Open access peer-reviewed chapter

Psoriasis and Genetics

Written By

Hız Meliha Merve, Kılıç Sevilay, Oymak Sibel, Büyük Başak, Canbey Göret Ceren, Tuba Demirci and Akı Cüneyt

Submitted: 23 September 2016 Reviewed: 06 March 2017 Published: 05 July 2017

DOI: 10.5772/intechopen.68344

From the Edited Volume

An Interdisciplinary Approach to Psoriasis

Edited by Anca Chiriac

Chapter metrics overview

1,714 Chapter Downloads

View Full Metrics

Abstract

Psoriasis is an erythematous, scaly chronic inflammatory dermatosis and occurs due to altered epidermal differentiation and hyperproliferation due to faulty signals that speed up the growth cycle of skin cells. Psoriasis reduces quality of life, and psoriatic patients generally have higher risk for metabolic disease. Psoriasis is associated with many burdening comorbidities, which often share similar pathogenic features and follow a progressive pattern. Genetic variation in human genome causes specific kind of disease, and nowadays, research is focused on metabolic pathways that trigger psoriasis and related comorbidities. In addition, genetic variations are also important for psoriasis treatment regime and response. The purpose of this section is to shown to genetic epidemiology, pharmacogenetics, immune genetics of psoriasis and related comorbidities.

Keywords

  • psoriasis
  • genetics
  • variations
  • SNP

1. Introduction

Psoriasis was first thought to originate from a biochemical imbalance due to excessive proliferation of keratinocytes; yet recent researches focused on the investigation of the genetic basis of this immunological condition.

Psoriasis, a chronic inflammatory disease, is thought to be closely related to metabolic and cardiovascular diseases; however, mortality rate of the disease is rare. Genetic and epidemiologic studies have also pointed out that cardiovascular and metabolic diseases are more prevalent in the community than psoriasis cases in psoriasis cases. Thus, determination of genetic variation on genome that triggers both psoriasis and related comorbidities is crucial. In addition, personal genomic variations are important for diagnosis and treatment of the psoriasis and related comorbidities effectively.

Advertisement

2. Epidemiology of psoriasis

Psoriasis can affect people of all ages, ethnic origins and genders around the world. There are very literatures showing the incidence of the disease, since there is no compulsory retention of psoriasis case recordings and reliable source constraints. On the other hand, prevalence varies among different countries in Europe [1]. Although psoriasis may affect 2% of the world population, the incidence of disease varies 0.09–11.4% between different countries and races [14]. The reasons for the variability of prevalence are that the epidemiological methods and limitations of the studies differ [2]. Psoriasis is not evident among Latin American indigenous, people of Samoa and New Zealand, and in contrast, the prevalence is increasing up to 12% in Kasachy [5]. The prevalence of psoriasis in Asian countries or Asians common countries is 0.4% in China, 0.3–1% in Japan, and 0.8% in India, while the prevalence of West African and American black population was reported as 0.3–0.7% and 0.7%, respectively [6].

The results have shown that there is a very weak correlation between geographical latitude and psoriasis [2, 7]. In contrast, psoriasis is more commonly seen in places where the upper latitudes of that are colder and Caucasians are more frequent[5, 810]. For example, in twin studies carried out in Australia, it has a higher prevalence of psoriasis in southern region of cold than the northern region where is warmer [8, 9]. When investigating the regional distribution of psoriasis cases in Norway, the incidence of disease has been found to increase inversely proportional to the temperature rise in the country [10].

According to the global disease burden study conducted in 2010 [11], it has been reported that the disability‐adjusted life year increases with age, especially the highest burden of disease shown between the ages of 50–69 [2, 11]. Psoriasis is associated with many burdening comorbidities, which often share similar pathogenic features and follow a progressive pattern, and thus, direct and indirect cost of psoriasis has significant effects on both patients and health care systems.

Advertisement

3. Psoriasis as a genetic disease

Genetic factors play a role in psoriasis development; yet, the exact mechanism that triggers psoriasis is still unknown. Twin studies, familial aggregation studies, linkage studies, and population‐based association studies are all important to understand the effect of the genes in heritability of the disease. Nowadays, genome‐wide association studies and linkage studies are also frequently used to identify disease mechanism and risk‐related genes. Association studies on genome‐wide and a meta‐analysis aim to reveal the potential risk genes and loci of genome that emerged comorbidities as a result of these diseases such as s psoriasis.

Psoriasis is a complex disease that not simply explained by Mendelian inheritance rules. Lomholt’s study that performed (1954) in an island has determined that 91% of psoriatic patients have a previous family history [12]. Earlier studies speculated psoriasis as autosomal dominant disease, but the truth is so different [13]. This speculation relies on the fact that human lymphocyte antigen gene located in a single locus thus its effects as dominant. That is why psoriasis is accepted as a dominant disease [13]. Although dominant inheritance firstly goes to foreground in studies to investigate the pattern of inheritance of psoriasis, it has been focused on the disease transition by heterogeneous or polygenic in broader studies.

The twin studies rely on the fact, if the disease occurs in monozygotic twins because of phenotypical differences between monozygotic twins. This means that environmental factors affect the disease because they have identical genetic background. In contrast, dizygotic twins have genetic background like brothers or sisters, and thus, phenotypical differences occur as a result of genetic or environmental factors or both.

The concordance of psoriasis has greater rates in monozygotic twins than dizygotic twins’; thus, genetic factors are important for disease development. In contrast, the rate is never reaching to 100% in monozygotic twins so environmental influences for disease development. Duffy et al. [8] have reported concordance for psoriasis higher in monozygotic twins (35%) when compared to dizygotic twins (12%) in Australian population. The data from National Twin Registry System of Denmark had been shown that the concordance of psoriasis in dizygotic twins was 15–30%; yet, the rates rises up to 65–72% in monozygotic twins [14]. The Danish Twin Registry System records have shown that monozygotic twins increased risk of psoriasis than in dizygotic twins [15]. A twin study has been shown that psoriasis risk has been rise up to 3 times in monozygotic twins when compared to fraternal twins [16]. Although twin studies have shown strongest genetic background of psoriasis, heritability rate of psoriasis never reaches to 100% among monozygotic twins. This missing heritability means environmental factors [15, 16] and epigenetic alterations [17] have effects on psoriasis development.

Psoriasis was diagnosed 6% in first‐degree relatives of patients with psoriasis [18, 19] generations, settlement in geographical regions and ethnic group studies related to population showed that psoriasis is a complex genetic‐based, multifactorial disease [6]. All these data showed why psoriasis was accepted as a heritable disease but not 100% as a genetic disease. Psoriasis was first associated with the major histocompatibility complex (MHC) region in the short arm of the 6th chromosome in 1972 [2022]. MHC is an intense region of the genome and contains HLA‐I and HLA‐class human leukocyte antigens. In fact, the 80–200 kb long region in the HLA‐C gene region has been accepted as the Psoriasis susceptibility gene region (PSOR1) [23]. HLA‐Cw6 in PSOR1 region was associated with psoriasis in many ethnic groups and geographical regions [2427]. However, the risk zones related to subclinical forms of the disease vary in patients with psoriasis. For example, HLA‐Cw6 positivity is associated with early onset and familial transition of psoriasis [28]. HLA‐CW6 gene region affects the response of ustekinumab using in the treatment of psoriasis [29]. Other gene regions (PSORS2 (17q25), PSORS3 (4q34), PSORS4 (1q), PSORS5 (3q21, PSORS6 (19p13), PSORS7 (1p) PSORS8 (16q12‐13), PSORS11 (5q31.1‐q33.1), PSORS12 (20q13), and PSORS13 (6q21)) related to psoriasis distribute to different chromosomes [13, 3037].

Advertisement

4. Immunogenetics of psoriasis

Psoriasis is an immune‐mediated disease and, for that reason, immune‐mediated mechanism has been extensively studied. Genetic studies focus on identification reasons that trigger uncontrolled proliferation of keratinocytes and recruitment of T cells into the skin. In healthy individuals, T cells are originated from embryonal pluripotent cells by cytokine induction. Immature thymocytes are transferred to thymus for proliferation through clonal expansion. T cells are specified to helper T cells or cytoxic T cells by adding specific surface antigens (CD4+ or CD8+) and differentiating to give response for antigenic signals. In psoriatic skin samples, Th1 cell differentiation is up‐regulated and that cause impaired Th1/Th2 balanced [38, 39]. Evidence indicates that Th1‐delivered cytokines IL‐1, IL‐2, IL‐6, IL‐8, tumor necrosis factor‐α (TNF‐ α), transforming growth factor (TGF) and Granulocyte‐macrophage colony‐stimulating factor are upregulated, while Th2‐delivered cytokines IL‐4, IL‐5, and IL‐10 are downregulated [4044]. The cytokines IL‐12 is critical for both T‐cell activation and differentiation also contributes to DC/T cell interactions [45]. TNIP1, NFKBIA, IL12B and LCE3D–LCE3E are known susceptibility loci for psoriasis [46], and the variation of IL‐12 is associated with psoriasis in different populations [4749]. Meta analyses have shown that IL‐12B variation (rs3212227, rs6887695) has been associated both psoriasis and psoriatic arthritis [50]. IL‐12 and IL‐23 are heterodynamic cytokines; both of them share same p40 subunit. IL‐12 consists of p35 and p40 subunit and IL‐23 consist of p19 and p40 subunits. The metabolic model for psoriasis suggests that dermal dendritic cells are secreted IL‐23 by that way trigger Th17 cell line activation. IL‐23 is crucial for naive T cell differentiation to Th17. Activated Th17 cells are secreted IL‐17A, IL‐17F, IL‐22 and IL‐26 and that proinflammatory cytokines effects epidermal and dermal cells gene expression and keratinocyte maturation. For that reason, IL12/IL 23 pathway seems important for psoriasis treatment and different studies are performed in that metabolic pathway [5154].

Advertisement

5. Immunohistochemical parameters

The demonstration of specific markers by immunohistochemical methods in histopathological tissue specimens diagnosed as psoriasis is important in terms of supporting and confirming the diagnosis. We have evaluated most of the cytokines referred to in the pathophysiology in immunogenetic section which can be used for immunohistochemical methods. In psoriatic tissue, sample Th1‐mediated cytokines IL‐1, IL‐2, IL‐6, IL‐8, tumor necrosis factor‐α (TNF‐α), transforming growth factor (TGF) and granulocyte‐macrophage colony‐stimulating factor increase and Th2‐mediated cytokines IL‐4, IL‐5, and IL‐10 decrease [4044].

IL‐17 plays an important role in psoriasis, and recently, both IL‐17 inhibitors and IL‐17receptor type A blockers are new monoclonal antibodies which are approved by US FDA [55]. Lee and colleagues have immunohistochemically evaluated IL‐17A expressions in different types of psoriasis, and the results have shown that samples from palmoplantar and pustular type psoriasis samples were stained with IL‐17A strongly, while plaque type samples were stained with IL‐17A intermittently [56].

Macrophage migration inhibitory factor (MIF) is directly related to the severity of clinical symptoms, and serum MIF levels are usually high in patients with psoriasis [57]. In contrast, immunohistochemical staining by anti‐MIF antibody in psoriatic lesions showed that MIF levels are very low [58]. Toll‐like receptors (TLRs) are a group of molecules that play a crucial role in innate immunity and also regulate antimicrobial defense. TLR2 is immunohistochemically staining uniformly in all the layers of the normal deep epidermis that does not lose its nuclei, yet TLR2 staining differs in psoriatic skin samples [59, 60]. Panzer and colleagues have shown that TLR2 staining is lower in granular and upper spinous, whereas it was strongly observed in the lower spinous and basal strata [59]. TLR4 is normally found in all the nuclear layers, while it is more pronounced in the upper layers of the basal layer. In psoriatic skin lesions, TLR4 was stained weak in the basal layer, yet TLR4 was stained strongly in the upper spinous and granular layers [59]. IL‐8 is produced in psoriatic lesions and immunohistochemical demonstration supports disease diagnosis [61]. In a study, evaluating the relationship between geographic tongue (GT) and psoriasis has been shown that anti‐CD4, CD8, CD20, CD68, S100, and Ki‐67 antibodies results were similar in tissue samples from both diseases, and according to that result, geographic tongue is speculates as an oral psoriasis lesion [62].

Advertisement

6. Pharmacokinetics of psoriasis in genetic aspect

Psoriasis is a chronic, non‐curative disease, and thus, effective psoriasis treatment is important for long‐term cure and inhibition of disease manifestation. Psoriasis can be treated by topical, photo(chemo)therapy, classic systemic drug(methotrexate, cyclosporine, acitretin) and biologic agents (infliximab, adalimumab, etanercept). The psoriasis treatment is affected by psoriasis type, severity, presence of comorbidities, previous response to other treatments and patients.

Ustekinumab is an anti‐p40 monoclonal antibody which is effective for psoriasis treatment via its inhibitor effect of interleukin‐12/23 pathway. By that way, ustekinumab inhibits T helper (Th)1 and Th17 pathway and inhibits immune cell activation. In addition, HLA‐Cw+ patients have better clinical response to ustekinumab than HLACw‐ ones [29]. Talamonti et al. state that HLACw+ patients response to ustekinumab is 9.8 times more efficient than mutant genotype. In contrast, TNFAIP3 polymorphism and LCE3B/3C gene deletions have no effect on ustekinumab response [29].

TNF‐inhibitors infliximab and adalimumab and the fusion protein, etanercept are used widely for psoriasis treatment. Nishikawa and colleagues [63] have shown that JAG2, ADRA2A and TLR10 polymorphisms are associated with TNFα response. Prieto‐Pérez and colleagues [64] have shown that PGLYR4, ZNF816A, CTNNA2, IL12B, MAP3K1, and HLA‐C genes are associated with anti‐TNF response at 3 months, while FCGR2A, HTR2A, and CDKAL1 are related anti‐TNF response at 6 months. TNF gene polymorphism is also associated with psoriasis and psoriatic arthritis due to its effects on TNF production. Murdaca and colleagues have demonstrated TNF‐α polymorphism on +489 position of the gene effects efficiency of etanercept treatment on psoriatic arthritis patients (PsA) [65]. In fact, the idea simply relies on the fact that gene polymorphism affects the biological properties of encoded proteins. TNF blockers are widely used for regulation of inflammatory cascade and by that way treatment of the immune‐related disease. If polymorphic product of the gene is your pharmaceutical target and has different molecular structure, then wild type its means pharmacodynamics of the drug varies due to personal variation.

Both IL‐17 antagonists and IL‐17 receptor blockers are accepted as effective and safe for psoriasis treatment [66]. Brodalumab is a human monoclonal antibody that binds to interleukin‐17 receptor and waits for US‐FDA approval for psoriasis treatment [55, 67]. IL‐17 expression is upregulated in psoriatic skin samples. Both secukinumab and ixekizumab are the cytokine antagonists used for psoriasis treatment [68]. Nowadays, recent studies are focused on the relationship between IL‐17 family cytokines (IL‐17A, IL‐17B, IL‐17C, IL‐17D, IL‐17E, and IL‐17F) and IL‐17 receptor polymorphisms with psoriasis susceptibility and treatment response [6971].

Advertisement

7. The link between psoriasis and comorbidities in view of genetics

Nowadays, psoriasis is accepted as a systemic disease because of higher rates of obesity, depression, psychiatric disorders, diabetes, hyperlipidemia, cardiovascular diseases, arthritis, uveitis, ulcerative colitis, Crohn’s disease, and metabolic syndrome among psoriatic patients. Even if, the mortality rate of the psoriasis is low, psoriatic patients over 65 have at least two comorbidities [72]. Thus, interdisciplinary approach should be an integral part of the routine, and also the dermatologist should be careful with psoriasis and related disorders. Although the systemic inflammation process during psoriasis development is still unclear, the chronic inflammatory process may trigger development of many burden comorbidities in psoriatic patients. Many epidemiological studies revealed relationship between psoriasis and diabetes [72, 73]. Aside, the studies have shown that cardiovascular risk increases among patient with psoriasis and psoriatic arthritis [74]. In that point, dermatologist should be careful about systemic drug usage for psoriasis treatment because medications can sometime trigger maintenance factor in the pathogenesis of psoriasis [7578].

In that section, we would like to evaluate the common mechanism between psoriasis and psoriatic comorbidities in molecular and genetic basis. For example, psoriatic patients are more prone to develop Crohn’s disease more than general population [79]. The molecular basis of Crohn’s disease is activation of CD4+ T cells by intestinal epithelial cells (IECs) and lamina propria immune cells and that activate both Th1 and Th17cells and cause elevated level of TNFα and Th17‐related cytokine. Th1 and Th17 differentiation is common both in psoriasis and Crohn’s disease; for that reason, TNF inhibitors are effective for treatment of Crohn’s disease as psoriasis [80].

7.1. Psoriatic arthritis

Joint involvements in patients with psoriasis were put forward for the first time by Alibert in 1818 [81, 82]. Psoriatic arthritis is a negative spondyloarthropathy [7183]. Important radiological findings of the psoriatic arthritis are peripheral joint involvement, destructive and proliferative changes, spinal involvement, interphalangeal joint ledge, fibroosteitis and acroostelolysis [84, 85]. Strong genetic relationship is remarkable between positive family history [81, 86] and human leukocyte antigen (HLA) class II alleles for psoriatic arthritis [8789].

Psoriatic arthritis (PsA) is diagnosed in 25–34% of psoriatic patients [90]. Psoriasis without joint involvement (PsO) and psoriatic arthritis (PsA) occur as a result of autoimmune responses that are triggered by genetic and environmental factors. The pathogenesis of psoriasis only (PsO) and psoriatic arthritis (PsA) may be explained impaired cytokine levels [91].

To clarify immungenetics mechanism of psoriatic arthritis has been focused on human leukocyte antigen (HLA). HLA gene region is located on the short arm of chromosome 6. This region is highly polymorphic depending on mutation and recombination [92]. Psoriatic arthritis associates with various HLA risk haplotype such as HLA‐B7 B57 B13, B16, B17, DR7, B38, B39, CW6, and B27 [89, 93, 94]. While CW6 HLA‐alleles were associated with early onset of the disease [95], HLA‐B38 and HLA‐B39 were associated with polyarthritis [87], and HLA‐B27 was associated with dorsal involvement. Positivity of human leukocyte antigen “HLA‐B27” in psoriatic arthritis patients makes a higher risk of arthritis compared to other HLA alleles [90, 96]. In Turkish population, we have found that HLA‐B27 positivity increased the risk of psoriasis and psoriatic arthritis 5 to 10 times more, respectively [97]. There are various molecular‐based hypotheses such as molecular mimicry, defective immunity, heavy chain folding incorrectly, and deterioration of HLA‐B27 allele for relationship between HLA‐B27 and psoriatic arthritis (PsA) [97].

7.2. Obesity

Storing energy in the form of triacylglycerols is considered to be the primary task of adipocytes; but adipocytes have physically some additional functions such as preservation of internal organs, regulating temperature, storage of lipid‐soluble vitamins, steroid metabolism, immunity, and being assigned in inflammatory processes [98, 99]. Adipose tissue runs like as an endocrine tissue as the sympathetic system regulators with hormone and cytokine production during the regulation of energy metabolism [100]. Adipose tissue plays also an important role in homeostasis besides production, secretion such as TNF‐alpha, IL‐8, monocyte chemoattractant protein 1, cytokines and chemokines and storing energy in the form of fatty acids, together with the adipokines, leptin, adiponectin, resistin, such as pro‐inflammatory and anti–inflammatory factors [98, 101].

The risk of diseases such as a high rate of insulin resistance, type 2 diabetes, dyslipidemia, gout, hypertension, and cardiovascular involvement is higher in obese individuals [102107, 110]. Adipokine tissue secretes cytokines thus increasing volume or amount of adipokine tissue cause increasing the amount of circulating cytokines. Then, elevated amount of cytokines causes arteriosclerosis and insulin resistance and to induce hypertension and type 2 diabetes [108].

In a study that investigated the risk of obesity in patients with psoriasis, same sex siblings and 88 psoriasis outpatients were compared by calculating the BMI. The result of this study has obtained a significant relationship between the severity of psoriasis and BMI [109]. Duarte et al. have found that it was a statistically significant relationship between obesity and psoriasis severity on patients with psoriasis by using different parameters, such as BMI and waist/hip ratio [110]. It was shown to be positive commensurate with psoriasis risk of weight gain and high BMI in another study performed in the United States with 809 psoriasis patients followed for 12 years [111]. Obesity was determined to be more than psoriasis only (PSO) patients [112114], and it was determined that obesity increases the risk of psoriatic arthritis in individuals under 18 years of age [115].

7.3. The impaired glucose tolerance and diabetes

Glucose intolerance, diabetes, and obesity are more frequent in patients with psoriasis [74, 75, 116119]. The study conducted by Onsun et al. has found that diabetes was significantly more frequent in patients with psoriasis when compared to the incidence of psoriasis in patients with diabetes [120]. Azfar and his colleagues have found that patients with severe psoriasis involvement have more risk of with diabetes; in contrast, they concluded that psoriasis was an independent risk factor for type 2 diabetes [121]. Armesto and colleagues have found that occurrence of risk of type 2 diabetes was found to have higher, late‐onset, nonfamilial transmission, but in patients with joint involvement [122]. Finley et al. have shown that expression of IL‐17 and IL‐20 is elevated in diabetic wounds, and they emphasized the psoriasis and diabetes link via cytokines [123]. Granata et al. have demonstrated that IL‐23/IL‐17, and IL‐18 are main cytokines that has central role for obesity, type 1 diabetes, and psoriasis [124]. Eirís et al. have demonstrated that IL12B rs6887695 and rs3212227 increase the type 2 diabetes risk approximately 3 and 6 times, respectively [125]. In addition, IL23 receptor polymorphism was also found significantly associated with diabetes in psoriatic patients [125]. Presta et al. have found that polymorphism in the 3′untranslated region of IL‐18 gene increases the insulin sensitivity risk [126]. In addition to literature, we have evaluated the diabetes risk among psoriasis patients; yet, we could not find any significant relation between TNF‐α‐238 G/A and TNF‐α‐308 G/A polymorphisms with diabetes [127].

7.4. Cardiovascular diseases

The epidemiological studies have shown that cardiovascular morbidity and mortality increased in patients with psoriasis in Refs. [77, 78, 128, 129]. Inflammation located in pathogenesis of psoriasis is known to cause metabolic and cardiovascular disease. The molecular relationship between psoriasis and cardiovascular disease may be explained by T cell activation and enhance of Th1 cytokine production (TNF‐α, IL‐1β, IL‐10, and IFN). The TNF‐α and IL‐1 trigger oxidative stress causes migration of leukocytes into atherosclerotic plaques [101], so TNF inhibitors have beneficial effects on prevention of cardiovascular events both in psoriasis and psoriatic arthritis [130]. Our study group has evaluated hypertension risk and psoriasis. eNOS Glu298Asp single‐nucleotide polymorphism has found to be increased in hypertensive psoriatic patients when compared with healthy volunteers [131]. In contrast, we could not find relationship between TNF‐α‐298 and TNF‐α‐308 polymorphisms with hypertension and cardiovascular disease [127].

7.5. Metabolic syndrome

Metabolic syndrome is associated with conditions such as physical inactivity and improper diet, occurring nutrition changes, and daily lifestyle. It is characterized as a disease of civilization by high mortality and morbidity [132].

It is a fact that metabolic syndrome and its components are often seen in patients with psoriasis. Also, polymorphism may be triggering both psoriasis and metabolic syndrome together. Abdel Hay and Rashed have shown that leptin gene 2548G/A polymorphism increases risk of both psoriasis and metabolic syndrome [133].

7.6. Psychological trauma and addiction

Psoriasis is a chronic skin disease that negatively affects the quality of life in many ways. Patients with psoriasis reduce quality of life with comorbidities like arthritis. Psoriasis can lead not only to physical health problems, but also to mental and social health problems. Psoriatic lesions that occur especially in hands, face, and on the scalp cause shame, embarrassment, and social inhibition in social life and by that way increase the risk of depression, anxiety, lack of self‐confidence, smoking and alcohol addiction, and trend of suicidal thoughts in patients with psoriasis [134].

Kurd et al. have shown that depression, anxiety, and suicidality risk increases 1.39, 1.31, and 1.44 times more in psoriatic patients than healthy people, respectively [135]. Psoriatic patients are more prone to psychiatric comorbidities and suicidal ideation than other dermatological diseases [136]. Deveci and et al. have found significantly higher depression rate and suicide attempts in cases with psoriasis when compared to patients with other dermatological problems [137].

Use of alcohol and tobacco makes difficult the therapeutic harmony and also increases the toxicity of the drug besides reducing the effectiveness of systemic anti‐psoriatic drug. Excessive alcohol intake can induce production of proinflammatory cytokine in various cell types and increase the cell lymphocyte proliferation and activation [138]. Serotonin may be triggered psoriasis as a growth factor that promotes keratinocyte proliferation [139]. The serotonin transporter gene promoter region and serotonin2A receptor gene have been found associated with psoriasis in Thai population [140, 141].

Advertisement

8. Conclusions

What would cause the metabolic alteration among psoriatic patients? Does systemic drug usage in long term causes comorbidities in patients with psoriasis? or Is it the domino effect of impaired metabolism? It has not reached a consensus about it.

Advances in understanding the immune‐mediated pathological mechanisms of psoriasis will be highlighted with the relationship between psoriasis and psoriasis‐related comorbidities. Much psoriatic comorbidity may be triggered by the cytokines and chemokines, which were secreted by T‐cells, keratinocytes, and antigens presenting cells after keratinocyte hyper proliferation. Hence, molecular approaches are important to identify the link immunogenic mechanism with psoriasis‐related comorbidities in view of molecular pathways.

Determinations of genomic risk profile of the psoriatic patients allow diagnosing of psoriasis and its related comorbidities at an early stage; hence, the life quality can be increased by treating them earlier. In addition, evidence‐based information helps physicians to make clear decision and help to minimize medical errors as well as unnecessary drug usage. As a sum, this information allows to increase treatment cost and work loss. In addition, pharmacogenomics of drugs among psoriatic patients is tightly related to personal variations on genome, and thus, complex genetic heterogeneity affects the treatment power.

References

  1. 1. Fernandez‐Obregon A. Psoriasis ‐ A systemic disease. In: Dr. O’ Daly J, editor. Psoriasis. InTech; 2012. pp. 159‐216. DOI: 10.5772/25894
  2. 2. WHO Library Cataloguing‐in‐Publication Data Global Report on Psoriasis. 1. Psoriasis—Epidemiology. World Health Organization. ISBN 978 92 4 156518 9
  3. 3. Gibbs S. Skin disease and socioeconomic conditions in rural Africa: Tanzania. International Journalof Dermatology. 1996;35(9):633‐639
  4. 4. Danielsen K, Olsen AO, Wilsgaard T, Furberg AS. Is the prevalence of psoriasis increasing? A 30–year follow‐up of a population–based cohort. British Journal of Dermatology. 2013;168:1303‐1310
  5. 5. Farber EM, Nall ML. Epidemiology: Natural history and genetics. In: Roenigk HH, Maibach HI, editors. Psoriasis. (3rd ed.). New York: Marcel; pp. 107‐158
  6. 6. Campalani E, Barker JNWN. The clinical genetics of psoriasis. Current Genomics. 2005;6(1):51‐60
  7. 7. Jacobson CC, Kumar S, Kimball AB. Latitude and psoriasis prevalence. Journal of the American Academy of Dermatology. 2011;65(4):870‐873
  8. 8. Duffy DL, Spelman LS, Martin NG. Psoriasis in Australian twins. Journal of the American Academy of Dermatology. 1993;29(3):428‐434
  9. 9. Neimann AL, Porter SB, Gelfand JM. The Epidemiology of Psoriasis. Expert Review of Dermatology. 2006;1(1):63‐75
  10. 10. Braathen DL, Botten G, Bjerkedal T. Prevalence of Psoriasis in Norway. ActaDermato‐Venereologica. 1989;142:5‐8
  11. 11. Institute for Health Metrics and Evaluation (IHME). Global Burden of Disease Study 2010: Results by Cause 1990-2010. Seattle: IHME; 2012
  12. 12. Lomholt G. Psoriasis on the Faroe Islands; a preliminary report. ActaDermato‐Venereologica. 1954;34(1-2):92
  13. 13. Tomfohrde J, Silverman A, Barnes R, Fernandez‐Vina MA, Young M, Lory D, Morris L, Wuepper KD, Stastny P, Menter A, et al. Gene for familial psoriasis susceptibility mapped to the distal end of human chromosome 17q. Science. 1994;264(5162):1141‐1145
  14. 14. Wuepper KD, Coulter SN, Haberman A. Psoriasis vulgaris: A genetic approach. Journal of Investigative Dermatology. 1990;95(5):2S‐4S
  15. 15. Lønnberg AS, Skov L, Skytthe A, Kyvik KO, Pedersen OB, Thomsen SF. Heritability of psoriasis in a large twin sample. British Journal of Dermatology. 2013;169(2):412‐416
  16. 16. AlShobaili HA, Shahzad M, Al‐Marshood A, Khalil A, SettinA, Barrimah I. Genetic background of psoriasis. International Journal of Health Sciences. 2010;4(1):23‐29
  17. 17. Gervin K, Vigeland MD, Mattingsdal M, Hammerø M, Nygård H, Olsen AO, Brandt I, Harris JR, Undlien DE, Lyle R. DNA methylation and gene expression changes in monozygotic twins discordant for psoriasis: Identification of epigenetically dysregulated genes. PLOS Genetics, 2012;8(1):e1002454
  18. 18. Li Y, Liao W, Chang M, Schrodi SJ, Bui N, Catanese JJ, Poon A, Matsunami N, Callis‐Duffin KP, Leppert MF, Bowcock AM, Kwok PY, Krueger GG, Begovich AB. Further genetic evidence for three psoriasis‐risk genes: ADAM33, CDKAL1, and PTPN22. Journal of Investigative Dermatology. 2009;129(3):629‐634
  19. 19. Bowcock AM, Cookson WO. The genetics of psoriasis, psoriatic arthritis and atopic dermatitis. Human Molecular Genetics. 2004;13(1):R43‐R55
  20. 20. Trembath RC, Clough RL, Rosbotham JL, Jones AB, Camp RD, Frodsham A, Browne J, Barber R, Terwilliger J, Lathrop GM, Barker JN. Identification of a major susceptibility locus on chromosome 6p and evidence for further disease loci. Human Molecular Genetics. 1997;6(5):813‐820
  21. 21. White SH, Newcomer VD, Mickey MR, Terasaki PI. Disturbance of HL‐Aantigen frequency in psoriasis. The New England Journal of Medicine. 1972;287(15):740‐743
  22. 22. Russell TJ, Schultes LM, Kuban DJ. Histocompatibility (HL‐A) antigens associated with psoriasis. The New England Journal of Medicine. 1972;287(15):738‐740
  23. 23. Zhu KJ, Lv YM, Yin XY, Wang ZX, Sun LD, He SM, Cheng H, Hu DY, Zhang Z, Li Y, Zuo XB, Zhou YW, Yang S, Fan X, Zhang XJ, Zhang FY. Psoriasis regression analysis of MHC loci identifies shared genetic variants with vitiligo. PLoS One. 2011;6(11):e23089. DOI: 10.1371/journal.pone.0023089
  24. 24. Fan X, Yang S, Huang W, Wang ZM, Sun LD, Liang YH, Gao M, Ren YQ, Zhang KY, Du WH, Shen YJ, Liu JJ, Zhang XJ. Fine mapping of the psoriasis susceptibility locus PSORS1 supports HLA‐C as the susceptibility gene in the Han Chin. 2008;4(3):e1000038. DOI: 10.1371/journal.pgen.1000038
  25. 25. Romphruk AV, Oka A, Romphruk A, Tomizawa M, Choonhakarn C, Naruse TK, Puapairoj C, Tamiya G, Leelayuwat C, Inoko H. Corneodesmosingene: No evidence for Psors 1 gene in North‐Eastern Thai psoriasis patients. Tissue Antigens. 2003;62(3):217‐224
  26. 26. Mallon E, Bunce M, Wojnarowska F, Welsh K. HLA‐CW*0602 is a susceptibility factor in type I psoriasis, and evidence ala‐73 is increased in male type I psoriatics. Journal of Investigative Dermatology. 1997;109(2):183‐186
  27. 27. Ikäheimo I Tiilikainen A, Karvonen J, Silvennoinen‐Kassinen S. HLA risk haplotype Cw6,DR7,DQA1*0201 and HLA‐Cw6 with reference to the clinical picture of psoriasis vulgaris. Archives of Dermatological Research. 1996;288(7):363‐365
  28. 28. Christophers E. Henseler T. Psoriasis type I and type II are subtypes of nonpustular psoriasis. Seminars in dermatology. 1992;11(4):261‐266
  29. 29. Talamonti M, Botti E, Galluzzo M, Teoli M, Spallone G, Bavetta M, Chimenti S, Costanzo A. Pharmacogenetics of psoriasis: HLA‐Cw6 but not LCE3B/3C deletion nor TNFAIP3 polymorphism predisposes to clinical response to interleukin 12/23 blocker ustekinumab. British Journal of Dermatology. 2013;169(2):458‐463. DOI: 10.1111/bjd.12331
  30. 30. Huffmeier U., Uebe S., Ekici AB, Bowes J, Giardina E, Korendowych E, Juneblad K, Apel M, McManus R, Ho P, Bruce IN, Ryan AW, Behrens F, Lascorz J, Böhm B, Traupe H, Lohmann J, Gieger C, Wichmann HE, Herold C, Steffens M, Klaresko L, Wienker TF, Fitzgerald O, Alenius GM, McHugh NJ, Novelli G, Burkhardt H, Barton A, Reis A. Common variants at TRAF3IP2 are associated with susceptibility to psoriatic arthritis and psoriasis. Nature Genetics. 2010;2(11):996‐999
  31. 31. Capon F, Bijlmakers MJ, Wolf N, Quaranta M, Huffmeier U, Allen M, Timms K, Abkevich V, Gutin A, Smith R, Warren RB, Young HS, Worthington J, Burden AD, Griffiths CE, Hayday A, Nestle FO, Reis A, Lanchbury J, Barker JN, Trembath RC. Identification of ZNF313/RNF114 as a novel psoriasis susceptibility gene. Human Molecular Genetics. 2008;17(13):1938‐1945
  32. 32. Asumalahti K, Laitinen T, Lahermo P, Suomela S, Itkonen‐Vatjus R, Jansen C, Karvonen J, Karvonen SL, Reunala T, Snellman E, Uurasmaa T, Saarialho‐Kere U, Kere J. Psoriasis susceptibility locus on 18p revealed by genome scan in Finnish families not associated with PSORS1. Journal of Investigative Dermatology. 2003;121(4):735‐740
  33. 33. Zhang XJ, He PP, Wang ZX, Zhang J, Li YB, Wang HY, Wei SC, Chen SY, Xu SJ, Jin L, Yang S, Huang W. Evidence for a major psoriasis susceptibility locus at 6p21(PSORS1) and a novel candidate region at 4q31 by genome‐wide scan in Chinese hans. Journal of Investigative Dermatology. 2002;119(6):1361‐1366
  34. 34. Veal CD, Clough RL, Barber RC, Mason S, Tillman D, Ferry B, Jones AB, Ameen M, Balendran N, Powis SH, Burden AD, Barker JN, Trembath RC. Identification of a novel psoriasis susceptibility locus at 1p and evidence of epistasis between PSORS1 and candidate loci. Journal of Medical Genetics. 2001;38(1):7‐13
  35. 35. Lee YA, Rüschendorf F, Windemuth C, Schmitt‐Egenolf M, Stadelmann A, Nürnberg G, Ständer M, Wienker TF, Reis A, Traupe H. Genomewide scan in German families reveals evidence for a novel psoriasis‐susceptibility locus on chromosome 19p13. The American Journal of Human Genetics. 2000;67(4):1020‐1024
  36. 36. Capon F, Novelli G, Semprini S, Clementi M, Nudo M, Vultaggio P, Mazzanti C, Gobello T, Botta A, Fabrizi G, Dallapiccola B. Searching for psoriasis susceptibility genes in Italy: genome scan and evidence for a new locus on chromosome 1. Journal of Investigative Dermatology. 1999;112(1):32‐35
  37. 37. Enlund F, Samuelsson L, Enerbäck C, Inerot A, Wahlström J, Yhr M, Torinsson A, Riley J, Swanbeck G, Martinsson T. Psoriasis susceptibility locus in chromosome region 3q21 identified in patients from Southwest Sweden. European Journal of HumanGenetics. 1999;7(7):783‐790
  38. 38. Jadali Z, Eslami MB, Mansouri P, Safari R, Bayatian P, Mirshafiey A,SalehiNodeh AR. Th1/Th2 cytokines in psoriasis. Iranian Journalof Public Health. 2007;36(2):87‐91
  39. 39. Craven NM, Jackson CW, Kirby B, Perrey C, Pravica V, Hutchinson IV, Griffiths CE. Cytokine gene polymorphisms in psoriasis. British Journal of Dermatology. 2001;144(4):849‐853
  40. 40. Grossman RM, Krueger J, Yourish D, Granelli‐Piperno A, Murphy DP, May LT, Kupper TS, Sehgal PB, Gottlieb AB. Interleukin 6 is expressed in high levels in psoriatic skin and stimulates proliferation of cultured human keratinocytes. Proceedings of the National Academy of Sciences. 1989;86:6367‐6371
  41. 41. Groves RW, Allen MH, Ross EL, Barker JN, MacDonald DM. Tumournecrosis factor alpha is pro‐inflammatory in normal human skin and modulates cutaneous adhesion molecule expression. British Journalof Dermatology. 1995;132(3):345‐352
  42. 42. Gomi T, Shiohara T, Munakata T, Imanishi K, Nagashima M. Interleukin 1a, Tumor Necrosis Factor a, and Interferon gamma in psoriasis. Archives of Dermatology. 1991;127(6):827‐830
  43. 43. Uyemura K, Yamamura M, Fivenson DF, Modlin RL, Nickoloff BJ. 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(5):701‐705
  44. 44. Konstantinova NV, Duong DM, Remenyik E, Hazarika P, Chuang A, Duvic M. Interleukin‐8 is induced in skin equivalents and is highest in those derived from psoriatic fibroblasts. Journal of Investigative Dermatology. 1996;107(4):615‐621
  45. 45. Lew W, Bowcock AM, Krueger JG. Psoriasis vulgaris: cutaneous lymphoid tissue sup-ports T‐cell activation and “Type 1” inflammatory gene expression. Trends in Immunology. 2004;25(6):295‐305
  46. 46. Zuo X, Sun L, Yin X, Gao J, Sheng Y, Xu J, Zhang J, He C, Qiu Y, Wen G, Tian H7, Zheng X, Liu S, Wang W, Li W, Cheng Y, Liu L, Chang Y, Wang Z, Li Z, Li L, Wu J, Fang L, Shen C, Zhou F, Liang B, Chen G, Li H, Cui Y, Xu A, Yang. Whole‐exome SNP array identifies 15 new susceptibility loci for psoriasis. Nature Communications. 2015;6:6793. DOI: 10.1038/ncomms7793
  47. 47. Haase O, Mosaad H, Eldarouti MA, Elramly AZ, Samir N, Abdelhady MM, Samir M, El‐Gharib I, Salah S, El‐Shennawy FA, Mosaad Y, Elwan N, Salem H, Abdelgaber S, Recke A, Möller S, Zillikens D, Ibrahim S. TNFAIP3 and IL12B gene polymorphisms associated with psoriasis vulgaris in an Egyptian cohort. Journal of the European Academy of Dermatology and Venereology.2015;29(7):1297‐1301
  48. 48. Oka A, Mabuchi T, Ikeda S, Terui T, Haida Y, Ozawa A, Yatsu K, Kulski JK, Inoko H. IL12B and IL23R gene SNPs in Japanese psoriasis. Immunogenetics. 2013;65(11):823‐828
  49. 49. Lee YH, Song GG. Associations between interleukin‐23R and interleukin‐12B polymorphisms and psoriasis susceptibility: A meta‐analysis. Immunological Investigations. 2013;42(8):726‐736
  50. 50. Zhu KJ, Zhu CY, Shi G, Fan YM. Author informationMeta‐analysis of IL12B polymorphisms (rs3212227, rs6887695) With Psoriasis and Psoriatic Arthritis. Rheumatology International. 2013;33(7):1785‐1790. DOI: 10.1007/s00296‐012‐2637‐4
  51. 51. Luu M, Cordoro KM. The Evolving Role of Biologics in The Treatment of Pediatric Psoriasis. Skin Therapy Letter. 2013;18(2):1‐4
  52. 52. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, Li S, Dooley LT, Gordon KB. Efficacy and Safety of Ustekinumab, a Human Interleukin‐12/23 Monoclonal Antibody, in Patients With Psoriasis: 76‐Week Results From a Randomise Double‐Blind, Placebo‐Controlled Trial (PHOENIX 1). Lancet. 2008;371(9625):1665‐1674
  53. 53. Kurzeja M, Rudnicka L, Olszewska M. New interleukin‐23 pathway inhibitors in dermatology: Ustekinumab, Briakinumab, and Secukinumab. American Journal of Clinical Dermatology. 2011;12(2):113‐125
  54. 54. Quatresooz P, Hermanns‐Lê T, Piérard GE, Humbert P, Delvenne P, Piérard‐Franchimont C. Ustekinumab in psoriasis immunopathology with emphasis on the Th17‐IL23 axis: A primer. Journal of Biomedicine and Biotechnology. 2012;2012:147413. DOI: 10.1155/2012/147413
  55. 55. Tong Y, Peranteau AJ, Nawas Z, Tyring SK. A review of brodalumab, an IL‐17 receptor antagonist, for moderate‐to‐severe plaque psoriasis. Skin Therapy Letter. 2017;22(1):1‐6
  56. 56. Lee E, Zarei M, LaSenna C, Villada G, Romanelli P. Psoriasis targeted therapy: Characterization of interleukin 17A expression in subtypes of psoriasis. Journal of Drugs in Dermatology. 2015;14(10):1133‐1136
  57. 57. Pazyar N, Feily A, Yaghoobi R. Macrophage migration inhibitory factor as an incriminating agent in dermatological disorders. Indian Journal of Dermatology. 2013;58(2):157. DOI:10.4103/0019‐5154.108068
  58. 58. Shimizu T, Nishihira J, Mizue Y, Nakamura H, Abe R, Watanabe H, Ishibashi T, Shimizu H. Histochemical analysis of macrophage migration inhibitory factor in psoriasis vulgaris. Histochemistry and Cell Biology. 2002;118(3):251‐257
  59. 59. Rudiger Panzer, Conrad Blobel, Regina Folster‐Holst, EhrhardtProksch. TLR2 and TLR4 expression in atopic dermatitis, contact dermatitis and psoriasis. Experimental Dermatology. 2014;23(5):364‐366. DOI: 10.1111/exd.12383
  60. 60. Salama DM, Abdel Halim DM, Fawzy N, Rashed L. Toll‐like receptor 2 (TLR2) gene expression in psoriasis vulgaris and its correlation with vitamin D3 serum level before and after NB‐UVB. The Medical journal of Cairo University. 2013;81(2):95‐101
  61. 61. Ishimoto T, Kataoka S, Shiga T, Sano S. Increase of psoriasis‐associated IL‐8 in the intralesional blood despite undetectable in the peripheral blood. Journal of Dermatological Science. 2016;84(1):e103‐e1. DOI: 10.1016/j.jdermsci.2016.08.312
  62. 62. Picciani BLS, Carneiro S, Cantisano MH, Avelleira JCR, Azulay DR, Pinto JMN, Dias EP. Geographic tongue or oral psoriasis?. Oral Surgery, Oral Medicine, Oral Pathology and Oral Radiology. 2012;114(4):e108‐e108
  63. 63. Nishikawa R, Nagai H, Bito T, Ikeda T, Horikawa T, Adachi A, Matsubara T, Nishigori C. Genetic prediction of the effectiveness of biologics for psoriasis treatment. The Journal of Dermatology. 2016;43(11):1273‐1277. DOI: 10.1111/1346‐8138.13412., 2016.
  64. 64. Prieto‐Pérez R, Solano‐López G, Cabaleiro T, Román M, Ochoa D, Talegón M, Baniandrés O, López‐Estebaranz JL, de la Cueva P, Daudén E, Abad‐Santos F. New polymorphisms associated with response to anti‐TNF drugs in patients with moderate‐to‐severe plaque psoriasis. The Pharmacogenomics Journal. 2016; DOI: 10.1038/tpj.2016.64, 2016
  65. 65. Murdaca G, Gulli R, Spanò F, Lantieri F, Burlando M, Parodi A, Mandich P, Puppo F. TNF‐α gene polymorphisms: association with disease susceptibility and response to anti‐TNF‐α treatment in psoriatic arthritis. Psoriasis and Genetics. 2014;134(10):2503‐2509. DOI: 10.1038/jid.2014.123
  66. 66. Wu D, Hou SY, Zhao S, Hou LX, Jiao T, Xu NN, Zhang N. Efficacy and safety of interleukin‐17 antagonists in patients with plaque psoriasis: A meta‐analysis from phase 3 randomized controlled trials. Journal of the European Academy of Dermatology and Venereology. 2017. DOI: 10.1111/jdv.14125. [Epub ahead of print]
  67. 67. FDA accepts AstraZeneca’s brodalumab BLA for plaque psoriasis, PDUFA date November 16Valeant Announces FDA Acceptance of BLA Submission for Brodalumab in Moderate‐to‐Severe Plaque Psoriasis. 2016
  68. 68. Burkett PR, Kuchroo VK. IL‐17 blockade in psoriasis. Cell. 2016;167(7):1669. DOI: 10.1016/j.cell.2016.11.044
  69. 69. Białecka M, Ostasz R, Kurzawski M, Klimowicz A, Fabiańczyk H, Bojko P, Dziedziejko V, Safranow K, Machoy‐Mokrzyńska A, Drozdzik M. IL17A and IL17F gene polymorphism association with psoriasis risk and response to treatment in a polish population. Dermatology. 2016;232(5):592‐596. DOI: 10.1159/000448090
  70. 70. Kim SY, Hur MS, Choi BG1, Kim MJ, Lee YW, Choe YB, Ahn KJ. A preliminary study of new single polymorphisms in the T helper type 17 pathway for psoriasis in the Korean population. Clinical & Experimental Immunology. 2017;187(2):251‐258. DOI: 10.1111/cei.12888
  71. 71. Prieto‐Pérez R, Solano‐López G, Cabaleiro T, Román M, Ochoa D, Talegón M, Baniandrés O, LópezEstebaranz JL, de la Cueva P, Daudén E, Abad‐Santos F. The polymorphism rs763780 in the IL‐17F gene is associated with response to biological drugs in patients with psoriasis. Pharmacogenomics. 2015;16(15):1723‐1731. DOI: 10.2217/pgs.15.107
  72. 72. Gullive WP. Importance of Screening for Comorbidities in Psoriasis Patients. Expert Review of Dermatology. 2008;3(2):133‐135
  73. 73. Brauchli YB, Jick SS, Meier CR. Psoriasis and the RISK of incident diabetes mellitus: A population‐based study. British Journal of Dermatology. 2008;159:1331‐1337
  74. 74. Cohen AD, Dreiher J, Shapiro Y, Vidavsky L, Vardy DA, Davidovici B, Meyerovitch J. Psoriasis and diabetes: A population‐based cross‐sectional study. Journal of the European Academy of Dermatology and Venereology. 2008;22(5):585‐589. DOI: 10.1111/j.1468‐3083.2008.02636.x
  75. 75. Horreau C, Pouplard C, Brenaut E, Barnetche T, Misery L, Cribier B, Jullien D, Aractingi S, Aubin F, Joly P, Le Maître M, Ortonne JP, Paul C, Richard MA. Cardiovascular morbidity and mortality in psoriasis and psoriatic arthritis: A systematic literature review. Journal of the European Academy of Dermatology and Venereology. 2013;3:12‐29. DOI: 10.1111/jdv.12163
  76. 76. Abuabara K, Lee H, Kimball AB. The effect of systemic psoriasis therapies on the incidence of myocardial infarction: A cohort study. British Journal of Dermatology. 2011;165:1066‐1073
  77. 77. Gelfand JM, Mehta NM, Langan SM. Psoriasis and cardiovascular risk:Strenght in numbers. JAMA. 2011;131(5):1007‐1010
  78. 78. Friedewald VE, Cather JC, Gelfand JM, Gordon KB, Gibbons GH, Grundy SM, Jarratt MT, Krueger JG, Ridker PM, Stone N, Roberts WC. AJC editor’s consensus: Psoriasis and coronary artery disease. American Journalof Cardiology. 2008;102(12):1631‐1643. DOI:10.1016/j.amjcard.2008.10.004
  79. 79. Najarian DJ, Gottlieb AB. Connections between psoriasis and Crohn’s disease. Journal of the American Academy of Dermatology. 2003;48(6):805‐821. quiz 822-4
  80. 80. Skroza N, IlariaProietti, Riccardo Pampena, Giorgio La Viola, Nicoletta Bernardini, Francesca Nicolucci, ErsiliaTolino, Sara Zuber, Valentina Soccodato, and Concetta Potenza. Correlations between Psoriasis and Inflammatory Bowel Diseases. BioMed Research International. vol. 2013;2013:8. Article ID 983902. DOI:10.1155/2013/983902
  81. 81. Moll JMH, Wright V. Psoriatic arthritis. Seminars in Arthritis and Rheumatism. 1973;3:55‐78
  82. 82. Erdem RH. PsoriatikArtrtitinKlinikÖzellikleri. Romatizma. 2000;15(1):31‐38
  83. 83. Gezer Albayrak I, Levendoğlu F, Özerbil ÖM. PsöriasisHastalarındaPsöriatikArtritGörülmeSıklığıvePsöriatikArtritinKlinikÖzellikleri. Genel Tip Dergisi. 2016;26(2):58‐61
  84. 84. Canpolati F, Akpinar H, Eskioğlu F. Mean Platelet Volume in Psoriasis and Psoriatic Arthritis. Clinical Rheumatology. 2010;29(3):325‐328
  85. 85. McHugh NJ, Laurent MR, Treadwell BLJ, Treadwell BL, Tweed JM, Dagger J. Other seronegative spondyloarthropathies. Medicine. 2009;38(4):190‐193
  86. 86. Pedersen OB, Svendsen AJ, Ejstrup L, Skytthe A, Junker P. On the Heritability of Psoriatic Arthritis. Disease Concordance Among Monozygotic And Dizygotic Twins. 2008;67(10):1417‐1421
  87. 87. Olivieri I, Padula A, D’Angelo S, Cutro MS. Psoriatic Arthritis Sine Psoriasis. The Journal of Rheumatology Supplement. 2009;83:28‐29. DOI: 10.3899/jrheum.090218
  88. 88. Bonfiglioli R, Conde RA, Sampaio‐Barros PD, Louzada‐Junior P, Donadi EA, Bertolo MB. Frequency of HLA‐B27 alleles in Brazilian patients with psoriatic arthritis. Clinical Rheumatology. 2008;27(6):709‐712
  89. 89. Gladman DD, Farewell VT, Kopciuk KA, Cook RJ. HLA Markers and progression in psoriatic arthritis. The Journal of Rheumatology. 1998;25(4):730‐733
  90. 90. Gladman DD, Antoni C, Mease P, Clegg DO, Nash P. Psoriatic arthritis: Epidemiology, clinical features, course and outcome. Annals of the Rheumatic Diseases. 2005;64(2):ii14‐7
  91. 91. Brockbank J. Gladman DD. Psoriatic arthritis. Expert Opinion on Investigational Drugs. 2000;9(7):1511‐1522
  92. 92. de Bakker PI, Mc Vean G, Sabeti PC, Miretti MM, Green T, Marchini J, Ke X, Monsuur AJ, Whittaker P, Delgado M, Morrison J, Richardson A, Walsh EC, Gao X, Galver L, Hart J, Hafler DA, Pericak‐‐Vance M, Todd JA, Daly MJ. A high‐resolution HLA and SNP haplotype map for disease association studies in the extended human MHC. Nature Genetics. 2006;38(10):1166‐1172
  93. 93. Eastmond CJ. Psoriatic arthritis. Genetics and HLA Antigens. Baillière’s Clinical Rheumatology. 1994;8(2):263‐276
  94. 94. Gladman DD, Anhorn KA, Schachter RK, Mervart H. HLA antigens in psoriatic arthritis. The Journal of Rheumatology. 1986;13(3):586‐592
  95. 95. Ho PY, Barton A, Worthington J, Plant D, Griffiths CE, Young HS, Bradburn P, Thomson W, Silman AJ, Bruce IN. Investigating the role of the HLA‐Cw*06 and HLA‐DRB1 genes in susceptibility to psoriatic arthritis: Comparison with psoriasis and undifferentiated inflammatory arthritis. Annals of the Rheumatic Diseases. 2008;67(5):677‐682
  96. 96. Veale DJ, Ritchlin C. ve FitzGerald O. Immunopathology of psoriasis and psoriatic arthritis. Annals of the Rheumatic Diseases. 2005;64(2):26‐29
  97. 97. Öğretmen Z, Hız MM, Sılan F, Koşar S, Özdemir Ö. Is the HLA B27 genotype a riscfaktor for psoriatic arthritis and psoriasis vulgaris? Türkderm 2014;48:131‐134
  98. 98. Fantuzzi G. Adipose tissue, adipokines, and inflammation. The Journal of Allergy and Clinical Immunology. 2005;115(5):911‐919
  99. 99. Kershaw EE, Flier JS. Adipose Tissue as an endocrine organ. The Journal of Clinical Endocrinology & Metabolism. 2004;89(6):2548‐2556
  100. 100. Frühbeck G, Gómez‐Ambrosi J, Muruzábal FJ, Burrell MA. The adipocyte: A model for integration of endocrine and metabolic signaling in energy metabolism regulation. American Journal of Physiology Endocrinology Metabolism. 280:E827‐E847
  101. 101. Russolillo A, Iervolino S, Peluso R, Lupoli R, Di Minno A, Pappone N, Di Minno MN. Obesity and psoriatic arthritis: From pathogenesis to clinical outcome and management. Rheumatology (Oxford). 2012;52(1):62‐67
  102. 102. Pessinaba S, Yayehd K, Pio M, Baragou R, Afassinou Y, Tchérou T, Damorou F. Obesity in cardiology consultation in lome: Prevalence and risk factors associated with cardiovascular disease study in 1200 patients. The Pan African Medical Journal. 2012;12:99
  103. 103. Blüher M. The Distinction of Metabolically ‘Healthy’ From ‘Unhealthy’ Obese Individuals. Current Opinion in Lipidology. 2010;21(1):38‐43
  104. 104. Spiotta RT, Luma GB. Evaluating obesity and cardiovascular risk factors in children and adolescents. American Family Physician. 2008;78(9):1052‐1058
  105. 105. Nguyen S, McCulloch C, Brakeman P, Portale A, Hsu CY. Being overweight modifies the association between cardiovascular risk factors and microalbuminuria in adolescents. Pediatrics. 2008;121(1):37‐45
  106. 106. Poirier P. Targeting abdominal obesity in cardiology: Can we be effective? Canadian Journal of Cardiology. 2008;24:13D‐17D
  107. 107. Wyatt SB, Winters KP, Dubbert PM. Overweight and obesity: Prevalence, consequences, and causes of a growing public health problem. The American Journal of the Medical Sciences. 2006;331(4):166‐174
  108. 108. Steinberger J, Daniels SR. Obesity, insulin resistance, diabetes, and cardiovascular risk in children. Circulation. 2003;107:1448‐1453
  109. 109. Hansson GK, Hermansson A. The immune system in atherosclerosis. Nature Immunology. 2011;12:204‐212
  110. 110. Murray ML, Bergstresser PR, Adams‐Huet B, Cohen JB. Relationship of psoriasis severity to obesity using same‐gender siblings as controls for obesity. Clinical and Experimental Dermatology. 2009;34(2):140‐144
  111. 111. Duarte GV, Oliveira Mde F, Cardoso TM, Follador I, Silva TS, Cavalheiro CM, Nonato W, Carvalho EM. Association between obesity measured by different parameters and severity of psoriasis. International Journal of Dermatology. 2012;52(2):177‐181
  112. 112. Kumar S, Han J, Li T, Qureshi AA. Obesity, waist circumference, weight change and the risk of psoriasis in US women. Journal of the European Academy of Dermatology and Venereology. 2012;10:1293‐1298. DOI: 10.1111/jdv.12001
  113. 113. Love TJ, Zhu Y, Zhang Y, Wall‐Burns L, Ogdie A, Gelfand JM, Choi HK. Obesity and the risk of psoriatic arthritis: A population‐based study. Annals of the Rheumatic Diseases. 2012;71(8):1273‐1277
  114. 114. Husted JA, Thavaneswaran A, Chandran Eder L, Rosen CF, Cook RJ, Gladman DD. Cardiovascular and other comorbidities in patients with psoriatic arthritis: Acomparison with patients with psoriasis. Arthritis Care & Research(Hoboken). 2011;63(12):17
  115. 115. Soltani‐Arabshahi R, Wong B, Feng BJ, Goldgar DE, Duffin KC, Krueger GG. Obesity in early adulthood as a risk factor for psoriatic arthritis. Archives of Dermatology. 2010;146(7):721‐726. DOI: 10.1001/archdermatol.2010.141
  116. 116. Yang YW, Keller JJ, Lin HC. Medical comorbidity associated with psoriasis in adults: A population based study. British Journal of Dermatology. 2011;165(5):1037‐1043
  117. 117. Davidovici BB, Sattar N, Prinz J, Puig L, Emery P, Barker JN, van de Kerkhof P, Ståhle M, Nestle FO, Girolomoni G, Krueger JG. Psoriasis and systemic inflammatory diseases: Potential mechanistic links between skin disease and co‐morbid conditions. Journal of Investigative Dermatology. 2010;130(7):1785‐1796. DOI: 10.1038/jid.2010.103
  118. 118. Qureshi AA, Choi HK, Setty AR, Curhan GC. Psoriasis and the risk of diabetes and hypertension: A prospective study of US female nurses. Archives of Dermatology. 2009;145(4):379‐382
  119. 119. Gibson SH, Perry HO. Diabetes and psoriasis. AMA Archives of Dermatology. 1956;74(5):487‐488
  120. 120. Onsun N, Su Ö, Eriş Z, Davutoğlu M, Cebeci F, Senocak M. PsoriasisteDiyabet, Diyabette Psoriasis Sıklığı. Türkderm. 2010;44(1):12‐14
  121. 121. Azfar RS, Seminara NM, Shin DB, Troxel AB, Margolis DJ, Gelfand JM. Increased risk of diabetes mellitus and likelihood of receiving diabetes mellitus treatment in patients with psoriasis. Archives of Dermatology. 2012;148(9):995‐1000
  122. 122. Armesto S, Santos‐Juanes J, Galache‐Osuna C, Martinez‐Camblor P, Coto E, Coto‐ Segura P. Psoriasis and type 2 diabetes risk among psoriatic patients in a spanish population. Australasian Journal of Dermatology. 2012;53(2):128‐130
  123. 123. Finley PJ, DeClue CE, Sell SA, DeBartolo JM, Shornick LP. Diabetic wounds exhibit decreased Ym1 and arginase expression with increased expression of IL‐17 and IL‐20. Advances in Wound Care(New Rochelle). 2016;5(11):486‐494
  124. 124. Granata M, Skarmoutsou E, Trovato C, Rossi GA, Mazzarino MC, D’Amico F. Obesity, type 1 diabetes, and psoriasis: An autoimmune triple flip. Pathobiology. 2016;17. [Epub ahead of print]
  125. 125. Eirís N, González‐Lara L, Santos‐Juanes J, Queiro R, Coto E, Coto‐Segura P. Genetic variation at IL12B, IL23R and IL23A is associated with psoriasis severity, psoriatic arthritis and type 2 diabetes mellitus. Journal of Dermatological Science. 2014;75(3):167‐172. DOI: 10.1016/j.jdermsci.2014.05.010. Epub 2014 Jun 11
  126. 126. Presta I, Andreozzi F, Succurro E, Marini MA, Laratta E, Lauro R, Hribal ML, Perticone F, Sesti G. IL‐18 gene polymorphism and metabolic syndrome. Nutrition, Metabolism & Cardiovascular Diseases. 2009;19(2):e5‐e6. DOI: 10.1016/j.numecd.2008.10.002. Epub 2009 Jan 26
  127. 127. Işik S, Hız MM, Kılıç S, Öğretmen Z, Silan F. Is there any increased risk of hypertension, diabetes and cardiac diseases in psoriatic patients with TNF‐α G238A and G308A polymorphism? Advances in Dermatology and Allergology/PostȩpyDermatologiiiAlergologii. 2016;33(6):440‐444. DOI:10.5114/pdia.2016.58384
  128. 128. Prodanovich S, Kirsner RS, Kravetz JD, Ma F, Martinez L, Federman DG. Asociation of psoriasis with coronaryartery, cerebrovascular and peripheral vascular diseases and mortality. 2009;145(6):700‐703
  129. 129. Driessen RJ, Boezeman JB, Van de Kerkhof PC, De Jong EM. Cardiovascular risk factors in high‐need psoriasis patientsand its implications for biological therapies. Journal of Dermatological Treatment. 2009;20(1):42‐47
  130. 130. Yang ZS, Lin NN, Li L, Li Y. The effect of TNF inhibitors on cardiovascular events in psoriasis and psoriatic arthritis: An Updated Meta‐Analysis. Clinical Reviews in Allergy & Immunology. 2016;51(2):240‐247
  131. 131. Ogretmen Z, Hiz MM, Silan F, Uludag A, Ozdemirc O. Association of endothelial nitric oxide synthase Glu298Asp gene polymorphism in psoriasis cases with hypertension. Annals of Saudi Medicine. 2014;34(4):340‐345. DOI: 10.5144/0256‐4947.2014.340
  132. 132. Bricker LA, Greydanus DE. The Metabolic Syndrome: a Gathering Challenge in a Time of Abundance. Adolescent Medicine: State of the Art Reviews. 2008;19:475‐497
  133. 133. Abdel Hay RM, Rashed LA. Association between the leptin gene 2548G/A polymorphism, the plasma leptin and the metabolic syndrome with psoriasis. Experimental Dermatology. 2011;20(9):715‐719
  134. 134. Cohen BE, Martires KJ, Ho RS. Psoriasis and the risk of depression in the US population: National Health and Nutrition Examination Survey 2009‐2012. JAMA Dermatology. 2016;152(1):73‐79
  135. 135. Kurd SK, Troxel AB, Crits‐Christoph P, Gelfand JM. The risk of depression, anxiety and suicidality in patients with psoriasis: A population‐based cohort study. Archives of Dermatology. 2010;146(8):891‐895. DOI:10.1001/archdermatol.2010.186
  136. 136. Pompili M, Innamorati M, Trovarelli S, Narcisi A, Bellini S, Orsini D, Forte A, Erbuto D, Botti E, Lamis DA, Girardi P, Costanzo A. Suicide risk and psychiatric comorbidity in patients with psoriasis. Journal of International Medical Research. 2016;44(1):61‐66
  137. 137. Deveci A, Ermertcan AT, Deniz F, Madak BG, Öztürkcan S. PsoriasisliHastalardaAlkolKullanımıveİntiharOlasılığıTürkiye’dePsikiyatri. 2007;9(2):103‐107
  138. 138. Farkas A, Kemény L. Psoriasis and alcohol: Is cutaneous ethanol one of the missing links? British Journal of Dermatology. 2010;162(4):711‐716
  139. 139. Younes SF, Bakry OA. Immunohistochemicalevaluation of role of serotonin in pathogenesis of psoriasis. Journal of Clinical and Diagnostic Research. 2016;10(10):EC05‐EC09
  140. 140. Tencomnao T, Wongpiyabovorn J. An investigation of the relationship between serotonin transporter gene promoter polymorphism and psoriasis susceptibility in a Thai population. Genetics and Molecular Research. 2010;9(4):2275‐2282
  141. 141. Ronpirin C, Tencomnao T, Wongpiyabovorn J. Association between the ‐1438A/G polymorphism of the serotonin 2A receptor gene and late‐onset psoriasis in a Thai population. Genetics and Molecular Research. 2010;9(1):208‐214

Written By

Hız Meliha Merve, Kılıç Sevilay, Oymak Sibel, Büyük Başak, Canbey Göret Ceren, Tuba Demirci and Akı Cüneyt

Submitted: 23 September 2016 Reviewed: 06 March 2017 Published: 05 July 2017