15
Current Drug Discovery Technologies, 2012, 9, 000-000 1 1570-1638/12 $58.00+.00 © 2012 Bentham Science Publishers Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool: A Contemporary Viewpoint Mahfoozur Rahman 1, *, Mohammad Zaki Ahmed 1 , Imran Kazmi 2 , Sohail Akhter 3 , Sarwar Beg 3 , Gaurav Gupta 2 , Muhammad Afzal 2 , Shakir Saleem 3 , Iqbal Ahmad 3 , Md.Adil Shaharyar 3 , Farhan Jalees Ahmed 3 and Firoz Anwar 2 1 Dreamz College of Pharmacy, Himachal Pradesh, India, 2 Siddhartha Institute of Pharmacy, Dehradun, Uttarakhand, India. 3 Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, Hamdard Nagar, New Delhi, India 110 062 Abstract: Psoriasis is a common chronic autoimmune skin disorder with T-cell mediated multifunctional complex pathogenesis along with genetic predisposition. Conventionally, many therapies are available for the management of psoriasis, but they have limited efficacy due to higher side effects. Over the last decade, one of the major efforts in psoriasis research has been made for the development of drug molecules by understanding the potential biomolecules/biomarkers associated with psoriasis. This approach aims to provide selective immunologically directed intervention with fewer side effects than conventional therapies. The present review aims to give an exhaustive account on various biomarkers including oxidative stress, peptide, biochemical and gene markers responsible for keratocyte hyper proliferation, inflammatory responses and abnormal differentiation in psoriasis. Effective targeting of these over expressed biomarkers can serve as the novel tool for anti-psoriatic drug development. In addition, this review also gives insights into several novel biomarkers targeted drugs under pre-clinical and clinical investigation or have been registered by FDA for psoriasis management. Keywords: Psoriasis, Biomarkers, Drug targeting, Biological agents, Gene marker. 1. INTRODUCTION Psoriasis is the most common chronic skin T-cell medi- ated multifactorial autoimmune disorder of skin marked by inflammation, redness and itching. Generally it is found in teens upto to 20 years of age [1-3]. At the world level ap- proximately 0.3 to 5% population is affected by this disorder [4]. In Europe it is estimated that 2-3% of the total popula- tion has psoriasis. In Asia and Africa nearly about 1% of the population is affected with psoriasis [5]. The keratinocyte hyper proliferation in psoriasis may be due to the activation of T-cells, which produce various in- flammatory mediators like cytokines and adhesion molecules etc. These mediators are produced via various pathways i.e., MAPK/AP-1, JAK–STAT and protein kinase-C (PKCs) pathways. After T-cell activation, all these mediators induce a cascade of inflammation, which results in psoriasis. These entire pathways are unique to psoriasis [6-9]. Psoriatic skin is rich in inflammatory cells, CD4+, CD8+ T-cell and infil- trating T-cells express markers such as IL-2 receptors, result- ing in epidermal proliferation and endothelial cell activation [10, 11]. The exact origin of psoriasis was not well defined and documented years ago, but now it is very well defined as *Address correspondence to this author at the Mahfoozur Rahman, Assistant Professor, Dreamz College of Pharmacy, H.P. India 175036; Tel: +91- 9625218477; Fax: ??????????????; E-mail: [email protected] several reports show predominance of cytotoxic CD8+ T cells in psoriatic lesion epidermis, whereas CD4+ cells are the predominant type in lesional dermis. As per psoriasis pathology these cells express CD45RO on the surface, indi- cating their effectors/memory status [12-14]. Environmental stress and antigens, cause stimulation of T-cell receptor by the major histocompatibility complex (MHC I or II) on the antigen presenting cell (APC). As a result adhesion of T-cell with the APC, mediated by interaction of surface molecules such as CD2 on the T-cell with leukocyte function associated antigen (LFA)-1 on the APC [15] is a another process of pathogenesis in psoriasis. Signal 1 for psoriasis is ultimately generated due to over all formation of antigen MHC com- plexes with the surface of APCs to interact with T-cell recep- tors and CD4/CD8 co-receptors on the surface of T-cells [12]. Signal 2, is initiated by interactions between CD28 and CD80, CD40 and CD40L, CD28 and CD86, and LFA3 and CD2 respectively. Presence of signal 1 and 2 is essential for T-cell activation [16, 17]. T-cell activation further breaks down into three steps 1) activation of T-cell; 2) the migration of T-cell into the lesion skin; 3) release of cytokines by acti- vated T-cell in the skin. If anyone of these signal is inhibited, than T-cell activation ceases. A number of new biological agents have been developed for psoriasis which inhibit T-cell activation, for example efalizumab, a humanized anti-CD11a monoclonal antibody, and alefacept, an LFA-3/IgG1 fusion protein. Activation of CD4+ and CD8+ T-cell, produces various inflammatory cytokines such as IL-5, IL-4, and IL-

Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Embed Size (px)

DESCRIPTION

Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Citation preview

Page 1: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Current Drug Discovery Technologies, 2012, 9, 000-000 1

1570-1638/12 $58.00+.00 © 2012 Bentham Science Publishers

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool: A Contemporary Viewpoint

Mahfoozur Rahman1,

*, Mohammad Zaki Ahmed1, Imran Kazmi

2, Sohail Akhter

3, Sarwar Beg

3,

Gaurav Gupta2, Muhammad Afzal

2, Shakir Saleem

3, Iqbal Ahmad

3, Md.Adil Shaharyar

3, Farhan

Jalees Ahmed3 and Firoz Anwar

2

1Dreamz College of Pharmacy, Himachal Pradesh, India,

2Siddhartha Institute of Pharmacy, Dehradun, Uttarakhand,

India. 3Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, Hamdard Nagar, New Delhi, India

110 062

Abstract: Psoriasis is a common chronic autoimmune skin disorder with T-cell mediated multifunctional complex

pathogenesis along with genetic predisposition. Conventionally, many therapies are available for the management of

psoriasis, but they have limited efficacy due to higher side effects. Over the last decade, one of the major efforts in

psoriasis research has been made for the development of drug molecules by understanding the potential

biomolecules/biomarkers associated with psoriasis. This approach aims to provide selective immunologically directed

intervention with fewer side effects than conventional therapies. The present review aims to give an exhaustive account on

various biomarkers including oxidative stress, peptide, biochemical and gene markers responsible for keratocyte hyper

proliferation, inflammatory responses and abnormal differentiation in psoriasis. Effective targeting of these over

expressed biomarkers can serve as the novel tool for anti-psoriatic drug development. In addition, this review also gives

insights into several novel biomarkers targeted drugs under pre-clinical and clinical investigation or have been registered

by FDA for psoriasis management.

Keywords: Psoriasis, Biomarkers, Drug targeting, Biological agents, Gene marker.

1. INTRODUCTION

Psoriasis is the most common chronic skin T-cell medi-ated multifactorial autoimmune disorder of skin marked by inflammation, redness and itching. Generally it is found in teens upto to 20 years of age [1-3]. At the world level ap-proximately 0.3 to 5% population is affected by this disorder [4]. In Europe it is estimated that 2-3% of the total popula-tion has psoriasis. In Asia and Africa nearly about 1% of the population is affected with psoriasis [5].

The keratinocyte hyper proliferation in psoriasis may be due to the activation of T-cells, which produce various in-flammatory mediators like cytokines and adhesion molecules etc. These mediators are produced via various pathways i.e., MAPK/AP-1, JAK–STAT and protein kinase-C (PKCs) pathways. After T-cell activation, all these mediators induce a cascade of inflammation, which results in psoriasis. These entire pathways are unique to psoriasis [6-9]. Psoriatic skin is rich in inflammatory cells, CD4+, CD8+ T-cell and infil-trating T-cells express markers such as IL-2 receptors, result-ing in epidermal proliferation and endothelial cell activation [10, 11]. The exact origin of psoriasis was not well defined and documented years ago, but now it is very well defined as

*Address correspondence to this author at the Mahfoozur Rahman, Assistant

Professor, Dreamz College of Pharmacy, H.P. India 175036; Tel: +91-

9625218477; Fax: ??????????????; E-mail: [email protected]

several reports show predominance of cytotoxic CD8+ T cells in psoriatic lesion epidermis, whereas CD4+ cells are the predominant type in lesional dermis. As per psoriasis pathology these cells express CD45RO on the surface, indi-cating their effectors/memory status [12-14]. Environmental stress and antigens, cause stimulation of T-cell receptor by the major histocompatibility complex (MHC I or II) on the antigen presenting cell (APC). As a result adhesion of T-cell with the APC, mediated by interaction of surface molecules such as CD2 on the T-cell with leukocyte function associated antigen (LFA)-1 on the APC [15] is a another process of pathogenesis in psoriasis. Signal 1 for psoriasis is ultimately generated due to over all formation of antigen MHC com-plexes with the surface of APCs to interact with T-cell recep-tors and CD4/CD8 co-receptors on the surface of T-cells [12]. Signal 2, is initiated by interactions between CD28 and CD80, CD40 and CD40L, CD28 and CD86, and LFA3 and CD2 respectively. Presence of signal 1 and 2 is essential for T-cell activation [16, 17]. T-cell activation further breaks down into three steps 1) activation of T-cell; 2) the migration of T-cell into the lesion skin; 3) release of cytokines by acti-vated T-cell in the skin. If anyone of these signal is inhibited, than T-cell activation ceases. A number of new biological agents have been developed for psoriasis which inhibit T-cell activation, for example efalizumab, a humanized anti-CD11a monoclonal antibody, and alefacept, an LFA-3/IgG1 fusion protein. Activation of CD4+ and CD8+ T-cell, produces various inflammatory cytokines such as IL-5, IL-4, and IL-

Page 2: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

2 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

10, IL-2, IFN- and TNF- , which stimulates keratinocytes, and dendritic cells [18, 19]. The cytokines and chemokines released in psoriasis lesional skin lead to the typical epider-mal hyperplasia and keratinocyte inflammation. It is clear from above stated mechanism that psoriasis is T-cell medi-ated disease and the activation generates various cytokines, adhesion molecules and chemokines which as a result initi-ates the keratinocyte hyperproliferation and inflammation in psoriasis.

As compared to normal skin, proliferative epidermal cell is higher by 26.6% of DNA synthesis in psoriasis. The rela-tive growth increases from 60 to100% and size of prolifera-tive cell is doubled. The cell cycle is shortened from 311hr to 36 hrs, which results in decrease in basal cells of stratum corneum from 27 to 4 days [20, 21]. Conventional treatment for psoriasis includes topical corticosteroids, tars, anthralin, vit D analogs, tazarotene and salicylic acids. All of these have severe side effects like hepatotoxicity. Nephrotoxicity is associated with methotrexate, cyclosporine etc, terato-genicity with oral retinoids and skin cancers with photo/chemotherapy [22, 23]. Modern approach with spe-cific target action, from biological agents, has fewer side effects than conventional therapy. Long term use of these agents has few side effects [24, 25]. According to national psoriasis foundation survey only 26% of patients were satis-fied with conventional treatment. Recently, a UK survey has been found that only 44% of patient prefers systemic con-ventional treatment than topical treatment [26]. According to a survey by European federations of psoriasis association, on 17,990 patients only 27% of patients were satisfaction with conventional treatment. The cause of dissatisfaction among the masses was due to severe side effect, time consuming, ineffective therapy which affected their quality of life [26-28]. Therefore etiology and pathogenesis is very important for understanding psoriasis. Its mechanism deciphered, the biomarkers at the cellular and molecular level involved in mediating inflammatory response and other related responses during development of psoriasis.

According to the National Institute of Health (NIH),

Biomarkers are indicators of pathogenic process in this dis-

ease. Psoriasis is caused by multi factorial (toxin, stress, mi-

croorganism, etc) or polygenic character, therefore it is es-

sential to identify the nature of biomarker for effective

treatment of psoriasis [29-33]. NIH classified it into three

types, first type is zero markers that is related to history of

disease, second is, type 1 marker which is related to action of

drug, and the last is type II marker which is associated with

end point i.e. clinical output [32]. From various research

studies it has been found that many markers may act as diag-

nostic tool for psoriasis such as oxidative stress, biochemical

factor or enzyme, peptides and gene.

2. OXIDATIVE STRESS MARKER IN PSORIASIS

Oxidative stress is prominently due to the reactive oxy-

gen species (ROS), its major targeting site is skin. ROS is

generated from endogenous and exogenous source. Endoge-

nously ROS is formed by multiple sources such as electron

transport chain (ETC) in mitochondria, radiations [34, 35],

enzyme phagocytic and non phagocytic NADPH oxidase

[36, 37], lipoxygenase [38] and cycloxygenase [39] are to

name a few. Exogenous sources for ROS are environmental

toxins [40], exposure to heavy metal [41], ionizing radiation

[42], UV irradiation [43] and antibiotics like adiramycin [44,

45]. ROS include molecular oxygen, hypochlorous acid, per-

oxynitrile, hydroxyl radical and superoxide anion, all these

are competent enough to produce an oxygen unpaired elec-

tron, which is known as reactive oxygen species (ROS) [46,

47]. Overproduction or inadequate removal of ROS from the

body results in development of oxidative stress, ultimately it

leads to various abnormal functions like lipid per oxidation,

damage of DNA, protein and production of various inflam-

matory cytokines [48]. ROS is one of the factors involved in

the pathogenesis of psoriasis and also for other inflammatory

diseases. ROS modulates the formation of various pro in-

flammatory cytokines mediators and triggers the mitogen-

activated protein kinase/activator protein 1 (MAPK/AP),

Janus kinase–signal transducers, activators of transcription

(JAK-STAT) pathway and also modulate other protein

kinase pathways like tyrosine kinase, lipid per oxidation,

causing keratinocyte hyper proliferation and inflammation of

psoriatic skin [49].

Pro- inflammatory cytokines in psoriasis like lymphoki-nes, monokines, interleukins, interferon’s and growth factor, which are chemically glycoproteins, binds to specific cell receptors at picomolar concentration, to produce inflamma-tion and hyper proliferation in psoriasis [50]. Development of psoriatic lesion involves epidermal keratinocyte and leu-kocyte cells [7]. Keratinocyte is the key centre for recruit-ment and activation of leukocyte in psoriatic lesion. They also play a role in the formation of various chemokines and growth regulated oncogene product. Further, these agent produces chemotactic agent, which attract the neutrophil cells to the cluster differentiation (CD11) and dendritic cell (DC) of skin. DC cell in psoriatic lesion produces the differ-ent interleukin IL-23, IL-20, IL-15, interferon-alpha (IFN-

), tumor necrosis factor (TNF) and inducible nitric oxide syntheses (iNOS) [51-53]. IL-23, IL-20 and IL-2 along with other many antigens further activate the T-cell and keratino-cyte [54, 55]. Among different interleukins, IL-2 is the strong growth factor for T-cell activation. IL-2 also produce TNF, IL-6, IL-2R, IL-15 and IL-2 itself [56]. IL-15 induce the expression of TNF, lymphotoxin and IL-17. IL-17 is also produced by CD4+ T helper cell [57-59], moreover IL-17 also produce the IL-6, IL-8 and chemo tactic agent linked with IFN gamma, IL-4 and TNF [60-62]. IL-8 increased 50 times in psoriatic scales as compared to a normal cell. IFN- induces a variety of cytokines- IL-1, IL-16, IL-8, IL-15 and interferon inducible protein-10 [63]. Cytokine mediator are released primarily from keratinocyte and T-cell of skin under the oxidative stress condition. All these mediators are in-volved in over expression of keratinocyte proliferation, dif-ferentiation, hyperplasia and inflammation of psoriatic skin [64, 65].

2.1. Induction of Mitogen Activated Protein

Kinase/Activator Protein -1 (MAPK/ AP-1) Pathway by

ROS

It is a signaling cascade, involving extracellular-regulated kinase (ERK1/2, ERK3/4), Jun- N- terminal kinase (JNK)

Page 3: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 3

[66-68], P38 kinase and big mitogen activated protein kinase(BMK1) pathways. All these are activated, when the ROS activates the MAPK/AP-1 [69-71]. As shown in Fig. (1).

2.1.1. Activation of Extracellular-Regulated Kinase (ERK)

Pathway

ROS activates the multiple factor such as, multiple recep-tor tyrosine kinase, G-protein coupled receptor an epidermal growth factor receptor [72-76], these all further activates the RAS/RAF/MEK signals of ERK pathways [77, 78]. Nitric oxide [79] and calcium ion [80-83] are also involved in the activation of ERK pathway via nitrosylation of cysteine resi-due at RAS and calcium- clamodulin protein kinase mecha-nism [84-87]. According to various researches, it has been found that RAS/RAF/MEK/ERK1/2 expression is increased in psoriatic skin than non psoriatic skin (Fig. 1). Therefore, it might be a potential target for treatment of psoriasis [88, 89].

2.1.2. Activation of Jun N-Terminal kinase (JNK Pathway)

It is activated by many physical and chemical agents, like UV radiation, osmotic pressure alteration [90], irradiation, heavy metals [19], chemotherapeutic agent and reactive oxy-gen species [44, 45, 91, 92]. ROS activate the JNK-pathway via activation of apoptosis signal-regulating kinase-1 (ASK1), TNF receptor and JNK-MEKK1. Further, its activa-tion causes C-Jun activation, which induces the keratinocyte hyper proliferation (Fig. 1). Many reports are available which proves that over expression of TNF, C-Jun, JNK, ASK1in psoriatic skin than non psoriatic skin [93-95].

2.1.3. Activation of P38 or CDC42 (Types of Protein) Pathway

Alike EPK and JNK pathway ,it is activated by ROS, reactive nitrogen species (RNS) and various growth factor receptor [96], these are responsible for activation of TNF family of receptor, and apoptosis signal regulating kinase-1(ASK1) [97], which results into activation of P38 pathway via cell division cycle 42 (CDC42) [98, 99]. Furthermore, it phosphorylates many transcription factor, including activat-ing transcription factor-1(ATF-1) [100], and cAMP-response element binding protein (CREB) [101], expresses the kerati-nocyte hyper proliferation in psoriasis [102] (Fig. 1).

2.2. Janus kinase–Signal Transducers and Activators of Transcription (JAK- STAT) Pathway

It is activated by IFN- and IL [103]. Recent studies have

shown and demonstrated that, ROS is also involved in the

activation of JAK-STAT pathway. This pathway plays an

important role in immune-inflammatory disorder [104]. JAK

composed of JAKI, JAK2, JAK3 and tyrosine kinase -2.

Various research revealed that JAK1 is associated with IFN-

, IL-2, IL-6 receptors, where as JAK-2 is associated with

IL-3 and IFN- receptors [105-107]. STAT family comprises

of seven members 1, 2, 3, 4, 5a, 5b, and 6, these members

consist of 750-900 amino acid with SH2 domain. Major por-

tion of STAT chains present in cytoplasm is in inactive form.

Receptor associated JAK are activated by legand attachment

which activates the SH2 domain of STAT via phosphoryla-

Fig (1). ROS-mediated activation of the MAPK/AP-1 signaling pathway. The major components of the MAPK/AP-1 cascade pathway are

shown along with the transcription factors. ROS,Ca2+

ion, nitrite ion, protein kinase, activate receptors, Ras, MEKK1, and ASK1, which

subsequently causes activation of gene expression , C-jun , B- jun ,CREB, operate in a cascade fashion and lead to many inflammatory me-

diators (as for PPAR ,) related gene expression.

Page 4: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

4 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

tion of tyrosine receptor residue. Further STAT dimerize and

translocate to the nucleus, leading to gene expression, this

activation give a signal for entire interleukin (IL-6 (IL-6, IL-

11, IL-31) and IL-10 (IL-10, IL-19, IL-20 IL-22, IL-24)

formation (Fig. 2). These all are mediator of inflammatory

response in psoriasis [108, 109].

2.3. Activation of Protein Kinase-C (PKCs) Pathways

Protein kinase is a serine threonine kinase [110], it is

divided into three groups, first is conventional protein

kinase, which includes (�,�II and form), second is novel

protein kinase, including ( , , , and form) and the third is

atypical form which have mainly form [111, 112] PKCs is

very sensitive to extracellular and intracellular ROS. Its acti-

vation result in transduction of TNF, CDId, human leukocyte

antigen-1(HLA-class-I) molecule, when combined with

keratinocyte natural- killer T-cell that mediate the cytokine

production. Phosphorylation at threonine centre of PKCs

finally translocate to nuclear membrane and form nuclear

factor b (NF-K�) by phosphorylation of RelA P65 protein.

NF-K� is a key molecule for DNA binding. Its binding

causes activation of other proinflammatory cytokines,

chemokines and different adhesion molecule in psoriasis

[112-114].

Fig. (2). ROS-mediated activation of the JAK–STAT signaling pathway. ROS, protein kinases, IFN- and IL are major trigger for stimula-

tion. These all may activate Jak2, Tyk2, and the MAPK pathway, which resultes in activation of monomerized JAK-STAT. Activated STAT

dimerize and translocate into nucleus, which leads to gene expression and induce formation of interleukin particularly (IL-6, IL-10) type.

Fig. (3). Cell types and responses mediated by exemplary kinases that may contribute to pathogenesis of psoriasis. Various markers involved

in psoriasis are vascular endothelial cell growth factor receptor (VEGFR) or epidermal growth factor receptor (EGFR) - induces prolifera-

tion, migration of cytokines, chemo tactic agent, survival, and adhesion of keratinocytes and endothelial cells. Janus kinase (JAK) -3 induces

T-cell to promoting its proliferation, activation, and development. A tyrosine kinase 2 (Tyk2) and (IFN) induces DC and T-helper cells acti-

vation, which produce abnormal keratinocyte proliferation, differentiation, gene expression and various other activities.

Page 5: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 5

2.4. Lipid Per Oxidation

NADPH-dependent oxidase/myleoperoxidase, and vari-ous proteolytic enzymes plays a key role in production of intracellular ROS, causing lipid per oxidation and oxidative damage of cell membrane and proteins [115, 116]. When

sensitive, psoriatic skin is oxidized to low density lipopro-teins (OX-LDL), malondialdehyde, and high amount of thio compound, their accumulation in psoriatic skin leads to many immune inflammatory events [117, 118]. Decrease amount of many antioxidant such as alpha- tocopherol and glutathione (Vit-E, GSH, etc) in the plasma, increases the

Fig. (4). Receptor tyrosine kinase (TK) activation. A. In absence of legend attachment receptor TKs exist in monomerized inactive form. B.

Legand binding to extracellular domain causes activation of receptor TKs, which leads to dimerization and autophosphorylation in cytoplas-

mic domain of TKs. As a result signal transduction is induced through mitogen-activated protein kinases (MAPKs), Akt, and STATs to pro-

ducethe effect. C. In presence of TK inhibitor (TKI), monomerized form of TKs receptor fails to change into dimerized effective form, as a

result cascade of reaction is inhibited.

Fig. (5). The origin of said mediators is from the tissues, blood vessels, and neurons under the influence of oxidative stress condition. This

excervate keratinocyte hyper proliferation and differentiation of psoriatic skin.

Page 6: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

6 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

level of malondialdehyde (MDA), which results in lipid per-oxidation and keratinocyte hyperproliferation of skin [119, 120]. A very recent study has found that under oxidative stress condition, psoriatic skin shows over expressed perox-isome proliferator-activated receptors (PpAR ), a member of nuclear steroid hormone receptor, it causes abnormal lipid protein metabolism and epidermal proliferation of skin. It is mostly activated by transcription factor AP-1, Jun-B and heparin binding EGF [121].

3. BIOCHEMICAL MARKER IN PSORIASIS

Enzyme keratinocyte transglutaminase type I, increases the keratinocyte proliferation in the skin. This result in rise

of involucrin, antileucoproteinase [122] and keratin protein such as k6 and k16 in tissues, which leads to skin hyperpro-liferatation in psoriasis [123]. Decreasing amount of k1and k10 causes terminal differentiation of keratinocyte. Many different biomarkers such as HSP60, HSP27 (heat shock protein), IL12, toll like receptor-4 (TLR4) [124, 125], con-nexins (Cx) [126, 127], epidermal growth factor(EGF) [128], transforming growth factor-alpha (TGF- ,) [129] and bone morphogenetic protein-6 (BMP-6) are found mostly in plaque psoriasis, guttae psoriasis and in other type of psoria-sis [130], plaque psoriasis is most common type of psoriasis. All these markers originates (Fig. 5) from tissue or blood [131]. As a biomarker, many natural death proteins such as Bcl-x and Bak are associated with psoriatic skin. Bax are

Table 1. Different Gene Involved in Psoriasis

Locus name Location Locus Marker Ref

PSORS1 6p21,6q,8q24,10q22-q23,14q31-q32 HLAC,D8S284 [179-182]

PSORS2 17q24-q25,19p13,20p D17S785,D19S916,D19S865 [179]

[180]

[183, 184]

PSORS3 4q34 D4S2982 [175]

PSORS4 1q21,2p D1S305,D1S1664 [178]

[179]

PSORS5 3q21,4q13,4q21 D3S1768,D3S2409 [175]

Table 2. Origin/Source of Different Biomarkers

Markers Origin

Thiobarbituric acid (TBA) Blood

Superoxide dismutase Blood

C-reactive protein, neutrophil function Blood

Fibrinogen Blood

TNF-alpha,IFN-y,IL-2,6,8,12,18 Blood

Malondialdehyde Tissue

Oxidized LDL Tissue

Cytokeratins Tissue

Heat shock protein Tissue

Connexins (26,30) Tissue

Bcl-x,Bax,p53,epideremal growth factor Tissue

TGF-alpha, involucrin Tissue

MRP-8 Tissue

SP,VIP,NGF,PACAP-38 Tissue

Keratin 6,16,MAPK Tissue

TNF, tumor necrosis factor IFN, interferon; IL, interleukin;LDL, low-density lipoprotein; TGF, transforming growth factor MRP, migration inhibitory factor-related protein SP,

substance p; VIP, vasoactive intestinal peptide. MAPK, mitogen-activated protein kinase;; NGF, nerve growth factor; PACAP,pituitary adenylate cyclase activating polypeptide;

MAPK, mitogen-activated protein kinase

Page 7: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 7

also found along with induction of p53and ki67 positive cell, all of these mediate cell proliferation [132-136]. Rocha –periera- et al. [137] has found many marker, C-reactive pro-tein (CRP), fibrinogen, C3, C4 and also deciphered the asso-ciation of CD4+ T cell, DCS and CD8+T-cell, that induces the formation of pro inflammatory cytokines such as IL-23, IL-15, IL-17, TNF alpha, IFN-alpha etc. Cumulative effect of these causes stimulation of T- cell and hyper proliferation in the skin [7, 138, 139]. Recently it has been demonstrated that, caspase is found in hyper proliferative keratinocyte cell of skin, which is a cysteine proteinases enzyme, consisting of Caspase 3, 6, 7, and 14. In the presence of external stimuli caspas-14 is cleaved and enters into keratinocyte cell nuclei to induce the keratinocyte hyper proliferation and differentia-tion [140-144]. Various research studies has demonstrated that tyrosine kinase is over expressed in many inflammatory disorders such as psoriasis, arthritis and and crohn disease [145-147]. Tyrosine kinases catalyzes the cellular event by reversible phosphorylation (phosphate group come from ATP or GTP) at the tyrosine residue on the protein substrate. TKs is divided into two parts first is receptor type and sec-ond is non receptor type [148]. Receptor type TKs is charac-teristically transmembrane protein, and it has two domains extracellular and intracellular. Extracellular domain have legand binding site, and intracellular consist of catalytic do-main. Receptor type TKs is divided into twenty families, including vascular endothelial growth factor (VEGF) recep-tor (VEGFR), epidermal growth factor (EGF) receptor (EGFR) (Fig. 3), and platelet-derived growth factor (PDGF) receptor (PDGFR), fibroblast growth factor receptor. They are rearranged during transfection (RET) kinase [149]. EGFR is classified into 4 groups, which are known as eryth-roblastic leukemia oncogene homolog [ErbB1], ErbB2, ErbB3, and ErbB4. ErbB2and ErbB3 causes mainly psoriatic cell proliferation [150]. All receptor type TKs (except insulin type TKs) are present in monomeric nonphosphorylated form. Keratinocyte cell produce many different type of ligand as EGF legends TGFa, and epiregulin [151]. This legend binds to extracellular domain of TKs, which results dimereization of receptor, leading to activation of intrinsic catalytic domain, present in cytoplasm and in nucleus. This induces various transduction signals to produce inflamma-tory mediator that is involved in pathogenesis of psoriasis (Fig. 4). Nonreceptor TKs composed of 10 families, like sarcoma (src), Janus kinase (JAK) and Abelson (Ab1) etc [149]. Under normal condition non receptor tyrosine kinase present in inactive form as like receptor TKs1 [151]. Non receptor TKs activation, activates the other family member of TKs like TK-JAK-3 [149], resulting in development of T-cell and various signals to the other cytokine mediator, which mediates keratinocyte hyper proliferation and differ-entiation in psoriasis (Fig. 3) [149-154]. Presently, many tyrosine kinase inhibitor as a drug has been approved and many are under clinical stage (Table 5). Recently polymorph nuclear leukocytes (PML) identified by histological exami-nation of psoriatic skin. PML activation lead to generation of potent chemo attractants, which are detected in the stratum corneum by immuneflourscence techniques, such as TNF and GM-CSF (Granulocyte-macrophages colony stimulating factors). PML inactivation prevents the activation or accu-mulation of PML in the skin and further inhibits the forma-

tion of chemo attractants and pro inflammatory mediators. All these biochemical markers might be targeted for effec-tive treatment of psoriasis [155, 156].

3.1. Arachidonic Acid Metabolite Biomarker

Every human cell membrane consist of phospholipids, made of archidonic acid (3, 5, 8, 11 tetraecisonapentanoic acid). Action of plasma membrane bound phospholipase A2 (PLA2) and lipoxygenase enzyme cleaves it into prosta-glandins (PGI2, PGE1, PGE2 etc) and leukotriens (LTB4, LTC4 etc). These are involved in the pathogenesis of psoria-sis [157, 158]. Many cycloxygenase and lipoxygenase in-hibitors like NSAIDS, Benoxaprofen and lonapalene etc has been used in psoriasis. Recently, it has been proved that in-travenous infusion of omega-3 fatty acid based lipid emul-sion containing EPA and DHA (like fish oil, linseed oil, and evening primrose oil), compete with archidonic acid as a substrate which leads to the formation of less psoriagenetic metabolites [158], and overall improvement in the severity of psoriasis.

3.2. Neuropeptides Biomarker in Psoriasis

They are released from cutaneous nerve fibres. Bio-chemical and histochemical examinations have revealed vasoactive intestinal peptide (VIP) and substance-P in deeper dermis around eccrine gland and also found in dermal papil-lae of psoriatic skin. Naukrinen et al has found that mast cell and mast cell nerve fiber are more frequently present in der-mis of psoriatic skin. Various stimuli stimulate these nerve fibers to release substance –P and VIP. This causes forma-tion of various pro inflammatory cytokines and degranulates the mast cell leading to releases of proinflammatory sub-stances [159, 160]. Moreover, many neuropeptides like pitui-tary adenylate cyclase activating polypeptide (PACAP)-38 [161], nerve growth factor (NGF), and calcitonin gene –related peptide also has been found in psoriatic plaque skin as a inflammatory agent by histochemical studies [162, 163].

3.3. Gene Marker Involved in Psoriasis

Various research studies have revealed that multiple genes are involved in the pathogenesis of psoriasis. Genome wide linkage scans is a technique used to identify a number of genetic loci such as PSORS1 in the major histocompati-bilty complex (MHC) on the arm of chromosome 6 [164, 165] and chromosome 17. Another technique to prove of genetic involvement is demographic, epidemiologic, sero-logic, family history and twin studies [166-168]. On the ba-sis of demography psoriasis mostly found in the age group of 20-30 (Type-1) years and found in 50-60 year (Type II). Type II is not very common in various ethnic groups. Type-1 is found in human leukocyte antigen (HLA) and is associated with family history. Type II is not associated with HLA and family history [168, 169]. HLA can be positive or negative. HLA +ve type is associated with low age patients and whereas HLA –ve is associated with high age group [170-172]. Recent study on 29 guttae psoriasis patients in UK confirmed that HLA-CW0602 alleles was responsible in guttae psoriasis and similar results were observed in chronic plaque psoriasis [173], chromosome 17q25 for PSORS2 and chromosome 4q for PSORS3 [174-176]. It has been found that many other linkages such as PSORS5 on 3q21 [177]

Page 8: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

8 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

Table 3. List of Major Targeting Drugs

Biological Actions Mechanism Involved FDA Approval Efficacy Adverse Effect Ref

Denilukin diftitox

Brand name: ontak

Act on IL-2,

Receptor decreases the

number of Pathogenic T-

cell.

approved Study on 24 patients with plaque type

psoriasis, administered dose of 2, 4, 6

and 9 mg/kg of Denilukin diftitox by i.v

route for continuous five days of 4 week

up to 6 month. In the end Psoriasis Area

and Severity Index (PASI) decrease 68%

with respect to baseline at higher dose >

50% reduction in PASI baseline in half

of all patient.

Fever, chill, vomiting [193]

Daclizumab

Brand name:

Zenapax

Act on IL-2,

Receptor decreases the

number of Pathogenic T-

cell.

under clinical

stage

In case of plaque psoriasis a trial on 19-

patients at a dose of 2mg/kg followed by

1mg/kg for 8 week. 30% patients showed

reduction in PASI.

No adverse effects [194]

Basilximab

Brand name:

Simulect

Act on IL-2,

Receptor decreases the

number of pathogenic T-

cell.

under clinical

stage

In the case of plaque type psoriasis two

cases has been effectively treated.

No adverse effects [195]

[196]

Efalizumab

Brand name: Rap-

tiva

Act on CD11a,causes

T-cell Inhibition

approved Study on 556 patients, at a dose of

1mg/kg for 12 week more than 75%

reduction in base line, PASI score will be

increased with increased in duration of

therapy.

may produce

Thrombocytopenia (Un-

der investigation)

[197]

Alefacept Brand

name : Amevive.

Act on CD2 unit of T-

cells. Block its hypersen-

sitiveness

approved A case study on 553 patients of plaque

type psoriasis at a dose of 7.5mg of I.V

bolus injection for a period of 12 weeks,

causes 75% reduction in base line in 28%

of patient after one course of treatment

and 40% of patient after second course of

treatment

dizziness, nausea, chills,

and cough

[198]

[199]

Siplizumab; Brand

name: Medimmune

Act on CD2 unit of T-

cells. Block its hypersen-

sitiveness

under clinical

stage

A case study on 26 patients of psoriasis

at a dose of 0.4-40% mg/kg for 8 weeks.

75% reduction from mean base line PASI

in 50% of the patient

chills, and headache [200]

rhIL-10 Brand

name: Tenovil

Act on cytokines such as

IL-4 IL-10 and IL-11-

antagonist action on

cytokines

under clinical

stage

A case study on 28 patients of psoriasis

at a dose of 20 mg/kg s.c three times a

day for 12 week causes 35% reduction

from baseline PASI.

headache, gastric pain [201]

Adalimumab

(Humira)

It act against TNF-a approved A case study on 148 patients of psoriasis

at a dose of 40 mg every week for 12

weeks as a consequence , 75% reduction

from base line PASI in 80% of the pa-

tient

Headache, nausea, ele-

vated triglycerides,

cough, site pain.

[202]

Oprelvekin Brand

name: Neumega

Antagonist action on

cytokines

Under clinical

stage

A case study on 12 patients of psoriasis

with a dose of 2.5 or 5.0 mg /kg for 8

weeks causes 30-80% decrease from

base line PASI in 91% patients.

no adverse effect [203]

Infliximab Brand

name: Remicade

Acts against TNF-a and

other cytokines

under clinical

stage

A case study on 249- patients with a dose

of 3 and 5 mg/kg for 6 weeks. 75% re-

duction from base line PASI in 72% of

the patients at 3 mg/kg, and 88% of the

patients at the 5mg/kg.

only Headache [204]

Page 9: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 9

Table 3. cont…

Etenercept Brand name: embrel

Act against TNF- alpha approved A case study on 672 patients of psoriasis with a dose of 25 mg/kg for every week.

25 mg/kg for twice a week or 50 mg/kg twice a week. It was observed that 75%

reduction from base line PASI in 44% of the patients at a 25mg/kg of every week,

59% of the patient at a dose of 50 mg/kg twice a week.

no adverse effect after long term it enhance

diabetes, multiple sclero-sis, and infection

[205]

ABX-IL8 It selectively act against IL-8.

clinical stage Not reported. still not reported [206]

Ustekinuma

(CNTO-1275)

It is a novel human

immunoglobulin IgG, binds with p40 subunit

of both IL-12 and IL-23.

FDA approved A study on 903 patients divided into three groups first 45 mg dose of usteki-

numab for 0 and 4 weeks, 90 mg ustekinumab for 0 and 4 weeks and 50

mg etanercept for 0 and 4 weeks. The maximum PASI reduction was observed

in 75% of patients with 90 mg of dose of ustekinumab with respect to etanercept.

upper respiratory infec-tion, nasopharyngitis,

arthralgia, headache, cough, and injection

Site reaction.

[207]

Briakinuma

(ABT-874):

Is a recombinant fully human IgG1 monoclonal

antibody that binds with high affinity to the

p40 subunit of both IL-

12 and IL-23 cytokines

clinical stage III ----------------- Nasopharyngitis and upper respiratory tract

infections

[208]

Certolizumab It is a inhibitor of TNF-.

clinical stage III A study on moderate to severe chronic plaque type psoriasis patient swere re-ceive 200 or 400 mg of certolizumab,

every 2 week for 12 week ,in the last of 12th week PASI 75% decrease from base

line.

headache, nasopharyngitis,

pruritus

[209]

Table 4. Drug Targeting on STAT Pathway

Therapeutic Agent Mechanism of Action Ref

Ethyl pyruvate inhibits STAT signal pathway [210]

N-acetyl-L-cysteine inhibits STAT signal pathway

Dimethyl fumarate (DMF) increased glutathione level in tissues and causes inhibition of ROS. [211]

Ascorbic acid (Vit-C) inhibits STAT signal pathway [212]

Uric acid Protects against oxidative damage by scavenging oxygen radicals,

•OH, and other cellular oxidants.[213]

Vit.E ( -tocopherol) it prevents lipid peroxidation and quenches free radicals [212]

PSORS4 on 1q21, PSORS8 on 16q12-q13, and PSORS7 on 1q35-p34 etc are limited to psoriasis [178]. Different loci of gene marker have been shown in Table 1. Genetic approach can decipher a novel biological pathway for the effective treatment of psoriasis. This may be very effective approach among than other targeting approaches. Advancement in genetic research results in discovery of novel targeted anti-proliferative agents. However identification of different loci of gene is in its juvenile stage.

4. DRUG TARGETING ON BIOMARKER

Healthy skin maintains many enzyme and cellular redox system in the body, which play a vital role in prohibition of inflammatory disease. Imbalance in these systems may re-sults in formation of various oxidative marker, biochemical marker and peptide marker, which in turn leads to psoriasis

[185]. So for its treatment or prevention it is necessary to target various signaling molecules. Many monoclonal anti-bodies [186] (Table 3), group of high molecular- weight an-tioxidant enzymes (HMWA), proteins and a group of non enzymatic low-molecular-weight antioxidants (LMWA) can be effective tools for its prevention and treatment. HMWA including superoxide dismutases (SODs) [187, 188], glu-tathione peroxidases (GPx), thioredoxin/thioredoxin reduc-tase (TR), glutathione reductase (GR), catalase, metal-lothioneins, and ferritin system, whereas the LMWA include

-tocopherol (vitamin E), reduced glutathione (GSH), ascor-bic acid (vitamin C), and uric acid [189-192] are to name a few for treatment of psoriasis. These drugs have been listed in Table 3-5. Most of the biological agents, which are pres-ently used in the treatment of cancer, have been shown very effective against psoriasis, through different markers. Many

Page 10: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

10 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

of these have been approved and many are under clinical trial at various stages as discussed in (Table 3, 5). The results of clinical trial show that these are good therapeutic targets, specific and selective in action, with fewer side effects. Some biological agents like, Daclizuumab, Etanercept, In-fliximab and Ustekinumab etc have found as immunosup-pressive agents; these are listed only in Table 3. Major exist-ing limitation with biological agents is cost and they are ad-ministered in injection form [24]. The cost can managed, if the dosage forms of these drugs are altered, formulating it into a gel, resulting in direct penetratation, thus reducing systemic availability of the drug, directly reducing the side effects. Moreover, optimum dosing regimens and administra-tion methods for many of these biologics must be estab-lished. Presently research is under process to search a phyto-constituents, which may show targeted action against these different markers responsible for psoriasis.

5. CONCLUSION

This review has provided expression of various mole-

cules or marker that monitor the initiation and progress of

psoriasis. Over or under expression of markers, like ROS,

biochemical and peptides markers exerts their action in nor-

mal and abnormal condition. In normal condition they per-

form normal cell function and immune function. Due to

various stimuli, the level of markers may increase and causes

induction of several cellular signal transduction pathways

such as MAPK/AP-1, NF-k , JAK-STAT, lipid per oxida-

tion and genes activation, which are involved in pathogene-

sis of psoriasis. This may pave the way to discover a novel

targeting drug molecules for psoriasis like monoclonal anti-

bodies, tyrosine kinase inhibitors, JAK-STAT pathway tar-

geting drugs and may exert their action as an antiprolifera-

tive and antioxidative effect. These all can help in better un-

derstanding and can open new avenues for effective treat-

ment of psoriasis, its identification to eliminate conventional

way of treatment and its serious adverse effects. The dis-

cussed markers may be involved in other inflammatory dis-

orders like arthritis/rheumatism. In the current scientific de-

velopment research work is in progress to establish the cor-

relation between tissue/serum marker and clinical examina-

tion to increase the linking between biomarkers and psoria-

sis.

ABBREVIATIONS

AP-1 = Activator protein 1

ASK1 = Apoptosis signal-regulating kinase-1

ATF = Activating transcription factor

CD = Cluster of differentiation

DC = Dendritic cell

DMF = Dimethyl fumarate

ERK = Extracellular-regulated kinase

GSH = Glutathione

IFN- = Interferon-

iNOS = Inducible nitric oxide synthase

IL = Interleukin

IKK = I B kinase

JAK–STAT = Janus kinase–signal transducers and activators of transcription

JNK = Jun N-terminal kinase

MAPK = Mitogen-activated protein kinase

MAPKK or MKK = MAP kinase kinase

MAPKKK = MAP kinase kinase kinase

MEK = MAP–ERK kinase

MEKK = MAP–ERK kinase kinase

MSK1/2 = Mitogen- and stress-activated pro-tein kinase 1/2

NADPH = Reduced nicotinamide adenined-inucleotide phosphate

NF- B = Nuclear factor B

PKC = Protein kinase C

ROS = Reactive oxygen species

SOD = Superoxide dismutase

TNF = Tumor necrosis factor

Table 5. Drug Targeting on Different Tyrosine Kinase

Tyrosine kinase Targeting on Tyrosine kinase (Generic Name) Status Ref

VEGFR1,VEGFR2VEGFR3,PDGFRb RET. NVP-BAW2881 (Novartis, Cambridge, MA). Preclinical stage 214]

JAK1, JAK3. R333, (Rigel Pharmaceuticals Inc, San Francisco, CA). Preclinical Stage 215]

EGFR, ErbB2. PD169540 (Pfizer Inc, New York, NY). Preclinical Stage 215]

VEGFR1,VEGFR2,VEGFR3, PDGFRa

PDGFRb, c-Fms,

c-Kit, FLT3, RET.

Sunitinib, (Pfizer Inc, New York, NY). Approved [216]

PDGFRa,PDGFRb

c-Fms,c-Abl, Lck.

Imatinib, (Novartis, Cambridge, MA) Approved [217]

[218]

EGFR, ErbB2. lapatinib, (GlaxoSmithKline, London), England) Approved [219]

Page 11: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 11

TRAF2 = TNF-receptor associated factor 2

VEGFR = Vascular endothelial growth factor receptor.

PDGFR = Platelet-derived growth factor re-ceptor.

RET = Rearranged during transfection.

JAK = Janus kinase.

EGFR = Epidermal growth factor receptor.

ErbB2 = Erythroblastic leukemia oncogene homolog 2

C-Fms = Colony-stimulating factor-1 recep-tor

FLT3 = Fms-like tyrosine kinase 3.

C-Abl = Abelson.

Lck = Lymphocyte-specific protein tyro-sine kinase

PASI = Psoriasis Area and Severity Index

REFERENCES

[1] Travis L, Weinberg JM. Medical backgrounder: psoriasis. Drugs Today 2002; 38: 847-65.

[2] Lebwohl M: Psoriasis. Lancet 2003; 361: 1197-04. [3] Christophers E. Psoriasis epidemiology and clinical spectrum. Clin

Exp Dermatol 2001; 26: 314-20. [4] Nevitt GJ, Hutchinson PE. Psoriasis in the community: prevalence,

severity and patients, beliefs and attitudes towards the disease. Br J Dermatol 1996; 135: 533-7.

[5] International psoriasis council IPC) Psoriasis Review: Focus on Asia-pacific, May 2010.

[6] Nickoloff BJ, Xin H, Nestle FO, Qin JZ. The cytokine and chemokines network in psoriasis. Clin Dermatol 2007; 25: 568-73.

[7] Lowes MA, Bowcock AM, Krueger, JG. Pathogenesis and therapy of psoriasis. Nature 2007; 445: 866-73.

[8] Shim JS, Kwon YY, Han YS, Hwang JK. Inhibitory effect of pan-duratin A on UV-induced activation of mitogen-activated protein

kinases (MAPKs) in dermal fibroblast cells. Planta Med 2008; 74: 1446-50.

[9] Kim AL, Labasi JM, Zhu Y, et al. Role of p38 MAPK in UVB-induced inflammatory responses in the skin of SKH-1 hairless

mice. J Invest Dermatol 2005; 124: 1318-25. [10] Lin WJ, Norris DA, Achziger M, Kotzin BL, Tomkinson B. Oligo-

clonal expansion of intraepidermal T cells in psoriasis skin lesions. J Invest Dermatol 2001; 117: 1546-53.

[11] Nickoloff BJ, Wrone-Smith T. Injection of pre-psoriatic skin with CD4+ T cells induces psoriasis. Am J Pathol 1999; 155: 145-58.

[12] Bos JD, Hagenaars C, Das PK, Krieg SR, Voorn WJ, Kapsenberg ML. Predominance of “memory” T cells (CD4+, CDw29+) over

“naive” T cells (CD4+, CD45R+) in both normal and diseased hu-man skin. Arch Dermatol Res 1989: 281: 24-30.

[13] Bos JD, Hulsebosch HJ, Krieg SR, Bakker PM, Cormane RH. Immunocompetent cells in psoriasis. In situ immunophenotyping

by monoclonal antibodies. Arch Dermatol Res 1983; 275: 181-9. [14] Morganroth GS, Chan LS, Weinstein GD, Voorhees JJ, Cooper

KD. Proliferating cells in psoriatic dermis are comprised primarily of T cells, endothelial cells, and factor XIIIa+ perivascular den-

dritic cells. J Invest Dermatol 1991: 96: 333-40. [15] Berridge MJ. Lymphocyte activation in health and disease. Crit

Rev Immunol 1997: 17: 155-178. [16] Wingren AG, Parra E, Varga M, et al. T cell activation pathways:

B7, LFA-3, and ICAM-1 shape unique T cell profiles. Crit Rev Immunol 1995; 15: 235-53.

[17] Racke MK, Stuart RW. Targeting T cell costimulation in autoim-mune disease. Expert Opin Ther Targets 2002; 6: 275-89.

[18] Uyemura K, Yamamura M, Fivenson DF, Modlin RL, Nickoloff

BJ. The cytokine network in lesional and lesionfree psoriatic skin is characterized by a T-helper type 1 cellmediated response. J Invest

Dermatol 1993: 101: 701-05. [19] Bonifati C, Ameglio F. Cytokines in psoriasis. Int J Dermatol 1999:

38: 241-251. [20] Austin LM, Ozawa M, Kikuchi T, Walters IB, Krueger JG. The

majority of epidermal T cells in Psoriasis vulgaris lesions can pro-duce type 1 cytokines interferongamma, IL-2 and tumor necrosis

factor-alpha defining TCI (cytotoxic T lymphocyte) and THI effec-tor populations a type 1 differentiation bias is also measured in cir-

culating blood T cells in psoriatic patients. J Invest Dermatol 1999; 113: 752-9.

[21] Bata-Csorgo Z, Hammerberg C, Voorhees JJ, Cooper KD. Intrale-sional T-lymphocyte activation as a mediator of psoriatic epidermal

hyperplasia. J Invest Dermatol 1995; 105 (Suppl): 89S-94S. [22] Lebwohl M, Ali S. Treatment of psoriasis Topical therapy and

phototherapy. J Am Acad Dermatol 2001; 45: 487-98. [23] Lebwohl M, Ali S. Part 2: Treatment of psoriasis Systemic thera-

pies. J Am Acad Dermatol 2001; 45: 649–61. [24] Gottlieb AB, Chao C, Dann F. Psoriasis comorbidities. J Dermatol

Treat 2008; 19: 5-21. [25] International psoriasis council IPC) Psoriasis Review – December

2010 issue. [26] Krueger G, Koo J, Lebwohl M, Menter A, Stern RS, Rolstad T.

The Impact of Psoriasis on Quality of Life. Results a 1998 Natl Psoriasis Foundation Patient-Membership Survey. Arch Dermatol

2001; 137: 280-84. [27] Richards HL, Fortune DG, O'Sullivan TM, Main CJ, Griffiths CE

Patients with psoriasis and their compliance with medication. J Am Acad Dermatol 1999; 41: 581-83.

[28] De Korte J, Mombers FM, Sprangers MA, Bos JD. The suitability of quality-of-life questionnaires for psoriasis research a systematic

literature review. Arch Dermatol 2002; 138: 1221-7. [29] Swanbeck G, Inerot A, Martinsson T, Wahlström J. A population

genetic study of psoriasis. Br J Dermatol 1994; 131: 32-9. [30] Tagami H. Triggering factors. Clin Dermatol 1997; 15: 677-85.

[31] Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin

Pharmacol Ther 2001; 69: 89-95. [32] de Vlam K, Gottlieb AB, Fitzgerald O. Biological biomarkers in

psoriatic disease. A review. J Rheumatol 2008; 35: 1443-8. [33] Hewitt SM, Dear J, Star RA. Discovery of protein biomarkers for

renal diseases. J Am Soc Nephrol 2004; 15: 1677–89. [34] Sander CS, Chang H, Hamm F, Elsner P, Thiele JJ. Role of oxida-

tive stress and the antioxidant network in cutaneous carcinogenesis. Int J Dermatol 2004; 43: 326-35.

[35] Genova ML, Pich MM, Bernacchia A, et al. The mitochondrial production of reactive oxygen species in relation to aging and

pathology. Ann N Y Acad Sci 2004; 1011: 86-100. [36] Babior BM. NADPH oxidase: an update. Blood 1999; 93: 1464-76.

[37] Van Heerebeek L, Meischl C, Stooker W, Meijer CJ, Niessen HW, Roos D. NADPH oxidase(s) new source(s) of reactive oxygen spe-

cies in the vascular system. J Clin Pathol 2002; 55: 561-68. [38] Kuhn H, Thiele BJ. The diversity of the lipoxygenase family many

sequence data but little information on biological significance. FEBS Lett 1999; 449: 7-11.

[39] Kuehl Jr FA, Egan RW. Prostaglandins arachidonic acid and in-flammation. Science. 1980; 210: 978–84.

[40] McCarthy S, Somayajulu M, Sikorska M, Borowy-Borowski H, Pandey S. Paraquat induces oxidative stress and neuronal cell death

neuroprotection by water-soluble coenzyme Q10. Toxicol Appl Pharmacol 2004; 201: 21-31.

[41] Leonard SS, Harris GK, Shi X. Metal-induced oxidative stress and signal transduction. Free Radic Biol Med 2004; 37: 1921-42.

[42] Ogawa Y, Kobayashi T, Nishioka A, et al. Radiation-induced oxi-dative DNA damage, 8-oxoguanine, in human peripheral T cells.

Int J Mol Med 2003; 11: 27-32. [43] Herrling T, Fuchs J, Rehberg J, Groth N. UV-induced free radicals

in the skin detected by ESR spectroscopy and imaging using nitrox-ides. Free Radic Biol Med 2003; 35: 59-67.

[44] Gewirtz DA. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics

adriamycin and daunorubicin. Biochem Pharmacol 1999; 57: 727-41.

Page 12: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

12 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

[45] Singal PK, Li T, Kumar D, Danelisen I, Iliskovic N. Adriamycin-

induced heart failure: mechanism and modulation. Mol Cell Bio-chem 2000; 207: 77-86.

[46] Bickers DR, Athar M. Oxidative stress in the pathogenesis of skin disease. J Invest Dermatol 2006; 126: 2565-75.

[47] Hampton MB, Kettle AJ, Winterbourn CC. Inside the neutrophil phagosome oxidants, myeloperoxidase and bacterial killing. Blood

1998; 92: 3007-17. [48] Trouba KJ, Hamadeh HK, Amin RP, Germolec DR. Oxidative

stress and its role in skin disease. Antioxid Redox Signaling 2002; 4: 665-73.

[49] Finkel T, Holbrook NJ. Oxidants oxidative stress and the biology of ageing. Nature 2000; 408: 239-47.

[50] Gilhar A, Ullmann Y, Kerner H, et al. Psoriasis is mediated by a cutaneous defect triggered by activated immunocytes: induction of

psoriasis by cells with natural killer receptors. J Invest Dermatol 2002; 119: 384-91.

[51] Arenberger P, Kemény L, Süss R, Michel G, Peter RU, Ruzicka T.Interleukin-8 receptors in normal and psoriatic polymorphonu-

clear leukocytes. Acta Derm Venereol 1992; 72: 334-36. [52] Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic

cells initiate psoriasis through interferon-alpha production. J Exp Med 2005; 202: 135-43.

[53] Lowes MA, Chamian F, Abello MV, et al. Increase in TNF-alpha and inducible nitric oxide synthase-expressing dendritic cells in

psoriasis and reduction with efalizumab (anti-CD11a). Proc Natl Acad Sci USA 2005; 102: 19057-62.

[54] Lee E, Trepicchio WL, Oestreicher JL, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with pso-

riasis vulgaris. J Exp Med 2004; 199: 125-130. [55] Wang F, Lee E, Lowes MA, et al. Prominent production of IL-20

by CD68+/ CD11c+ myeloid-derived cells in psoriasis gene regula-tion and cellular effects. J Invest Dermatol 2006; 126: 1590-99.

[56] Kemmett D, Symons JA, Colver GB, Duff GW. Serum-soluble interleukin 2 receptor in psoriasis failure to reflect clinical im-

provement. Acta Derm Venereol 1990; 70: 264-66. [57] Pietrzak AT, Zalewska A, Chodorowska G, et al. Cytokines and

anticytokines in psoriasis. Clin Chim Acta 2008; 394: 7-21. [58] Elder JT. IL-15 and psoriasis another genetic link to Th17. J Invest

Dermatol 2007; 127: 2495-7. [59] Haider AS, Lowes MA, Suarez-Farinas M, et al. Identification of

cellular pathways of type 1q Th17 T cells, and TNF- and inducible nitric oxide synthase-producing dendritic cells in autoimmune in-

flammation through pharmacogenomic study of cyclosporine A in psoriasis. J Immunol 2008; 180: 1913-20.

[60] Teunissen MB, Koomen CW, de Waal Malefyt R, Wierenga EA, Bos JD. Interleukin-17 and interferon-gamma synergize in the en-

hancement of proinflammatory cytokine production by human keratinocytes. J Invest Dermatol1998; 111: 645-9.

[61] Albanesi C, Cavani A, Girolomoni G. IL-17 is produced by nickel-specific T lymphocytes and regulates ICAM-1 expression and

chemokine production in human keratinocytes synergistic or an-tagonist effects with IFN-gamma and TNF-alpha. J Immunol 1999;

162: 494-502. [62] Albanesi C, Scarponi C, Cavani A, Federici M, Nasorri F, Girolo-

moni G. Interleukin-17 is produced by both Th1 and Th2 lympho-cytes and modulates interferon-gamma- and interleukin-4-induced

activation of human keratinocytes. J Invest Dermatol 2000; 115: 81-7.

[63] Xie QW, Whisnant R, Nathan C. Promoter of the mouse gene en-coding calcium-independent nitric oxide synthase confers induci-

bility by interferon gamma and bacterial lipopolysaccharide. J Exp Med 1993; 177: 1779-84.

[64] Grossman RM, Krueger J, Yourish D, et al. Interleukin 6 is ex-pressed in high levels in psoriatic skin and stimulates proliferation

of cultured human keratinocytes. Proc Natl Acad Sci USA 1989; 86: 6367-71.

[65] Finch PW, Murphy F, Cardinale I, Krueger JG. Altered expression of keratinocyte growth factor and its receptor in psoriasis. Am J

Pathol 1997; 151: 1619-28. [66] Seternes OM, Mikalsen T, Johansen B, et al. Activation of

MK5/PRAK by the typical MAP kinase ERK3 defines a novel sig-nal transduction pathway. EMBO J 2004; 23: 4780-91.

[67] Cano E, Mahadevan LC. Parallel signal processing among mam-malian MAPKs. Trends. Biochem Sci 1995; 20: 117-22.

[68] Davis RJ, MAPKs new JNK expands the group. Trends Biochem

Sci 1994; 19: 470-3. [69] Takahashi H, Ibe M, Nakamura S, Ishida-Yamamoto A, Hashimoto

Y, Iizuka H. Extracellular regulated kinase and c-Jun N-terminal kinase are activated in psoriatic involved epidermis. J Dermatol Sci

2002; 30: 94-99. [70] Johansen C, Kragballe K, Westergaard M, Henningsen J, Kristian-

sen K, Iversen L. The mitogen-activated protein kinases p38 and ERK1/2 are increased in lesional psoriatic skin. Br J Dermatol

2005; 152: 37-42. [71] Yu XJ, Li CY, Dai HY, et al. Expression and localization of the

activated mitogen-activated protein kinase in lesional psoriatic skin. Exp Mol Pathol 2007; 83: 413-18.

[72] Burgering BM, de Vries-Smits AM, Medema RH, van Weeren PC, Tertoolen LG, Bos JL. Epidermal growth factor induces phos-

phorylation of extracellular signal-regulated kinase 2 via multiple pathways. Mol Cell Biol 1993; 13: 7248-56.

[73] Kribben A, Wieder ED, Li X, van, et al. AVP-induced activation of MAP kinase in vascular smooth muscle cells is mediated through

protein kinase. C Am J Physiol 1993; 265: 939-45. [74] Gardner AM, Vaillancourt RR, Johnson GL. Activation of mito-

gen-activated protein kinase/extracellular signal-regulated kinase kinase by G protein and tyrosine kinase oncoproteins. J Biol Chem

1993; 268: 17896-901. [75] Johnson GL, Gardner AM, Lange-Carter C, Qian NX, Russell M,

Winitz S.How does the G protein Gi2 transduce mitogenic signals. J Cell Biochem1994; 54: 415-22.

[76] Kavelaars A, Broeke D, Jeurissen F, et al. Activation of human monocytes via a non-neurokinin substance-P receptor that is cou-

pled to G protein calcium phospholipaseD MAP kinase and IL-6 production. J Immunol 1994; 153: 3691-9.

[77] Seger R, Biener Y, Feinstein R, Hanoch T, Gazit A, Zick Y. Dif-ferential activation of mitogen-activated protein kinase and S6

kinase signaling pathways by 12-O-tetradecanoylphorbol-13-acetate (TPA) and insulin: evidence for involvement of a TPA-

stimulated protein-tyrosine kinase. J Biol Chem 1995; 270: 28325-30.

[78] Gamou S, Shimizu N. Hydrogen peroxide preferentially enhances the tyrosine phosphorylation of epidermal growth factor receptor.

FEBS Lett 1995; 357: 161-4. [79] Lander HM, Milbank AJ, Tauras JM, et al. Redox regulation of cell

signalling. Nature 1996; 381: 380-1. [80] Chao TS, Byron KL, Lee KM, Villereal M, Rosner MR. Activation

of MAP kinases by calcium-dependent and calcium-independent pathways stimulation by thapsigargin and epidermal growth factor.

J Biol Chem 1992; 267: 19876-83. [81] Franklin RA, Tordai A, Mazer B, Terada N, Lucas JJ, Gelfand EW.

Activation of MAP2-kinase in B lymphocytes by calcium ionopho-res. J Immunol 1994; 153: 4890-98.

[82] Lev S, Moreno H, Martinez R, et al. Protein tyrosine kinase PYK2 involved in Ca(2+)- induced regulation of ion channel and MAP

kinase functions. Nature 1995; 376: 737-45. [83] Rosen LB, Ginty DD, Weber MJ, Greenberg ME. Membrane depo-

larization and calcium influx stimulate MEK and MAP kinase via activation of Ras. Neuron 1994; 12: 1207-21.

[84] Atherfold PA, Norris MS, Robinson PJ, Gelfand EW, Franklin RA. Calcium-induced ERK activation in human T lymphocytes. Mol

Immunol 1999; 36: 543-49. [85] Franklin RA, Atherfold PA, McCubrey JA. Calcium-induced ERK

activation in human T lymphocytes occurs via p56 (Lck) and CaM-kinase. Mol Immunol 2000; 37: 675-83.

[86] Ginnan R, Singer HA. CaM kinase II-dependent activation of tyro-sine kinases and ERK1/2 in vascular smooth muscle. Am J Physiol

Cell Physiol 2002; 282: C754-C61. [87] Schmitt JM, Wayman GA, Nozaki N, Soderling TR. Calcium acti-

vation of ERK mediated by calmodulin kinase I. J Biol Chem 2004; 279: 24064-72.

[88] Takahashi H, Ibe M, Nakamura S, Ishida-Yamamoto A, Hashimoto Y, Iizuka H. Extracellular regulated kinase and c-Jun N-terminal

kinase are activated in psoriatic involved epidermis. J Dermatol Sci 2002; 30: 94-9.

[89] Johansen C, Kragballe K, Westergaard M, Henningsen J, Kristian-sen K, Iversen L. The mitogen-activated protein kinases p38 and

ERK1/2 are increased in lesional psoriatic skin. Br J Dermatol 2005; 152: 37-42.

Page 13: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 13

[90] Galcheva-Gargova Z, Derijard B, Wu IH, Davis RJ. An osmosens-

ing signal transduction pathway in mammalian cells. Science1994; 265: 806-8.

[91] Conde de la, Rosa L, Schoemaker MH, et al. Superoxide anions and hydrogen peroxide induce hepatocyte death by different

mechanisms: involvement of JNK and ERK MAP kinases. J Hepa-tol 2006; 44: 918-29.

[92] Murakami T, Takagi H, Suzuma K, et al. Angiopoietin-1 attenuates H2O2-induced SEK1/JNK phosphorylation through the phosphati-

dylinositol 3-kinase/Akt pathway in vascular endothelial cells. J Biol Chem 2005; 280: 31841-49.

[93] Matsukawa J, Matsuzawa A, Takeda K, Ichijo H. The ASK1–MAP kinase cascades in mammalian stress response. J Biochem 2004;

136: 261-65. [94] Shrivastava A, Aggarwal BB. Antioxidants differentially regulate

activation of nuclear factor-kappa B, activator protein-1, c-jun amino-terminal kinases, and apoptosis induced by tumor necrosis

factor: evidence that JNK and NF-kappa B activation are not linked to apoptosis. Antioxid Redox Signaling 1999; 1: 181-91.

[95] Schmidt KJ, Büning J, Jankowiak C, Lehnert H, Fellermann K. Crohn´s Targeted Therapy: Myth or Real Goal?Current Drug

Discovery Technologies l 2009: 6; 290-8. [96] Schieke SM, Briviba K, Klotz LO, et al. Activation pattern of

mitogen Activated Protein kinases elicited by peroxynitrite attenua-tion by selenite supplementation. FEBS Lett 1999; 448: 301-3.

[97] Matsuzawa A, Nishitoh H, Tobiume K, Takeda K, Ichijo H. Physiological roles Of ASK1-mediated signal transduction in oxi-

dative stress- and endoplasmic reticulum stress-induced apoptosis: advanced findings from ASK1 knockout mice. Antioxid Redox

Signaling 2002; 4: 415-25. [98] Teramoto H, Coso OA, Miyata H, Igishi T, Miki T, Gutkind JS.

Signaling from the small GTP-binding proteins Rac1 and Cdc42 to the c-Jun N-terminal kinase/stress-activated protein kinase pathway

a role for mixed lineage kinase 3/protein-tyrosine kinase 1 a novel member of the mixed lineage kinase family. J Biol Chem 1996;

271: 27225-28. [99] Kyriakis JM, Avruch J. Protein kinase cascades activated by stress

and inflammatory cytokines. Bioessays 1996; 18: 567-77. [100] Wiggin GR, Soloaga A, Foster JM, Murray-Tait V, Cohen P, Ar-

thur JS. MSK1 and MSK2 are required for the mitogen- and stress-induced phosphorylation of CREB and ATF1 in fibroblasts. Mol

Cell Biol 2002; 22: 2871-81 [101] Deak M, Clifton AD, Lucocq LM, Alessi DR. Mitogen and stress-

activated protein kinase-1 (MSK1) is directly activated by MAPK and SAPK2/p38 and may mediate activation of CREB. EMBO J

1998; 17: 4426-41. [102] Funding AT, Johansen C, Kragballe K, et al. Mitogen- and stress-

activated protein kinase 1 is activated in lesional psoriatic epider-mis and regulates the expression of pro-inflammatory cytokines. J

Invest Dermatol 2006; 126: 1784-91. [103] Wang F, Lee E, Lowes MA, et al. Prominent production of IL-20

by CD68+/ CD11c+ myeloid-derived cells in psoriasis gene regula-tion and cellular effects. J Invest Dermatol 2006; 126: 1590-99

[104] Kishimoto T, Taga T, Akira S. Cytokine signal transduction. Cell 1994; 76: 253-62.

[105] Kisseleva T, Bhattacharya S, Braunstein J, Schindler CW. Signal-ing through the JAK/STAT pathway recent advances and future

challenges. Gene 2002; 285: 1-24. [106] O'Shea JJ, Gadina M, Schreiber RD. Cytokine signaling in 2002:

new surprises in the Jak/Stat pathway. Cell (Suppl) 2002; 109: S121-31.

[107] Wilks AF. Two putative protein-tyrosine kinases identified by application of the polymerase chain reaction. Proc Natl Acad Sci

USA 1989; 86: 1603-7. [108] Mertens C, Zhong M, Krishnaraj R, Zou W, Chen X, Darnell JE Jr.

Dephosphorylation of phosphotyrosine on STAT1 dimers requires extensive spatial reorientation of the monomers facilitated by the

N-terminal domain. Genes Dev 2006; 20: 3372-81. [109] Schindler C, Levy DE, Decker T. JAK–STAT signaling from inter-

ferons to cytokines. J Biol Chem 2007; 282: 20059-63. [110] Bouwman RA, Musters RJ, van Beek-Harmsen BJ, de Lange JJ,

Lamberts RR, Loer SA, Boer C. Sevoflurane-induced cardio pro-tection depends on PKC alpha activation via production of reactive

oxygen species. Br J Anaesth 2007; 99: 639-45.

[111] Martelli AM, Evangelisti C, Nyakern M, Manzoli FA. Nuclear

protein kinase C. Biochim Biophys Acta 2006; 1761: 542-51. [112] Moscat J, Rennert P, Diaz-Meco MT. PKC zeta at the crossroad of

NF-kappaB and Jak1/Stat6 signaling pathways. Cell Death Differ 2006; 13: 702-11.

[113] Zhao Y, Fishelevich R, Petrali JP, et al. Activation of keratinocyte protein kinase C zeta in psoriasis plaques. J Invest Dermatol 2008;

128: 2190-97. [114] Duran A, Diaz-Meco MT, Moscat J. Essential role of RelA Ser311

phosphorylation by zetaPKC in NF-kappaB transcriptional activa-tion. EMBO J 2003; 22: 3910-8.

[115] Rocha-Pereira P, Santos-Silva A, Rebelo I, Figueiredo A, Quin-tanilha A, Teixeira F. Dislipidemia and oxidative stress in mild and

in severe psoriasis as a risk for cardiovascular disease. Clin Chim Acta 2001; 303: 33-9.

[116] Tekin NS, Tekin IO, Barut F, Sipahi EY. Accumulation of oxidized low density lipoprotein in psoriatic skin and changes of plasma

lipid levels in psoriatic patients. Mediators Inflamm 2007: 2007: 78454.

[117] Yildirim M, Inaloz HS, Baysal V, Delibas N. The role of oxidants and antioxidants in psoriasis. J Eur Acad Dermatol Venereol 2003;

17: 34-6. [118] Piskin S, Gurkok F, Ekuklu G, Senol M. Serum lipid levels in

psoriasis. Yonsei Med J 2003; 44: 24-6. [119] Drewa G, Krzyzynska-Malinowska E, Wozniak A, et al. Activity

of superoxide dismutase and catalase and the level of lipid peroxi-dation products reactive with TBA in patients with psoriasis. Med

Sci Monit 2002; 8: BR338-BR43. [120] Kokcam I, Naziroglu M. Antioxidants and lipid peroxidation status

in the blood of patients with psoriasis. Clin Chim Acta 1999; 289: 23-31.

[121] Romanowska M, al Yacoub N, Seidel H, et al. PPARdelta en-hances keratinocyte proliferation in psoriasis and induces heparin-

binding EGF-like growth factor. J Invest Dermatol 2008; 128: 110-24.

[122] Schroeder WT, Thacher SM, Stewart-Galetka S, et al. Type I ker-antinocyte transglutaminase: expression in human skin and psoria-

sis. J Invest Dermatol 1992; 99: 27-34. [123] Gilhar A, Ullmann Y, Kerner H, et al. Psoriasis is mediated by a

cutaneous defect triggered by activated immunocytes induction of psoriasis by cells with natural killer receptors. J Invest Dermatol

2002; 119: 384-91. [124] Seung NR, Park EJ, Kim CW, et al. Comparison of expression of

heat-shock protein 60, Toll-like receptors 2 and 4 and T-cell recep-tor gammadelta in plaque and guttate psoriasis. J Cutan Pathol

2007; 34: 903-11. [125] Curry JL, Qin JZ, Bonish B, et al. Innate immune-related receptors

in normal and psoriatic skin. Arch Pathol Lab Med 2003; 127: 178-86.

[126] Lucke T, Choudhry R, Thom R, Selmer IS, Burden AD, Hodgins MB. Upregulation of connexin 26 is a feature of keratinocyte dif-

ferentiation in hyperproliferative epidermis vaginal epithelium and buccal epithelium. J Invest Dermatol 1999; 112: 354-61.

[127] Lemaitre G, Sivan V, Lamartine J, et al. Connexin 30 a new marker of hyperproliferative epidermis. Br J Dermatol 2006; 155: 844-6.

[128] Nanney LB, Stoscheck CM, Magid M, King LE Jr. Altered [125i]epidermal growth factor binding and receptor distribution in

psoriasis. Biull Eksp Biol Med 1993; 115: 618-20. [129] Nickoloff BJ, Karabin GD, Barker JN et al. Cellular localization of

interleukin-8 and its inducer, tumor necrosis factor-alpha in psoria-sis. Am J Pathol 1991; 138: 129-40.

[130] Blessing M, Schirmacher P, Kaiser S. Overexpression of bone morphogenetic protein-6 (BMP-6) in the epidermis of transgenic

mice: inhibition or stimulation of proliferation depending on the pattern of transgene expression and formation of psoriatic lesions. J

Cell Biol 1996; 135: 227-39. [131] Grove T, Mulfinger L. The pathogenesis of psoriasis: biochemical

aspects. J Young Invest 2001; 4. [132] Wrone-Smith T, Mitra RS, Thompson CB, Jasty R, Castle VP,

Nickoloff BJ. Keratinocytes derived from psoriatic plaques are re-sistant to apoptosis compared with normal skin. Am J Pathol 1997;

151: 1321-9. [133] Batinac T, Zamolo G, Hadzisejdic I, et al. Expression of Bcl-2

family proteins in psoriasis. Croat Med J 2007; 4: 319-26.

Page 14: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

14 Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 Rahman et al.

[134] Wrone-Smith T, Johnson T, Nelson B, et al. Discordant expression

of Bcl-x and Bcl-2 by keratinocytes in vitro and psoriatic keratino-cytes in vivo. Am J Pathol 1995; 146: 1079-88.

[135] Tomkova H, Fujimoto W, Arata J. Expression of bcl-2 antagonist bak in inflammatory and neoplastic skin diseases. Br J Dermatol

1997; 137: 703-8. [136] Tomkova H, Fujimoto W, Arata J. Expression of the bcl-2 homo-

logue Bax in normal human skin, psoriasis vulgaris and non-melanoma skin cancers. Eur J Dermatol 1998; 8: 256-60.

[137] Rocha-Pereira P, Santos-Silva A, Rebelo I, Figueiredo A, Quin-tanilha A, Teixeira F. The inflammatory response in mild and in

severe psoriasis. Br J Dermatol 2004; 150: 917-28. [138] Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic

cells initiate psoriasis through interferonalpha production. J Exp Med 2005; 202: 135-43

[139] Zheng Y, Danilenko DM, Valdez P, et al. Interleukin-22 a TH17 cytokine mediates IL-23-induced dermal inflammation and acan-

thosis. Nature 2007; 445: 648-51. [140] Chien AJ, Presland RB, Kuechle MK. Processing of native

caspase-14 occurs at an atypical cleavage site in normal epidermal differentiation. Biochem Biophys Res Commun 2002; 296: 911-7.

[141] Hu S, Snipas SJ, Vincenz C, Salvesen G, Dixit VM. Caspase-14 is a novel developmentally regulated protease. J Biol Chem 1998;

273: 296 48-53. [142] Van de Craen M, Van Loo G, Pype S, et al. Identification of a new

caspase homologue: caspase-14. Cell Death Differ 1998; 5: 838-46. [143] Lippens S, VandenBroecke C, Van Damme E, Tschachler E, Van-

denabeele P, Declercq W. Caspase-14 is expressed in the epidermis the choroid plexus the retinal pigment epithelium and thymic Has-

sall’s bodies. Cell Death Differ 2003; 10: 257-9. [144] Lippens S, Kockx M, Knaapen M, et al. Epidermal differentiation

does not involve the pro-apoptotic executioner caspases but is as-sociated with caspase-14 induction and processing. Cell Death Dif-

fer 2000; 7: 1218-24. [145] D’Aura Swanson C, Paniagua RT, Lindstrom TM, Robinson WH.

Tyrosine kinases as targets for the treatment of rheumatoid arthritis. Nat Rev Rheumatol 2009; 5: 317-24.

[146] Eklund KK, Lindstedt K, Sandler C, et al. Maintained efficacy of the tyrosine kinase inhibitor imatinib mesylate in a patient with

rheumatoid arthritis. J Clin Rheumatol 2008; 14: 294-6. [147] Paniagua RT, Sharpe O, Ho PP, et al. Selective tyrosine kinase

inhibition by imatinib mesylate for the treatment of autoimmune ar-thritis. J Clin Invest 2006; 116: 2633-42.

[148] Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 2002;

298: 1912-34. [149] Robinson DR, Wu YM, Lin SF. The protein tyrosine kinase family

of the human genome. Oncogene 2000; 19: 55 48-57. [150] Pastore S, Mascia F, Mariani V, Girolomoni G. The epidermal

growth factor receptor system in skin repair and inflammation. J Invest Dermatol 2008; 128: 1365-74.

[151] Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med 2005; 353: 172-87.

[152] Hubbard SR, Till JH. Protein tyrosine kinase structure and func-tion. Annu Rev Biochem 2000; 69: 373-98.

[153] Ghoreschi K, Laurence A, O’Shea JJ. Selectivity and therapeutic inhibition of kinases to be or not to be. Nat Immunol 2009; 10:

356-60. [154] Schindler T, Bornmann W, Pellicena P, Miller WT, Clarkson B,

Kuriyan J. Structural mechanism for STI-571 inhibition of Abelson tyrosine kinase. Science 2000; 289: 1938-42.

[155] Gilhar A, Ullmann Y, Kerner H, et al. Psoriasis is mediated by a cutaneous defect triggered by activated immunocytes induction of

psoriasis by cells with natural killer receptors. J Invest Dermatol 2002; 119: 384-91.

[156] Hegemann L, Hatzelmann A, Grewig S, Schmidt BH. Potent an-tagonism of calmodulin activity in vitro, but lack of antiprolifera-

tive effects on keratinocytes by the novel leukotriene biosynthesis inhibitor MK-886. Br J Dermatol1995; 133: 41-7.

[157] Mayser P, Grimm H, Grimminger F. N-3 fatty acids in psoriasis. Br J Nutrition 2002; 87(Suppl): 577-82.

[158] Mommers JM, Van Rossum MM, Kooijmans-Otero ME, Parker GL, van de Kerkhof PC. VML 295 (LY-293111) a novel LTB4 an-

tagonist is not effective in the prevention of relapse in psoriasis. Br J Dermatol 2000; 142: 259-66.

[159] Mozzanica N, Cattaneo A, Vignati G, Finzi A. Plasma neuropep-

tide levels in psoriasis. Acta Dermatovenereol 1994; 186; 67-8. [160] Naukkarinen A, Järvikallio A, Lakkakorpi J, Harvima IT, Harvima

RJ, Horsmanheimo M. Quantitative histochemical analysis of mast cells and sensory nerves in psoriatic skin. J Pathol 1996; 180: 200-

5. [161] Steinhoff M, Ständer S, Seeliger S, Ansel JC, Schmelz M, Luger T.

Modern aspects of cutaneous neurogenic inflammation. Arch Der-matol 2003; 139: 1479-88.

[162] Saraceno R, Kleyn CE, Terenghi G, Griffiths CE. The role of neu-ropeptides in psoriasis. Br J Dermatol 2006; 155: 876-82.

[163] Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic keratinocytes express high levels of nerve growth factor. Acta Derm Venereol

1998; 78: 84-6. [164] Elder JT, Henseler T, Christophers E, Voorhees JJ, Nair RP. Re-

view of genes and antigens the inheritance of psoriasis. J Invest Dermatol 1994; 103: 150S-3S.

[165] Elder JT, Nair RP, Henseler T, et al. The genetics of psoriasis 2001 the odyssey continues. Arch Dermatol 2001; 137: 1447-54.

[166] Farber EM, Nall L, Watson W. Natural history of psoriasis in 61 twin pairs. Arch Dermatol 1974; 109: 207-11.

[167] Brandrup F, Holm N, Grunnet N, Henningsen K, Hansen HE. Pso-riasis in monozygotic twins variations in expression in individuals

with identical genetic constitution. Acta Dermatol 1982; 62: 229-36.

[168] Henseler T, Christophers E. Psoriasis of early and late onset char-acterization of two types of psoriasis vulgaris. J Am Acad Derma-

tol 1985; 13(3): 450-6. [169] Swanbeck G, Inerot A, Martinsson T, Wahlström J. A population

genetic study of psoriasis. Br J Dermatol 1994; 131: 32—9. [170] Guedjonsson JE, Karason A, Antonsdottir AA, et al. HLA-CW6-

positive and HLA-CW6-negative patients with psoriasis vulgaris have distinct clinical features. J Invest Dermatol 2002; 118: 362-5.

[171] Gottlieb AB, Krueger JG. HLA region genes and immune activa-tion in the pathogenesis of psoriasis. Arch Dermatol 1990; 126:

1083-6. [172] Tiilikainen A, Lassus A, Karvonen J, Vartiainen P, Julin M. Psoria-

sis and HLA-Cw6.Br J Dermatol 1980; 102: 179-84. [173] Mallon E, Bunce M, Savoie H, et al. HLA-C and guttate psoria-

sis.Br J Dermatol 2000; 143: 1177-82. [174] Nair RP, Henseler T, Jenisch S, et al. Evidence for two psoriasis

susceptibility loci (HLA and 17q) and two novel candidate regions (16q and 20p) by genome wide scan. Hum Mol Genet 1997; 6:

1349-56. [175] Matthews D, Fry L, Powles A, et al. Evidence that a locus for

familial psoriasis maps to chromosome 4q. Nat Genet 1996; 14: 231-3.

[176] Speckman RA, Daw J, Helms C, et al. Novel cluster of immuno-globulin superfamily members mapping to a region of 17q25.1

linked to psoriasis susceptibility. Hum Genet 2002; 112: 34-41. [177] Enlund F, Samuelsson L, Enerback C, et al. Psoriasis susceptibility

locus in chromosome region 3q21 identified in patients from southwest Sweden. Eur J Hum Genet 1999; 7: 783-90.

[178] Capon F, Novelli G, Semprini S, et al. Searching for psoriasis susceptibility genes in Italy Genome scan and evidence for a new

locus on chromosome 1. J Invest Dermatol 1999; 112: 52-5. [179] Trembath RC, Clough RL, Rosbotham JL, et al. Identification of a

major susceptibility locus on chromosome 6p and evidence for fur-ther disease loci revealed by a two stage genome-wide search in

psoriasis. Hum Mol Genet 1997; 6: 813-20. [180] Enlund F, Samuelsson L, Enerback C, et al. Analysis of three sug-

gested psoriasis susceptibility loci in a large Swedish set of families confirmation of linkage to chromosome 6p (HLA region) and to

17q, but not to 4q. Hum Hered 1999; 49: 2-8. [181] Nair RP, Stuart P, Henseler T, et al. Localization of psoriasis-

susceptibility locus PSORS1 to a 60-kb interval telomeric to HLA-C. Am J Hum Genet 2000; 66:1 833-44.

[182] Oka A, Tamiya G, Tomizawa M, et al. Association analysis using refined microsatellite markers localizes a susceptibility locus for

psoriasis vulgaris within a 111 kb segment telomeric to the HLA-C gene. Hum Mol Genet 1999; 8: 2165-70.

[183] Tomfohrde J, Silverman A, Barnes R, et al. Gene for familial pso-riasis susceptibility mapped to the distal end of human chromo-

some 17q. Science 1994; 264: 1141-5.

Page 15: Insight Into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery

Insight into the Biomarkers as the Novel Anti-Psoriatic Drug Discovery Tool Current Drug Discovery Technologies, 2012, Vol. 9, No. 2 15

[184] Lee YA, Roschendorf F, Windemuth C, et al. Genome wide scan in

German families reveals evidence for a novel psoriasis-susceptibility locus on chromosome 19p13. Am J Hum Genet 2000;

67: 1020-4. [185] Portugal M, Barak V, Ginsburg I, Kohen R. Interplay among oxi-

dants, antioxidants, and cytokines in skin disorders present status and future considerations. Biomed Pharmacother 2007; 61: 412-22.

[186] Kunz , Manfred . Current Treatment of Psoriasis with Biologics. Current Drug Discovery Technologies, 2009; 6: 231-40.

[187] Fridovich I. Superoxide radical and superoxide dismutases. Annu Rev Biochem1995; 64: 97-112.

[188] Thérond P, Gerbaud P, Dimon S, Anderson WB, Evain-Broin D, Raynaud F. Antioxidant enzymes in psoriatic fibroblasts and eryth-

rocytes. J Invest Dermatol 1996; 106: 1325-28. [189] Yildirim M, Inaloz HS, Baysal V, Delibas N. The role of oxidants

and antioxidants in psoriasis. J Eur Acad Dermatol Venereol 2003; 17: 34–36.

[190] Brigelius-Flohe R. Tissue-specific functions of individual glu-tathione peroxidases. Free Radic Biol Med 1999; 27: 951-65.

[191] Arner ES, Holmgren A. Physiological functions of thioredoxin and thioredoxin reductase. Eur J Biochem 2000; 267: 6102-9.

[192] Schallreuter KU, Wood JM. Thioredoxin reductase, its role in epidermal redox status. J Photochem Photobiol 2001; 64: 179-84.

[193] Foss FM. DAB (389) IL-2 (denileukin diftitox, ONTAK) a new fusion protein technology. Clin Lymphoma 2000; 1(Suppl. 1): S27-

31. [194] Krueger JG, Walters IB, Miyazawa M, et al. Successful in vivo

blockade of CD25 (high-affinity interleukin 2 receptor) on T cells by administration of humanized anti-Tac antibody to patients with

psoriasis. J Am Acad Dermatol 2000; 43: 448-58. [195] Salim A, Emerson RM, Dalziel KL. Successful treatment of severe

generalized pustular psoriasis with basiliximab (interleukin-2 re-ceptor blocker). Br J Dermatol 2000; 143: 1121-2.

[196] Owen CM, Harrison PV. Successful treatment of severe psoriasis with basiliximab, an interleukin-2 receptor monoclonal antibody.

Clin Exp Dermatol 2000; 25: 195-7. [197] Gordon KB, Papp KA, Hamilton TK, et al. Efalizumab for patients

with moderate to severe plaque psoriasis: a randomized controlled trial. JAMA 2003; 290: 3073-80.

[198] Da Silva AJ, Brickelmaier M, Majeau GR, et al. Alefacept an im-munomodulatory recombinant LFA-3/IgG1 fusion protein induces

CD16 signaling and CD2/CD16-dependent apoptosis of CD2 ( ) cells. J Immunol 2002; 168: 4462-71.

[199] Krueger G. Clinical response to alefacept results of a phase 3 study of intravenous administration of alefacept in patients with chronic

plaque psoriasis. J Eur Acad Dermatol Venereol 2003; 17(Suppl. 2): 17-24.

[200] Langley R, Roenigk H, McCall C. Phase I results of intravenous MEDI-507 an anti-T-cell antibody for the treatment of psoriasis. J

Invest Dermatol 2001; 117: 817. [201] Kimball AB, Kawamura T, Tejura K, et al. Clinical and immu-

nologic assessment of patients with psoriasis in a randomized dou-ble-blind placebo-controlled trial using recombinant human inter-

leukin -10. Arch Dermatol 2002; 138: 1341-6. [202] Chen DM, Gordon K, Leonardi C, et al. Adalimumab efficacy and

safety in patients with moderate to severe chronic plaque psoriasis preliminary findings from a 12 week dose ranging trial. J Am Acad

Dermatol 2004; 50(Pt II): PS 491.

[203] Trepicchio WL, Bozza M, Pedneault G, Dorner AJ Recombinant

human IL-11 attenuates the inflammatory response through down-regulation of proinflammatory cytokine release and nitric oxide

production. J Immunol 1996; 157: 3627-34. [204] Gottlieb AB, Evans R, Li S, et al. Infliximab induction therapy for

patients with severe plaque-type psoriasis a randomized double-blind placebo-controlled trial. J Am Acad Dermatol 2004; 51: 534-

42. [205] Leonardi CL, Powers JL, Matheson RT, et al. Etanercept as mono-

therapy in patients with psoriasis. N Engl J Med 2003; 349: 2014-22.

[206] Zhou X, Krueger JG, Kao MC. Abgenix discontinues treatment after disappointing psoriasis study results. East Bay Business

Times 2002. [207] Griffiths CE, Strober BE, Van De Kerkhof P, et al. Comparison of

ustekinumab and etanercept for moderate-to-severe psoriasis. N Engl J Med 2010; 362: 118-28.

[208] Kimball AB, Gordon KB, Langley RG, Menter A, Chartash EK, Valdes J. Safety and efficacy of ABT- 874, a fully human interleu-

kin 12/23 monoclonal antibody in the treatment of moderate to se-vere chronic plaque psoriasis results of a randomized placebo-

controlled phase 2 trials. Arch Dermatol 2008; 144: 200-7. [209] Safety and efficacy of subcutaneous certolizumab pegol, a new

anti-TNFa monoclonal antibody, in patients with moderate-to-severe chronic plaque psoriasis: Preliminary results from a double-

blind, placebo-controlled trial. Jean-Paul Ortonnne, University of Nice-Sophia Antipolis, Nice, France; Chantal Tasset.

[210] Kim HS, Cho IH, Kim JE, Shin, et al. Ethyl pyruvate has an anti-inflammatory effect by inhibiting ROS-dependent STAT signaling

in activated microglia. Free Radic Biol Med 2008; 45: 950-63. [211] Nelson KC, Carlson JL, Newman ML, et al. Effect of dietary in-

ducer dimethylfumarate on glutathione in cultured human retinal pigment epithelial cells. Invest Ophthalmol Visual Sci 1999; 40:

1927-35. [212] Burton G, Ingold K. Mechanisms of Antioxidant Action Preventive

and Chain- Breaking Antioxidants. CRC press, Boca Raton; 1989. [213] Becker BF, Reinholz N, Leipert B, Raschke P, Permanetter B,

Gerlach E. Role of uric acid as an endogenous radical scavenger and antioxidant. Chest 1991; 100: 176S-81S.

[214] Varani J, Lateef H, Fay K, Elder JT. Antagonism of epidermal growth factor receptor tyrosine kinase ameliorates the psoriatic

phenotype in organ-cultured skin. Skin Pharmacol Physiol 2005; 18: 123-31.

[215] Keshtgarpour M, Dudek AZ. SU-011248 a vascular endothelial growth factor receptor-tyrosine kinase inhibitor controls chronic

psoriasis. Transl Res 2007; 149: 103-6. [216] Paniagua RT, Sharpe O, Ho PP, et al. Selective tyrosine kinase

inhibition by imatinib mesylate for the treatment of autoimmune ar-thritis. J Clin Invest 2006; 116: 2633-42.

[217] Distler JH, Jungel A, Huber LC, et al. Imatinib mesylate reduces production of extracellular matrix and prevents development of ex-

perimental dermal fibrosis. Arthritis Rheum 2007; 56: 311-22. [218] Miyagawa S, Fujimoto H, Ko S, Hirota S, Kitamura Y.. Improve-

ment of psoriasis during imatinib therapy in a patient with a metas-tatic gastrointestinal stromal tumor. Br J Dermatol 2002; 147: 406-

7. [219] Cameron D, Stein S. Drug insight intracellular inhibitors of HER2

clinical development of lapatinib in breast cancer. Nat Clin Pract Oncol 2008; 5: 512-20

Received: August 25, 2011 Revised: September 05, 2011 Accepted: September 28, 2011