleukin-1 (IL-1), present in healthy epidermis in inactive form [57], when released autocrinely, activates NFκB and C/EBPβ, thus initiating KC activation. Among the characteristics of activated KCs is the production of tumor necrosis factor-α (TNF-α) [58], which maintains activated NFκB and C/EBPβ. Activated KCs produce ligands of the EGF receptor that cause activation of AP1, such as transforming growth factor-α (TGF-α), amphiregulin and HB-EGF. Interestingly, all these cytokines and growth factors IL-1, TNF-α, TGF-α and EGF, induce expression of the K16 genes, albeit through separate pathways [54, 59, 60, 61]. Study has shown that biosynthesis of AP1 protein (c-Jun and c-Fos) through Ras activation are actively participate in the transcriptional regulation for EGF which induced keratin 16 gene expression in KCs [62], this implies that EGF induces Keratin 16 expression in psoriasis (Figure 4).

Figure 4.

Transcriptional regulation of the human keratin 16 gene in HaCaT cells.

2.4 Keratin 17

Keratin 17 (K17) is a type I intermediate filament mainly expressed in the basal cells of epithelial in hair follicles. As a complex cytoskeletal protein, K17 regulates a numereous biological processes in epithelial layer, including cell proliferation and growth, skin inflammation and hair follicle cycling. Abnormal expression of K17 is found in various diseases ranging from psoriasis to malignancies such as breast, cervical, oral squamous and gastric carcinomas [63, 64].

Many studies have showed the aberrant expression of K17 in psoriatic epidermis [35, 65, 66]. This aberrant expression is mainly due to interferon-γ (IFN-γ) produced by activated T cell [67]. Various inflammatory mediators like IFN-γ, IL-6, IL-17 A, IL-22 derived from T cells induced transcription of keratin K17 through STAT3- and ERK1/2-dependent mechanisms [64, 68, 69]. Thus, all these cytokines are involved in the induction of K17/T cell/cytokine autoimmune loop and play an important role in the progression of psoriasis [68].

Advertisement

3. Keratinocyte activation cycle (KAC)

Basal KC have two alternative pathways to end up. In normal skin, KCs undergo proliferation in basal layer and differentiation in both spinosus, granular layer, finally it end up as anucleate corneocytes in cornified layers. During all these process, various factors like calcium, retinoic acid, vitamin D3 and protein kinase C (PKC) activators are required to induce proliferation and differentiation in epidermis [70, 71, 72, 73, 74]. For proliferation and differentiation, Epidermal KCs required a pair of kertain proteins, for instance, in normal skin Keratin pair K5/K14 expressed in basal layer and K1/K10 are expressed in spinosus layer [75].

However, in pathological conditions like epidermal injury, cancer and psoriasis an alternative KCs pathway is get activated by inflammatory mediators. This alternative pathway disturbed normal proliferation and migrating phenotype in KCs [76, 77]. This alternative pathway is considered as a unusual cycle formed by the interaction of KCs-immunocytes, which is precisely elucidated as KC activation cycle [78]. This activation cycle begins with leakage of interleukin-1 beta (IL-1 β) from KC after any injury or disease condition. This inflammatory mediator initiate activation of KCs by changing the keratin pattern from K5/14 to K6/16 and firmly maintained by TNF-α and TGF-α [60, 61, 75, 79]. After any treatment or lesional healing stage, IFN-γ was released by KCs which act as a signal for healing process and also induces the expression of K17 [69, 80]. To normalize the healed skin, a transforming growth factor-β (TGF-β) synthesized from dermal fibroblasts were found to aid KCs phenotype by producing K5/14 keratin in basal layer (Figure 5) [81].

Figure 5.

The KC activation cycle. In normal KC, Basal layer produces K5 and K14 and differentiate to K1 and K10 with the help of ca2+, D3. Whereas in psoriatic KCs, K5 and K14 produce K6 and K16 by IL-1. TNF-α help to maintains activated KCs. Then, IFN-γ prompts K17 expression which stimulates KCs contractility. TGF-γ,a de-activating signal is not produced during psoriass, thereby hyperproliferation of epidermis in psoriasis. Symbol ↑ indicates increase in psoriasis; symbol ↓ indicates decrease in psoriasis.

3.1 Initiator of activation

The most common initiator of KC activation is IL-1. In pathological conditions like psoriasis, KC process and release IL-1, allowing the surrounding cells to perceive it [82, 83, 84]. The released IL-1 serves as a paracrine signal to dermal endothelial cells to become activated, express selectins, and slow down the circulating lymphocytes [8586]. IL-1 also serves as a chemoattractant for lymphocytes, causing them to extravasate and migrate to the site of lesion [87]. Furthermore, IL-1 is an activator of dermal fibroblasts, enhancing their migration, proliferation, and production of dermal extracellular matrix components [88]. IL-1 is also an autocrine signal that activates KC. IL-1 causes them to proliferate, become migratory, and express an activation-specific set of genes [76]. Thus, IL-1 initiates KC activation not only by triggering additional signaling events, but also by inducing directly the synthesis of K6/K16 in epidermal KC, and thus changing the composition of their cytoskeleton.

3.2 Maintenance of activation

TNF-α induced by IL-1 can maintain KCs in an activated state [75]. In psoriasis, a wide variety of cells produce TNF-α, primarily macrophages and monocytes but also epithelial cells including KCs [89]. TNF-α activates immune responses by inducing production of additional signaling molecules, cytokines, growth factors, their receptors, and adhesion proteins (e.g., amphiregulin, TGF-α, IL-1 α, IL-1 receptor antagonist, epidermal growth factor receptor (EGFR), and intercellular adhesion molecule (ICAM-1) [90]. In response to the activation of the EGFR, KCs proliferate, degrade components of the extracellular matrix, and become migratory [91].

3.3 The activated phenotype

Once activated, KCs synthesize additional signaling growth factors and cytokines including TGF-α IL-3, IL-6, IL-8, G-CSF, GM-CSF, and M-CSF [91, 92]. These signaling molecules produced by KCs act as paracrine signal to white blood cells, lymphocytes, fibroblasts, and endothelial cells in dermis. Apart from paracrine, it also acts as an autocrine signal for KCs in epidermis. Numerous cell surface proteins and integrins are also found to act as secondary moiety to activate epidermal KCs [93, 94].

3.4 The contractile keratinocyte

Psoriasis is associated with high levels of IFN-γ in epidermis [91]. IFN-γ strongly and specifically induced the promoter of the K17 (abnormal marker) gene. K17 is exceptional because it is not found in healthy interfollicular epidermis, but it is expressed in certain pathologic states, psoriasis [28]. The function of K17 in epidermis therefore may be to promote or allow KC contractility and/or frequent changes in shape [95]. Indeed, expression of K17 has been used to evaluate the course of treatment of psoriatic patients [63]. Due to failure, to resolve the deregulated inflammatory response in psoriasis leads to the persistent activation of KCs, which is characterized by prolonged K17 expression [13].

3.5 Back to normal basal phenotype

To revert to the basal cell phenotype, KCs need a signal. This signal comes from the dermal fibroblasts in the form of TGF-β. Cell kinetics study by Van Ruissen et al. [96] clearly indicate that the TGF-β donot control KCs normal proliferation, but it controls the abnormal proliferation of KCs (antihyperproliferative). Whereas in psoriasis, the expression of TGF-β is low [97]. So the activation cycle is not reverted to normal basal phenotype, therefore hyperproliferation of KC takes place in psoriasis.

Advertisement

4. Methotrexate

MTX is the most commonly used systemic agent for psoriasis and, because it has been available for 35 years, most dermatologists are comfortable with its use. It is considered as an longstanding effective oral medication for the treatment of various types of psoriasis [98, 99]. Generally psoriasis treatment is mainly depends on the PASI score. For mild psoriasis patients, doctors prescribed topical therapy and phototherapy. For moderate to severe disease who have at least 5% of their skin covered with psoriasis are treated with oral medications like MTX, cyclosporine. [100]. Folate supplementation along with MTX reduces the incidence of megaloblastic anemia, hepatotoxicity, and gastrointestinal intolerance [101].

4.1 History

Anti-metabolites mimic substances required for normal biochemical reactions and thus interfere with normal functions of the cell, including cell division. They may masquerade as purines (e.g., azathioprine), pyrimidines (e.g., 5-flourouracil) and folic acid analogs essential for purine and pyrimidine synthesis, (e.g., MTX). Methotrexate, formerly known as aminopterin, has been widely used in the treatment of cancer and autoimmune diseases [102]. It was first developed in the 1940s when scientists were investigating the effects of folic acid on cancer, particularly childhood leukemia [103]. MTX is an analog of folic acid that inhibits cellular proliferation inducing folate coenzyme deficiencies [104]. When MTX was incidentally noted to improve psoriatic lesions in the 1960s it became clear that it possessed anti-inflammatory properties in addition to its antiproliferative effects [105, 106, 107]. Anti-inflammatory actions of MTX is accomplished by inhibiting dihydrofolate reductase which eventually diminishes the de novo synthesis of purines and pyrimidines and increases the catabolism of AMP and adenosine to IMP and inosine. Increased catabolism leads to accumulation of adenosine which confers anti-inflammatory effects of MTX [108].

4.2 Chemical structure of methotrexate

(2S)-2-[[4-[[2,4-bis (azanyl)pteridin-6-yl]methyl-methylamino]phenyl]carbonylamino] pentanedioic acid.

Advertisement

5. Clinical efficacy of methotrexate on psoriasis

Even though MTX has been used extensively for the treatment of psoriasis, so far its efficacy has not been supported by any clinical datas. A non-randomized controlled trial study done by our research team showed PASI 75 was achieved in 75% of psoriasis patients after 7.5 mg of MTX orally per week for a period of 12 weeks. PASI 75 is defined as a reduction from baseline PASI score of >75%. PASI 75 is used as the benchmark of primary end points in assessing therapies for psoriasis. Patients reaching PASI 75 represent very meaningful changes in psoriasis severity [109, 110, 111, 112]. A randomized controlled clinical trial using 15 mg of MTX for 16 weeks in moderate to severe plaque has showed 60% of patients achieved a PASI-75 response (no significant difference, P = 0.29), [113, 114]. For safety consideration, MTX has found to be administered along with 5 mg of folic acid to minimize the serious toxicity include hematological disorder, hepatotoxicity and gastrointestinal toxicity, MTX has also affect lymphocytes and also cause autoimmune reaction at extreme usage [115, 116, 117]. But, several studies has shown that 5–10 mg of MTX cause less side-effect on liver of psoriasis patients [118, 119, 120, 121]. However more than 15 mg will cause severe side effects in psoriasis patients [121]. Clinical improvement of psoriatic patients before and after treatment of 7.5 mg of MTX per week for 12 weeks were depicted in (Figure 6).

Figure 6.

Clinical efficacy of MTX on psoriatic patients.

Overall, the clinical data from our research study and also from other clinical trial has strongly supports that using low dose of MTX is well and safe for the treatment of moderate to severe psoriasis patients, and also frequent expert supervision along with laboratory monitoring is necessary.

Advertisement

6. Metabolic activity of methoxerate on psorisis

MTX mechanisms of action are likely to account for its antiproliferative and immunosuppressive effects [122, 123]. The key feature of psoriasis is KC hyperproliferation. MTX was found to induce maturation and inhibition of KC proliferation through its metabolic action of kerationocyte. The putative effects of MTX are listed below:

  1. Reduction of cell proliferation

  2. Increase of apoptosis of T cells

  3. Increase of endogenous adenosine release

  4. Alteration of expression of cellular adhesion molecules and

  5. Influence on production of cytokines, humoral responses, and bone formation.

6.1 Effect of methotrexate on T cells

Evidence supports that activated T cells are key players in the immunopathogenesis of psoriasis. The following components involving T cells are considered crucial in the pathogenesis of psoriasis. Activated endothelium of psoriatic skin has shown adhesion molecules like E-selectin, CLA, ICAM-1 and ICAM-3, which promoted the activation of KCs and T-cells in psoriasis. Study by Sigmundsdottir et al. showed that treatment of 5−25 mg of MTX in 16 moderate to severe psoriasis patients has showed decreased E-selectin and CLA expression in psoriatic skin. Thus, the downregulation of peripheral Tcell-adhesion interaction by MTX in psoriasis patients implies its therapeutic action on psoriatic skin lesion [124]. Invitro flowcytometric and immunohistochemistry studies have shown that peripheral T cells were found to show less interaction with CLA and ICAM-1 after MTX administration 10−9 M to 10−5 M for 5 days. Follow-up experiments revealed that MTX suppression of CLA expression could be reversed by folinic acid (leucovorin) supplementation [123].

MTX has also target T cell by inducing cytolysis. Some studies has shown that MTX induce Tcell apoptosis in more sensitive manner [125, 126, 127]. MTX may induce cell death via free radical oxygen species. Phillips et al. [126] inhibited MTX induced T-cell death with the addition of the antioxidant glutathione and its precursor, Nacetylcysteine. Accumulating evidence suggests that MTX alters T-cell production of several cytokines, including IL-1, IL-2, IL-4, IL-8, INF-γ and TNF-α [128, 129, 130, 131]. As a key element in psoriasis pathogenesis, the cytokine TNF-α was found at higher levels in psoriasis plaques and the synovial fluid of patients with psoriatic arthritis [129, 132, 133]. Associations between MTX and TNF-α levels have been observed since the 1990s. Studies by Seitz et al. [132], Neurath et al. [134] and Hildner et al. [135] found that MTX had reduced TNF-α production in the peripheral blood mononuclear cells (PBMCs) of psoriasis patients.

Action of MTX on reducing serum and synovial TNF-α has been widely established in both Psoriasis and rheumatoid arthritis patients showed its immunomodulatory effects [136, 137, 138]. Based on the stage and route of T-cell activation, it has been evident that MTX inhibits T-cell TNF-α production [129, 130, 135]. These all findings suggest that MTX can diminish TNF-α produced by activated T cells showed its immunomodulatory action.

6.2 Effects of methotrexate on endothelial cells

T-cell migration from the intravascular space into the dermis is a crucial step in the pathogenesis of psoriasis, and this process is dependent on interactions between endothelial cells and T cells. Endothelial expression of appropriate adhesion ligands such as E-selectin and ICAM-1, are necessary for successful T-cell adhesion and migration [124, 139]. Studies have shown that MTX treatment firmly decreased the CLA, ICAM-1 and E-selectin expression in the endothelial cells [24]. Histologically, hypervascularity is noted in psoriatic skin, which contributes to the grossly observed erythema. When used at the high dosages necessary for chemotherapy, MTX is capable of inhibiting angiogenesis. MTX exerted its therapeutic effects in psoriasis by inhibiting angiogenesis.In 2003 Yamasaki et al. [140] found that MTX had an inhibitory effect on endothelial cell growth. Two years later, in 2005, Yazici et al. [25] employed immunohistochemistry on lesional skin biopsies to study the effects of MTX on angiogenesis, reporting a statistically significant decrease in the endothelial marker CD31 after treatment with MTX.

Dendritic cells (DCs) are considered as a key player in the pathogenesis of psoriasis. Interplay between DCs, T-cells and cytokine are main and complex in psoriasis. DCs are well recognized has antigen-presenting cells (APC) in the skin. Any modulation in APC interaction with other cells may significantly influence development of psoriatic lesions. Many studies has showed that MTX showed its immunomodulatory effect by suppressing APCs activity [141, 142]. Recently, the use of T-cell targeted therapy confirmed the critical role of lymphocytes [143]. On the other hand, the clinical phenotype observed in psoriasis is mostly accounted for by several alterations in epidermal KCs [144].

Advertisement

7. Action of methotrexate on activated KC-immunocyte cycle in psoriasis

The epidermis is a multilayered epithelium consisting mainly of proliferating and differentiated, postmitotic KCs [145]. The latter derive from transit amplifying cells originating from stem cells that represent a restricted number of basal KCs [146, 147]. The proliferation and differentiation of epidermal KCs is regulated by a multitude of signaling cascades and transcription factors including Wnt/b-catenin [148], growth factors of the EGF and FGF family [149], TGF-β, members of the NFkB family [150], and c-Myc [151].

7.1 Activation and deactivation signals of KAC

In psoriasis, KCs undergo activation pathway. This activation process is governed by growth factors and cytokines, such IL-1, TNF-α, IFN-γ and TGF-β [78].

7.1.1 Interleukin-1

IL-1 is likely to be an important mediator in the initiation and maintenance of psoriatic plaques and may represent an attractive therapeutic target [152].

7.1.1.1 Interleukin-1 alpha

Our studies in IL-l α levels and its action in psoriasis had clearly showed that IL-l α level is reduced in psoriatic skin as well as in plasma [153], which was further supported by several studies [154, 155, 156, 157]. Mechanism behind this reduction is mainly depend on the upstream level of nerve growth factor, a IL-1 α down-regulator [158, 159] in psoriasis.

The second messenger cyclic adenosine monophosphate (cAMP) has been regarded as a regulator for cell growth and proliferation [160]. The action of IL-l α is mainly depends on its phosphorylation by cyclic cAMP-dependent protein kinase. Thus, phosphorylation converts it into active form and enhanced its susceptibility to tryptic digestion, which may allow its release into the extracellular milieu [161]. Reduced cAMP levels has been reported in psoriasis [162]. This also could lead to reduced IL-l α levels in lesional skin biopsies.

Studies showed that MTX significantly increased IL-l α levels in plasma and skin biopsies of psoriasis patients. This may due to increase in the levels of cAMP by MTX through adenosine release [153, 163] and that led to an increase in IL-1 α level.

7.1.1.2 Interleukin-1 beta and caspase-1

Many studies have reported the importance of IL-1 β and caspase-1 (IL-1 β converting enzyme) in pathogenesis of psoriasis [153, 156, 157, 164]. Increased expression of IL-1 β in psoriatic epidermal cells, related to the activated KC-immunocyte in psoriasis. Psoriatic plaques express increased IL-1 β mRNA relative to non-lesional skin [161]. IL-1 β has been shown to induce the expression of adhesion molecules on various cell types and contributes to inflammatory responses. Activation of ERK, JNK, AP-1, and NFκB are leads to IL-1 β-induced ICAM-1 expression and leukocyte adhesion [165], thereby increase in ICAM-1 cause hyperproliferation of KC in psoriasis.

Normal KCs do not contain a biologically active form of caspases-1 [166], whereas in psoriatic epidermis, caspase-5 act as an upstream activator of caspase-1 [167]. Caspase-5 mRNA is induced by IFN-γ in vitro in both KCs and PBMCs and that this induction is most likely mediated through the NFκB pathway [168].

Mizutani et al. [169] found that IL-1 β levels in PBMC of psoriatic patients were decreased after 2 weeks of MTX treatment, due to suppression of KC paracrine system by MTX. Also, MTX effectively reduced IL-1 β levels in plasma and skin biopsies of psoriasis patients [153]. There are two possible mechanism of MTX decrease IL-1 β is elucidated above. One is direct mechanism and another one is indirect mechanism. The direct mechanism is as follows: MTX treatment inhibits the enzyme5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase (ATIC), which leads to accumulation of the substrate AICAR. The increased AICAR inhibit the enzymes, AMP deaminase and adenosine deaminase, which are essential for the catabolism of AMP and adenosine. This inhibitory action eventually increased adenosine in circulation. Thus, increased extracellular adenosine firmly increased cAMP in skin which finally inhibits the production of proinflammatory cytokine IL-1 β [163].

The indirect mechanism of MTX on reducing IL-1 β levels in psoriasis is mainly depend on reduction of infiltration of lymphocytes and monocytes in psoriatic dermis [162, 170]. MTX also decreased g the circulating and systemic levels of IL-1 β, by blocking the binding of IL-1 β to its respective receptor on monocytes, lymphocytes and granulocytes [171]. Studies have shown that IL-l α down-regulates IL-1 β via prostaglandin E2 synthesis [172]. Therefore increase in IL-1 α levels by MTX, definitely downregulates IL-1 β by above said mechanisms. Another mechanisms of MTX reduced IL-1 β, its action on caspase-1 expression in lesional skin biopsy. This may due to IFN-γ reduction by MTX [173, 174], which leads to decrease in caspase-5 expression that causes reduction in caspase-1 expression.

7.1.2 Tumor necrosis factor alpha

TNF-α is a pivotal proinflammatory cytokine of the innate immune response and a key for skin inflammation [175]. TNF-α induce the expression of adhesion molecule ICAM-1 on KC through the mediation of p55 and ICAM-1 induces the infiltration of MNCs in the dermis, which promotes the development and progression of psoriasis vulgaris [176]. Thereby it causes hyperproliferation in psoriasis.

Many studies have shown that plasma concentration of TNF-α was significantly higher in psoriatic patients compared to the control group [35, 177, 178]. Johansen [179] showed increased TNF-α protein expression, but similar TNF-α mRNA levels, in lesional compared with nonlesional psoriatic skin, this results showed that TNF-α is regulated posttranscriptionally. Increased activation of MAPK-activated protein kinase 2 (MK2) is responsible for the elevated and posttranscriptionally regulated TNF-α protein expression in psoriatic skin. IL-1 β also amplified TNF-α protein expression by causing activation of p38 MAPK and MK2.

Action of MTX to overwhelm TNF activity is by suppressing TNF-induced nuclear factor-κB activation in vitro, in part related to a reduction in the degradation and inactivation of an inhibitor of this factor, IκBα, and probably related to the release of adenosine [180]. Gerards et al. [130] showed that Adenosine or adenosine receptor agonists inhibit production of TNF-α. MTX reduces TNF-α level and also reduces the adhesion molecules like E-selctin and ICAM-1, thereby it reduces TNF-α induced hyperproliferation in psoriasis. Decrease in TNF-α by MTX shows its anti-proliferative and anti-inflammatory effects.

7.1.3 Interferon-γ

IFN-γ is believed to be an important mediator in psoriasis. Accumulated evidence from both in vivo and in vitro studies show that IFN-γ is a critical element in the induction of KC hyperproliferation in psoriasis [172, 181]. Abdallah et al. [182] showed that serum IFN-γ is a psoriasis severity and prognostic marker.

Some authors showed that serum IFN-γ levels in psoriatic patients were 15-fold and in blister fluid 17-fold higher than those in the control group. They correlated with the clinical severity of psoriasis expressed as PASI score [183, 184, 185, 186]. In lesional skin, expression of IFN-γ is induced by some cytokines like IL-12, IL-18, and IL-23 are termed as IFN-γ stimulator [187]. Apart from above said stimulator, IL-7 also indirectly increased IFN-γ levels in both the psoriatic skin and serum of psoriatic patients. Inflammatory mediator IL-7 interact with both IL-2 and IL-12, which indirectly induced the synthesis of IFN-γ in psoriatic skin [188].

MTX treatment effectively reduced IFN-γ levels in serum and skin biopsy of psoriatic patients. This may due to the action of MTX on reducing serum IL-7 levels and is found to correlate with disease in RA patients [20]. Similar to this result, studies have shown that MTX decreases the expression of IFN-γ in RA patients [135, 136]. Adenosine or adenosine receptor agonists inhibit production of IFN-γ [130]. In 1990, Nesher and Moore [189] proposed that MTX might inhibit polyamine synthesis in monocytes, thereby polyamines failed to restore IFN-γ production.

As we discussed earlier in this chapter about the MTX action on T cell cytolysis, the same concept is applicable in this mechanism. The primary source of activated T cell is IFN-γ [127]. MTX induce the cytolysis of activated T-cell which directly reduced the IFN-γ in psoriasis. Invitro studies showed that inhibition of NFκB led to the complete blockade of extracellular IL-17A, IL-22, IFN-γ, and TNF-α production in CD4+ cells [190]. It is well known that MTX directly inhibit NFκB [191]. Thus, MTX declines IFN-γ levels by inhibiting NFκB in psoriatic skin which control the hyperproliferation of KCs.

7.1.4 Transforming growth factor-β1

In the skin, TGF-β has been found to inhibit the growth of KCs but stimulate the growth of fibroblasts [192]. Plasma TGF-β1 is considered as a biomarker of psoriasis activity and treatment efficacy [193].

Gene and protein expression of TGF-β1 in lesional skin biopsies was found to be reduced compared to nonlesional skin biopsies [35, 97, 193]. Reduction in TGF-β1 in psoriasis may be due to increase in activated Akt levels. Activated Akt inhibit the phosphorylation of Smad2/3, is essential for TGF-β1 production [194, 195]. IFN-γ and NF-kappaB induces the expression of Smad7, an antagonistic Smad, which prevents the interaction of Smad3 with the TGF-β receptor [196, 197] leading to TGF-β1 production. Other factors that can suppress TGF-β1 productions are Th1 cytokines like TNF-α, IL-1, IL-6 [196]. All these factors could have lead to reduction in TGF-β1 levels in psoriasis. Reduction in TGF-β1 leads to increase in proliferation of KCs in psoriasis. Adhesiveness of T lymphocytes to dermal microvascular endothelial cells can be blocked by TGF-β1, so reduction of its expression and function may contribute to lymphocyte infiltration into psoriatic plaques [198].

MTX treatment causes overexpression of protein and mRNA level of TGF-β1 in lesional skin biopsy. Possible mechanism of MTX is accompanied by decreasing Ras methylation in psoriasis. This hypomethylation is accompanied by a mislocalization of Ras to the cytosol and a 4-fold decrease in the activation of Akt [199]. Decrease in Akt may lead to Smad 3 activation, which in turn increase TGF-β1 expression. Also MTX reduces Th1 cytokines like IL-1, IFN-γ, TNF-α, IL-6 [111, 135, 136, 188] which may lead to increase in TGF-β1 expression. Increase in TGF-β1 expression induced inhibition of KCs and is characterized by reversible retention of proliferation, or stopping of cell division cycle in the G1 phase [96]. Moreover, TGF-β causes transcriptional induction of K5 and K14 keratin genes [81]. Thus, TGF-β promotes the basal cell phenotype in stratified epithelia such as the epidermis, and that the effects of TGF-β are not anti-proliferative, but merely anti-hyperproliferative [78].

7.2 Keratin changes in psoriatic skin after MTX action

The pathogenesis of psoriasis is mainly depend on KCs and immune cells interaction. These interaction causes changes in the epidermal layers which destined the healthy skin to lesional psoriatic skin is critical one. Due to the epidermal change, acanthosis is caused by disruption in the transition of KC to anucleate corneocytes. This disrupts transition leads to hyperproliferation and immatured differentiation of KC in psoriasis. Thus, an alternate or regenerative pathway is activated by abnormal immune response along with abnormal keratins in psoriatic epidermis [200].

7.2.1 Keratin 14

K14 is an important IF expressed in the basal KCs which decide the fate of epidermis by protecting it from any mechanical or chemical disturbances. Normallay, basal layer contains epidermal stem cells and TA cells that proliferate into KCs. The epidermal stem cells and TA cells expressed keratin K5/K14 which destined the cells to proliferate into KC [32].

In psoriatic skin biopsy, K14 expression was considerably higher than in normal epidermis [33, 34, 35]. However, MTX reduced the expressions of K14 in lesional skin biopies by increasing Phosphorylated form of p53 expression [201]. Studies have shown that p53 enhance the notch signaling [202], which leads to modulation and normalization of K14 expression to K10 differentiation marker. Thus, decrease in K14 expression leads to reduction in cell proliferation, decrease in phospho-Akt levels, increase in activated Notch1 levels, and increase in levels of KC differentiation markers [39] and also increased in p21 and p27 levels, which are known to be direct targets of Notch1 signals. Activation of p21 and p27 leads to cell cycle arrest [191, 203] which leads to inhibition of hyperproliferating KCs in psoriasis.

7.2.2 Keratin 10

When mitotically active basal epidermal KCs withdrawn from the cell cycle are committed to terminal differentiation, they switch off K14 expression and induce the expression of K10 [27]. K10 are the major structural proteins of the epidermis and belong to the large family of IF proteins [29, 41, 42]. K10 is also involved in the control of cell proliferation [43]. Decreased K10 protein and gene expression were observed in lesional psoriatic skin biospies [33, 34, 35]. Subsequently, hyperproliferation of KCs in psoriasis is mainly depend on the expression of K10. The possible mechanism is as follows, decrease in differentiation marker K10 leads to increased phosphorylation of Rb protein, which leads to increased epidermal cell cycle proteins, Cyclin D and E. The increased epidermal cell cycle proteins activate the phosphorylation of antiapoptotic protein Akt. Thus activation of Akt by decreased K10 leads to hyperproliferation of KCs in psoriasis [52].

E-cadherin is specific markers expressed in the endothelial cells are important for differentiation process. MTX promptly increased E-cadherin expression [201] which deliberately causes extracellular calcium concentration-dependent KC differentiation by increasing K10 in psoriatic skin. IL-22 which is considered to have an inhibitory effect on the expression of K10 [205], is reduced by MTX in psoriasis patients [206]. Also MTX increase c-fos expression which activate AP-1 transcription factor regulates K10 expression [207]. Thus MTX action on K10 expression proves the controlled differentiation in psoriatic KC.

The main therapeutic action of MTX is inhibiting DNA methylation by interfering folate metabolism [207]. Studies have shown that inhibition of DNA methylation directly increased transcription of K10 gene [208]. This shows the therapeutic action of MTX on psoriatic skin.

7.2.3 Keratin 16

Keratin 16 expressions, reduces the fraction of cells in G1 while increasing that in S phase [53]. Many authors also shown an upregulation of K6 and K16 [33, 34, 35] in psoriatic epidermis. Action of MTX on reducing Keratin 16 protein and mRNA expression in lesional skin biopsies is interesting. Ras signaling is also induces K16 in psoriasis. The action of MTX in decreasing K16 level is depends on the enzyme isoprenylcysteine carboxyl methyltransferase (Icmt). Inhibition of Icmt by MTX, indicates the link between antifolates and Ras. Icmt inhibition leads to decrease in carboxyl methylation of Ras [199], which induces EGF. Induced EGF directly decreased K16 expression. Overall mechanism indicates the action of MTX on K16 reduction. Apart from this, MTX also inhibits the IL-1, TNF-α levels [135, 153, 169], which also might leads to decrease in K16 expression in psoriasis. All these leads to inhibition of cell proliferation in psoriasis. Decrease in Keratin 16 by MTX indicates the strong anti-proliferative effect of MTX.

7.2.4 Keratin K17

Keratin K17, the myoepithelial keratin, which is not expressed in normal skin except for hair follicles, sweat and sebaceous glands and basal cells of the interfollicular epidermis in the scalp, is over-expressed in psoriatic epidermis [209, 210]. Keratin 17 is considered as a therapeutic target and marker of anti-psoriatic therapies used for the treatment of psoriasis [63, 64].

MTX substantially reduced abnormal K17 protein and gene expression in psoriasis by reducing circulatory inflammatory mediators like IL-22 [205] and IL-6 levels [111] in psoriasis patients. Many studies in Rheumatoid Arthritis patients showed that the production of IL-17 at the mRNA level and IFN-γ were reduced after MTX treatment [35, 135, 211]. Altogether MTX reduces K17 expression by decreasing IFN-γ inducers. Decrease in Keratin 17 by MTX indicates its therapeutic efficacy which helps to maintain the normal phenotype in KC.

Advertisement

8. Conclusion

When we put all these data and review together, we attain at a consistent outline for the action of keratins, growth factors and cytokines in psoriasis. MTX has been used as a effective agent in the treatment of psoriasis from the decades of 1960s. But still its mechanisms of clearing psoriatic remains ill-defined. In conclusion, we strongly thought that MTX inhibits the hyperproliferation of KCs by decreasing the levels of IL-1 and caspases-1 (activating signals), TNF-α, IFN-γ and by increasing deactivation signal through various effective pathway. Also it efficiently regulate abnormal keratins by upregulating K10 and downregulating K14, K16 and K17, thereby maintaining the normal phenotype in KC (Figure 7). Mutation in keratins filaments were also observed in psoriasis [31]. Thereby, understanding keratin functions and related regulatory mechanisms will help to design new therapeutic interventions for keratin-related skin diseases. We strongly concluded that MTX roles on controlling the KC-immunocyte cycle by activating important keratins and deactivating abnormal kertains showed its well-organized therapeutic effect in psoriasis patients.

Figure 7.

Effect of MTX on KC activation cycle. MTX normalizing or reversing the phenotype of psoriatic KC but altering the inflammatory mediators as well as keratin proteins in KCs. Symbol ↑ indicates increase in psoriasis; symbol ↓ indicates decrease in psoriasis.

Advertisement

Acknowledgments

We thank all patients who have assisted for all our studies.

Advertisement

Conflict of interests

The authors declare that they have no conflict of interests.

Abbreviation

KACkeratinocyte activation cycle
KCkeratinocyte
ILinterleukin
MTXmethotrexate
TNFtumor necrosis factor
IFNinterferon
TGFtransforming growth factor
K14keratin 14
K10keratin 10
K16Keratin 16
K17keratin 17

References

  1. 1. Van Onselen J. Psoriasis in general practice. Nursing Standard. 1998;12:32-33
  2. 2. Ortonne JP. Etiology and pathogenesis of psoriasis. The British Journal of Dermatology. 1996;135(49S):1-5
  3. 3. 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):369-375
  4. 4. Nickoloff BJ, Nestle FO. Recent insights into the immunopathogenesis of psoriasis provide new therapeutic opportunities. Journal of Clinical Investigation. 2004;113(12):1664-1675
  5. 5. Hussein MR, Al-Badaiwy ZH, Guirguis MN. Analysis of p53 and bcl-2 protein expression in the non-tumorigenic, pre-tumorigenic and tumorigenic keratinocyte hyper proliferative lesions. Journal of Cutaneous Pathology. 2004;31(10):643-651
  6. 6. Gottlieb AB. Infliximab for psoriasis. Journal of the American Academy of Dermatology. 2003;49(Suppl. 2):S112-S117
  7. 7. Carlen LM et al. Proteome analysis of skin distinguishes acute guttate from chronic plaque psoriasis. Journal of Investigative Dermatology. 2005;124(1):63-69
  8. 8. Zhou X et al. Novel mechanisms of T-cell and dendritic cell activation revealed by profiling of psoriasis on the 63,100-element oligonucleotide array. Physiological Genomics. 2003;13(1):69-78
  9. 9. Waikel RL, Kawachi Y, Waikel PA. Deregulated expression of c-Myc depletes epidermal stem cells. Nature Genetics. 2001;28:165-168
  10. 10. Zibert JR, Lovendorf MB, Litman T, et al. MicroRNAs and potential target interactions in psoriasis. Journal of Dermatological Science. 2010;58:177-185
  11. 11. Pasquinelli AE. NON-CODING RNA MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship. Nature Reviews. Genetics. 2012;13:271-282
  12. 12. Selbach M, Schwanhausser B, Thierfelder N, et al. Widespread changes in protein synthesis induced by microRNAs. Nature. 2008;455:58-63
  13. 13. Coronnello C, Benos PV. ComiR: Combinatorial microRNA target prediction tool. Nucleic Acids Research. 2013;41:W159-W164
  14. 14. Lindsay MA. microRNAs and the immune response. Trends in Immunology. 2008;29:343-351
  15. 15. Baumjohann D, Ansel KM. MicroRNA-mediated regulation of T helper cell differentiation and plasticity. Nature Reviews Immunology. 2013;13:666-678
  16. 16. Ichihara A, Jinnin M, Yamane K, et al. microRNA-mediated keratinocyte hyperproliferation in psoriasis vulgaris. The British Journal of Dermatology. 2011;165:1003-1010
  17. 17. de Cid R, Riveira-Munoz E, Zeeuwen PL, et al. Deletion of the late cornified envelope LCE3B and LCE3C genes as a susceptibility factor for psoriasis. Nature Genetics. 2009;41(2):211-215
  18. 18. Zhang XJ, Huang W, Yang S, et al. Psoriasis genome-wide association study identifies susceptibility variants within LCE gene cluster at 1q21. Nature Genetics. 2009;41(2):205-210
  19. 19. Lane EB, McLean WH. Keratins and skin disorders. Journal of Pathology. 2004;204(4):355-366
  20. 20. Herman S, Zurgil N, Langevitz P, et al. Methotrexate selectively modulates TH1/TH2 balance in active rheumatoid arthritis patients. Clinical and Experimental Rheumatology. 2008;26:317-323
  21. 21. Bonish B, Jullien D, Dutronc Y, et al. Overexpression of CD1d by keratinocytes in psoriasis and CD1d-dependent IFN-gamma production by NK-T cells. Journal of Immunology. 2000;165(7):4076-4085
  22. 22. Sigmundsdóttir H, Gudjónsson JE, Jónsdóttir I, Lúdvíksson BR, Valdimarsson H. The frequency of CLA+ CD8+ T cells in the blood of psoriasis patients correlates closely with the severity of their disease. Clinical and Experimental Immunology. 2001;126(2):365-369
  23. 23. Saleh A, Abuhilal M, Cheung B. Methotrexate in psoriasis: From A to Z. Journal of the Turkish Academy of Dermatology. 2010;4:04101r
  24. 24. Dahlman-Ghozlan K, Ortonne JP, Heilborn JD, Stephansson E. Altered tissue expression pattern of cell adhesion molecules, ICAM-1, E-selectin and VCAM-1, in bullous pemphigoid during methotrexate therapy. Experimental Dermatology. 2004;13(2):65-69
  25. 25. Yazici AC, Tursen U, Apa DD, et al. The changes in expression of ICAM-3, Ki-67, PCNA, and CD31 in psoriatic lesions before and after methotrexate treatment. Archives of Dermatological Research. 2005;297(6):249-255
  26. 26. Johnston A, Gudjonsson JE, Sigmundsdottir H, et al. The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clinical Immunology. 2005;114(2):154-163
  27. 27. Fuchs E, Green H. Changes in keratin gene expression during terminal differentiation of the keratinocyte. Cell. 1980;19:1033-1042
  28. 28. Moll R, Moll I, Franke WW. Differences of expression of cytokeratin polypeptides in various epithelial skin tumors. Archives of Dermatological Research. 1984;276(6):349-363
  29. 29. Hesse M, Magin TM, Weber K. Genes for intermediate filament proteins and the draft sequence of the human genome: Novel keratin genes and a surprisingly high number of pseudogenes related to keratin genes 8 and 18. Journal of Cell Science. 2001;114:2569-2575
  30. 30. Machesney M, Tidman N, Waseem A, et al. Activated keratinocytes in the epidermis of hypertrophic scars. The American Journal of Pathology. 1998;152(5):1133-1141
  31. 31. Elango T, Sun J, Zhu C, et al. Mutational analysis of epidermal and hyperproliferative type I keratins in mild and moderate psoriasis vulgaris patients: A possible role in the pathogenesis of psoriasis along with disease severity. Human Genomics. 2018;12(1):27
  32. 32. Coulombe PA, Kopan R, Fuchs E. Expression of keratin K14 in the epidermis and hair follicle: Insights into complex programs of differentiation. Journal of Cell Biology. 1989;109:2295-2312
  33. 33. Castelijns FA, Gerritsen MJ, van Vlijmen-Willems IM, et al. Proliferation is the main epidermal target in the treatment of psoriatic plaques with once daily application of tacalcitol ointment. Acta Dermato-Venereologica. 1999;79(2):111-114
  34. 34. Thewes M, Stadler R, Korge B, Mischke D. Normal psoriatic epidermis expression of hyperproliferation-associated keratins. Archives of Dermatological Research. 1991;283(7):465-471
  35. 35. Elango T, Thirupathi A, Subramanian S, Dayalan H, Gnanaraj P. Methotrexate normalized keratinocyte activation cycle by overturning abnormal keratins as well as deregulated inflammatory mediators in psoriatic patients. Clinica Chimica Acta. 2015;451(Pt B):329-337
  36. 36. Cai BH, Hsu PC, Hsin IL, et al. p53 acts as a co-repressor to regulate keratin 14 expression during epidermal cell. PLoS One. 2012;7(7):e41742
  37. 37. Xiaolian G, Elisabet N, Philip JC. Effect of narrow-band ultraviolet B phototherapy on p63 and microRNA (miR-21 and miR-125b) expression in psoriatic epidermis. Acta Dermato-Venereologica. 2011;91:392-397(6)
  38. 38. Thélu J, Rossio P, Favier B. Notch signaling is linked to epidermal cell differentiation level in basal cell carcinoma, psoriasis and wound healing. BMC Dermatology. 2002;2:7
  39. 39. Alam H, Sehgal L, Kundu ST, et al. Novel function of keratins 5 and 14 in proliferation and differentiation of stratified epithelial cells. Molecular Biology of the Cell. 2011;22(21):4068-4078
  40. 40. Truzzi F, Marconi A, Atzei P, et al. p75 neurotrophin receptor mediates apoptosis in transit-amplifying cells and its overexpression restores cell death in psoriatic keratinocytes. Cell Death and Differentiation. 2011;18(6):948-958
  41. 41. Herrmann H, Hesse M, Reichenzeller M, Aebi U, Magin TM. Functional complexity of intermediate filament cytoskeletons: From structure to assembly to gene ablation. International Review of Cytology. 2003;223:83-175
  42. 42. Hesse M, Zimek A, Weber K, Magin TM. Comprehensive analysis of keratin gene clusters in humans and rodents. European Journal of Cell Biology. 2004;83:19-26
  43. 43. Reichelt J, Furstenberger G, Magin TM. Loss of keratin 10 leads to mitogen-activated protein kinase (MAPK) activation, increased keratinocyte turnover, and decreased tumor formation in mice. Journal of Investigative Dermatology. 2004;123(5):973-981
  44. 44. Li ZX, Ji FP, Peng ZH, Wang K. Expression of E-cadherin, beta-catenin and cyclin D1 in epidermal skin lesions of patients with active psoriasis vulgaris. Nan Fang Yi Ke Da Xue Xue Bao. 2008;28(4):545-547
  45. 45. Boniface K, Bernard FX, Garcia M, et al. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. Journal of Immunology. 2005;174(6):3695-3702
  46. 46. Wolk K, Witte E, Wallace E, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: A potential role in psoriasis. European Journal of Immunology. 2006;36(5):1309-1323
  47. 47. Sa SM, Valdez PA, Wu J, et al. The effects of IL-20 subfamily cytokines on reconstituted human epidermis suggest potential roles in cutaneous innate defense and pathogenic adaptive immunity in psoriasis. Immunology. 2007;178:2229-2240
  48. 48. Nograles KE, Zaba LC, Guttman-Yassky E, et al. Th17 cytokines interleukin (IL)-17 and IL-22 modulate distinct inflammatory and keratinocyte-response pathways. The British Journal of Dermatology. 2008;159(5):1092-1102
  49. 49. Johansen C, Kragballe K, Henningsen J, et al. 1a,25-dihydroxyvitamin D3 stimulates activator protein 1 DNA-binding activity by a phosphoinositol 3-kinase/Ras/MEK/extracellular signal regulated kinase 1/2 and c-jun N-terminal kinase 1-dependent increase in cfos,fra1, and c-jun expression in human keratinocytes. Journal of Investigative Dermatology. 2003;120:561-570
  50. 50. Schmuth M, Elias PM, Hanley K, et al. The effect of LXR activators on AP-1 proteins in keratinocytes. Journal of Investigative Dermatology. 2004;123:41-48
  51. 51. Eckert RL, Crish JF, Efimova T, et al. Regulation of involucrin gene expression. Journal of Investigative Dermatology. 2004;123:13-22
  52. 52. Basset-Seguin N, Escot C, Moles JP, et al. C-fos and c-jun proto-oncogene expression is decreased in psoriasis: An in situ quantitative analysis. Journal of Investigative Dermatology. 1991;97:672-678
  53. 53. Paramio JM, Casanova ML, Segrelles C, et al. Modulation of cell proliferation by cytokeratins K10 and K16. Molecular and Cellular Biology. 1999;19(4):3086-3094
  54. 54. Komine M, Rao LS, Kaneko T, et al. Inflammatory vs. proliferative processes in the epidermis: Tumor necrosis factor alpha induces K6b keratin synthesis through a transcriptional complex containing NFkB and C/EBPb. Journal of Biological Chemistry. 2000;275:32077-32088
  55. 55. van de Kerkhof PCM, Jorn Bovenschen H, et al. In vivo effects of topical anti-psoriatic treatments on cutaneous inflammation, epidermal proliferation and keratinisation. Current Drug Therapy. 2007;2:21-26
  56. 56. Reichelt J, Magin TM. Hyperproliferation, induction of c-Myc and 14-3-3σ, but no cell fragility in keratin-10-null mice. Journal of Cell Science. 2002;115:2639-2650
  57. 57. Kupper TS. The activated keratinocyte: A model for inducible cytokine production by non-bone marrow-derived cells in cutaneous inflammatory and immune responses. Journal of Investigative Dermatology. 1990;94:146S-150S
  58. 58. Barker JN, MacDonald DM. Cutaneous lymphocyte trafficking in the inflammatory dermatoses. British Journal of Dermatology. 1992;126:211-215
  59. 59. Pittelkow MR, Lindquist PB, Abraham RT, et al. Induction of transforming growth factor-alpha expression in human keratinocytes by phorbol ester. Journal of Biological Chemistry. 1989;264:5164-5171
  60. 60. Jiang CK, Magnaldo T, Ohtsuki M, et al. Epidermal growth factor and transforming growth factor alpha specifically induce the activation- and hyperproliferation associated keratins 6 and 16. Proceedings of the National Academy of Sciences. 1993;90:6786-6790
  61. 61. Komine M, Rao LS, Freedberg IM, et al. Interleukin-1 induces transcription of keratin K6 in human epidermal keratinocytes. Journal of Investigative Dermatology. 2001;116:330-338
  62. 62. Wang YN, Chang WC. E Induction of disease-associated keratin 16 gene expression by epidermal growth factor is regulated through cooperation of transcription factors Sp1 and c-Jun. Journal of Biological Chemistry. 2003;278(46):45848-45857
  63. 63. de Jong EMGJ, van Vlijmen IMMJ, et al. Keratin 17: A useful marker in anti-psoriatic therapies. Archives of Dermatological Research. 1991;283(7):480-482
  64. 64. Vogel U, Denecke B, Sergey M, et al. Transcriptional activation of psoriasis-associated cytokeratin K17 by interferon-γ Analysis of γ-interferon activation sites. European Journal of Biochemistry. 1995;227:143-149
  65. 65. de Jong EMG, Ferrier CM, de Zwart A, et al. Effects of topical treatment with budesonide on parameters for epidermal proliferation, keratinization and inflammation in psoriasis. Journal of Dermatological Science. 1995;9:185-194
  66. 66. Baker BS, Swain AF, Fry L, Valdmarsson H. Epidermal T lymphocytes and HLA-DR expression in psoriasis. British Journal of Dermatology. 1984;110:555-564
  67. 67. Jiang CK, Flanagan S, Ohtsuki M, et al. Disease-activated transcription factor: Allergic reactions in human skin cause nuclear translocation of stat-91 and induce synthesis of keratin K17 molecular effects of T lymphocytes on the regulation of keratin gene expression. A cluster of five nuclear proteins regulates keratin gene transcription. Molecular and Cellular Biology. 1994;14(7):4759-4769
  68. 68. Zhang W, Dang E, Shi X, Jin L, Feng Z, et al. The pro-inflammatory cytokine IL-22 up-regulates Keratin 17 expression in keratinocytes via STAT3 and ERK1/2. PLoS One. 2012;7(7):e40797
  69. 69. Komine M, Freedberg IM, Blumenberg M. Regulation of epidermal expression of keratin K17 in inflammatory skin diseases. Journal of Investigative Dermatology. 1996;107(4):569-575
  70. 70. Hennings H, Michael D, Cheng C. Calcium regulation of growth and differentiation of mouse epidermal cells in culture. Cell. 1980;19(1):245-254
  71. 71. Fuchs E, Green H. Regulation of terminal differentiation of cultured human keratinocytes by vitamin A. Cell. 1981;25(3):617-625
  72. 72. Hosomi J, Hosoi J, Abe E, et al. Regulation of terminal differentiation of cultured mouse epidermal cells by 1 alpha,25-dihydroxyvitamin D3. Endocrinology. 1983;113(6):1950-1957
  73. 73. Parkinson EK, Pera MF, Emmerson A. “Differential effects of complete and second-stage tumor promoters in normal but not transformed human and mouse keratinocytes,” Carcinogenesis. 1984;5(8):1071-1077
  74. 74. Eckert RL, Crish JF, Robinson NA. The epidermal keratinocyte as a model for the study of gene regulation and cell differentiation. Physiological Reviews. 1997;77(2):397-424
  75. 75. Darmon M, Blumenberg ML. Molecular Biology of the Skin: The Keratinocyte. San Diego: Academic Press; 1993
  76. 76. Nickoloff BJ, Turka LA. Keratinocytes: Key immunocytes of the integument. The American Journal of Pathology. 1993;143(2):325-331
  77. 77. Tomic-Canic M, Komine M, Freedberg IM, et al. Epidermal signal transduction and transcription factor activation in activated keratinocytes. Journal of Dermatological Science. 1998;17(3):167-181
  78. 78. Freedberg IM, Tomic-Canic M, Komine M, et al. Keratins and the keratinocyte activation cycle. Journal of Investigative Dermatology. 2001;116(5):633-640
  79. 79. Ma S, Rao L, Freedberg IM, Blumenberg M. Transcriptional control of K5, K6, K14, and K17 keratin genes by AP-1 and NF-kappaB family members. Gene Expression. 1997;6(6):361-370
  80. 80. Jiang CK, Flanagan S, Ohtsuki M. Disease-activated transcription factor: Allergic reactions in human skin cause nuclear translocation of STAT-91 and induce synthesis of keratin K17. Molecular and Cellular Biology. 1994;14(7):4759-4769
  81. 81. Jiang CK, Tomic-Canic M, Lucas DJ, et al. TGF beta promotes the basal phenotype of epidermal keratinocytes: Transcriptional induction of K#5 and K#14 keratin genes. Growth Factors. 1995;12(2):87-97
  82. 82. Zepter K, Haffner A, De Soohoo LF, et al. Induction of biologically active IL-1 beta converting enzyme and mature IL-1 beta in human keratinocytes by inflammatory and immunologic stimuli. Journal of Immunology. 1997;159(12):6203-6208
  83. 83. Corsini E, Primavera A, Marinovich M, Galli CL. Selective induction of cell associated interleukin-1alpha in murine keratinocytes by chemical allergens. Toxicology. 1998;129(2-3):193-200
  84. 84. Murphy JE, Robert C, Kupper TS. Interleukin-1 and cutaneous inflammation: A crucial link between innate and acquired immunity. Journal of Investigative Dermatology. 2000;114(3):602-608
  85. 85. Romero LI, Zhang DN, Herron GS, Karasek MA. Interleukin-1 induces major phenotypic changes in human skin microvascular endothelial cells. Journal of Cellular Physiology. 1997;173(1):84-92
  86. 86. Wyble CW, Hynes KL, Kuchibhotla J, et al. TNF alpha and IL-1 upregulate membrane-bound and soluble E-selectin through a common pathway. Journal of Surgical Research. 1997;73(2):107-112
  87. 87. Santamaria Babi LF, Moser R, Perez Soler MT. Migration of skin-homing T cells across cytokine-activated human endothelial cell layers involves interaction of the cutaneous lymphocyte-associated antigen (CLA), the very late antigen-4 (VLA-4), and the lymphocyte function associated antigen-1 (LFA-1). Journal of Immunology. 1995;154(4):1543-1550
  88. 88. Maas-Szabowski N, Fusenig NE. Interleukin-1-induced growth factor expression in postmitotic and resting fibroblasts. Journal of Investigative Dermatology. 1996;107(6):849-855
  89. 89. Kolde G, Schulze-Osthoff K, Meyer H, et al. Immunohistological and immunoelectron microscopic identification of TNF alpha in normal human and murine epidermis. Archives of Dermatological Research. 1992;284(3):154-158
  90. 90. Griffiths TW, Griffiths CE, Voorhees JJ. Immunopathogenesis and immunotherapy of psoriasis. Dermatologic Clinics. 1995;13(4):739-749
  91. 91. Nickoloff BJ, Griffiths CE, Barker JN. The role of adhesion molecules, chemotactic factors, and cytokines in inflammatory and neoplastic skin disease. Journal of Investigative Dermatology. 1990;94(Suppl. 6):151S-157S
  92. 92. Kupper TS. Role of epidermal cytokines. In: Oppenheim JJ, Shevach EM, editors. Immunophysiology. The Role of Cells and Cytokines in Immunity and Inflammation. London and New York: Oxford University Press; 1990. pp. 285-305
  93. 93. Marinkovich MP, Lunstrum GP, Keene DR, et al. The dermal-epidermal junction of human skin contains a novel laminin variant. Journal of Cell Biology. 1992;119(3):695-703
  94. 94. Burgeson RE. Type VII collagen, anchoring fibrils, and epidermolysis bullosa. Journal of Investigative Dermatology. 1993;101(3):252-255
  95. 95. Troyanovsky SM, Leube RE, Franke WW. Characterization of the human gene encoding cytokeratin 17 and its expression pattern. European Journal of Cell Biology. 1992;59(1):127-137
  96. 96. van Ruissen F, van Erp PEJ, de Jongh GJ. Cell kinetic characterization of growth arrest in cultured human keratinocytes. Journal of Cell Science. 1994;107(8):2219-2228
  97. 97. Nockowski P, Szepietowski JC, Ziarkiewicz M. Serum concentrations of transforming growth factor beta 1 in patients with psoriasis vulgaris. Acta Dermatovenerologica Croatica. 2004;12(1):2-6
  98. 98. Ryan TJ, Vickers HR, Salem SN, et al. The treatment of psoriasis with folic acid antagonist. The British Journal of Dermatology. 1964;76:555-564
  99. 99. Boffa M, Chalmers R. Methotrexate for psoriasis. Clinical and Experimental Dermatology. 1996;21:399-408
  100. 100. Bright R. Methotrexate in the treatment of psoriasis. Cutis. 1999;64:332-334
  101. 101. Strober BE, Menon K. Folate supplementation during methotrexate therapy for patients with psoriasis. Journal of the American Academy of Dermatology. 2005;53:652-659
  102. 102. Gubner R, August S, Ginsberg V. Therapeutic suppression of tissue reactivity. II. Effect of aminopterin in rheumatoid arthritis and psoriasis. The American Journal of the Medical Sciences. 1951;221:176-182
  103. 103. Ward JR. Historical perspective on the use of methotrexate for the treatment of rheumatoid arthritis. Journal of Rheumatology. 1985;12:3-6
  104. 104. Cronstein BN. Molecular therapeutics: Methotrexate and its mechanism of action. Arthritis and Rheumatism. 1996;39:1951-1960
  105. 105. Lynch JP III, McCune J. Immunosuppressive and cytotoxic pharmacotherapy for pulmonary disorders. American Journal of Respiratory Critical Care Medicine. 1997;155:395-420
  106. 106. Kremer JM. Methotrexate update. Scandinavian Journal of Rheumatology. 1996;25(6):341-344
  107. 107. Goodman TA, Polisson RP. Methotrexate: Adverse reactions and major toxicities. Rheumatic Diseases Clinics of North America. 1994;20:513-528
  108. 108. Chan ES, Cronstein BN. Methotrexate—how does it really work? Nature Reviews Rheumatology. 2010;6:175-178
  109. 109. Griffiths CEM, Clark CM, Chalmers RJG, et al. A systematic review of treatments for severe psoriasis. Health Technology Assessment. 2000;4(40):1-125
  110. 110. Callis KP, Chadha A, Vaishnaw A, et al. Reduction of CD45RO+ effector T lymphocytes is not observed in the treatment of psoriasis with methotrexate. Journal of Investigative Dermatology. 2002;119:244 (abstract 220)
  111. 111. Elango T, Dayalan H, Subramanian S, Gnanaraj P, Malligarjunan H. Serum interleukin-6 levels in response to methotrexate treatment in psoriatic patients. Clinica Chimica Acta. 2012;413(19-20):1652-1656
  112. 112. Elango T, Dayalan H, Gnanaraj P, Malligarjunan H, Subramanian S. Impact of methotrexate on oxidative stress and apoptosis markers in psoriatic patients. Clinical and Experimental Medicine. 2014;14(4):431-437
  113. 113. Heydendael VM, Spuls PI, Opmeer BC, et al. Methotrexate versus cyclosporine in moderate-to-severe chronic plaque psoriasis. The New England Journal of Medicine. 2003;349(7):658-665
  114. 114. Sandhu K, Kaur I, Kumar B, et al. Efficacy and safety of cyclosporine versus methotrexate in severe psoriasis: A study from north India. Journal of Dermatology. 2003;30(6):458-463
  115. 115. Gawkrodger DJ. On behalf of the Therapy Guidelines and Audit Subcommittee of the British Association of Dermatologists. Current management of psoriasis. Journal of Dermatological Treatment. 1997;8:27-55
  116. 116. Said S, Jeffes EW, Weinstein GD. Methotrexate. Clinics in Dermatology. 1997;15(5):781-797
  117. 117. Roenigk HH Jr, Auerbach R, Maibach HI. Methotrexate in psoriasis: Revised guidelines. Journal of the American Academy of Dermatology. 1988;19(1 pt1):145-156
  118. 118. Roenigk HH Jr, Auerbach R, Maibach HI, et al. Methotrexate in psoriasis: Consensus conference. Journal of the American Academy of Dermatology. 1998;38(3):478-485
  119. 119. Dogra S, Krishna V, Kanwar AJ. Efficacy and safety of systemic methotrexate in two fixed doses of 10. Clinical and Experimental Dermatology. 2012;37(7):729-734
  120. 120. Yeo CM, Chong VH, Earnest A. Prevalence and risk factors of methotrexate hepatoxicity in Asian patients with psoriasis. World Journal of Hepatology. 2013;5(5):275-280
  121. 121. Zachariae H. Have methotrexate-induced liver fibrosis and cirrhosis become rare? A matter for reappraisal of routine liver biopsies. Dermatology. 2005;211(4):307-308
  122. 122. Kremer JM. Methotrexate treatment of rheumatic diseases: Can we do better? Arthritis and Rheumatism. 2008;58:3279-3282
  123. 123. Wessels JA, Huizinga TW, Guchelaar HJ. Recent insights in the pharmacological actions of methotrexate in the treatment of rheumatoid arthritis. Rheumatology (Oxford). 2008;47:249-255
  124. 124. Sigmundsdottir H, Johnston A, Gudjonsson JE, et al. Methotrexate markedly reduces the expression of vascular E-selectin, cutaneous lymphocyte-associated antigen and the numbers of mononuclear leucocytes in psoriatic skin. Experimental Dermatology. 2004;13(7):426-434
  125. 125. Jeffes EW III, McCullough JL, Pittelkow MR, et al. Methotrexate therapy of psoriasis: Differential sensitivity of proliferating lymphoid and epithelial cells to the cytotoxic and growth-inhibitory effects of methotrexate. Journal of Investigative Dermatology. 1995;104(2):183-188
  126. 126. Phillips DC, Woollard KJ, Griffiths HR. The anti-inflammatory actions of methotrexate are critically dependent upon the production of reactive oxygen species. British Journal of Pharmacology. 2003;138(3):501-511
  127. 127. Herman S, Zurgil N, Deutsch M. Low dose methotrexate induces apoptosis with reactive oxygen species involvement in T lymphocytic cell lines to a greater extent than in monocytic lines. Inflammation Research. 2005;54(7):273-280
  128. 128. Magari K, Miyata S, Nishigaki F. Differential effects of FK506 and methotrexate on inflammatory cytokine levels in rat adjuvant-induced arthritis. Journal of Rheumatology. 2003;30(10):2193-2200
  129. 129. Partsch G, Steiner G, Leeb BF. Highly increased levels of tumor necrosis factor-alpha and other proinflammatory cytokines in psoriatic arthritis synovial fluid. Journal of Rheumatology. 1997;24(3):518-523
  130. 130. Gerards AH, de Lathouder S, de Groot ER. Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis. Rheumatology (Oxford, England). 2003;42(10):1189-1196
  131. 131. Barsig J, Yam G, Lehner MD, Beume R. Methotrexate treatment suppresses local cytokine and chemokine production in rat adjuvant arthritis. Drugs under Experimental and Clinical Research. 2005;31(1):7-11
  132. 132. Seitz M, Loetscher P, Dewald B, et al. Methotrexate action in rheumatoid arthritis: Stimulation of cytokine inhibitor and inhibition of chemokine production by peripheral blood mononuclear cells. British Journal of Rheumatology. 1995;34(7):602-609
  133. 133. Partsch G, Wagner E, Leeb BF, et al. T cell derived cytokines in psoriatic arthritis synovial fluids. Annals of the Rheumatic Diseases. 1998;57(11):691-693
  134. 134. Neurath MF, Hildner K, Becker C et al. “Methotrexate specifically modulates cytokine production by T cells and macrophages in murine collagen-induced arthritis (CIA): A mechanism for methotrexate-mediated immunosuppression”. Clinical and Experimental Immunology. 1999;115(1):42-55
  135. 135. Hildner K, Finotto S, Becker C, et al. Tumor necrosis factor (TNF) production by T cell receptor primed T lymphocytes is a target for low dose methotrexate in rheumatoid arthritis. Clinical and Experimental Immunology. 1999;118(1):137-146
  136. 136. Constantin A, Loubet-Lescoulie P, Lambert N, et al. Antiinflammatory and immunoregulatory action of methotrexate in the treatment of rheumatoid arthritis: Evidence of increased interleukin-4 and interleukin-10 gene expression demonstrated in vitro by competitive reverse transcriptase polymerase chain reaction. Arthritis and Rheumatism. 1998;41(1):48-57
  137. 137. Dolhain RJ, Tak PP, Dijkmans BA, et al. Methotrexate reduces inflammatory cell numbers, expression of monokines and of adhesion molecules in synovial tissue of patients with rheumatoid arthritis. British Journal of Rheumatology. 1998;37(5):502-508
  138. 138. Kraan MC, Reece RJ, Barg EC, et al. Modulation of inflammation and metalloproteinase expression in synovial tissue by leflunomide and methotrexate in patients with active rheumatoid arthritis. Findings in a prospective, randomized, double-blind, parallel design clinical trial in thirty-nine patients at two centers. Arthritis and Rheumatism. 2000;43(8):1820-1830
  139. 139. Bouma MG, van den Wildenberg FA, Buurman WA. Adenosine inhibits cytokine release and expression of adhesion molecules by activated human endothelial cells. The American Journal of Physiology. 1996;270(2Pt1):C522-C529
  140. 140. Yamasaki E, Soma Y, Kawa Y, Mizoguchi M. Methotrexate inhibits proliferation and regulation of the expression of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 by cultured human umbilical vein endothelial cells. The British Journal of Dermatology. 2003;149(1):30-38
  141. 141. Morhenn VB, Orenberg EK, Kaplan J, et al. Inhibition of a Langerhans cell mediated immune response by treatment modalities useful in psoriasis. Journal of Investigative Dermatology. 1983;81(1):23-27
  142. 142. Liu HN, Wong CK. In vitro immunosuppressive effects of methotrexate and azathioprine on Langerhans cells. Archives of Dermatological Research. 1997;289(2):94-97
  143. 143. Griffiths CE. T-cell-targeted biologicals for psoriasis. Current Drug Targets. Inflammation and Allergy. 2004;3(2):157-161
  144. 144. Wrone-Smith T, Mitra RS, Thompson CB, et al. Keratinocytes derived from psoriatic plaques are resistant to apoptosis compared with normal skin. The American Journal of Pathology. 1997;151(5):1321-1329
  145. 145. Watt FM, Hogan BL. Out of Eden: Stem cells and their niches. Science. 2000;287:1427-1430
  146. 146. Taylor G, Lehrer MS, Jensen PJ, et al. Involvement of follicular stem cells in forming not only the follicle but also the epidermis. Cell. 2000;102(4):451-461
  147. 147. Oshima H, Rochat A, Kedzia C, et al. Morphogenesis and renewal of hair follicles from adult multipotent stem cells. Cell. 2001;104(2):233-245
  148. 148. Huelsken J, Vogel R, Erdmann B, et al. β-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell. 2001;105(4):533-545
  149. 149. Werner S, Smola H. Paracrine regulation of keratinocyte proliferation and differentiation. Trends in Cell Biology. 2001;11(4):143-146
  150. 150. Kaufman CK, Fuchs E. It’s got you covered. NF-kappaB in the epidermis. Journal of Cell Biology. 2000;149(5):999-1004
  151. 151. Arnold I, Watt FM. c-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny. Current Biology. 2001;11(8):558-568
  152. 152. Mee JB, Cork M, di Giovineb FS, et al. Interleukin-1: A key inflammatory mediator in psoriasis? Cytokine. 2006;33(2):72-78
  153. 153. Tamilselvi E, Haripriya D, Hemamalini M, Pushpa G, Swapna S. Association of disease severity with IL-1 levels in methotrexate-treated psoriasis patients. Scandinavian Journal of Immunology. 2013;78(6):545-553
  154. 154. Takematsu H, Terui K, Ohkohchi H, et al. Interleukin-1-like activity in horny layer extracts: Decreased activity in scale extracts of psoriasis and sterile dermatoses. Dermatologica. 1986;172:236
  155. 155. Takematsu H, Tagam H. Lack of correlation interleukin 1 levels between interleukin 6 in psoriatic lesional skin. Tohoku Journal of Experimental Medicine. 1994;172:243-252
  156. 156. Yoshinaga Y, Higaki M, Terajima S. Detection of inflammatory cytokines in psoriatic skin. Archives of Dermatological Research. 1995;287:158-164
  157. 157. Debets R, Hegmans JP, Croughs P, et al. The IL-1 system in psoriatic skin. IL-1 antagonist sphere of influence in lesional psoriatic epidermis. Journal of Immunology. 1997;158(2955):± 63
  158. 158. Fantini F, Magnoni C, Bracci-Laudiero L, Pincelli C. Nerve growth factor is increased in psoriatic skin. Journal of Investigative Dermatology. 1995;105:854-855
  159. 159. Pincelli C. Nerve growth factor and keratinocytes: A role in psoriasis. European Journal of Dermatology. 2000;10(2):85-90
  160. 160. Ockenfels HM, Nussbaum G, Schneck B, et al. The effect of cyclosporin A and FK 506 on the cAMP content in psoriatic keratinocytes. Skin Pharmacology and Physiology. 1996;9(4):281-288
  161. 161. Kobayashi Y, Appella E, Yamada M, et al. Phosphorylation of Intracellular Precursors of Human IL-1. Journal of lmmunology. 1988;14:2279
  162. 162. Barrera P, Boerbooms AM, Demacker PN, et al. Circulating concentrations and production of cytokines and soluble receptors in rheumatoid arthritis patients: Effects of a single dose Methotrexate. British Journal of Rheumatology. 1994;33:1017-1024
  163. 163. Stamp LK, Hazlett J, Roberts RL, et al. Adenosine receptor expression in rheumatoid synovium: A basis for methotrexate action. Arthritis Research & Therapy. 2012;14:138
  164. 164. Debets R, Hegmans JP, Troost RJ, et al. Enhanced production of biologically active interleukin-1a and interleukin-1b by psoriatic epidermal cells ex vivo: Evidence of increased cytosolic interleukin-1b levels and facilitated interleukin-1 release. European Journal of Immunology. 1995;25:1624-1630
  165. 165. Yang CM, Luo SF, Hsieh HL, et al. Interleukin-1beta induces ICAM-1 expression enhancing leukocyte adhesion in human rheumatoid arthritis synovial fibroblasts: Involvement of ERK, JNK, AP-1, and NF-kappaB. Journal of Cellular Physiology. 2010;224(2):516-526
  166. 166. Pietrzak AT, Zalewska A, Chodorowska G, et al. Invited critical review: Cytokines and anticytokines in psoriasis. Clinica Chimica Acta. 2008;394:7-21
  167. 167. Martinon F, Tschopp J. Inflammatory caspases and inflammasomes: Master switches of inflammation. Cell Death and Differentiation. 2007;14:10-22
  168. 168. Salskov-Iversen ML, Johansen C, Kragballe K. Caspase-5 expression is upregulated in lesional psoriatic skin. Journal of Investigative Dermatology. 2011;131(3):670-676
  169. 169. Mizutani H, Ohmoto Y, Mizutani T, et al. Role of increased production of monocytes TNF-alpha, IL-1beta and IL-6 in psoriasis: Relation to focal infection, disease activity and responses to treatments. Journal of Dermatological Science. 1997;14(2):145-153
  170. 170. Chang DM, Weinblatt ME, Schur PH. The effects of methotrexate on interleukin 1 in patients with rheumatoid arthritis. Journal of Rheumatology. 1992;19:1678-1682
  171. 171. Brody M, Böhm I, Bauer R. Mechanism of action of methotrexate: Experimental evidence that methotrexate blocks the binding of interleukin 1 beta to the interleukin 1 receptor on target cells. European journal of clinical chemistry. 1993;31(10):667-674
  172. 172. Beta-Csorgo Z, Hammerberg C, Voorhees JJ, Cooper KD. Intralesional T-lymphocyte activation as a mediator of psoriatic hyperplasia. Journal of Investigative Dermatology. 1995;105:89S-894S
  173. 173. Dinarello CA, Ikejima T, Warner SJ, et al. Interleukin 1 induces interleukin 1. I. Induction of circulating interleukin 1 in rabbits in vivo and in human mononuclear cells in vitro. Journal of Immunology. 1987;139:1902
  174. 174. Thirupathi A, Elango T, Subramanian S, Gnanaraj P. Methotrexate regulates Th-1 response by suppressing caspase-1 and cytokines in psoriasis patients. Clinica Chimica Acta. 2016;453:164-169
  175. 175. Bradley JR. TNF-mediated inflammatory disease. Journal of Pathology. 2008;214:149-160
  176. 176. Shifa Z, Junping L, Yakun W, et al. Expressions of TNF receptors, ICAM-1 and LFA-1 in lesional skin of psoriasis vulgaris. Chinese Journal of Dermatology. 1998;31(2):84-86
  177. 177. Mussi A, Bonifati C, Carducci M, et al. Serum TNF-alpha levels correlate with disease severity and are reduced by effective therapy in plaque-type psoriasis. Journal of Biological Regulators and Homeostatic Agents. 1997;11(3):115-118
  178. 178. Chodorowska G. Plasma concentrations of IFN-gamma and TNF-alpha in psoriatic patients before and after local treatment with dithranol ointment. Journal of the European Academy of Dermatology and Venereology. 1998;10(2):147-151
  179. 179. Johansen C, Funding AT, Otkjaer K, et al. Protein expression of TNF-alpha in psoriatic skin is regulated at a posttranscriptional level by MAPK-activated protein kinase 2. Journal of Immunology. 2006;176(3):1431-1438
  180. 180. Majumdar S, Aggarwal BB. Methotrexate suppresses NfkappaB activation through inhibition of IkappaBalpha phosphorylation and degradation. Journal of Immunology. 2001;167:2911-2920
  181. 181. Christophers E. The immunopathology of psoriasis. International Archives of Allergy and Immunology. 1996;110:199-206
  182. 182. Abdallah MA, Abdel-Hamid MF, Kotb AM, Mabrouk EA. Serum interferon-gamma is a psoriasis severity and prognostic marker. Cutis. 2009;84(3):163-168
  183. 183. Uyemura K, Yamamura M, Fivenson DF, Modlin RL. The cytokine network in lesional and lesion-free psoriatic skin is characterized by a T-helper type 1 cell-mediated response. Journal of Investigative Dermatology. 1993;101:701-705
  184. 184. Schlaak JF, Buslau M, Jochum W, et al. T cells involved in psoriasis vulgaris belong to the Th1 subset. Journal of Investigative Dermatology. 1994;102:145-149
  185. 185. El Barnawi NY, Ciasuddin ASM, Ziu MM, Singh M. Serum cytokine levels in psoriasis vulgaris. British Journal of Biomedical Science. 2001;58:40-44
  186. 186. Arican O, Aral M, Sasmaz S, et al. Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediators of Inflammation. 2005;5:273-279
  187. 187. Piskin G, Koomen CW, Picavet D. Ultraviolet-B irradiation decreases IFN-γ and increases IL-4 expression in psoriatic lesional skin in situ and in cultured dermal T cells derived from these lesions. Experimental Dermatology. 2003;12:172-180
  188. 188. Bonifati C, Trento E, Cordiali-Fei P, et al. Increased interleukin-7 concentrations in lesional skin and in the sera of patients with plaque-type psoriasis. Clinical Immunology and Immunopathology. 1997;83:41-44
  189. 189. Nesher G, Moore TL. The in vitro effects of methotrexate on peripheral blood mononuclear cells. Modulation by methyl donors and spermidine. Arthritis and Rheumatism. 1990;33:954-959
  190. 190. Kagami S, Rizzo HL, Lee JJ, et al. Circulating Th17, Th22, and Th1 cells are increased in psoriasis. Journal of Investigative Dermatology. 2010;130(5):1373-1383
  191. 191. Rangarajan A, Talora C, Okuyama R, et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. The EMBO Journal. 2001;20(13):3427-3436
  192. 192. Cutroneo KR. TGF-beta-induced fibrosis and SMAD signaling: Oligo decoys as natural therapeutics for inhibition of tissue fibrosis and scarring. Wound Repair and Regeneration. 2007;15(1):S54-S60
  193. 193. Flisiak I, Porebski P, Flisiak R, Chodynicka B. Plasma transforming growth factor beta1 as a biomarker of psoriasis activity and treatment efficacy. Biomarkers. 2003;8(5):437-443
  194. 194. Conery AR, Cao Y, Thompson EA, et al. Akt interacts directly with Smad3 to regulate the sensitivity to TGF-beta induced apoptosis. Nature Cell Biology. 2004;6(4):366-372
  195. 195. Song K, Wang H, Krebs T, Danielpour D. Novel roles of Akt and mTOR in suppressing TGF-b/ALK5-mediated Smad3 activation. The EMBO Journal. 2006;25:58-69
  196. 196. Ulloa L, Doody J, Massagué J. Inhibition of transforming growth factor-beta/SMAD signaling by the interferon-gamma/STAT pathway. Nature. 1999;397(6721):710-713
  197. 197. Bitzer M, von Gersdorff G, Liang D, et al. A mechanism of suppression of TGF-beta/SMAD signaling by NF-kappa B/RelA. Genes & Development. 2000;14(2):187-197
  198. 198. Cai JP, Falanga V, Taylor JR, Chin YH. Transforming growth factor beta receptor binding and function are decreased in posriatic dermal endothelium. Journal of Investigative Dermatology. 1996;106:225-231
  199. 199. Winter-Vann AM, Kamen BA, Bergo MO, et al. Targeting Ras signaling through inhibition of carboxyl methylation: An unexpected property of Methotrexate. Proceedings of the National Academy of Sciences. 2003;100(11):6529-6534
  200. 200. Simpson CL, Patel DM, Green KJ. Deconstructing the skin: Cytoarchitectural determinants of epidermal morphogenesis. Nature Reviews. Molecular Cell Biology. 2011;12:565-580
  201. 201. Huang WY, Yang PM, Chang YF, Marquez VE, Chen CC. Methotrexate induces apoptosis through p53/p21-dependent pathway and increases E-cadherin expression through downregulation of HDAC/EZH2. Biochemical Pharmacology. 2011;81(4):510-517
  202. 202. Yugawa T, Handa K, Narisawa-Saito M, Ohno S, Fujita M, Kiyono T. Regulation of Notch1 gene expression by p53 in epithelial cells. Molecular and Cellular Biology. 2007;27(10):3732-3742
  203. 203. Sriuranpong V, Borges MW, Ravi RK, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Research. 2001;61(7):3200-3205
  204. 204. Miyoshi K, Takaishi M, Nakajima K, et al. Stat3 as a therapeutic target for the treatment of psoriasis: A clinical feasibility study with STA-21, a Stat3 inhibitor. Journal of Investigative Dermatology. 2011;131(1):108-117
  205. 205. Meephansan J, Ruchusatsawat K, Sindhupak W, Thorner PS, Wongpiyabovorn J. Effect of methotrexate on serum levels of IL-22 in patients with psoriasis. European Journal of Dermatology. 2011;21(4):501-504
  206. 206. Spurlock CF, Aune ZT, Tossberg JT, et al. Increased sensitivity to apoptosis induced by methotrexate is mediated by JNK. Arthritis and Rheumatism. 2011;63(9):2606-2616
  207. 207. Neradil J, Pavlasova G, Veselska R. New mechanisms for an old drug; DHFR- and non-DHFR-mediated effects of methotrexate in cancer cells. Klinická Onkologie. 2012;2:S87-S92
  208. 208. Paradisi A, Pasquariello N, Barcaroli D, Maccarrone M. Anandamide regulates keratinocyte differentiation by inducing DNA methylation in a CB1 receptor-dependent manner. Journal of Biological Chemistry. 2008;283(10):6005-6012
  209. 209. Leigh IM, Navsaria H, Purkis PE, et al. Keratins (K16 and K17) as markers of keratinocyte hyperproliferation in psoriasis in vivo and in vitro. The British Journal of Dermatology. 1995;133:501-511
  210. 210. Fu M, Wang G. Keratin 17 as a therapeutic target for the treatment of psoriasis. Journal of Dermatological Science. 2012;67(3):161-165
  211. 211. Li Y, Jiang L, Zhang S, Yin L, et al. Methotrexate attenuates the Th17/IL-17 levels in peripheral blood mononuclear cells from healthy individuals and RA patients. Rheumatology International. 2012;32(8):2415-2422

Written By

Tamilselvi Elango, Anburaj Jeyaraj, Haripriya Dayalan, Pushpa Gnanaraj, Xinghui Li and Xuejun Zhang

Submitted: 19 December 2021 Reviewed: 21 January 2022 Published: 08 April 2022