genetic and environmental risk factors for psoriatic ... · genetic and environmental risk factors...

210
Genetic and Environmental Risk Factors for Psoriatic Arthritis among patients with Psoriasis by Lihi Eder A thesis submitted in conformity with the requirements for the degree of PhD Institute for Medical Science University of Toronto © Copyright by Lihi Eder 2011

Upload: buikien

Post on 31-Jul-2019

217 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

Genetic and Environmental Risk Factors for Psoriatic Arthritis among patients with Psoriasis

by

Lihi Eder

A thesis submitted in conformity with the requirements for the degree of PhD

Institute for Medical Science University of Toronto

© Copyright by Lihi Eder 2011

Page 2: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

ii

Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis

Lihi Eder

PhD

Institute of Medical Science University of Toronto

2011

Abstract

Aim: Most of the patients with Psoriatic Arthritis (PsA) develop arthritis following the onset

of psoriasis. The aim of the project is to identify genetic and environmental risk factors for

PsA among psoriasis patients.

Methods: PsA and psoriasis patients from two prospective cohorts were analyzed. The

incidence of PsA among a prospective cohort of psoriasis patients was assessed. The

distribution of Human Leukocyte Antigen (HLA) alleles and Killer Cell Immunoglobulin

like Receptors (KIRs) and their combinations was compared between PsA, psoriasis and

healthy controls. In addition, the association between a wide range of environmental

exposures and PsA was evaluated by comparing the frequencies of exposed individuals

among patients with recent onset PsA and psoriasis. The association between smoking and

PsA was further investigated. The prevalence of smoking was in PsA, psoriasis and the

general population. The interaction between HLA-C*06 and smoking was also tested.

Page 3: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

iii

Results: The genetic analysis revealed several HLA-B alleles and HLA haplotypes that are

associated with PsA compared to psoriasis and can potentially serve as independent markers

for PsA. Furthermore, several combinations of KIR genes and their respective HLA ligands

were also found to be associated with PsA compared to psoriasis. The incidence of PsA

among psoriasis patients was found to be higher than previously reported and its rate was

constant over time. HLA-C*06 was associated with increased interval between psoriasis

onset and PsA. Several environmental factors including occupational exposures, infections,

injuries and smoking were associated with development of PsA. The prevalence of smoking

was decreased among PsA patients compared to psoriasis. The interaction between HLA-

C*06 and smoking was found to be significant.

Conclusions: Genetic and environmental factors are associated with the development of PsA

in patients with psoriasis. These factors may serve as specific markers to identify psoriasis

patients at increased risk for PsA.

Page 4: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

iv

Acknowledgments

I would like to express my gratitude and appreciation to Dr. Dafna Gladman,

my thesis supervisor for introducing me to the great world of research. Her

leadership, support, hard work, and scholarship have set an example I hope to

match some day. She is a true role model to me as a researcher, physician and

as a person.

I would like to thank the other members of my committee, Dr. Cheryl Rosen,

for the insights and patience particularly in reviewing my thesis, and Dr.

Shelley Bull for her direction and guidance in analyzing the data and insightful

comments.

Gratitude is also expressed to all of the people whom without their help this

project would not have been possible. I would like to thank Dr. Vinod Chandran

who helped me understand better the world of genetic epidemiology. My

sincere thanks also go to Sutha Shanmugarajah for her immeasurable help in

recruiting patients and managing the data for this project. I would also like to

thank Fawnda Pellet and Remy Pollock for their hard work in the laboratory

performing the HLA and KIR genotyping. I wish to thank Dr. Richard Cook,

Hua Shen and Arane Thavaneswaran for the meaningful analysis and guidance

in performing the statistical analysis of this project. Special thanks to Anne

MacKinnon for her help “behind the scene” in getting things done. I would like

to thank the Abbott PsA Fellowship and the Canadian Arthritis Network for

their financial support.

Last but not least, I would like to thank my family, Ben, Adi and Shani Eder,

who joined me on this journey. Without their encouragement and understanding

it would have been impossible for me to finish this work.

Page 5: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

v

Table of Contents

Abstract ..................................................................................................................................... ii

Table of Contents ...................................................................................................................... v

List of Tables .......................................................................................................................... vii

List of Figures ........................................................................................................................... x

List of Appendices ................................................................................................................... xi

Chapter 1. Literature Review ........................................................................................ 1

1.1. The Phenotype - Psoriasis....................................................................................... 2

1.2. The Phenotype – Psoriatic Arthritis ........................................................................ 5

1.3. The Etiology of Psoriasis and PsA ......................................................................... 9

1.4. Environmental risk factors for Psoriasis and PsA ................................................ 11

1.5. Genetics of Psoriasis and PsA .............................................................................. 16

Chapter 2. Hypothesis and Aims ................................................................................. 31

2.1. Rationale ............................................................................................................... 32

2.2. Hypothesis ............................................................................................................ 33

2.3. Aims...................................................................................................................... 33

Chapter 3. Study population and data collection......................................................... 34

3.1. Study population ................................................................................................... 35

3.2. Data collection ...................................................................................................... 37

Chapter 4. The incidence of arthritis in a prospective cohort of psoriasis patients ..... 39

4.1. Background ........................................................................................................... 40

4.2. Methods ................................................................................................................ 41

4.3. Results .................................................................................................................. 44

4.4. Discussion ............................................................................................................. 49

Chapter 5. HLA risk alleles for PsA among psoriasis patients ................................... 51

5.1. Background ........................................................................................................... 52

5.2. Methods ................................................................................................................ 54

5.3. Population stratification ........................................................................................ 61

5.4. Results .................................................................................................................. 64

5.5. Discussion ............................................................................................................. 91

Chapter 6. The effect of HLA risk alleles on the rate of development of PsA among psoriasis patients ................................................................................................................. 96

Page 6: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

vi

6.1. Background ........................................................................................................... 97

6.2. Methods ................................................................................................................ 98

6.3. Results ................................................................................................................ 100

6.4. Discussion ........................................................................................................... 106

Chapter 7. The association of KIRs and their HLA ligands with PsA ...................... 108

7.1. Background ......................................................................................................... 109

7.2. Methods .............................................................................................................. 112

7.3. Results ................................................................................................................ 116

7.4. Discussion ........................................................................................................... 124

Chapter 8. The association between environmental factors and onset of PsA in patients with psoriasis ....................................................................................................... 128

8.1. Background ......................................................................................................... 129

8.2. Methods .............................................................................................................. 130

8.2.1. Study population ............................................................................................. 130

8.2.2. Data collection ................................................................................................ 130

8.2.3. Statistical analysis ........................................................................................... 132

8.3. Results ................................................................................................................ 133

8.3.1. Study population characteristics ..................................................................... 133

8.3.2. Proportion of exposure to the different factors ............................................... 133

8.3.3. Smoking and alcohol consumption ................................................................. 134

8.4. Discussion ........................................................................................................... 141

Chapter 9. Smoking is inversely associated with development of psoriatic arthritis among psoriasis patients ................................................................................................... 145

9.1. Background ......................................................................................................... 146

9.2. Methods .............................................................................................................. 147

9.3. Results ................................................................................................................ 151

9.4. Discussion ........................................................................................................... 158

Chapter 10. General Discussion .................................................................................. 163

10.1. Conclusions ..................................................................................................... 171

10.2. Future directions ............................................................................................. 172

References ......................................................................................................................... 174

Appendices ........................................................................................................................ 196

Appendix 1 - Questionnaire for the assessment of exposure to environmental factors 196

Page 7: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

vii

List of Tables

Table 4.1 - Characteristics of the study population and the incident cases of PsA ................ 48

Table 5.1 - Population stratification within the Caucasian study sample ............................... 62

Table 5.2 - Demographic and clinical characteristics of the study population ...................... 64

Table 5.3 - HLA-A allele distribution – Psoriatic disease vs. Controls .................................. 67

Table 5.4 - HLA-B allele distribution – Psoriatic disease vs. Controls .................................. 68

Table 5.5 - HLA-C allele distribution – Psoriatic disease vs. Controls .................................. 69

Table 5.6 - HLA-DRB1 allele distribution – Psoriatic Disease vs. Controls ......................... 69

Table 5.7 - HLA-DQ allele distribution – Psoriatic Disease vs. Controls .............................. 70

Table 5.8 - The association of HLA alleles and PsD compared to healthy controls using

multivariate logistic regression analysis ................................................................................. 71

Table 5.9 - HLA-A allele distribution – PsA vs. Psoriasis ..................................................... 73

Table 5.10 - HLA-B allele distribution – PsA vs. Psoriasis ................................................... 74

Table 5.11 - HLA-C allele distribution – PsA vs. Psoriasis ................................................... 75

Table 5.12 - HLA-DRB1 allele distribution – PsA vs. Psoriasis ............................................ 75

Table 5.13 - HLA-DQ allele distribution – PsA vs. Psoriasis ................................................ 76

Table 5.14 - Odds ratios comparing HLA allele frequencies in PsA to psoriasis using logistic

regression analysis .................................................................................................................. 77

Table 5.15 - Odds ratios comparing HLA-C*06 allele frequencies in PsA to psoriasis by age

at onset of psoriasis ................................................................................................................. 78

Table 5.16 - Odds ratios comparing HLA allele frequencies in Axial PsA vs. psoriasis using

logistic regression analysis ..................................................................................................... 80

Table 5.17 - Odds ratios comparing HLA allele frequencies in peripheral PsA vs. psoriasis

using logistic regression analysis ............................................................................................ 81

Table 5.18 - Odds ratios comparing extended HLA haplotype frequencies in PsD to healthy

controls using logistic regression analysis .............................................................................. 83

Table 5.19 - Linkage Disequilibrium of selected HLA-B and –C alleles ............................... 85

Table 5.20 - Odds ratios comparing HLA-B/HLA-C haplotype frequencies in PsA to

psoriasis using logistic regression analysis ............................................................................ 86

Table 5.21 - Detailed family structure of the study population .............................................. 88

Page 8: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

viii

Table 5.22 - Family based association test Affected (PsA probands) - Unaffected (psoriasis)

sib-pairs and Trios................................................................................................................... 89

Table 5.23 - Family based association test Affected (PsD)-Unaffected (Healthy controls) sib-

pairs and Trios......................................................................................................................... 90

Table 6.1 - Characteristics of the study population .............................................................. 102

Table 6.2 - The association between HLA alleles and the risk of PsA in analysis of time from

onset of psoriasis by parametric proportional hazard model ................................................ 104

Table 6.3 - The association between HLA alleles and risk of PsA in analysis of time from

birth by parametric proportional hazard model .................................................................... 105

Table 7.1 - KIR ligand specificities ...................................................................................... 110

Table 7.2 - KIR gene distribution – Psoriatic disease vs. Healthy controls .......................... 119

Table 7.3 - KIR gene distribution – PsA vs. Psoriasis .......................................................... 119

Table 7.4 - The frequency of HLA-C group 1/HLA-C group 2 alleles in PsA, psoriasis and

healthy controls ..................................................................................................................... 120

Table 7.5 – The association between combinations of KIR2D/HLA based on activation

model and PsA vs. Psoriasis ................................................................................................. 121

Table 7.6 - The frequencies of KIR2DS2 and KIR2DL2 in PsA and psoriasis.................... 121

Table 7.7 - The association between combinations of KIR2D/HLA based on activation model

and PsD vs. Controls ............................................................................................................. 122

Table 7.8 - The frequency of KIR3DS1 in PsA and psoriasis .............................................. 122

Table 7.9 - The association between combinations of KIR3D/HLA based on activation model

and PsA vs. Psoriasis ............................................................................................................ 123

Table 8.1 - Clinical characteristics of the study population.................................................. 137

Table 8.2 – The association between environmental exposures and PsA............................. 138

Table 8.3 – The Association between female hormonal exposures and PsA ....................... 139

Table 8.4 - Full regression model adjusted for age, sex, education level, duration and severity

of psoriasis ............................................................................................................................ 140

Table 9.1 – Demographic and clinical characteristics of the study population .................... 154

Table 9.2 – Smoking characteristics among psoriasis and PsA patients .............................. 155

Table 9.3 – Smoking status at the time of the diagnosis by matched pairs .......................... 155

Page 9: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

ix

Table 9.4 – The association between smoking status and PsA compared to psoriasis alone by

logistic regression analysis ................................................................................................... 156

Page 10: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

x

List of Figures

Figure 1.1 - Genes within the MHC region ............................................................................ 24

Figure 4.1 - Follow-up summary of the study population ...................................................... 46

Figure 4.2 - PsA incidence over time estimated with a Kaplan-Meir curve from time of first

clinic visit to diagnosis of PsA................................................................................................ 47

Figure 6.1- The probability of developing PsA among psoriasis patients from the onset of

psoriasis assuming an exponential model. ............................................................................ 103

Figure 7.1 - Illustration of the structure of the different type of KIRs ................................. 110

Figure 7.2 - The 10 most frequent KIR genotypes in PsA, psoriasis and healthy controls .. 120

Figure 9.1 - Smoking status among PsA and psoriasis patients by HLA-C*06 status ........ 110

Page 11: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

xi

List of Appendices

Appendix 1 - Questionnaire for the assessment of exposure to environmental factors ........... 206

Page 12: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

xii

List of abbreviations

AS - Ankylosing Spondylitis

BSA - Body Surface Area

CASPAR – Classification of Psoriatic Arthritis

CDSN - Corneodesmosin

CI – Confidence Interval

DMARDs - Disease Modifying Anti-Rheumatic Drugs

FDR - False Discovery Rate

GWAS - Genome Wide Association Study

HLA - Human Leukocyte Antigen

IBD – Inflammatory Bowel Disease

KIR - Killer Immunoglobulin-like Receptor

LR – Likelihood Ratio

LD - Linkage Disequilibrium

mSASSS - modified Stoke Ankylosing Spondylitis Spine Score

mNAPSI - modified Nail Psoriasis Severity Index

NK - Natural Killer

NSAIDs - Non-Steroidal Anti-Inflammatory Drugs

OR - Odds Ratio

PsA – Psoriatic Arthritis

PASI – Psoriasis Area and Severity Index

PSORS - PSORiasis Susceptibility locus

PsD - Psoriatic Disease

ReA - Reactive arthritis

SNP - Single Nucleotide Polymorphisms

SpA – Spondyloarthropathies

SPR – Standardized Prevalence Ratio

TNF - Tumor Necrosis Factor

ToPAS - Toronto Psoriatic Arthritis Screen

Page 13: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

1

Chapter 1. Literature Review

Page 14: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

2

1.1. The Phenotype - Psoriasis

Psoriasis is a common skin disease affecting 1-3% of the population [1-3]. The antiquity of the

disease dates back to biblical times. It has long been recognized that most cases of biblical leprosy

were actually psoriasis [4]. Lepers were considered unclean and were subject to regulation and

segregation from the normal population. Only in the 18th century a detailed description that

distinguished these skin lesions by the British dermatologist Robert Willan, ended hundreds of

years of confusion and laid the foundation for establishing psoriasis as an independent disease [5].

1.1.1. Epidemiology of psoriasis

Psoriasis affects men and women equally, and is seen in all races. Although psoriasis can

begin at any age, there seem to be two peaks in onset: one between ages 20 and 30 and

another between ages 50 and 60 [6]. Psoriasis has been described as two types depending

on the age of onset. Patients with early-onset or Type I psoriasis (before the age of 40) tend

to have more severe disease with a familial history. Patients with late-onset or Type II

psoriasis (after the age of 40) tend to have a milder disease [7].

1.1.2. Clinical manifestations of psoriasis

Psoriasis is usually manifested as raised, erythematous plaques with adherent silvery scales.

It is usually easily recognized, but atypical or non-classic forms are more difficult to

identify. There are several clinical types of psoriasis; the most common one is chronic

plaque psoriasis or psoriasis vulgaris that affects 85-90% of all patients with the disease.

This type usually presents in young adults with symmetrically distributed plaques involving

the scalp, extensor elbows, knees, and back. Other types include flexural psoriasis, guttate

psoriasis, pustular psoriasis and erythroderma [8]. Approximately 40% of patients with

psoriasis have nail lesions. One of the typical nail abnormalities in psoriasis is pitting,

consisting of a few to multiple tiny pits scattered over the nail plate. The pits reflect

abnormal nail plate growth resulting from psoriatic involvement of the nail matrix. These

changes produce friable areas of nail plate that erode away with normal friction. Another

Page 15: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

3

typical psoriatic nail lesion is onycholysis, that occurs as a result of a separation of the nail

plate from its underlying attachment to the nail bed [6].

1.1.3. Diagnosis of psoriasis

Although the differential diagnosis of psoriasis is broad, a skin biopsy is rarely needed. The

diagnosis is usually made by history and physical examination. There are no laboratory

tests that confirm or exclude the diagnosis. A detailed physical examination should focus

on typical sites of involvement such as knees and elbows with a special attention to subtle

findings in the scalp, umbilicus, intergluteal cleft, and nails.

1.1.4. Clinical course and co-morbidities of psoriasis

Psoriasis tends to be a chronic disease. However, its course is unpredictable. There may be

marked variability in severity over time, and remissions at some stage are seen in up to

40% of cases [9]. Although generally not life threatening, psoriasis may be associated with

important morbidity and disability. It can range from a very mild disease with few small

hidden plaques that do not interfere with daily life to severe wide-spread skin lesions that

may lead to disability and poor quality of life. Patients with psoriasis, like those with other

major medical disorders, have reduced levels of employment as well as decreased quality

of life [10, 11].

Psoriasis is also associated with other co-morbidities including obesity and other related

metabolic abnormalities such as diabetes mellitus and dyslipidemia [12, 13]. These in turn

lead to increased cardiovascular morbidity and mortality [14]. In addition, approximately

30% of the patients with psoriasis develop an inflammatory arthritis termed Psoriatic

Arthritis (PsA) [15].

Page 16: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

4

1.1.5. Measurement tools in Psoriasis

The Psoriasis Area and Severity Index (PASI) is the most widely used tool for the

measurement of severity of psoriasis. PASI combines the assessment of the severity

of lesions and the area affected into a single score in the range of 0 (no disease) to 72

(maximal disease) [16]. Another measurement tool for psoriasis severity is the Body

Surface Area (BSA) which is an estimate of the percentage of the body surface affected by

psoriasis. Patients with severe psoriasis are those with BSA>10% or PASI>10 [17].

1.1.6. Treatment of psoriasis

Treatment in psoriasis can generally be categorized into topical and systemic therapies. The

treatment choice is dictated by the severity, type, and location of psoriasis. Patients with

mild psoriasis can usually be managed with topical agents including: corticosteroids, tar,

retinoids and vitamin D derivates. Moderate to severe psoriasis requires phototherapy or

systemic therapies such as methotrexate, retinoids, cyclosporine or the biologic immune

modifying agents including alefacept, the anti- Tumor Necrosis Factor (TNF) agents or

anti-IL12/23 monoclonal antibody (ustekinumab) [18].

Page 17: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

5

1.2. The Phenotype – Psoriatic Arthritis

Psoriatic arthritis has been defined as an inflammatory arthritis associated with psoriasis,

usually seronegative for rheumatoid factor [19]. The association between psoriasis and

arthritis was first described in 1818 by the French physician, Baron Jean Louis Alibert [20].

However, only in 1964 did the American Rheumatology Association recognize PsA as a

unique disease entity that is separate from rheumatoid arthritis [21]. Furthermore, only in

2006, were classification criteria for PsA developed and allowed a better definition of cases

for research purposes [22].

Psoriatic arthritis is classified among the seronegative spondyloarthropathies (SpA). This

term refers to a family of diseases that share certain clinical features. The most

distinguishing features are inflammation of the axial joints, asymmetric oligoarthritis, and

enthesitis (inflammation at sites of ligamentous or tendon attachment to bone). Additional

features are genital and skin lesions, eye and bowel inflammation, an association with

preceding or ongoing infectious disorders, and a strong association with the Human

Leukocyte Antigen (HLA) -B*27. The SpA group consists of the following disorders:

Ankylosing Spondylitis (AS), Reactive arthritis (ReA) , PsA , Undifferentiated

spondyloarthritis, SpA associated with Inflammatory Bowel Disease (IBD) and Juvenile

onset spondyloarthritis [23].

1.2.1. Epidemiology of PsA

The most recent estimate of the prevalence of PsA in North America is 0.25% (95% CI:

0.18%, 0.31%) [24]. The reported incidence of PsA in the general population ranges from

3.4-8 per 100,000 [25-28]. PsA has been reported in 7-42% of patients with psoriasis, with

a recent estimate being approximately 30% [15, 29, 30]. The marked variability in reported

prevalence and incidence rates is probably related to different definitions of the disease as

well as variable sources of populations. Approximately 67% of the patients develop

psoriasis before arthritis and in 16% arthritis and psoriasis present within 12 month of each

other [31]. There is limited information about the incidence of PsA among patients with

Page 18: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

6

psoriasis. A retrospective study from Germany reported that the cumulative incidence of

PsA among psoriasis patients reached 20.5% after 30 years from the diagnosis of psoriasis

[32]. Another retrospective study from Rochester, Minnesota has found a lower cumulative

incidence of 3.1% cases of PsA among psoriasis patients after 10 years from the onset of

the skin disease [33]. No study to date has prospectively assessed the incidence of PsA

among psoriasis patients. The prevalence of the disease is equal among males and females.

PsA usually occurs in the third or fourth decade of life [15]. There is very little information

about racial and ethnic associations as most epidemiological studies has been performed in

Caucasians.

1.2.2. Clinical manifestations

Five patterns of PsA have been described: the symmetric polyarticular pattern being the

most common [34], distal arthritis that involves the distal interphalangeal joints,

asymmetric oligoarthritis in which less than 5 joints are affected, arthritis mutilans that is

characterized by deforming and destructive arthritis, and spondyloarthritis that includes

sacroiliitis and spondylitis [35]. Some of the patients present with more than one pattern or

change their pattern during the course of their disease. Another common feature of PsA is

enthesitis, an inflammation at the site of tendon insertion into the bone, that often affects

the Achilles tendon, plantar fascia and pelvis bones [36]. Dactylitis is characterized by

diffuse swelling of the entire finger or toe; it affects about half of the patients and is

associated with radiographic joint damage [37]. Similarly to the other

spondyloarthropathies, PsA is also associated with inflammation in other extra-articular

sites including the eye (uveitis) and the gastrointestinal tract (inflammatory bowel disease).

1.2.3. Relationship between skin and joint disease

PsA may be considered as a disease within a disease as most of the patients with PsA also

have psoriasis, although according to the new classification criteria, patients with PsA do

not have to have established psoriasis [22]. Most of the patients develop PsA after or at the

Page 19: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

7

same time as the skin disease, however, 15% of patients with PsA present with arthritis

before psoriasis [35]. Since most patients develop PsA after the onset of psoriasis, the skin

disease serves as a marker for the development of PsA. The paradigm that patients with

severe psoriasis are the ones who develop arthritis is controversial. Several studies reported

a higher prevalence of PsA among patients with severe psoriasis [24, 38]. However, the

fact that PsA can present before psoriasis as well as recent observations of no relationship

between the severity of the skin and joint manifestations [39, 40], suggest there is no direct

link between psoriasis severity and arthritis. Nail lesions are more common among PsA

patients than in psoriasis and the presence of nail lesions has been suggested as marker of

increased risk for PsA among psoriasis patients [41, 42].

1.2.4. Diagnosis of PsA

The diagnosis of PsA is based on a typical combination of clinical, laboratory and

radiographic findings. The ClASsification of Psoriatic ARthritis (CASPAR) criteria are a

set classification criteria for PsA that were published in 2006. They may be used for

diagnosis and allow a uniform definition of cases for research purposes. The CASPAR

criteria showed high sensitivity and specificity for PsA (91.4% and 98.7%, respectively)

[22].

The CASPAR criteria consist of the following terms:

Required: The presence of an inflammatory arthritis, enthesitis, or spondylitis.

Plus 3 points from the following:

1. Skin psoriasis (present) (2 points), previously present by history (1 point), or a

family history of psoriasis (1 point)

2. Psoriatic nail lesions (1 point)

3. Dactylitis (1 point)

4. Negative rheumatoid factor (1 point)

Page 20: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

8

5. Juxta-articular bone formation on radiographs (1 point)

1.2.5. Clinical course of PsA

In most patients PsA runs a course of a chronic, progressive disease, although some

patients can achieve a complete remission. In our cohort, 17.6% of the patients achieved a

remission, however, periods of remission lasted on average 2.6 years and most patients

experienced a relapse [43]. PsA is more severe than previously thought [44, 45]. It can lead

to severe joint damage and disability that are comparable to those that occur in rheumatoid

arthritis [46].Patients with PsA demonstrate clinical and radiographic progression in the

course of follow-up [47, 48] and have an increased mortality risk compared to the general

population, although this risk has decreased over the past two decades [49, 50]. In addition

to the disability that is related to the joint disease, PsA patients can suffer from the same

co-morbidities as psoriasis patients [51]. Thus, PsA poses a major health burden, in

addition to that caused by psoriasis alone.

1.2.6. Treatment of PsA

The treatment in PsA is aimed at controlling both skin and joint inflammation in order to

reduce the symptoms and to prevent joint damage. Treatment usually begins with Non-

Steroidal Anti-Inflammatory Drugs (NSAIDs) that can control mild arthritis, enthesitis and

spondylitis. Second line therapies are indicated when arthritis does not respond to NSAIDs.

Many of the Disease Modifying Anti-Rheumatic Drugs (DMARDs), such as methotrexate,

leflunomide, sulfasalazine, cyclosporine and azathioprine, were “borrowed” from

rheumatoid arthritis for the treatment of peripheral arthritis in PsA. Although there are

limited clinical trials that evaluated their efficacy in PsA, they are often effective in

controlling articular symptoms. The new targeted biologic therapies particularly the anti-

TNF agents, are the most effective treatments currently available to control all aspects of

the disease and to prevent the progression of joint damage [52].

Page 21: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

9

1.3. The Etiology of Psoriasis and PsA

1.3.1. Immunologic mechanisms in Psoriasis and PsA

The role of immunologic mechanisms is suggested by the inflammatory response in the

psoriatic skin lesions and by the synovial lesions, which are at times indistinguishable from

rheumatoid arthritis [53]. T cells and pro-inflammatory cytokines have important roles in

the pathogenesis of both diseases. CD8+ T cells clones from the synovial fluid and skin

lesions of patients expand in a presumptive autoantigen-driven manner [54-56]. Patients

with PsA have a pattern of pro-inflammatory cytokines in the synovium, particularly from

the Th1 milieu including: TNF-alpha, IL-1, IL-6, IL-8 and more [57-59]. The effectiveness

of T cell-targeting and cytokine modulating treatments including the anti TNF-α agents,

alefacept and efalizumab in psoriasis and PsA support the important role of T cells in the

pathogenesis of these conditions. The innate immune system provides an early response

mechanism against external threats, mainly infectious agents and includes dendritic cells,

keratinocytes, macrophages and Natural Killer (NK) cells. Dysregulation of the innate

immune system is thought to play a role in the pathogenesis of psoriasis and PsA [60].

Activation of dendritic cells through antimicrobial peptides leads to an increased

production of interferon alpha which is a major inducer of psoriasis. This mechanism

provides a potential explanation for the link between environmental triggers that may break

the immunologic tolerance and lead to psoriasis and PsA [61].

Natural Killer cells and NK-T cells, which are part of the innate immune system, have been

implicated in the pathogenesis of psoriasis [62] and PsA [63-65]. Both NK and NK-T cells

have been described in increased numbers in psoriatic plaques [66] and in synovial tissues

from PsA patients [63]. NK cell activity is partially controlled through interaction between

Killer Immunoglobulin-like Receptors (KIRs) on NK cells and their respective HLA class I

ligands [67]. It has been proposed that direct activation of NK cells bearing KIRs for MHC

class I may play a role in the pathogenesis of PsA [68, 69].

The concept of synovio-entheseal complex is a proposed theory that can provide a link

between mechanical stress and the development of PsA. The synovium provides

nourishment and lubrication to the entheseal fibrocartilage. During mechanical stress and

Page 22: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

10

injury to the enthesis, the associated inflammatory reaction would be manifested in the

juxtaposed synovium. The damaged tissue triggers the previously discussed immune

mechanisms [70]. Microanatomical studies provide support for the theory by demonstrating

an anatomical connection of the nail and the enthesis, that may explain the observation that

psoriatic nail lesions are associated with higher risk of developing PsA [71, 72].

Page 23: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

11

1.4. Environmental risk factors for Psoriasis and PsA

Psoriasis and PsA are considered complex diseases in which the interaction between

genetic and environmental risk factors is thought to play a major role [73]. Genetic factors

cannot solely account for all cases. One of the suggested pathogenic models for PsA is that

psoriasis patients who carry susceptibility genes for arthritis develop PsA after being

exposed to triggering environmental factors [74]. These environmental factors are largely

unknown.

Several environmental factors have been associated with psoriasis including: infections,

particularly streptococcal pharyngitis, smoking, trauma, stressful life events, obesity and

certain medications [75]. Only a limited number of studies have investigated environmental

risk factors for PsA. Pattison et al. compared the prevalence of environmental exposures

among 98 British PsA and 163 psoriasis patients over a window of exposure that ranged

from 5 to 10 years prior to the onset of arthritis. Information about environmental

exposures was collected through questionnaires. In their study, physical trauma, rubella

vaccination, oral ulcers and moving house were found to be associated with PsA [76].

Another study by Thumboo et al. used an administrative database from Rochester,

Minnesota and retrospectively evaluated exposure to several environmental risk factors

among 60 PsA and 120 psoriasis patients [77]. They did not define a specific window of

exposure. In that study, pregnancy was found to be protective of PsA, while steroid use was

associated with higher risk of the disease.

Several case reports and case series have highlighted the role of physical trauma as a

potential triggering environmental exposure for PsA. In these reports, patients with

psoriasis developed arthritis following local trauma, such as injury, surgery or fracture [78-

80]. The pathogenic process that may lead from local trauma to joint inflammation in PsA

is unclear. However, the hypothesis that these two events are linked is supported by a

similar process in the skin that was termed the “Koebner phenomenon”, in which local skin

trauma induces certain skin lesions including psoriasis. This process has been reported in

24-52% of psoriasis patients [81-83]. Many different injuries can induce Koebner response

including: insect bites, burns, laceration, venipuncture among others. The time period from

Page 24: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

12

injury to psoriasis is diverse; in general the interval ranges between 10 and 20 days [84].

Koebnerization can trigger a cascade of events that switch the feature of the disease, from

localized to generalized psoriasis. Psoriasis patients who carry the HLA-C*06 allele have a

higher incidence of Koebnerization. Local trauma to the skin induces release of

proliferation factors and proliferation of inflammatory cells that initiate an inflammatory

process. Similar events may take place in a joint following local trauma and was termed

“Deep Koebner phenomenon” [85].

Reactive arthritis is an episode of an acute articular and periarticular sterile inflammation

occurring in a genetically predisposed individual secondary to a primary infection

elsewhere in the body [86]. Urogenital and gastrointestinal infections are well established

triggering events for ReA [87]. Reactive arthritis occurs in 1% to 4% of patients with

preceding bacterial gastrointestinal infections with enterobacteria or urogenital infections

with Chlamydia trachomatis, however, this rate increases to 20% to 30% in patients

infected with one of these bacteria who are positive for HLA-B*27 [86]. Reactive arthritis

belongs to the SpA group and has some common features with PsA including: similar

pattern of joint involvement, psoriasiform skin lesions, enthesitis, eye involvement and an

association with the HLA-B*27 allele.

Infections also play a role in the pathogenesis of psoriasis. Streptococcal infection is a

common trigger for guttate psoriasis particularly in children and young adults [88] and the

reported incidence of streptococcal infections preceding this type of psoriasis ranges

between 56% and 97% [89]. Chronic plaque psoriasis is also exacerbated after such

infections and psoriasis patients were found to have a ten-fold higher frequency of

streptococcal throat infections than age-matched household controls [90]. Psoriatic lesional

T cells are oligoclonal, and T cells recognizing determinants common to streptococcal M-

protein and keratin have been detected in patients’ blood [91]. It has been suggested that

the association between streptococcal infection and psoriasis may reflect an autoimmune

process that is triggered by the presence of a superantigen, possibly the streptococcal M-

protein. However, the role of autoimmunity in the pathogenesis of psoriasis remains

controversial. The association between streptococcal infection and PsA was suggested by

elevated levels of streptococcal RNA in PsA peripheral blood [92, 93]. However, although

Page 25: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

13

the immunoreactivity to streptococcal antigens is accepted, it is unclear if the infection

triggers PsA or if the breakage of skin barrier because of psoriasis leads to streptococcal

exposure and finally to a form of reactive arthritis.

Infection with Human Immunodeficiency Virus (HIV) is also associated with the

development of psoriasis and PsA particularly in HIV endemic populations [94, 95]. It has

been reported that in Sub-Saharan Africa the great majority of PsA patients were HIV

positive [96]. This association may hint at a viral trigger of PsA or that HIV increases the

risk for other infections that may trigger PsA.

Obesity is associated with psoriasis. Multiple studies have demonstrated that patients with

psoriasis are more frequently overweight or obese compared with patients without psoriasis

[97-99] .Obesity occurs prior to the onset of psoriasis and is now thought to be a risk factor

for the development of the disease. A large prospective cohort from the US demonstrated a

‘dose-response’ relationship for obesity on the risk of developing incident psoriasis [100].

Obesity (BMI≥30) compared with normal body weight was associated with two-fold

increased risk for psoriasis onset. Obesity is also associated with more severe psoriasis

[101] and poor response to therapy [102]. A recent case-control study from Utah

demonstrated an association between obesity and PsA. The results showed that obesity at

age 18 was associated with an increased risk and an earlier onset of PsA among psoriasis

patients [103], however the study was based on a recall weight and height at age 18 and

was not a prospective study.

The role of smoking as a risk factor for psoriasis is well established. The first studies drew

attention to the linkage between smoking and palmo-plantar pustular psoriasis [104]. Since

then several case-control studies have established the association between smoking and

psoriasis vulgaris [105-108], the most common type of psoriasis. The most conclusive

epidemiological evidence for the causative role of smoking in the pathogenesis of psoriasis

was provided by the Nurses’ Health Study II that showed, for the first time, a strong

association between smoking and incident psoriasis in a large population-based prospective

cohort study [109]. In this study the relative risk of psoriasis was 1.8 for current smokers

and 1.4 for past smokers. The dose-effect relation between smoking intensity and psoriasis

Page 26: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

14

risk also supports its etiological role. Smoking has also been associated with more severe

psoriasis and poor response to treatment [110, 111]. The independent association between

smoking and psoriasis remained significant even after adjustment for another potential

confounder, excessive alcohol consumption, that is also increased among psoriasis patients

[112, 113]. Rakkhit et al. found a temporal association between psoriasis, PsA and

smoking [114]. They reported that the duration of time from the onset of psoriasis to

development of PsA decreases with a history of smoking prior to psoriasis onset and

increases with smoking after psoriasis onset.

Lastly, stressful life events are thought to have an effect on the course of psoriasis. In a

case-control study, psoriasis patients were more likely to experience a stressful event that

preceded the onset and the exacerbation of their disease compared to patients with other

skin disorders [115]. There is some evidence that psychological stress may modulate

immune function in humans and experimental animals [116]. It has been reported that

stress induced anxiety is related to T helper type 1 response. A study that applied

psychological stress prior to immunization suggested that stress exerts an adjuvant effect on

dendritic cells by promoting enhanced migration to lymph nodes and resulting in increased

antigen-specific T cell responses [117]. This effect may be mediated by the release of

norepinephrine from the sympathetic nervous system.

1.4.1. Limitations of previous studies of environmental risk factors in

PsA

There were only two studies that assessed environmental exposures among PsA and

psoriasis patients and one that compared PsA to rheumatoid arthritis. These studies were

limited in several aspects [76, 77, 118].First, due to their small sample size these studies

were underpowered to detect important associations. Secondly, the use of an administrative

database as a source of information [77] may have led to an underestimation of events that

are often not coded or are coded incorrectly. Lastly, as PsA is a common condition among

psoriasis patients, a potential misclassification of cases and controls might have occurred

Page 27: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

15

since psoriasis patients were not assessed to rule out the presence of inflammatory arthritis

[76, 77]. It has been shown that PsA is underestimated among psoriasis patients, as

approximately 18% of the psoriasis patients that were found to have inflammatory arthritis

were unaware of their condition and had never been seen by a rheumatologist [119]. The

problem of misclassification of patients and controls may significantly affect the results in

case-control studies where the outcome (PsA in this case) is not rare among the control

group. Misclassification may decrease the power of the study to detect significant

associations

Page 28: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

16

1.5. Genetics of Psoriasis and PsA

1.5.1. The Approach to identification of genetic determinants of a disease

A series of steps is required for the identification of susceptibility genes for a disease [120].

The first step involves determination of a familial aggregation. Segregation analysis is the

next step, which examines the patterns of disease in families and determines if the patterns

are indicative of traditional genetic inheritance models (such as autosomal dominant or

recessive) or are more consistent with multi-factorial (polygenic and environmental)

models [121]. Finally, once enough evidence of a genetic component in the cause of the

disease has been obtained, the next step is to locate and identify any causative gene [122].

Two basic study designs are used to identify susceptibility loci: linkage analysis and

association studies. Genetic linkage analysis relies on the tendency for short haplotypes to

pass intact, without recombination events, to the next generation within families. A genetic

marker that passes down though a family accompanied by the disease of interest, suggests

that the susceptibility gene for the disease is located close to that marker [123, 124]. An

association analysis is based on a putative functional genetic variant in the

pathophysiologic mechanism of the disease of interest. Association studies look for a

significant increase or decrease in frequency of a marker allele, genotype or haplotype

within a disease trait than would be expected by chance if there were no association

between markers and phenotype. This method can be applied to data obtained from families

or populations of healthy individuals and patients with the disease of interest [125, 126].

For complex diseases, segregation and linkage analyses have not proven to be generally

useful [127, 128]mainly due to low statistical power and the need to recruit large samples.

Therefore, for these diseases, once familial aggregation is demonstrated, it may be

appropriate to proceed with association studies, either candidate gene or genome-wide, to

determine susceptibility loci/genes. Once a gene is identified, functional studies in

biological systems are required to characterize the gene.

Page 29: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

17

1.5.2. Familial aggregation of Psoriasis and PsA

A study on familial aggregation is the first step in pursuing a possible genetic etiology to a

disease. Familial aggregation is assessed by the recurrence risk ratio (λ). The recurrence

risk ratio (λR) is defined as the prevalence of the disease in relatives of type R of affected

cases divided by the prevalence in the general population [129-131].

Family and twin studies have clearly demonstrated that psoriasis has a strong genetic basis.

Two large scale epidemiological studies revealed a substantially higher incidence of

psoriasis in relatives of patients with psoriasis compared to the general population. The

recurrence risk for affected siblings (λs) was estimated to be between 4 and 10 [132-134].

Twin studies reveal a concordance rate for monozygotic twins to be between 62 to 70%

compared to 21 to 23% for dizygotic twins [135-137]. A segregation study concluded that a

polygenic or a multifactorial pattern is the most likely mode of inheritance [138, 139].

The genetics of PsA has been much less investigated compared to that of psoriasis.

However, epidemiological studies have implicated a strong genetic basis to PsA. Four

family studies have investigated the recurrence risk of PsA within families. The first study

that demonstrated familial aggregation of PsA was published in 1973 and found that the

prevalence of PsA among first-degree relatives of probands with PsA was 5.5% compared

to the estimated prevalence in the UK population of 0.1%. The calculated recurrence risk

ratio in first degree relatives (λ1) was 55, compared with estimates ranging from 5–10 in

cutaneous psoriasis [140]. Another study from the UK, reported a PsA prevalence of 14.3%

among siblings and recurrence risk ratio (λs) of 47 (based on estimated PsA prevalence of

0.3% in the general population) [141]. Recently, the risk ratio for PsA was estimated using

the Icelandic genealogical database. First degree relatives to fourth-degree relatives of

patients with PsA had risk ratios of 39, 12, 3.6 and 2.3, respectively (all P-values < 0.0001),

reflecting a strong familial aggregation, whereas the fifth-degree relatives had an RR of 1.2

(P = 0.236) [142]. Finally, our PsA cohort was assessed for familial aggregation. All

Page 30: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

18

available first degree relatives of 100 PsA patients were assessed for evidence of PsA and

psoriasis. Among the 289 first degree relatives that were evaluated, 7.6% had PsA leading

to a calculated recurrence risk ratio in first degree relatives (λ1) of 30.4 [143]. There is

only one twin study in PsA from Denmark that evaluated 36 complete twins that included

one proband with PsA. In this study no difference was found in the concordance rate

between mono- and dizygotic twins [144]. However, this study may have been

underpowered to detect a genetic effect.

1.5.3. Mode of inheritance

Psoriasis and PsA are consistent with a multifactorial pattern of inheritance [145]. Several

studies have suggested an autosomal dominant [146] and autosomal recessive pattern [147]

of inheritance for psoriasis, however, even in these studies; a multifactorial model could

not be ruled out.

Genomic imprinting, a non-Mendelian mode of transmission, has been suggested to play a

role in the inheritance of psoriasis and PsA [148, 149]. Genomic imprinting refers to an

epigenetic effect that causes differential expression of a gene depending on the sex of the

transmitting parent. The imprinting process allows gene expression from only the

maternally or paternally derived chromosome [150]. A family study from the Faroe Island

reported a higher penetrance of psoriasis if the father was affected or a presumed gene

carrier [151].

A similar phenomenon has also been reported in PsA patients. In our cohort, the proportion

of PsA patients with an affected father was significantly higher than the expected

proportion (0.65 vs. 0.5 p=0.001) [148]. These findings were supported by a linkage study

in PsA that noted a significant linkage on chromosome 16q only after conditioning for

paternal transmission [152]. Thus, there are some evidence that this epigenetic

phenomenon may play a role in the inheritance pattern of psoriasis and PsA.

Page 31: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

19

1.5.4. Genetic model for the relationship between PsA and Psoriasis

There is no doubt that there is a close relationship between psoriasis and PsA however the

nature of the relationship is not completely clear. Several models have been suggested to

describe the genetic and clinical association between psoriasis and PsA [153].

The first model suggests that PsA and psoriasis should be viewed as two distinct conditions

with different risk factors with the psoriasiform skin manifestation common to each. The

model explains the families with large numbers of psoriasis cases without any PsA, on the

other hand families with cases of both PsA and psoriasis or only PsA. This model predicts

that case series ascertained by their psoriasis will show genetic heterogeneity. However

case series ascertained by their PsA will share certain susceptibility genes and all PsA will

be genetically distinct from psoriasis.

The second model is based on the hypothesis that the genetic determinants of the skin and

the joint diseases are independent. However, the presence of one of these factors leads to a

lower threshold for the development of the other. This model explains the presence of

undifferentiated spondyloarthritis which is very similar to PsA but does not involve the

skin. This model predicts that case series ascertained by their psoriasis will be genetically

heterogeneous and those ascertained by the presence of PsA will be homogeneous and

share only some of the genes with psoriasis.

The third model views PsA as a “disease within a disease” with psoriasis as the parent

disease. PsA is considered a more severe phenotype of psoriasis that occurs due to a greater

number of susceptibility genes or environmental factors. The additional genes or

environmental risk factors on the background of psoriasis will lead to development of PsA.

This model predicts that case series ascertained by psoriasis will be genetically

homogenous and that case series ascertained by PsA will completely overlap those of

psoriasis apart from several additional distinct genes.

Page 32: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

20

1.5.5. Gene identification studies - Linkage analysis studies

Genetic linkage analysis can be used to identify regions of the genome that contain genes

that predispose to disease. The aim of this method is to isolate the disease gene by its

chromosomal location without any prior knowledge of the position or function of the gene.

Linkage analysis requires collection of families with multiple affected individuals. There

are two methods for linkage analysis: parametric (model-based) linkage analysis and non-

parametric linkage analysis. Parametric linkage analysis requires constructing a model to

explain the inheritance of a disease in the pedigrees and then estimating the recombination

fraction for a given pedigree. For multifactorial diseases, where several genes (and

environmental factors) might contribute to disease risk, there is no clear mode of

inheritance [123]. Methods to investigate linkage have therefore been developed that do not

require specification of a disease model. Such methods are referred to as non-parametric, or

model-free. In this method, affected sibling pairs are assessed for allele sharing. The

premise for the non-parametric method is the fact that in the presence of linkage between a

marker and disease, sets of relatives who share the same disease status are more likely to

share alleles at the marker locus than the value of 50% that would be predicted by chance

[154].

1.5.6. Linkage analysis in psoriasis

Both parametric and non-parametric methods have been used to investigate the genetics of

psoriasis. The psoriasis susceptibility loci that have been mapped using linkage methods

include: PSORiasis Susceptibility locus (PSORS)1 on 6p21.3 [151, 152], PSORS2 on 17q

[155, 156],PSORS3 on 4q [157], PSORS4 on 1q21 [158], PSORS5 on 3q21 [159],

PSORS6 on 19p [160], PSORS7 on 1p [161], PSORS8 on 16q [162], PSORS9 on 4q [163]

and PSORS10 on 18p11 [164]. By far the strongest association is with a locus within the

MHC on chromosome 6p21 (PSORS1). Additional putative psoriasis candidate loci have

been reported on 16q and 20p [165]. The loci on 6p and 17q have been replicated with

independent linkage studies and a meta-analysis of previous studies has found an increased

Page 33: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

21

allele sharing for 16q [155, 163, 165]. However, the rest of the loci were found to be

difficult to replicate.

With regard to PsA, however, only one genome-wide linkage scan has been conducted

[152]. This study identified a locus on 16q close to the PSORS8 locus identified for

psoriasis, but only when conditioned on paternal inheritance. Thus, it is unclear whether

there are distinct susceptibility loci for psoriasis and PsA. Maximum LOD score (MLS)

analysis of affected sibling pairs yielded allele sharing of 60% for markers within the MHC

region. These results emphasize the importance of the MHC region as a candidate

susceptibility region in psoriasis. In that study, evidence of allele sharing was also found on

16q and 10q22-q23.

1.5.7. Gene identification studies - Association Studies

Genetic association studies aim to detect associations between one or more genetic

polymorphisms and a trait. Traditionally, linkage analysis studies were performed for

coarse mapping as they have a limited genetic resolution of 1 cM. Association studies were

the next step for fine mapping. However, association studies have several advantages over

linkage studies. No assumption about the mode of inheritance is required, they have greater

power to detect small effects and they do not require large affected families. Therefore,

association studies are often used initially for the investigation of complex diseases [126].

There are two basic approaches. The first one is to determine the association between a

candidate gene and a trait. This approach is based on prior knowledge from linkage studies

or the biology of the disease. The other approach is to perform “hypothesis free” Genome

Wide Association Study (GWAS), where thousands of genetic markers, single nucleotide

polymorphisms (SNPs), throughout the genome are tested individually for their association

with the disease.

The association between genotype and phenotype can be explained either by direct

biological action of the polymorphism or by allelic association between the marker and a

susceptibility gene. The term Linkage Disequilibrium (LD) is used to refer to allelic

association between linked loci. Linkage disequilibrium is the hallmark of association

Page 34: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

22

studies. The assumption is that the genetic marker studied is close enough to the actual

disease gene. This will result in an allelic association at the population level. The

magnitude of LD is affected by many factors, but the most important one is the

physical/genetic distance between the disease gene and the marker allele; the closer they

are the stronger the LD [125]. The MHC region on chromosome 6 is an example to a region

that contains a large number of genes that are in strong LD.

1.5.8. Candidate genes within MHC region (chromosome 6p)

1.5.8.1. Human Leukocyte Antigen in Psoriasis

Numerous case-control association studies and later GWAS have found a strong

association between psoriasis and the MHC region [166-170]. The strongest association has

been found with a 300kb-segment in the MHC-I region on chromosome 6p21.3 known as

PSORS1 [171, 172]. This region contains HLA genes that are associated with autoimmune

diseases. Case-control studies identified the class I antigens HLA-B*13, B*17 and its split

B*57, C*06, C*07 as associated with psoriasis [168, 173-176]. Several studies

demonstrated an association with the class II antigens, HLA-DRB1*04 and HLA-

DRB1*07 [167, 177]. The largest and most consistently reported association is with HLA-

C*06, with a relative risk of 22. The presence of HLA-C*0602 is associated with an earlier

onset and more severe psoriasis [178, 179]. The association with HLA-B and the Class II

antigens, HLA-DR, was later determined to be due to extended haplotypes and LD with

HLA-C [180]. Several studies have shown that the strongest link with psoriasis is with the

57.1 ancestral haplotype (C*06-B*57-DRB1*07-DQ*03). These findings are consistent

with the association of individual components of this haplotype with psoriasis [181, 182].

Since this region is in strong LD, the true risk allele has been difficult to determine.

Candidate genes just telomeric to HLA-C, such as CDSN and HCR, seemed like good

candidate genes since they are expressed in the skin [183, 184]. However, none of these

candidate genes were consistently associated with psoriasis independently of HLA-C [185,

186]. In order to determine the psoriasis susceptibility locus within the PSORS1 region, a

Page 35: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

23

study that involved sequencing the putative 300-kb risk segment of PSORS1 from just

telomeric to HLA-B to beyond CDSN thus including HLA-C was conducted. After

sequencing this segment in 2 risk and 5 non-risk chromosomes, then examining

recombinant haplotypes retaining HLA-C*06 but lacking risk alleles in CDSN, the authors

concluded that HLA-C*06 is the PSORS1 risk variant that confers susceptibility to

psoriasis [180]. Two GWAS among Caucasians and Chinese psoriasis patients confirmed

previous findings. In these studies, by far, the most significant associations were of SNPs

that were in tight LD with HLA-C*0602 [170, 187].

1.5.8.2. Human Leukocyte Antigen in PsA

Case-control studies identified the HLA region as containing potential susceptibility loci

for PsA. HLA-B*13, B*17 and its split B*57 and C*06 are associated with psoriasis across

various population [41, 188-192]. While HLA-C*06 is also increased in PsA patients

compared to the general population, this association is stronger with psoriasis itself than

with PsA [193]. Several studies reported an association between HLA-B*13, B*57,

DRB1*07 and PsA. However, these results are most likely secondary to the presence of

these alleles on the recognized ancestral haplotypes: AH.13 and AH.57 both contain HLA-

C*06 [181, 182]. HLA B*27 and B*07 have been specifically associated with PsA in case-

control studies that compared patients with PsA to psoriasis [41]. Several of the HLA

antigens have been related to specific manifestations of PsA. HLA-B*27 is more common

among PsA patients with axial disease while B*38 and B*39 are increased among those

with peripheral polyarthritis [41, 189, 192]. Within the HLA Class I region the

associations are stronger with HLA-B than HLA-C alleles [194]. It has been assumed that

associations with HLA-C alleles are related to the skin disease and are not a specific

marker for the joint disease. Among PsA patients, HLA-C*06 is associated only with early

onset psoriasis and no association was observed with PsA patients with late onset psoriasis

[193]. HLA-DRB1*04 was reported to confer a risk of PsA [189, 195, 196], but several

investigators found no such associations [41, 197]. The role of HLA genes in susceptibility

to PsA was demonstrated when increased sharing of HLA haplotypes was documented

among sib pairs concordant for PsA but not among those concordant for psoriasis only

[198]. Since almost all patients with PsA have psoriasis, it is unclear whether the HLA

Page 36: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

24

associations described above are related to psoriasis, PsA, or both. The HLA alleles that

may be specific to PsA are HLA-B*27 and possibly B*07, B*38 and B*39.

1.5.9. Other candidate genes within MHC region in psoriasis

The MHC region contains more than 160 genes that have strong LD and span over 4

megabases. Many of the genes within this area are implicated in the pathogenesis of

autoimmune diseases including: HLA, TNF, C4 and others (Figure 1.1) [199].

Figure 1.1 - Genes within the MHC region

Since the PSORS1 region is included in the MHC region, several candidate genes in the

proximal MHC class I region have been assessed for their association with psoriasis. The

Page 37: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

25

following genes have been found to be associated with psoriasis: HLA-B [200], HLA-C

[180, 201, 202], PSORS1C3 [201, 203], OTF3 [204], HCR [205, 206], SEEK1[207],

corneodesmosin (CDSN) [203, 208, 209] and TNF-α [210-212]. Since all of these genes

have a potential role in the pathogenesis of the disease, the precise identity of the PSORS1

determinant has not been determined due to the strong LD within this region. A detailed

analysis of genomic DNA sequences and recombinant haplotypes strongly suggested that

HLA-C*0602 is the disease allele at PSORS1 [180]. In a recent detailed analysis of GWAS

data, two additional SNPs within the MHC region were found to be significantly associated

with psoriasis after adjusting for HLA-C*0602. One SNP was located within c6orf10, a

potential downstream effecter of TNF-alpha, and the other SNP located between HLA-B

and MHC class I Chain- related sequence A (MICA) locus [170].

1.5.10. Other candidate genes within MHC region in PsA

The MHC region has also been investigated for susceptibility loci to PsA. Unlike psoriasis,

CDSN was not associated with PsA [213]. The region 100 Kb centromeric to the HLA-B

locus has been associated with PsA [214-216]. The MICA gene, located in this region, is a

candidate susceptibility gene for PsA due to its functional relevance, being involved in

activation of NK cells. In a Spanish population, a MICA allele with 9 GCT repeats (A9)

corresponding to MICA*002 was associated with PsA, but not with psoriasis [214],

independently of HLA-C*06. Similar associations have been shown in Jewish [215] and

Croatian [217] patients. However, other studies could not replicate these findings [218].

We have recently compared the distribution of 55 MICA alleles among patients with

psoriasis, PsA and healthy controls. The results showed that most MICA allele associations

with psoriasis and PsA were dependent on LD with HLA-B and HLA-C risk alleles.

Independent of HLA, only homozygosity for MICA*00801 increased the risk of

developing PsA [219].

TNF-α is increased in the skin and synovial tissue of patients with PsA, and the

inflammation in both sites is dramatically reduced following treatment with anti-TNF-α

agents [220-222]. The TNF-α gene lies just centromeric to MICA. Associations have been

Page 38: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

26

reported with TNF-α -308 polymorphism in the promoter region of the gene and PsA [210,

212]. A meta-analysis confirmed an association between TNF-α -238 polymorphism and

PsA [223]. Thus it appears that the susceptibility locus for PsA lies more centromeric than

that of psoriasis, closer to HLA-B, MICA and TNF.

1.5.11. Candidate genes outside the MHC region

1.5.11.1. KIR genes on 19q and their interaction with HLA

NK cells play an essential role in innate immunity, particularly against tumors and viral

infections [67]. Dysregulation of the innate immune system has been involved in the

development of psoriasis and PsA. NK cells accumulate in active psoriatic skin plaques

[63] and inflamed synovium in PsA [60, 61] and loss of their inhibition may exacerbate

damage to the tissue. The activity of NK cells is determined by a balance between

activating and inhibitory signals transmitted by a range of receptors including KIRs,

Leukocyte immunoglobulin like receptor and the CD94/NKG2 family of receptors

expressed on the surface of NK cells [224]. KIRs recognize HLA class I molecules and due

to the association of the latter with psoriasis and PsA, KIRs have been investigated as

potential candidate susceptibility loci.

The KIR gene cluster spans approximately 150 kb in the leukocyte receptor complex on

chromosome 19q13. The KIR region is highly diverse; different KIR haplotypes exhibit

differences in both gene content and allelic polymorphism. The KIR gene family currently

consists of 15 genes. A particular KIR haplotype encodes a distinct set of receptors for an

individual’s NK cells. Each NK cell has a combination of inhibitory and activating

receptors that interact with certain HLA alleles to result in an immune response. KIRs are

classified into activating and inhibitory receptors. The engagement of a long-tailed

cytoplasmic KIR (L) with HLA class I allotypes transmits an inhibitory signal while the

short-tailed KIR (S) leads to an activating signal. KIRs are also categorized on the basis of

external immunoglobulin-like domain into 2 groups (2D or 3D) [225]. The specificity of

the different KIRs to HLA class I molecules is determined by their extracellular structure.

KIR2DL2 and 2DL3 bind to HLA-C group 2 molecules that contain the amino acids Asn77

and Lys80, while KIR2DL1 binds to HLA-C group 1 molecules that contain the amino

Page 39: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

27

acids Ser77 and Asn80. KIR3DL1 binds the HLA-Bα1 helix around residues 76-80 with

specificity for all Bw4 alleles containing isoleucin at heavy chain residue 80. Studies have

shown that binding of inhibitory KIRs to specific HLA molecules correlates well with their

ability to inhibit NK cytolysis of target cells bearing those HLA allotypes. Ligands for the

activating KIRs are largely unknown despite possessing similar extracellular regions,

although it has been suggested that they recognize the same HLA molecules that are

recognized by their corresponding inhibitory KIRs [68, 226].

Overall, this system provides a mechanism by which an individual’s HLA repertoire may

directly influence the type and extent of the immune response in the context of a particular

KIR haplotype. Thus, the biologic interaction between KIRs and their specific HLA ligands

may predispose to the development of psoriasis and PsA. The interaction between HLA and

KIR genes may explain the differential susceptibility to these diseases.

Several studies have shown that the presence of genes encoding activating KIRs (KIR2DS1

and/or KIR2DS2) is associated with PsA, particularly in the absence of HLA-C alleles that

encode corresponding ligands for the inhibitory KIRs (KIR2DL1 and KIR3DL2/3) [227,

228]. Furthermore, KIR2DS1 but not KIR2DS2 was associated with psoriasis in Japanese

and Polish patients [229, 230] and among a group of North American patients a decrease in

KIR2DS1 in psoriasis, but an increase in patients with PsA has been noted [231]. It is

unclear whether these findings represent an association that is independent of the skin

disease, since PsA patients were not compared directly to those with psoriasis.

Additional KIR – HLA interactions may play a role in the pathogenesis of PsA. The

inhibitory receptor, KIR3DL1, binds HLA-B molecules with Bw4 motif [232]. The

activating receptor, KIR3DS1, shares approximately 97% sequence similarity in their extra-

cellular domain with KIR3DL1, suggesting that they may share similar ligands [233]. A

study from Spain have found that the inhibitory KIR3DL1 allele was decreased while the

activating KIR3DS1 allele was increased in AS patients compared to HLA-B*27 positive

healthy controls, suggesting that the interaction between the activating receptor and its

specific ligand may increase the susceptibility to the disease [234]. However, another large

study among AS patients was unable to replicate these findings [235]. The interaction

Page 40: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

28

between KIR3DS1 and its putative ligand HLA-Bw4 leads to activation of NK cells, which

may be beneficial for the clearance of viral infections however may also lead to the

development of autoimmune diseases. It has been shown that the combination of KIR3DS1

and HLA-Bw4 was associated with slower progression from HIV infection to AIDS, lower

viral load and protection against opportunistic infections [236]. HIV infection has been

linked to increased risk for developing psoriasis and PsA [95]. The association between

KIR3DL1/KIR3DS1 and PsA has not been assessed in the past.

1.5.12. Other candidate genes

A number of candidate genes outside chromosome 6p have been associated with psoriasis

including IL-10 on 1q31–q32, IL-12B/p40 subunit on 5q31.1–q33.1, IL-19/IL-20/IL-24 on

1q32, IL-23R on 1p31.3, IRF2 on 4q35.1, MGST2 on 4q28.3, PTPN22 on 1p13.3–p13.1,

RAPTOR on 17q25, SLC12A8 on 3q21, SLC9A3R1/NAT9 on 17q25.1 and SUMO4 on

6q25.1 [62]. Polymorphisms of interferon regulatory factor (IRF5) interacting with Class I

MHC genes were found to be associated with psoriasis [237]. Candidate genes outside 6p

have also been evaluated for a possible association with PsA. CARD 15 (NOD2) on 16q

and several SNPs from the IL-1 cluster were associated with PsA patients from

Newfoundland, however these associations have not been replicated in other populations

[238-243]. Additionally, an IL-23 receptor polymorphism on chromosome 1p31 was

associated with PsA [244]. However, this locus was also associated with psoriasis in

several studies [245-247] and does not confer an independent risk for arthritis [248].

Recently, polymorphisms within IL-13 gene on chromosome 5q31 have been associated

with PsA [249]. This association was independent of psoriasis and was replicated in two

additional populations [250, 251]. Therefore, IL-13 gene polymorphism may be one of the

first specific genetic markers for PsA that does not confer a risk for psoriasis.

Page 41: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

29

1.5.13. Genome-wide Association studies

Several GWAS among Caucasian and Chinese psoriasis patients have been performed in

recent years. In all of the studies the strongest associations were with SNPs within the

PSORS1 region. Several of these SNPs were in strong LD with known HLA-C*0602,

confirming its role as the major susceptibility genetic marker for psoriasis. A recent GWAS

from the UK found an interaction between HLA-C and ERAP1 loci. ERAP1 is involved in

MHC class I peptide processing. ERAP1 polymorphisms were associated with

susceptibility for psoriasis only among patients carrying the HLA-C risk allele [252].

Additional novel associations from GWAS provide an insight into several important

pathogenic pathways in psoriasis. Susceptibility loci for psoriasis included genes from the

NFkB signaling pathway: TNFAIP3, TNIP1, NFKBIA, REL [246, 252, 253], genes from

the Th17 signaling pathway: IL-23A, IL-23R, IL-12B, TYK2 and TRAF3IP2 [187, 245,

246, 252-254], and genes from the epidermal differentiation complex (PSORS4): LCE3B,

LCE3D and LCE3C [187, 246, 253, 254]. A recent GWAS among PsA patients from

Germany identified HLA-C, IL-12B and TRAF3IP2 as susceptibility loci for PsA

compared to healthy controls [255]. However, all these loci have already been identified as

susceptibility genes for psoriasis and therefore cannot be considered as independent

markers for PsA. Another GWAS among PsA patients from North America is underway.

The density of markers used in GWAS will however not be able to distinguish between the

various genes in the MHC region as this region has high gene density and LD and would

therefore require a much higher density of markers to distinguish the various genes

associated with disease susceptibility.

1.5.14. Limitations of previous genetic studies

The great challenge in investigating the genetics of PsA is the ability to differentiate the

genes that confer an independent risk for the joint disease from those that are associated

with the skin disease. Studies for that purpose require a large sample size, careful

characterization of clinical phenotype and appropriate controls.

Page 42: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

30

Most of the previous genetic studies in PsA included only small numbers of participants

and therefore were underpowered to detect significant associations. In addition, many of

the published genetic studies in PsA have used only healthy individuals as controls;

therefore it is difficult to conclude whether the reported associations are related to the skin

or the joint manifestation of PsA. Furthermore, those studies that have used patients with

psoriasis as controls do not provide details about whether the presence of inflammatory

arthritis was ruled out. PsA is often under diagnosed among psoriasis patients. It has been

shown that up to 18% of the psoriasis patients that are thought to have only skin disease,

actually also suffer from inflammatory arthritis after careful rheumatologic evaluation

[119]. Therefore, misclassification of cases and controls can be a major problem in genetic

studies in PsA and psoriasis if a proper rheumatologic evaluation is not performed. Lastly,

the problem of a clear definition of phenotype is complicated by the fact that until 2006,

there were no accepted classification criteria for PsA [22] and previous genetic studies in

PsA have used different definitions of phenotypes. All of these factors support the need for

further studies of the genetics of PsA.

In summary, psoriasis and PsA have substantive genetic determinants. Some susceptibility

genes are probably to be shared by psoriasis and PsA; however, it is likely that there are

some distinct genes that confer an independent risk for PsA. In PsA, several candidate

genes have been replicated in studies of disease susceptibility, most of them within the

MHC region. Past genetic studies in PsA have been limited by their small sample size,

potential misclassification of cases and controls, lack of appropriate control groups and

lack of uniform definition of phenotype.

Page 43: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

31

Chapter 2. Hypothesis and Aims

Page 44: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

32

2.1. Rationale

The nature of relationship between PsA and psoriasis is not completely clear. In one model,

since most patients with PsA develop psoriasis at some point in their disease, PsA can be

viewed as a “disease within a disease”. In this model, PsA is considered a more severe

phenotype of psoriasis that occurs due to a greater number of susceptibility genes or

environmental factors. The additional genes or environmental risk factors on the background

of psoriasis will lead to development of PsA. This model predicts that case series ascertained

by psoriasis will be genetically homogenous and that case series ascertained by PsA will

completely overlap those of psoriasis apart from several additional distinct genes. Although

other models have been suggested to describe the relation between psoriasis and PsA, the fact

that most of the susceptibility genes for psoriasis were also found to be associated with PsA

supports this model.

Approximately 85% of the patients develop PsA after or at the same time as the skin disease

[35]. Therefore, psoriasis serves as a marker for the development of PsA. Currently, there is

limited information about the epidemiology, clinical, environmental and genetic risk factors

for PsA among patients with psoriasis. The few previous epidemiological studies were

limited by their small sample size and potential misclassification of cases and controls and

lack of uniform definition of phenotype. Furthermore, studies to date have looked either at

psoriasis cohorts or at PsA cohorts, but there has been no systematic evaluation of psoriasis

patients without arthritis and patients with PsA from the same ethnic and geographic

background. The lack of direct comparison between PsA and psoriasis patients does not

allow conclusions as to whether the reported associations were related to the skin or the joint

disease. Therefore, the inclusion of both groups in one study will allow identifying markers

for PsA among psoriasis patients.

The HLA genes are associated with numerous immune-mediated diseases including psoriasis

and spondyloarthritis. GWAS among psoriasis patients have shown that by far the most

significant associations are located in this region. KIR genes have also been associated with

SpA and PsA in the past and their binding to HLA genes provide a compelling functional

Page 45: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

33

mechanism for this association. Therefore in this thesis I have decided to focus on these two

genetic regions and to assess their association with PsA.

2.2. Hypothesis

I hypothesize that genetic factors in the MHC region, most likely HLA genes on

chromosome 6p, and KIR genes on chromosome 19q, predispose patients with psoriasis to

develop PsA. Genes that differentiate patients with PsA from those with psoriasis alone serve

as markers associated with the development of PsA in patients with psoriasis.

In addition, I hypothesize that environmental factors play a role in the pathogenesis of PsA

among psoriasis patients and their interaction with susceptibility genes for PsA may lead to

the development of the disease among psoriasis patients.

2.3. Aims

1) To identify HLA alleles that confer a risk for PsA among patients with psoriasis. (Chapter

5)

2) To determine the association between KIR genes and PsA and to assess the joint statistical

association of KIR genes and their corresponding HLA alleles with PsA. (Chapter 7)

3) To identify environmental risk factors for PsA among patients with psoriasis. (Chapter 8)

4) To determine whether there is an interaction between the selected environmental risk

factors and genetic markers for PsA in the susceptibility for PsA. (Chapter 9)

5) To determine the rate of PsA among psoriasis patients and whether it is affected by HLA

risk alleles. (Chapter 4, 6)

Page 46: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

34

Chapter 3. Study population and data collection

Page 47: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

35

3.1. Study population

3.1.1. Group 1- Psoriasis without arthritis

This group includes individuals from a recently established, ongoing prospective cohort of

adult psoriasis patients without arthritis. The cohort forms the basis of a long term

prospective study that aims to assess genetic and clinical risk factors for the development of

inflammatory arthritis among patients with psoriasis. All potential study subjects have a

diagnosis of psoriasis confirmed by a dermatologist. The sources of recruitment are varied

and include patients with a range of psoriasis types and severity. Patients were mainly

recruited from dermatology clinics and phototherapy centers in the Greater Toronto Area and

also from family practice clinics and through advertisement in flyers posted in several

hospitals and local media. By approaching patients with wide range of disease severity we

aimed to minimize a selection bias of patients with more severe psoriasis. The cohort

includes different ethnic groups although most of the patients (86%) are Caucasians.

The inclusion criteria required a diagnosis of psoriasis confirmed by a dermatologist. The

exclusion criteria were the presence of inflammatory arthritis or spondylitis in the past or at

the time of the assessment. All subjects were evaluated by experienced rheumatologists from

the PsA research team to exclude inflammatory arthritis and spondylitis before enrollment.

Subjects were interviewed and examined according to a standardized protocol similar to that

used in the PsA clinic [35]. Information about demographics, co-morbidities and

rheumatologic symptoms was collected. Each subject underwent a comprehensive

musculoskeletal examination that included joint assessment for tenderness, swelling and

deformities, evaluation for the presence of enthesitis, tendonitis and dactylitis, and

assessment for restriction of movement in the spine. If there were definite clinical findings of

inflammatory arthritis, enthesitis or spondylitis, the patient was excluded from the study. In

cases of doubt, imaging studies, including radiographs, ultrasound or MRI, were performed

as indicated to investigate the nature of the abnormality. Patients thus diagnosed with

psoriasis that have no current evidence or past history of PsA were eligible for the study. If a

non-inflammatory condition, such as osteoarthritis, was diagnosed, the subject was included

Page 48: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

36

in the study. This process ensured that all participants do not have clinical inflammatory

arthritis at enrollment. The recruitment to the cohort started in January 2006. Thus far, 564

psoriasis patients were screened by a rheumatologist and to date (March 2011), 499 patients

are included in the cohort. Following the screening assessment, 65 individuals were found

ineligible to participate in the study, 34 of them due to PsA.

3.1.1.1. Follow-up in the Psoriasis cohort

All study participants are re-assessed annually according to the same protocol regardless of

their symptoms. Patients who fail to come to the yearly assessment are requested by

telephone and by mail to complete the Toronto Psoriatic Arthritis Screen (ToPAS)

questionnaire, a screening questionnaire designed to detect PsA among patients with

psoriasis as well as the general population. The ToPAS has been validated in different

populations of patients with skin conditions other than psoriasis, PsA and healthy subjects

[256]. Subjects scoring ≥8 points on the ToPAS screen were requested to come to the clinic

for an assessment.

The diagnosis of inflammatory arthritis or spondylitis was determined by the PsA research

team that included several experienced rheumatologists, after reviewing the clinical,

laboratory and imaging data. Patients were classified as having PsA if they fulfilled the

CASPAR criteria [22]. Patients diagnosed with PsA were considered to have developed the

outcome of interest. For the purpose of analysis in the case-control study designs these

incident cases were included in the PsA group.

3.1.2. Group 2 - Patients with PsA

This group includes adult prevalent PsA patients. All patients are from the University of

Toronto Psoriatic Arthritis clinic database, which currently (March 2011) numbers 1143

patients. The PsA clinic was established in 1978 as part of an ongoing prospective study.

Patients are registered in the clinic if they are diagnosed as having PsA. The patients in the

Page 49: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

37

PsA clinic represent a wide spectrum of disease that is related to the broad referral base of

the clinic. The clinic serves as a primary, secondary and tertiary referral centre for PsA

patients from the Greater Toronto Area and Southern Ontario. Furthermore, patients are

followed regularly, irrespective of their disease activity. Many of the patients were referred

from dermatology clinics for the assessment of PsA, thus minimizing ascertainment bias

between the PsA and the psoriasis groups, since most of the psoriasis patients were also

recruited from dermatology clinics.

Since classification criteria for PsA were not available until 2006, the diagnosis of PsA was

based on the presence of psoriasis and inflammatory arthritis and exclusion of other types of

arthritis. However, 98% of the patients in the clinic satisfy the CASPAR criteria for

classification of PsA. The patients are reviewed at initial clinic entry and at 6-12 month

intervals according to a standard protocol [35] using validated clinical and radiographic

measurement tools.

3.1.3. Group 3-Healthy Controls

Control DNA was obtained from 707 healthy Caucasian volunteers, cadaveric organ donors

from laboratories at the University Health Network (usually young previously healthy

individuals appropriate for organ donation) and from a commercial biobank. Blood samples

were used for DNA typing however no additional clinical information is available.

3.2. Data collection

The following information was collected as part of the standardized protocol for each subject

in groups 1 and 2: demographic information, race and ethnicity (based on self-report),

country of origin, year of diagnosis of psoriasis and PsA, family history of psoriasis, PsA and

other SpA, alcohol and smoking habits, co-morbidities and medication history. Additional

information was collected through physical examination: tender and swollen joint count,

number of dactylitic digits, active entheseal sites, clinically damaged joint counts and

measurements of spinal mobility. An “actively inflamed” joint was defined as the presence of

Page 50: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

38

stress pain and/or effusion. Enthesitis was defined as tenderness at sites of insertion of

tendons and ligaments into bones (initially including only the Achilles tendon and plantar

fascia and since 2006 according to the SPARCC enthesitis index [257]). Dactylitis was

defined as the swelling of a whole digit with tenderness. Clinically damaged joint was

defined as the presence of limitation of range of movement of >20% of the range not related

to the presence of joint effusion, presence of joint deformity, subluxation, loosening, or

ankylosis. Measurements of spinal mobility were made according to a standard protocol. At

the lumbar spine these included forward flexion (modified Schober's test), lateral flexion

(Domjan and INSPIRE methods) [257]. Chest expansion, cervical rotation and Occiput-to-

wall distance were also measured. Psoriasis activity was determined using the PASI score

[16]and modified Nail Psoriasis Severity Index (mNAPSI) [258] which are validated tools

for assessment of severity of skin and nail involvement, respectively. Information about

concomitant use of DMARDs or biologic medications was also collected.

Radiographic damage was assessed according to a modification of the Steinbrocker method

[259]. Radiographic lumbar and cervical spine damage was scored according to the modified

Stokes Ankylosing Spondylitis Spine Score(mSASSS) [260]. Sacroiliitis was defined

according to the New York criteria [261].

All information is tracked and on a computerized database.

Page 51: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

39

Chapter 4. The incidence of arthritis in a prospective cohort of psoriasis patients

Page 52: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

40

4.1. Background

The estimated prevalence of inflammatory arthritis among patients with psoriasis has varied

from 6% to 42% [31]. Approximately 67% of the patients develop psoriasis before arthritis

and in 16% arthritis and psoriasis present within 12 month of each other [31]. There is

limited information about the incidence of PsA among patients with psoriasis. A

retrospective study from Germany reported that the cumulative incidence of PsA among

psoriasis patients reached 20.5% after 30 years from the diagnosis of psoriasis [32]. Another

retrospective study from Rochester, Minnesota has found a lower cumulative incidence of

3.1% cases of PsA among psoriasis patients after 10 years from the onset of the skin disease

[33]. A prospective study is considered the preferred study design to determine the incidence

of a disease; however, no study to date has prospectively assessed the incidence of PsA

among psoriasis patients.

Our group has recently established a cohort of patients with psoriasis uncomplicated by

arthritis. The patients are followed prospectively to detect patients that develop inflammatory

arthritis. This report summarizes the results of the first four years of follow-up and reports

the cumulative incidence of PsA.

Page 53: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

41

4.2. Methods

4.2.1. Setting

The Toronto Psoriasis Cohort was established in 2006 and enrolls psoriasis patients without

inflammatory arthritis or spondylitis. The cohort forms the basis of a long term prospective

study that aims to assess genetic, clinical and environmental risk factors for inflammatory

arthritis among psoriasis patients. The prospective design of the study provides the

opportunity to determine the incidence of inflammatory arthritis in psoriasis patients. The

recruitment for this ongoing study commenced in January 2006.

4.2.2. Patients and Assessments

A detailed description of patients’ recruitment and assessment is provided in the Methods

section (Chapter 3.1 – Study population – Group 1 psoriasis without arthritis). Briefly, all

potential study subjects have a diagnosis of psoriasis confirmed by a dermatologist. The

sources of recruitment are varied and include patients with a range of psoriasis types and

severity. Patients are mainly recruited from dermatology clinics and phototherapy centers in

the Greater Toronto Area and also from family practice clinics and through advertisement in

flyers posted in several hospitals and local media. The exclusion criteria are the presence of

inflammatory arthritis or spondylitis in the past or at the time of the assessment. All subjects

are evaluated by rheumatologists from the PsA research program to exclude inflammatory

arthritis and spondylitis before enrollment. If there are clinical findings of inflammatory

arthritis, enthesitis or spondylitis, the patient is excluded from the study. Where the diagnosis

of PsA is not clear, imaging studies are performed as indicated to investigate the nature of the

abnormality. If a non-inflammatory condition, such as osteoarthritis, is diagnosed, the subject

was included in the study. This process ensured that no participants have clinical

inflammatory arthritis at enrollment.

Page 54: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

42

All study participants are then re-assessed annually regardless of their symptoms. Patients

who failed to come to the yearly assessment were contacted and requested to fill out the

ToPAS questionnaire [256], a screening questionnaire designed to detect PsA among patients

with psoriasis. Subjects scoring ≥8 points on the ToPAS screen were requested to come to

the clinic for an assessment.

4.2.3. Case definition

The diagnosis of inflammatory arthritis or spondylitis was determined by the PsA research

team that included several experienced rheumatologists, after reviewing the clinical,

laboratory and imaging data. Patients were classified as having PsA if they fulfilled the

CASPAR criteria [22]. Patients diagnosed with PsA were considered to have developed the

outcome of interest. Their years at risk were the time since entry into the cohort to the

diagnosis of PsA. Patients not found to have developed PsA were considered at risk from

entry into the cohort to the last contact.

4.2.4. Statistical Analysis

Descriptive statistics were computed with continuous variables summarized by their means

and standard deviations and categorical variables summarized by proportions.

The person-years at risk was calculated as the time between enrollment date and last

assessment date (in subjects not developing PsA) or the date of PsA diagnosis, whichever

came first. This duration was used to estimate the cumulative incidence of PsA among

patients with psoriasis. Some patients did not have any follow-up visits after their initial

screening visit and therefore did not contribute to follow-up time. However, on the

assumption that these patients would have contacted the clinic if they developed symptoms

of arthritis, we performed a secondary analysis that included these patients, assuming that

they did not develop PsA by December 31, 2009.

Page 55: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

43

There is a common notion among PsA researchers that the greatest risk of developing the

disease is during the first years following the onset of psoriasis. We have used two

parametric models to evaluate whether the risk of developing PsA, as modeled by the hazard

function, changes over time. A time homogeneous (exponential) model, that assumes a

constant hazard function, was used to estimate the rate of PsA among psoriasis patients. This

model was compared to a Weibull model that assumes a trend in hazard function. The lack of

a significant difference between the two models, as assessed by the likelihood ratio (LR) test,

was taken as lack of evidence for time-dependent rate. A Kaplan-Meier curve was used to

obtain a nonparametric estimate of the cumulative incidence of PsA over time.

Page 56: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

44

4.3. Results

The outcome of the 313 patients that were recruited from January 2006 until December 2008

is summarized in Figure 4.1. The sources of recruitment were largely from dermatology

clinics and phototherapy centers (289 patients (92.3%). The rest were recruited through local

advertisement (20 patients (6.4%)) and from a general rheumatology clinic (4 patients

(1.3%)). The patients recruited from the rheumatology clinic had been referred for an

assessment of musculoskeletal pain; however they were found to have non-inflammatory

articular problems and therefore were included in the cohort. In total, 10 subjects developed

PsA during 4 years of the prospective study follow-up; 253 participants had at least one

follow-up visit and were free of inflammatory arthritis. Twenty patients filled out the ToPAS

questionnaire. None of them scored 8 or more, therefore, they were all considered as not

having PsA at that time point.

Overall, 50 subjects did not have any follow-up assessment. 17 declined to return for a

follow up visit or to fill out the ToPAS questionnaire. We were unable to contact 30, two

moved from the region and one patient had died. This cohort had a total of 534.5 person-

years of follow-up with a mean of 1.71+1.01 years per person. Among the 10 incident cases,

the duration of time from enrolment to development of PsA ranged from 4 to 42 months,

with 3 patients that developed PsA within the first year of follow-up.

4.3.1. Incidence of PsA among patients with psoriasis

The annual incidence rate was 1.87 [95% confidence interval (CI) (0.71-3.03)] PsA cases per

100 psoriasis patients when patients with no follow-up visits were assumed not to have

developed PsA by December 31, 2009. The incidence rate increased to 2.53 (95% CI 0.96,

4.1) when only the 253 participants with at least one follow-up assessment were included. A

Kaplan-Meier curve and the estimated cumulative probability of developing PsA among the

study participants are shown in Figure 4.1. The distribution of the time to the development of

PsA was fit with an exponential model, which has a constant hazard rate. Tests for trend did

Page 57: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

45

not suggest a departure from the constant hazard, so there was insufficient evidence to claim

a trend in the risk of disease.

4.3.2. Characteristics of the PsA cases

Ten patients developed new onset PsA during the study follow-up period. The characteristics

of the study population and the incident cases of PsA are presented in Table 4.1. The mean

age at onset of psoriasis and arthritis were 34.6±14.2, 52.2±8.8 years, respectively. Four of

the patients were male, 6 had a family history of psoriasis; however, none reported a family

history of PsA. The most common type of psoriasis was chronic plaque psoriasis (8 patients)

followed by guttate psoriasis (2 patients). The mean PASI score at presentation of arthritis

was 6.7 and 6 patients had psoriatic nail lesions.

At the time of the diagnosis, 8 patients had oligoarthritis and 2 had polyarthritis, and 3

patients had additional evidence of spondylitis. All of the patients were symptomatic at the

time of the diagnosis of PsA. The median number of actively inflamed joints was 3 (range 0-

27) with 1 swollen joint (range 0-3). Three patients had evidence of enthesitis, the most

common site being the plantar fascia and none had dactylitis. Only one patient had evidence

of a clinically damaged joint (1 joint). All of the patients were seronegative for rheumatoid

factor. Only three patients had elevated inflammatory markers, either ESR or CRP. Two

patients had evidence of definite radiographic erosions with a total modified Steinbrocker

score of 2 and 13, respectively. Two patients had grade 3 bilateral sacroiliitis and none had

evidence of syndesmophytes (mSASSS score was 0). These two patients did not have back

pain nor any limitation of spinal mobility at baseline assessment, therefore, no radiographic

studies were performed. Furthermore, none of these patients had back pain at the time of the

diagnosis of PsA and they were diagnosed based on findings of peripheral oligoarthritis.

Page 58: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

46

Figure 4.1 - Follow-up summary of the study population

(January 2006 – December 2009)

Free of arthritis at last assessment: 253 patients

Developed PsA:

10 patients

No Follow-up visit:

50 patients

313 psoriasis patients recruited from

Jan. 2006-Dec. 2008

20 patients were free of arthritis

based on the ToPAS questionnaire - Declined to return to visit or to fill-out ToPAS: 17 patients - Unable to contact: 30 patients - Moved from the region: 2 patients - Died: 1 patient

Page 59: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

47

Figure 4.2 - PsA incidence over time estimated with a Kaplan-Meir (KM) curve (95% CI) from time of first clinic visit to diagnosis of PsA, along with an estimated cumulative incidence curve based on an exponential model (Exp).

Page 60: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

48

Table 4.1 - Characteristics of the study population and the incident cases of PsA

Variable Psoriasis

(N=313)

Incident cases of PsA

(N=10)

Age (Mean ± SD) Sex: Male (%) Ethnicity: Caucasian (%) Family history of psoriasis (%) Family history of PsA (%) Duration of psoriasis (Years) Psoriasis type

- Chronic plaque psoriasis - Guttate - Pustular - Flexural

PASI score (Mean ± SD) Psoriatic nail lesion (%) Current use of medications DMARDs Biologic agents

46.5±12.8 183 (58.5%) 269 (86.2%) 127 (40.6%)

7 (2.2%) 15.8±14.0

270 (86.5%) 48 (15.4%) 13 (4.1%) 9 (2.9%) 5.4±5.7

249 (81%)

21 (18.8%)

4 (4.1%)

50.4±9.9 4 (40%) 9 (90%) 6 (60%) 0 (0%)

17.6 ±10.3

8 (80%) 2 (20%)

- -

6.7±6.5 6 (60%)

2 (20%)

0 (0%)

PsA – Psoriatic Arthritis, PASI – Psoriasis Area and Severity Index, DMARDs – Disease Modifying Anti Rheumatic Drugs.

Page 61: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

49

4.4. Discussion

The study presents results from an ongoing prospective longitudinal cohort of psoriasis

patients. To my knowledge, this is the first study that has prospectively assessed the

incidence of PsA among psoriasis patients. After 4 years of follow-up, the annual incidence

rate was 1.87 cases among 100 psoriasis patients.

In contrast to the prevailing notion that the greatest risk of developing PsA is during the first

years following the onset of psoriasis, the present report and another retrospective cohort

study from Germany have shown that the rate of development of PsA among psoriasis patient

is constant and unrelated to the duration of psoriasis [32]. Although several epidemiologic

studies have assessed the incidence of PsA in the general population, there is limited

information about the incidence of PsA among psoriasis patients. In a population based

retrospective study that used medical records to confirm the diagnosis of PsA, Wilson et al.,

from Rochester, estimated that the cumulative incidence of PsA was 3.1% after 10 years

from the onset of psoriasis and 5.1% after 20 years [33]. Several reasons, other than

geographic and ethnic differences, may explain the differences in the cumulative incidence

between our study and those of the group from Rochester. First, Wilson et al. identified cases

based on a retrospective review of computerized medical records, while in our study each

subject was examined prospectively by a rheumatologist. Underestimation and

misclassification of cases are major threats in epidemiological studies that rely on

computerized data registries, particularly since widely accepted classification criteria for PsA

were unavailable until recently.

Furthermore, underestimation of PsA among psoriasis patients may be even more

pronounced. In a German study, approximately 18% of the psoriasis patients that were found

to have inflammatory arthritis were unaware of their condition and had never seen by a

rheumatologist [119]. A referral bias may also account for the difference, since by relying

on medical records some of the mild cases of arthritis may not come to medical attention.

The prospective design of the study and careful assessment of each subject allowed us to

diagnose early and milder cases that potentially could have escaped medical attention.

Page 62: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

50

Finally, the differences in the source populations of psoriasis patients may explain some of

the differences. The present study included mainly patients with moderate to severe psoriasis

from dermatology clinics, while the Rochester study assessed patients with a wider range of

psoriasis severities. Several studies in the past suggested that the severity of psoriasis is

associated with higher risk of developing PsA [24, 262]. A recently published study from

Europe that assessed the presence of PsA among psoriasis patients attending dermatology

clinics reported a cumulative incidence of PsA that was closer to that found in this study

(20.5% of patients developed PsA after 30 years from the diagnosis of psoriasis) [32].

In this study, most of the patients had oligoarthritis at the time of the diagnosis and only a

few had developed clinical or radiographic joint damage, which indicates an early diagnosis

of the disease. These findings are in accordance with Kane et al. that have found that 27% of

129 PsA patients assessed within 5 months of onset of symptoms had joint erosions [263].

The course of these patients with mild forms of arthritis cannot be predicted, although our

previous studies suggest that male patients with a smaller number of involved joints fare

better [43].

In summary, the results of 4 years of follow-up of a newly established cohort of psoriasis

patients without arthritis were described. The annual incidence rate of arthritis among the

cohort population was 1.87 cases among 100 psoriasis patients. These figures suggest that the

incidence rate of PsA may be higher than previously reported, particularly among patients

with moderate to severe psoriasis.

Page 63: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

51

Chapter 5. HLA risk alleles for PsA among psoriasis patients

Page 64: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

52

5.1. Background

PsA can be considered as a “disease within a disease”, as almost all patients with PsA will

develop psoriasis at some point in their lifetime. The strong familial aggregation, as

demonstrated by high recurrence rate of PsA and psoriasis in relatives supports the genetic

basis of these diseases [264].

Candidate gene and genome wide studies have shown a strong association between psoriasis

and the MHC region [166-170] particularly with a 300kb-segment in the MHC class I region

on chromosome 6p21.3 known as PSORS1 [171, 172]. Within this region the largest and

most consistently reported association is with the HLA-C*06 [180] allele that is linked to

earlier onset and more severe psoriasis [178, 179].

The MHC region, particularly HLA genes, has also been associated with PsA [41, 188-192].

While HLA-C*06 is more frequent in PsA patients compared to the general population, this

association is stronger with psoriasis than with PsA [193]. Several studies reported an

association between HLA-B*13, B*57 and DRB1*07 and PsA. However, these results are

most likely secondary to the presence of these alleles on the recognized ancestral haplotypes:

AH.13 and AH.57, both contain HLA-C*06 [181, 182]. In a case-control study that

compared PsA to psoriasis, HLA-B*27 and B*07 frequencies were higher in PsA [41]. HLA-

DRB1*04 was reported to confer a risk for PsA [189, 195, 196], but several investigators

found no such association [41, 197]. Evidence for the role of HLA in susceptibility to PsA

has also been demonstrated by increased sharing of HLA haplotypes among sib pairs

concordant for PsA but not those concordant for psoriasis [198].

The great challenge in investigating the genetics of PsA is related to the difficulty in

differentiating the genes that confer risk for the joint disease from those associated with the

cutaneous disease. Many previous genetic studies in PsA have used only healthy individuals

as controls, preventing the determination of whether any significant association was related

to cutaneous disease or to joint manifestations of PsA. Additional limitations of previous

studies include small sample sizes, potential misclassification of cases and controls and the

lack of uniform classification criteria for the clinical definition of the PsA phenotype.

Page 65: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

53

Genes that differentiate patients with PsA from those with psoriasis alone may serve as

markers for the development of PsA in patients with psoriasis. In this study I aimed to

identify HLA alleles that confer a risk for PsA in psoriasis patients.

Page 66: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

54

5.2. Methods

5.2.1. Study population

In this population-based case-control study three groups of individuals from the same

geographic region were compared. All participants were Caucasians (by self-report) from the

Greater Toronto Area.

The PsA group included 712 adult PsA patients recruited from the University of Toronto PsA

cohort. All patients were carefully phenotyped by a rheumatologist and satisfied the

CASPAR criteria for the classification of PsA [22].

The psoriasis group included 335 psoriasis patients who were recruited from a prospective

cohort of psoriasis patients. These patients had psoriasis confirmed by a dermatologist and

were assessed by a rheumatologist to rule out inflammatory arthritis. Therefore at the time of

analysis, all psoriasis patients were free of arthritis.

Healthy Controls DNA was available from the laboratory control biobank as well as from

ethnically matched unrelated healthy volunteers from the Toronto area.

The groups of PsA and psoriasis patients were combined to create a Psoriatic Disease group

(PsD) that was compared to the healthy control group.

A population based case-control study design is the most commonly used method for

investigating susceptibility genes of a disease. The main advantages of this method are the

lower costs compared to a cohort study and the fact that the predictor variable (the gene

effect) is not subjected to a recall bias. Furthermore, the method is generally considered to

have more power to identifying susceptibility genes for a disease compared to a family-based

case-control study and other types of study designs. A potential limitation of a case-control

design, that considers psoriasis patients as controls, is that the outcome of interest (PsA) may

not be rare among the “controls”. This may result in misclassification of cases and controls as

a proportion of the controls may become cases in the future. Despite the previously described

limitation I have used the case-control study design as the primary method to identify

Page 67: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

55

susceptibility genes for PsA among psoriasis patients. Other types of analyses will be used to

determine the associations that will be identified in the population based case-control study.

5.2.2. HLA typing

DNA was extracted from peripheral blood using a modified salting out procedure (Gentra®

Puregene® Blood Kit). Extracted genomic DNA was amplified by PCR using locus specific

primers for each of the HLA-A, -B, -C, -DR and DQ loci. PCR amplicons were identified by

Sequence Specific Oligonucleotide (SSO) probes using the reverse line blot technique.

(RELITM SSO HLA typing kits) [265]. Ambiguous results were resolved using Sequence

Specific Primers (PCR-SSP).

5.2.3. Sample size

Power calculations show that a study comparing 712 PsA patients to 335 psoriasis patients

has >80% power to detect an odds ratio of 2.5, 2.0, 1.7 and 1.6, for the frequencies of the risk

allele of 0.05, 0.1, 0.2 and 0.3, respectively (p<0.01). Power calculations were performed

using PS Power and Sample Size Calculations version 3.0 [266].

5.2.4. Statistical methods

5.2.4.1. Single locus analysis

Since HLA antigens are expressed in a co-dominant manner, the frequency of each of the

HLA alleles was used for comparisons. Since the study was underpowered to detect the

association of rare alleles, only those with a frequency of > 1% in the study population were

included in the analysis. Overall, the presence of each of 105 HLA alleles at 5 HLA loci was

tested, of these, 11 rare alleles were excluded. A Likelihood Ratio (LR) test was used to

assess the differences in allelic distribution between the PsA vs. psoriasis and PsD vs. healthy

controls. To avoid false positive results due to multiple testing (type I error) the False

Page 68: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

56

Discovery Rate (FDR) approach was employed [267] to account for multiple testing. FDR is

the expected proportion of false discoveries among all discoveries, limiting false positives to

a specific proportion. This method is less conservative than the Bonferroni correction and

increases the power to detect significant associations [268]. Each HLA locus was assessed

separately. Significance criteria of p < 0.01 (p=0.05 divided by the number of tested HLA

loci (5)) were applied.

5.2.4.2. Multi-locus analysis

Following the initial comparisons of allele frequencies, multivariate logistic regression

analysis was used to identify key genetic differences between PsA and psoriasis while

accounting for LD between loci and dependence among allele frequencies within a locus. A

targeted approach was employed to select variables for inclusion in the full regression model.

Previous literature showed that alleles with the strongest PsA associations are within the

HLA-B and –C loci [41, 188-192]. Therefore, in a screening step, a separate regression

model for each of these two HLA loci was constructed by backward selection. In the second

step, a single full logistic regression model was constructed for both loci. This model

included the following alleles: HLA-B*27, B*57, B*08, B*38, C*01, C*02, C*06, C*07,

C*12 and DRB1*07. A stepwise logistic regression was used to identify HLA alleles

occurring more or less frequently in PsA compared to psoriasis patients. Allelic associations

that were considered statistically significant were retained in the multivariate regression if the

p-value from the 2-sided Wald-test was less than 0.05. Statistical analysis was performed

using SAS 9.2© statistical software.

5.2.4.3. The association between HLA and sub-phenotypes of PsA

As in many complex diseases, there are large variations in the phenotype of PsA. It has been

shown that refining the phenotype may strengthen genetic associations by decreasing the

heterogeneity of the cases [269]. Furthermore, different mechanisms may lead to the

development of different phenotypes, as in the case of seropositive vs. seronegative

Page 69: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

57

rheumatoid arthritis [270]. To facilitate the identification of susceptibility genes in complex

diseases such as PsA, one approach is to split the disorder based on different phenotypes.

This approach is one of the preferred methods to reduce genetic heterogeneity. The goal is to

split the disorder into subgroups based on clinical characteristics known to differentiate the

disorder into more homogenous groups. These subgroups are then analyzed separately for

association with candidate genes. For this study the PsA group was subdivided into axial and

peripheral PsA and each group was then compared separately to patients with psoriasis. This

classification has been shown in the past to be associated with different HLA alleles in PsA

[41, 189, 192]. Axial PsA was defined by radiological evidence of either bilateral at least

grade 2 sacroiliitis or unilateral grade 3 or 4 sacroiliitis at any point during the follow-up,

with or without peripheral arthritis. Peripheral PsA was defined on the basis of clinical and

radiographic evidence of peripheral joint involvement and no evidence of axial involvement

as defined above. Patients with unilateral sacroiliitis grade 2 were excluded from the

analysis. A similar statistical approach as used for the multi-locus analysis was employed to

identify the association of HLA alleles with subtypes of PsA.

Since HLA-C*06 is strongly associated with age at onset of psoriasis [9], the PsA and

psoriasis groups were sub-divided into patients with early and late onset psoriasis (early

onset: age ≤ 40, late onset: age > 40). The frequencies of HLA-C*06 were compared in these

sub-groups using the LR test.

Calculation of Positive Predictive Values (PPV) of HLA risk alleles

Sensitivity, specificity were calculated for each of the identified HLA risk alleles. Assuming

a 30% prevalence of PsA among psoriasis patients, the calculated specificity and sensitivity

were used to calculate the PPV of the respective HLA alleles to assess their in identifying

PsA patients among patients with psoriasis.

Page 70: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

58

5.2.4.4. Haplotype Analysis

A haplotype is a combination of alleles at multiple loci on a single chromosome. Based on

the initial single locus analysis of our HLA data in psoriasis and PsA patients it appears that

several HLA-B and HLA-C alleles confer a risk for PsA among psoriasis patients. However,

due to the strong LD between the alleles in the different HLA loci it is difficult to conclude

which one imparts the primary association. Haplotype analysis has several advantages over a

single locus analysis. It is considered to be more powerful to detect significant association

compared to allele or genotype associations since haplotypes might include two or more

causative sites whose combined effect is measurable, particularly if they show synergistic

interaction [125]. Another reason is that fewer tests need to be performed. Lastly, the

causative locus may reside between the two tested loci; however, it is less likely in this case,

since there is no known gene that lies between HLA-B and HLA-C.

Haplotype analysis was performed only for HLA-C and HLA-B alleles as these loci have

shown the strongest association with PsA in the single locus analysis. Haplotype information

was inferred among PsA and psoriasis patients using the Expectation-Maximization (EM)

algorithm (SAS-Genetics©) [271] generating Maximum Likelihood Estimates given a multi-

locus sample of HLA-C and HLA-B genotypes [272-274]. Because pedigree data was

unavailable for most patients and each individual linkage phase is unknown, there might have

been two pairs of haplotypes which were consistent with the observed genotype. Posterior

probabilities of all possible haplotypes for an individual, conditional on the observed

genotypes, were estimated using EM algorithm implemented in SAS-Genetics using a

maximal number of 20 iteration and convergence criteria of 0.00001. The resulting posterior

probabilities made it possible to test the haplotype-disease association while accounting for

the uncertainly in haplotype estimation. Because of the imprecision involved in estimating

the effects of low-frequency haplotypes, only those with an estimated frequency of more than

1% in the study population were included in the analysis. Stepwise logistic regression was

used to identify haplotypes occurring at higher or lower frequencies in PsA compared to

psoriasis and PsD compared to healthy controls. Haplotype associations were considered

statistically significant if the p-value from the 2-sided Wald-test was less than 0.05. Linkage

disequilibrium between HLA-C and HLA-B alleles was calculated using Lowenstein D’

Page 71: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

59

statistics. D’ weights the contribution of specific allele pairs to the LD using the product of

their allelic frequencies. The measure is normalized to fall between 0 and 1, with higher

values indicating a stronger LD contribution.

5.2.5. The association between HLA alleles and PsA - Family based association study

HLA alleles that were identified in the population based study as conferring a risk for PsA

were further assessed in a family based association study. The advantage of this type of study

design is that it avoids population stratification bias which can be a major threat to the

validity of any population based genetic study.

5.2.5.1. Study population and methods

PsA and psoriasis probands and their 1st degree family members (parents and siblings) were

included in this analysis. Altogether, 178 PsA probands, 30 psoriasis probands and 561 first

degree family members belonging to 208 families were analyzed.

The PsA probands were part of the University of Toronto PsA cohort. One hundred of them

were recruited consecutively to participate in a previous study that assessed familial

aggregation of PsA. The remaining patients participated in a linkage study and were recruited

due to a positive family history of psoriasis and PsA. The psoriasis probands were part of the

psoriasis cohort and were recruited consecutively. All family members were evaluated for the

presence of psoriasis and inflammatory arthritis using a screening questionnaire, physical

examination by a rheumatologist and additional tests if clinically indicated as described in

details in the Methods section. All family members were genotyped for all HLA loci as

described previously in section 5.2.2 of this chapter.

Page 72: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

60

5.2.5.2. Definitions of traits

In the population based study three groups were assessed: PsA, psoriasis, and healthy

individuals. Since the Family Based Association Test (FBAT) method allows direct

comparison of only two traits the data were analyzed in two ways.

1. In order to compare PsA and psoriasis, PsA patients were coded as affected and

psoriasis as unaffected. Healthy siblings were excluded from the analysis. Since the disease

status of the parents is not relevant for calculation of the FBAT test statistics they were

included in the analysis irrespective of their trait.

2. In order to compare PsD and healthy controls, all available information was included.

PsA and psoriasis probands and siblings were coded as affected. Healthy siblings were coded

as unaffected. In this analysis probands with PsD were compared to their healthy siblings.

5.2.5.3. Statistical analysis - Family based association test

Only HLA alleles that were identified as associated with PsA in the population based study

(HLA- B*27, B*38, B*08, B*39, C*01, C*02, C*06, C*07 and C*12) were analyzed. FBAT

version 2.0.3 software was used for the analysis [275]. FBAT uses a unified approach to

family-based tests of association, introduced by Rabinowitz and Laird [276]. The method is

based on the original Transmission Disequilibrium Test (TDT) method [277] in which

alleles transmitted to affected offspring are compared with the expected distribution of alleles

among offspring under Mendelian transmission. The FBAT test statistic is based on the

distribution of the offspring genotypes conditional on any trait information and on the

parental genotypes. If one of the parental genotypes in a trio is not observed, the test statistic

is conditioned on the sufficient statistics for the offspring distribution. FBAT handles

pedigrees by breaking each pedigree into all possible nuclear families, and evaluating their

contribution to the test statistic independently.

A family based test statistic (Z value) and p value were calculated. Z of more than 0 indicates

over-transmission of the allele, while Z of less than 0 indicates under-transmission. An

additive model was specified. The test statistic was computed using the empirical variance (-

Page 73: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

61

e) [278]. This option is used since we were testing for an association in an area of known

linkage with multiple sibs in a family.

The power of a family based study to detect an association is related in part to the number of

informative families. FBAT requires at least one heterozygote parent per family; otherwise

the family cannot be used, since it is not possible to determine which of the two alleles was

transmitted. Such families are called non-informative families. In this study, the number of

families that contributed to a specific allele test depended on whether the allele of interest

was observed in the family. Since each HLA locus is multiallelic, the frequency of each

allele in the study population was relatively low resulting in a relatively low number of

informative families for each allele.

5.3. Population stratification

Population stratification refers to a situation in which cases and controls differ with respect to

their ethnic background or another variable that may have resulted in a pattern of nonrandom

mating [279]. There are two basic conditions that must be met before population stratification

can be considered a threat to internal validity. The first condition is the presence of

significant population subgroup differences with respect to the genetic variable in question

within the sample of a particular study (i.e., the frequency of the tested allele must vary

across population subgroups within the study sample). The second condition is that there

must be significant differences across the same population subgroups with respect to the

outcome variable (i.e., the population subgroups that differ in allele frequency must also

differ significantly with respect to the outcome variable).

In the present study the likelihood that population stratification poses a major threat to

internal validity is low for several reasons:

1. The study was limited to Caucasians from the same geographic area.

2. In moderate scale studies that assess candidate genes; the effect of population

stratification is not large unless subjects are sampled from a few very distinct strata

[280]. Within the Caucasian population in the Greater Toronto Area there are

Page 74: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

62

multiple subpopulations. Information about grandparental country of origin was

available for most participants and stratification within Caucasians was compared

based on categorization of country of origin to: North and South Europe, North

America and Others (mostly from Australia and South Africa). There were no

significant differences in the distribution of the sub-populations between PsA and

psoriasis patients (Table 5.1).

Table 5.1 - Population stratification within the Caucasian study sample

Grandparental country of origin PsA (N=529) Psoriasis (N=328)

North Europe 377 (71.2%)

215 (65.6%)

South Europe 106 (20%) 65 (19.8%)

North America 37 (7%) 35 (10.6%)

Other

9 (1.8%) 13 (4%)

3. Several studies have questioned the magnitude of the effect of population

stratification in moderate scale studies [281, 282]. Khlat et al. performed a simulation

study that quantified the bias and the probability of false association (Type I error) in

different scenarios of population sub-structures. They concluded that in moderate

sample size, such as in the current study, the magnitude of bias is small when the

differences in the allele frequency in question between the sub-populations do not

exceed 20% [283]. The difference in HLA allelic distribution within European

subpopulations is not that large. For example, the prevalence of HLA-C*06 among

Caucasians from European origin ranges from 4.6% to 11.5% [284].

Page 75: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

63

4. Population stratification does not threaten the validity of family study designs since

these studies use within family information. Therefore, in the present study

significant associations were further assessed in a family based design.

In summary, in this study several measures have been taken to minimize the effect of

population stratification bias. Furthermore, based on a previous simulation study the

sample size and the population frequencies of the tested alleles indicate that the bias,

if present at all, is negligible. Therefore, population stratification bias does not pose a

threat to the validity of the present study.

Page 76: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

64

5.4. Results Demographic and clinical characteristics of the study population are presented in Table 5.2.

The psoriasis patients were slightly older than the PsA (46.3 vs. 42.4 years, p<0.001).

Unexpectedly, the PsA patients had more severe psoriasis as measured by maximal PASI

score in the first 3 years of follow-up (8.7 vs. 6.1, p<0.001). This finding may be partially

explained by the fact that psoriasis patients who were recruited from phototherapy centers

were evaluated subsequent to their phototherapy treatments that most likely improved their

psoriasis. There was no difference in the frequency of family history of psoriasis, however,

more patients in the PsA group reported a family history of PsA, AS or Inflammatory Bowel

Disease.

Table 5.2 - Demographic and clinical characteristics of the study population

PsA

(N=712)

Psoriasis

(N=335)

Controls

(N=713)

P value

Age Sex: Male (%)

42.4±12.7 402 (58.2%)

46.3±13.3 188 (55.9%)

43.6±17.2 283 (50.7%)

<0.001 0.49

Age at diagnosis of psoriasis 27.9±14.4 29.8±16.7 0.06 Age at diagnosis of PsA 36.3±13

Duration of Psoriasis 14.4±12.5 16.4±14.5 0.02 Duration of PsA Early onset Psoriasis (age at onset<40 years) Psoriasis type -Chronic Plaque psoriasis (%)

PASI (Max. in 3 years) Severe Psoriasis (PASI>10) Psoriatic nail lesions (%) Family history of: - psoriasis (%) - PsA (%)

- Ankylosing Spondylitis (%)

- IBD (%)

6.1±8.0 547 (79.6%)

604 (90.3%) 8.7±9.9

192 (26.9%) 533 (82.9%)

329 (44.8%) 73 (10.6%) 12 (1.7%) 28 (6%)

--- 250 (74.8%)

288 (86.6%) 6.1±5.1

51 (15.2%) 169 (51.6%)

144 (43.4%)

11 (3.3%) 1 (0.2%)

24 (3.5%)

0.09

0.09 <0.001 <0.001 <0.001

0.18

<0.001 0.02 0.04

Page 77: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

65

5.4.1. The association between HLA alleles and Psoriatic Disease

Allele frequencies in the Psoriatic disease (PsD) group were compared to those in the healthy

control group to identify susceptibility alleles that confer a risk for PsD. The results are

presented in Tables 5.3-5.7. FDR adjusted P values were not calculated for rare alleles.

Significant associations are bolded (FDR – p <0.01).

The following HLA alleles were associated with PsD compared to healthy controls: HLA-

A*03 (OR 0.66, p=0.0003, FDR-p=0.004), HLA-B*07 (OR 0.57, p=3x10-6, FDR-p<0.0001),

HLA-B*13 (OR 2.16, p=0.0001, FDR-p=0.0004), HLA-B*27 (OR 2.15, p=3x10-6, FDR-

p<0.0001), HLA-B*38 (OR 5.30, p=9x10-13, FDR-p<0.0001), HLA-B*51 (OR 0.50,

p=0.0001, FDR-p=0.0004), HLA-B*57 (OR 2.19, p=2x10-7, FDR-p<0.0001), HLA-B*60

(OR 0.54, p=0.0006, FDR-p=0.002), HLA-C*03 (OR 0.67, p=0.001, FDR-p=0.003), HLA-

C*06 (OR 2.18, p=7x10-12, FDR-p<0.0001), HLA-C*07 (OR 0.61, p=4x10-7, FDR-

p<0.0001), HLA-C*12 (OR 2.1, p=2x10-7, FDR-p<0.0001), HLA-C*15 (OR 0.48, p=0.002,

FDR-p=0.005), HLA-DRB1*15 (OR 0.66, p=0.0003, FDR-p=0.004), HLA-DQB1*0303

(OR 1.61, p=0.001, FDR-p=0.007) and HLA-DQB1*0602 (OR 0.64, p=8x10-5, FDR-

p=0.001).

The frequencies of HLA-B*27 and HLA-C*06 were compared between the group of

psoriasis alone and healthy controls. There was no difference in the frequency of HLA-B*27

between psoriasis patients and healthy controls (4.5% vs. 7.2%, p=0.09, respectively), while

the association between HLA-C*06 and psoriasis was stronger for the psoriasis alone groups

vs. controls (44.2% vs. 18.1%, p=3x10-19, respectively).

5.4.2. The association between HLA alleles and PsD compared to Healthy controls – Multivariate analysis

Due to the strong LD between the alleles at the different HLA loci, multivariate logistic

regression analysis was employed to identify the key HLA associations. The model included

all HLA alleles that were found to be significantly associated with PsD in the univariate

analysis. The results are presented in Table 5.8. The following alleles were more frequent in

Page 78: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

66

the PsD group compared to healthy controls: HLA-B*27 (OR 2.36, p<0.0001), HLA-B*38

(OR 6.01, p<0.0001) and HLA-C*06 (OR 2.23, p<0.0001). While the following alleles were

less frequent in the PsD group compared to healthy controls: HLA-A*03 (OR 0.72, p=0.005),

HLA-B*51 (OR 0.55, p=0.001), HLA-B*60 (OR 0.59, p=0.006) and HLA-DQ*0602 (OR

0.71, p=0.007).

Page 79: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

67

^HLA-A*28 includes HLA-A*68 and A*69, PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Table 5.3 - HLA-A allele distribution – Psoriatic disease vs. Controls

Allele PsD (N=1047) Control (N=712) OR 95% CI P value FDR-p

A*01 357

(34.1%) 226 (31.7%) 1.11 0.9-1.4 0.3 0.46 A*02 589 (56.3%) 355 (49.8%) 1.29 1.1-1.6 0.008 0.03

A*03

210 (20.1%) 195 (27.4%) 0.66 0.5-0.8 0.0003 0.004

A*11 116 (11.1%) 78 (10.9%) 1.01 0.7-1.4 0.93 0.93

A*23 23 (2.2%) 30 (4.2%) 0.51 0.3-0.9 0.01 0.04

A*24 158 (15.1%) 90 (12.6%) 1.22 0.9-1.6 0.14 0.28

A*25 33 (3.2%) 28 (3.9%) 0.79 0.5-1.3 0.37 0.51

A*26 86 (8.2%) 36 (5.0%) 1.68 1.1-2.5 0.01 0.04

A*28 59 (5.6%) 61 (8.6%) 0.64 0.3-4.1 0.02 0.05

A*29 70 (6.7%) 69 (9.6%) 0.67 0.5-0.9 0.02 0.05

A*31 49 (4.7%) 34 (4.7%) 0.98 0.6-1.5 0.92 0.93

A*32 62 (5.9%) 44 (6.1%) 0.95 0.6-1.4 0.82 0.93

A*33 36 (3.4%) 18 (2.5%) 1.37 0.8-2.4 0.27 0.46

A*66 A*68

9 (0.9%) 8 (1.1%) 0.76

0.3-2.0 0.58 0.73

Page 80: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

68

PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Table 5.4 - HLA-B allele distribution – Psoriatic disease vs. Controls

Allele PsD (N=1047) Control (N=713) OR 95% CI P value FDR-p

B*07

176 (16.8%) 185 (25.9%) 0.57 0.4-0.7 3x10-6 <0.0001 B*08 212 (20.2%) 173 (24.3%) 0.79 0.6-1.0 0.05 0.1

B*13 102 (9.7%) 34 (4.8%) 2.16 1.4-3.2 0.0001 0.0004

B*14 89 (8.5%) 61 (8.6%) 0.99 0.7-1.4 0.96 0.96 B*18 B*

76 (7.3%) 57 (8.0%) 0.90 0.6-1.3 0.56 0.64

B*27 152 (14.5%) 52 (7.3%) 2.15 1.6-3.0 3x10-6 <0.0001

B*35 148 (14.1%) 122 (17.1%) 0.79 0.6-1.0 0.09 0.16

B*37 33 (3.2%) 23 (3.2%) 0.98 0.6-1.7 0.93 0.96

B*38 125 (11.9%) 18 (2.5%) 5.30 3.2-8.7 9x10-13 <0.0001

B*39 60 (5.7%) 22 (3.1%) 1.90 1.2-3.1 0.009 0.02

B*41 23 (2.1%) 10 (1.4%) 1.58 0.7-3.3 0.23 0.36

B*42 0 (0%) 2 (0.3%) 0.08

B*44 233 (22.3%) 185 (25.9%) 0.82 0.7-1.0 0.07 0.14

B*45 8 (0.8%) 7 (0.9%) 0.78 0.3-2.2 0.63

B*47 7 (0.7%) 4 (0.6%) 1.20 0.3-4.1 0.76

B*48 1 (0.1%) 1 (0.1%) 0.68 0.04-10.9 0.78

B*49 15 (1.4%) 23 (3.2%) 0.44 0.2-0.8 0.01 0.03

B*50 30 (2.9%) 9 (1.3%) 2.31 1.1-4.8 0.02 0.05

B*51 60 (5.7%) 77 (10.8%) 0.50 0.4-0.7 0.0001 0.0004

B*52 19 (1.8%) 17 (2.4%) 0.76 0.4-1.5 0.41 0.54

B*53 9 (0.9%) 2 (0.3%) 3.10 0.7-14.3 0.13

B*55 30 (2.9%) 24 (3.3%) 0.85 0.5-1.5 0.54 0.64

B*56 12 (1.2%) 5 (0.7%) 1.65 0.6-4.7 0.34

B*57 176 (16.8%) 60 (8.4%) 2.19 1.6-3.0 2x10-7 <0.0001

B*58 12 (1.2%) 11 (1.5%) 0.73 0.3-1.7 0.47 0.59

B*60 60 (5.7%) 72 (10.1%) 0.54 0.4-0.8 0.0006 0.002

B*61 20 (1.9%) 20 (2.8%) 0.67 0.4-1.3 0.22 0.32

B*62 76 (7.3%) 67 (9.4%) 0.75 0.5-1.1 0.11 0.18

B*63 19 (1.8%) 7 (1.0%) 1.86 0.8-4.5 0.15 0.34

B*70 16 (1.5%) 10 (1.4%) 1.10 0.5-2.4 0.83 0.91 B*71 B*73

1 0

(0.1%) (0%)

0 1

(0 %) (0.1%)

0.41 0.22

Page 81: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

69

PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Table 5.5 - HLA-C allele distribution – Psoriatic disease vs. Controls

Allele PsD (N=1044) Control (N=708) OR 95% CI P value FDR-p

C*01 91 (8.7%) 42 (5.9%) 1.51 1.0-2.2 0.03 0.05 C*02 109 (10.4%) 49 (6.9%) 1.56 1.1-2.2 0.01 0.02 C*03

166 (15.9%) 156 (22.0%) 0.67 0.5-0.9 0.001 0.003

C*04 176 (16.9%) 145 (20.5%) 0.78 0.6-1.0 0.05 0.08

C*05 158 (15.1%) 101 (14.3%) 1.07 0.8-1.4 0.61 0.66

C*06 342 (32.8%) 129 (18.2%) 2.18 1.7-2.7 7x10-12 <0.0001

C*07 461 (44.2%) 402 (56.8%) 0.61 0.5-0.7 4x10-7 <0.0001

C*08 89 (8.5%) 61 (8.6%) 0.99 0.7-1.4 0.94 0.94

C*12 203 (19.4%) 73 (10.3%) 2.10 1.6-2.8 2x10-7 <0.0001

C*15 32 (3.1%) 43 (6.1%) 0.48 0.3-0.8 0.002 0.005

C*16 67 (6.4%) 61 (8.6%) 0.73 0.5-1.0 0.08 0.11

C*17 23 (2.2%) 10 (1.4%) 1.57 0.7-3.3 0.23 0.27

C*18 4 (0.4%) 1 (0.1%) 2.72 0.3-24.3 0.35

Table 5.6 - HLA-DRB1 allele distribution – Psoriatic Disease vs. Controls

Allele PsD (N=1047) Control (N=711) OR 95% CI P value FDR-p

DRB1*01

250 (23.9%) 132 (18.6%) 1.36 1.1-1.7 0.008 0.03 DRB1*02

0 (0%) 2 (0.3%) 0.09 DRB1*03

222 (21.2%) 184 (25.8%) 0.77 0.6-1.0 0.02 0.05

DRB1*04

307 (29.3%) 226 (31.8%) 0.89 0.7-1.1 0.27 0.43

DRB1*07

354 (33.8%) 199 (28.0%) 1.31 1.1-1.6 0.01 0.03

DRB1*08 B*

56 (5.3%) 36 (5.1%) 1.06 0.7-1.6 0.8 0.8

DRB1*09

15 (1.4%) 14 (2.0%) 0.72 0.3-1.5 0.38 0.54

DRB1*10

15 (1.4%) 13 (1.8%) 0.78 0.4-1.7 0.51 0.62

DRB1*11

135 (12.9%) 126 (17.7%) 0.69 0.5-0.9 0.005 0.03

DRB1*12

50 (4.8%) 21 (2.9%) 1.65 1.0-2.9 0.06 0.13

DRB1*13

207 (19.8%) 149 (20.9%) 0.92 0.7-1.8 0.54 0.62

DRB1*14

67 (6.4%) 41 (5.8%) 1.13 0.7-1.7 0.58 0.62

DRB1*15

201 (19.2%) 189 (26.6%) 0.66 0.5-0.8 0.0003 0.004

DRB1*16

50 (4.8%) 24 (3.3%) 1.44 0.9-2.4 0.15 0.27

Page 82: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

70

PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Table 5.7 - HLA-DQ allele distribution – Psoriatic Disease vs. Controls

Allele PsD (N=1045) Control (N=711) OR 95% CI P value FDR - p

DQB1*02

434 (41.5%) 303 (42.3%) 0.96 0.8-1.2 0.69 0.79 DQB1*05

0 (0%) 1 (0.1%) 0.22 DQB1*0301

307 (29.5%) 237 (33.2%) 0.83 0.7-1.0 0.08 0.26

DQB1*0302

205 (19.6%) 146 (20.5%) 0.94 0.7-1.2 0.63 0.79

DQB1*0303 B*

161 (15.5%) 72 (10.3%) 1.61 1.2-2.2 0.001 0.007

DQB1*0304

4 (0.4%) 1 (0.1%) 2.73 0.3-24.5 0.35 DQB1*0305

6 (0.6%) 1 (0.1%) 4.10 0.5-34.1 0.16 DQB1*0401

2 (0.2%) 1 (0.1%) 1.36 0.1-15.0 0.08 DQB1*0402

49 (4.7%) 33 (4.6%) 1.01 0.6-1.6 0.96 0.96

DQB1*0502

53 (5.1%) 25 (3.5%) 1.47 0.9-2.4 0.12 0.31

DQB1*0503

64 (6.1%) 39 (5.4%) 1.12 0.7-1.7 0.57 0.79

DQB1*0504

0 (0%) 3 (0.4%) 0.04 DQB1*0601

22 (2.1%) 18 (2.5%) 0.83 0.4-1.6 0.57 0.79

DQB1*0602

178 (17%) 176 (24.7%) 0.64 0.5-0.8 8x10-5 0.001

DQB1*0603

123 (11.8%) 72 (10.1%) 1.18 0.9-1.6 0.28 0.52

DQB1*0604

55 (5.3%) 49 (6.9%) 0.75 0.5-1.1 0.16 0.34

DQB1*0605

2 (0.2%) 6 (0.8%) 0.23 0.04-1.1 0.05 DQB1*0609 DQB1*0501

12 253

(1.2%) (24.2%)

7 143

(1.0%) (20.1%)

1.17 1.26

0.5-3.0 1.0-1.6

0.73 0.04

0.79 0.17

Page 83: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

71

Table 5.8 - The association of HLA alleles and PsD (N=1042) compared to healthy

controls (N=704) using multivariate logistic regression analysis

Univariate analysis Multivariate analysis (reduced model)

Allele OR 95% CI P value OR 95% CI P Value

HLA-A*03

HLA-B*07

HLA-B*13

0.66

0.57

2.14

0.5-0.83

0.45-0.73

1.43-3.19

0.0003

<0.0001

0.0002

0.72 0.57-0.91 0.005

HLA-B*27 2.12 1.52-2.96 <0.0001 2.36 1.67-3.33 <0.0001

HLA-B*38 5.51 3.28-9.23 <0.0001 6.01 3.56-10.15 <0.0001

HLA-B*51

HLA-B*57

0.49

2.17

0.35-0.71

1.59-2.95

0.0001

<0.0001

0.55 0.39-0.81 0.001

HLA-B*60 0.53 0.37-0.77 0.0007 0.59 0.41-0.86 0.006

HLA-C*06

HLA-C*07

HLA_C*12

HLA-C*15

2.18

0.59

2.12

0.49

1.73-2.74

0.49-0.72

1.59-2.83

0.31-0.78

<0.0001

<0.0001

<0.0001

0.003

2.23 1.76-2.84 <0.0001

HLA_DR*15

HLA_DQ*0303

0.65

1.62

0.52-0.82

1.20-2.18

0.0003

0.002

HLA-DQ*0602 0.62 0.49-0.78 <0.0001 0.71 0.55-0.91 0.007

PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Page 84: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

72

5.4.3. The association between HLA alleles and PsA compared to psoriasis

Allele frequencies in the PsA group were compared to those in the psoriasis group to identify

susceptibility alleles that confer a risk for PsA. The results are presented in Tables 5.9-5.13.

The following HLA alleles were more frequent in PsA compared to psoriasis: HLA-B*27

(OR 5.07, p=2x10-10, FDR-p<0.0001), HLA-C*01 (OR 2.55, p=0.0009, FDR-p=0.0036) and

HLA-C*02 (OR 2.41, p=0.0005, FDR-p=0.003), while HLA-C*06 (OR 0.48, p=8x10-8,

FDR-p<0.0001), HLA-B*57 (OR 0.58, p=0.001, FDR-p=0.01) and HLA-DRB1*07 (OR

0.62, p=0.0004, FDR-p=0.005) were less frequent in PsA compared to psoriasis.

5.4.4. Association between HLA alleles and PsA – Multivariate analysis

In a multivariate regression analysis the following alleles were more frequent in PsA

compared to psoriasis: HLA-B*27 (OR 5.17, p<0.0001), HLA-B*08 (OR 1.61, p=0.009) and

HLA-B*38 (OR 1.65, p=0.026) (Table 5.14). Two of these alleles were also more frequent in

PsA compared to healthy controls (single locus analysis): HLA-B*27 (OR 3.05, p=2x10-11,

FDR-p<0.0001) and HLA-B*38 (OR 5.9, p=5x10-14, FDR-p<0.0001). In contrast, HLA-

C*06 was less frequent in the PsA group compared to psoriasis (OR 0.58, p=0.0002), but

more frequent in the PsA group compared to the healthy controls (single locus analysis) (OR

1.71, p=2x10-5, FDR-p<0.0001). The remaining HLA alleles were not found to be associated

with PsA in the multivariate regression, suggesting that they were part of extended

haplotypes with the more strongly associated alleles.

Page 85: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

73

^ HLA-A*28 includes also HLA-A*68, *A69. PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

Table 5.9 - HLA-A allele distribution – PsA vs. Psoriasis

Allele PsA (N=712) Psoriasis (N=334) OR 95% CI P value FDR- p

A*01 233 (32.7%) 124 (37.1%) 0.82 0.6-1.1 0.16 0.65 A*02 404 (56.7%) 185 (56.5%) 1.05 0.8-1.4 0.69 0.88

A*03

141 (19.8%) 69 (20.6%) 0.95 0.7-1.3 0.74 0.88

A*11 84 (11.8%) 32 (9.6%) 1.26 0.8-1.9 0.28 0.65

A*23 15 (2.1%) 8 (2.4%) 0.87 0.4-2.1 0.76 0.89

A*24 117 (16.4%) 41 (12.2%) 1.41 1.0-2.1 0.08 0.65

A*25 19 (2.6%) 14 (4.2%) 0.63 0.3-1.3 0.19 0.65 0.65 A*26 63 (8.8%) 23 (6.9%) 1.31 0.8-2.2 0.28 0.65

A*28^ 39 (5.5%) 21 (6.3%) 0.97 0.6-1.7 0.53 0.88

A*29 49 (6.9%) 21 (6.3%) 1.10 0.6-1.9 0.72 0.88

A*31 33 (4.6%) 16 (4.6%) 0.98 0.5-1.8 0.92 0.92

A*32 41 (5.8%) 21 (6.3%) 0.91 0.5-1.6 0.73 0.88

A*33 20 (2.8%) 16 (4.8%) 0.57 0.3-1.1 0.10 0.65

A*34

1 (0.1%) 0 (0%) 0.49 A*36 1 (0.1%) 0 (0%) 0.49 A*66 A*68

6 (0.8%) 3 (1.1%) 0.93 0.2-3.7 0.92 0.92

Page 86: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

74

Table 5.10 - HLA-B allele distribution – PsA vs. Psoriasis

Allele PsA (N=712) Psoriasis (N=335) OR 95% CI P value FDR – p

B*07

119 (16.7%) 57 (17.1%) 0.97 0.7-1.4 0.89 0.95 B*08 159 (22.2%) 54 (16.1%) 1.48 1.1-2.1 0.02 0.12

B*13 58 (8.1%) 44 (13.1%) 0.58 0.4-0.9 0.01 0.08

B*14 53 (7.4%) 36 (10.8%) 0.66 0.4-1.0 0.07 0.24

B*18 B*

58 (8.1%) 18 (5.4%) 1.55 0.9-2.7 0.1 0.27

B*27 137 (19.2%) 15 (4.5%) 5.07 2.9-8.8 2*10-10 <0.0001

B*35 95 (13.3%) 31 (9.3%) 0.82 0.6-1.2 0.28 0.51

B*37 18 (2.5%) 15 (4.5%) 0.55 0.3-1.1 0.09 0.27

B*38 94 (13.2%) 31 (9.3%) 1.48 1.0-2.3 0.07 0.24

B*39 44 (6.2%) 16 (4.8%) 1.31 0.7-2.4 0.36 0.51

B*41 14 (1.9%) 9 (2.6%) 0.72 0.3-1.7 0.45 0.57

B*42 0 (0%) 2 (0.3%) 0.16 B*44 152 (21.3%) 81 (24.2%) 0.85 0.6-1.2 0.29 0.51

B*45 4 (0.6%) 4 (1.2%) 0.46 0.1-1.9 0.29 0.51

B*47 4 (0.6%) 3 (0.9%) 0.62 0.1-2.8 0.53 B*48 1 (0.1%) 0 (0%) 0.49 B*49 8 (1.1%) 7 (2.1%) 0.53 0.2-1.5 0.22 0.51

B*50 20 (2.8%) 10 (3%) 0.94 0.4-2.0 0.87 0.94

B*51 38 (5.3%) 22 (6.6%) 0.80 0.5-1.4 0.42 0.56

B*52 15 (2.1%) 4 (1.2%) 1.77 0.6-5.4 0.30 0.51

B*53 8 (1.1%) 1 (0.3%) 3.79 0.5-30.3 0.17 B*55 20 (2.8%) 10 (3%) 0.93 0.4-2.0 0.87 0.94

B*56 7 (1.0%) 5 (1.5%) 0.65 0.2-2.1 0.46 B*57 102 (14.3%) 75 (22.4%) 0.58 0.4-0.8 0.001 0.01

B*58 8 (1.1%) 4 (1.2%) 0.93 0.3-3.1 0.91 0.94

B*60 44 (6.2%) 16 (4.8%) 1.31 0.7-2.4 0.36 0.51

B*61 16 (2.2%) 4 (1.2%) 1.89 0.6-5.7 0.24 0.51

B*62 48 (6.4%) 28 (8.4%) 0.79 0.5-1.3 0.34 0.51

B*63 13 (1.8%) 6 (1.7%) 1.01 0.4-2.7 0.97 0.97

B*70

8 (1.1%) 9 (2.7%) 0.41 0.1-1.0 0.06 0.24

PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

Page 87: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

75

Table 5.11 - HLA-C allele distribution – PsA vs. Psoriasis

Allele PsA (N=710) Psoriasis (N=334) OR 95% CI P value FDR-p

C*01 76 (10.7%) 15 (4.5%) 2.55 1.4-4.5 0.0009 0.0036 C*02 90 (12.7%) 19 (5.7%) 2.41 1.4-4.0 0.0006 0.003

C*03

112 (15.8%) 54 (16.2%) 0.97 0.7-1.7 0.87 0.88

C*04 115 (16.2%) 61 (18.3%) 0.86 0.6-1.2 0.41 0.63

C*05 111 (15.6%) 47 (14.1%) 1.13 0.8-1.7 0.51 0.65

C*06 195 (27.5%) 147 (44%) 0.48 0.4-0.6 8*10-8 <0.0001

C*07 330 (46.5%) 131 (39.2%) 1.34 1.0-1.8 0.03 0.09

C*08 53 (7.6%) 36 (10.8%) 0.66 0.4-1.0 0.07 0.16

C*12 147 (20.7%) 56 (16.8%) 1.29 0.9-1.8 0.13 0.28

C*15 21 (2.9%) 11 (3.3%) 0.89 0.4-1.9 0.76 0.84

C*16 41 (5.8%) 26 (7.8%) 0.72 0.4-1.2 0.21 0.37

C*17 15 (2.1%) 8 (2.4%) 0.87 0.4-2.1 0.77 0.84

C*18 3 (0.4%) 1 (0.3%) 1.41 0.1-13.6 0.76

Table 5.12 - HLA-DRB1 allele distribution – PsA vs. Psoriasis

Allele PsA (N=712) Psoriasis (N=335) OR 95% CI P value FDR -p

DRB1*01

182 (25.5%) 68 (20.3%) 1.34 1.0-1.9 0.06 0.19 DRB1*03

167 (23.4%) 55 (16.4%) 1.56 1.1-2.2 0.009 0.06 DRB1*04

211 (29.6%) 96 (28.6%) 1.04 0.8-1.4 0.75 0.82 DRB1*07

216 (30.3%) 139 (41.5%) 0.62 0.5-0.8 0.0004 0.005 DRB1*08 B*

35 (4.9%) 21 (6.3%) 0.77 0.4-1.3 0.36 0.78 DRB1*09

11 (1.5%) 4 (1.2%) 1.29 0.4-4.1 0.65 0.82 DRB1*10

11 (1.5%) 4 (1.2%) 1.29 0.4-4.1 0.65 0.82 DRB1*11

89 (12.5%) 47 (14%) 0.87 0.6-1.3 0.48 0.82 DRB1*12

35 (4.9%) 15 (4.5%) 1.10 0.6-2.0 0.76 0.82 DRB1*13

142 (19.9%) 65 (19.4%) 1.02 0.7-1.4 0.84 0.84 DRB1*14

60 (7%) 17 (5.0%) 1.41 0.8-2.5 0.23 0.59 DRB1*15

134 (18.8%) 67 (20%) 0.92 0.7-2.3 0.64 0.82 DRB1*16

40 (5.6%) 10 (3.0%) 1.93 1.0-3.9 0.06 0.19

PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

Page 88: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

76

Table 5.13 - HLA-DQ allele distribution – PsA vs. Psoriasis

Allele PsA (N=712) Psoriasis (N=335) OR 95% CI P value FDR - p

DQB1*02

292 (41.1%) 142 (42.4%) 0.94 0.7-1.2 0.68 0.91 DQB1*0301

201 (28.3%) 107 (31.9%) 0.84 0.6-1.1 0.22 0.61

DQB1*0302

141 (19.8%) 64 (19.1%) 1.04 0.8-1.5 0.78 0.91

DQB1*0303 B*

100 (14.1%) 62 (18.5%) 0.72 0.5-1.0 0.06 0.42

DQB1*0304

3 (0.4%) 1 (0.3%) 1.41 0.1-13.6 0.76 DQB1*0305

6 (0.8%) 0 (0%) 0.09 DQB1*0401

2 (0.3%) 0 (0%) 0.33 DQB1*0402

29 (4.1%) 20 (6.0%) 0.67 0.4-1.2 0.17 0.59

DQB1*0501

183 (25.7%) 70 (20.9%) 1.31 0.9-1.8 0.09 0.42

DQB1*0502

42 (5.9%) 11 (3.3%) 1.85 0.9-3.6 0.07 0.42

DQB1*0503

47 (6.6%) 17 (5.1%) 1.32 0.7-2.3 0.33 0.66

DQB1*0504

0 (0%) 3 (0.4%) 0.08 DQB1*0601

15 (2.1%) 7 (2.1%) 1.0 0.4-2.5 0.98 0.99

DQB1*0602

121 (17%) 57 (17%) 1.0 0.7-1.4 0.99 0.99

DQB1*0603

85 (11.9%) 38 (11.3%) 1.06 0.7-1.6 0.77 0.91

DQB1*0604

40 (5.6%) 15 (4.5%) 1.27 0.7-2.3 0.44 0.77

DQB1*0605

2 (0.3%) 0 (0%) 0.33 DQB1*0609 DQB1*0613

9 1

(1.3%) (0.1%)

3 0

(0.9%) (0%)

1.42 0.4-5.2 0.59 0.32

0.91

PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

Page 89: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

77

PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

Table 5.14 - Odds ratios comparing HLA allele frequencies in PsA (N=710) to psoriasis

(N=334) using logistic regression analysis

Univariate analysis Multivariate analysis (reduced model)

Allele OR 95% CI P value OR 95% CI P Value

HLA-B*08

HLA-B*27

HLA-B*38

HLA-B*57

1.53

4.99

1.49

0.58

1.08-2.15

2.89-8.66

0.97-2.29

0.42-0.82

0.015

<0.0001

0.067

0.001

1.61

5.17

1.65

1.12-2.30

2.95-9.04

1.06-2.57

0.009

<0.0001

0.026

HLA-C*01 2.54 1.44-1.51 0.001

HLA-C*02 2.41 1.44-2.02 0.0008

HLA-C*06

HLA-C*07

0.48

1.35

0.37-0.63

1.03-1.75

<0.0001

0.027

0.58 0.44-0.77 0.0002

HLA-C*12

HLA-DRB1*07

1.29

0.62

0.92-1.82

0.50-0.80

0.13

0.0004

Page 90: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

78

5.4.5. Subgroup analysis based on age at onset of psoriasis

HLA-C*06 is associated with early onset psoriasis, family history of the disease and with

more severe psoriasis [178, 179]. A subgroup analysis was performed to determine whether

the association between HLA-C*06 and PsA is related to differences in the age at onset of

psoriasis between the two groups. The results are presented in Table 5.15.

The association between PsA and HLA-C*06 remained significant in the early onset

psoriasis group (OR 0.41, p<0.0001). These findings suggest that the lower frequency of

HLA-C*06 in the PsA group compared to psoriasis is not related to differences between the

two groups in the age at onset of psoriasis. In the late onset psoriasis group, the frequency of

HLA-C*06 was slightly lower in the PsA compared to the psoriasis group, however not

significantly, possibly due to the small number of individuals in each group.

Table 5.15 - Odds ratios comparing HLA-C*06 allele frequencies in PsA (N=687)* to

psoriasis (N=334) by age at onset of psoriasis

Age at onset of

Psoriasis PsA Psoriasis OR 95% CI P value

Early onset (<40)

162 (29.6%)

127 (50.8%)

0.41

0.30-0.55

<0.0001

Late onset (≥40)

26 (18.7%)

20 (23.8%)

0.74

0.38-1.42

0.36

All

188 (27.4%)

147 (47.1%)

0.47

0.36-0.63

<0.0001

PsA - Psoriatic Arthritis, CI - Confidence Interval, OR - Odds Ratio

*The reduced number of PsA patients is due to lack of information about age at onset of psoriasis for some of the patients.

Page 91: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

79

5.4.6. The association of HLA alleles with Axial and Peripheral PsA

A subgroup analysis was employed to identify new HLA alleles associated with subtypes of

PsA (Tables 5.16-5.17). One additional HLA allele was identified as being associated with a

PsA subtype but not with the whole group of PsA patients. After multivariate analysis, the

frequency of HLA-B*39 was found to be increased in axial PsA compared to psoriasis (OR

2.51, 95% CI 1.25-5.01, p=0.009). This allele was not associated with peripheral PsA (OR

0.9, 95% CI 0.43-1.87, p=0.77). The remaining alleles associated with axial PsA were: HLA-

B*27, HLA-B*08 and HLA-B*38. Analysis of patients with peripheral PsA did not reveal

new associations as only HLA-C*06 and HLA-B*27 met significance criteria for association

with this subgroup compared to psoriasis.

Page 92: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

80

Table 5.16 - Odds ratios comparing HLA allele frequencies in Axial PsA (N=255) vs.

psoriasis (N=334) using logistic regression analysis

Univariate analysis Multivariate analysis (reduced model)

Allele OR 95% CI P value OR 95% CI P Value

HLA-B*08 HLA-B*27

HLA-B*38

HLA-B*39 HLA-B*07 HLA-B*13 HLA-B*14 HLA-B*18 HLA-B*35 HLA-B*37 HLA-B*41 HLA-B*44 HLA-B*50 HLA-B*51 HLA-B*52 HLA-B*55 HLA-B*57 HLA-B*58 HLA-B*4001 HLA-B*4002 HLA-B*1501 HLA-B*1517 HLA-B*1509/10 HLA-C*01 HLA-C*02 HLA-C*03 HLA-C*04 HLA-C*05 HLA-C*06 HLA-C*07 HLA-C*08 HLA-C*12 HLA-C*15 HLA-C*17

1.75 7.34

1.80

1.86 0.86 0.57 0.59 0.94 0.75 0.51 0.43 0.72 0.91 0.82 1.97 0.64 0.71 1.30 0.89 1.97 0.58 1.08 0.14 2.94 3.26 0.69 0.83 0.85 0.54 1.25 0.63 1.56 0.83 0.48

1.17-2.64 4.09-13.23

1.09-2.97

0.96-3.62 0.56-1.35 0.32-0.97 0.32-1.07 0.45-1.94 0.47-1.21 0.19-1.33 0.11-1.59 0.48-1.07 0.34-2.42 0.41-1.63 0.55-7.06 0.22-1.91 0.46-1.07 0.32-5.27 0.41-1.49 0.55-7.06 0.29-1.14 0.33-3.59 0.02-1.12 1.55-5.58 1.85-5.77 0.43-1.11 0.54-1.28 0.52-1.37 0.38-0.76 0.90-1.74 0.35-1.13 1.04-2.34 0.32-2.17 0.13-1.84

0.007 <0.0001

0.02

0.07 0.54 0.04 0.08 0.85 0.24 0.17 0.20 0.10 0.84 0.56 0.29 0.42 0.10 0.71 0.76 0.29 0.11 0.89 0.06

0.0009 <0.0001

0.13 0.41 0.49

0.0005 0.18 0.12 0.03 0.70 0.28

2.46 9.08

2.55

2.51

1.59-3.79 4.95-16.65

1.50-4.34

1.25-5.01

<0.0001 <0.0001

0.0006

0.009

Page 93: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

81

Table 5.17 - Odds ratios comparing HLA allele frequencies in peripheral PsA (N=323) vs.

psoriasis (N=334) using logistic regression analysis

Univariate analysis Multivariate analysis (reduced model)

Allele OR 95% CI P value OR 95% CI P Value

HLA-B*08 HLA-B*27 HLA-B*38 HLA-B*39 HLA-B*07 HLA-B*13 HLA-B*14 HLA-B*18 HLA-B*35 HLA-B*37 HLA-B*41 HLA-B*44 HLA-B*50 HLA-B*51 HLA-B*52 HLA-B*55 HLA-B*57 HLA-B*58 HLA-B*4001 HLA-B*4002 HLA-B*1501 HLA-B*1517 HLA-B*1509/10 HLA-C*01 HLA-C*02 HLA-C*03 HLA-C*04 HLA-C*05 HLA-C*06 HLA-C*07 HLA-C*08 HLA-C*12 HLA-C*15 HLA-C*17

1.51 3.80 1.07 0.90 1.11 0.60 0.69 1.47 0.92 0.40 0.57 0.97 0.93 0.79 1.82 1.03 0.55 1.03 1.66 1.83 0.92 1.03 0.68 2.25 1.64 1.18 0.94 1.22 0.46 1.47 0.66 1.04 1.13 0.64

1.04-2.29 2.09-6.93 0.64-1.81 0.43-1.87 0.74-1.65 0.36-0.99 0.41-1.18 0.78-2.75 0.61-1.41 0.15-1.05 0.19-1.71 0.68-1.39 0.37-2.31 0.41-1.51 0.53-6.30 0.42-2.52 0.36-0.83 0.25-4.17 0.87-3.18 0.53-6.30 0.52-1.61 0.33-3.24 0.24-1.94 1.19-4.27 0.89-2.98 0.79-1.77 0.63-1.40 0.80-1.87 0.33-0.64 1.07-1.99 0.38-1.14 0.69-1.56 0.49-2.61 0.21-1.97

0.03 <0.0001

0.78 0.77 0.61

0.048 0.18 0.22 0.72 0.06 0.31 0.90 0.87 0.47 0.33 0.93

0.005 0.96 0.12 0.33 0.76 0.95 0.47 0.01 0.11 0.41 0.76 0.34

<0.0001 0.015 0.13 0.84 0.76 0.43

3.66

0.47

1.99-6.72

0.34-0.66

<0.0001

<0.0001

Page 94: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

82

5.4.7. Positive Predictive Value of the identified HLA risk alleles

To assess the utility of HLA risk alleles in identifying PsA patients among patients with

psoriasis, the PPV of each of the identified HLA risk allele was calculated as follows: HLA-

B*08 PPV=0.42, HLA-B*27 PPV=0.64, HLA-B*38 PPV=0.43, HLA-C*06 PPV=0.35.

5.4.8. Haplotype analysis - Psoriatic disease vs. Healthy controls The association between the extended haplotype of HLA-A-B-C-DR-DQ and PsD was

investigated. A total of 1560 possible extended haplotypes were imputed, however, only 9 of

them with a prevalence of >1% were included in the analysis. The results are presented in

Table 5.18. The following haplotypes were significantly increased in the PsD group

compared to healthy controls: HLA-A*01-B*57-C*06-DR*07-DQ*0303, HLA-A*02-B*13-

C*06-DR*07-DQ*02 and HLA-A*02-B*57-C*06-DR*07-DQ*0303. As expected, all of

these haplotypes included HLA-C*06, the strongest susceptibility allele for psoriasis. HLA-

DR*07 was also present in each of the risk haplotype given its LD with HLA-C*06 (D’=0.5).

Page 95: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

83

PsD - Psoriatic Disease, CI - Confidence Interval, OR - Odds Ratio

Table 5.18 - Odds ratios comparing extended HLA haplotype frequencies in PsD to healthy

controls using logistic regression analysis

Haplotype Univariate analysis Multivariate analysis

(reduced model)

HLA-A-B-C-DR-DQ OR 95% CI P value OR 95% CI P value

*01-*08-*07-*03-*02 0.53 0.31-0.92 0.02

*01-*57-*06-*07-*0303 3.97 1.48-10.66 0.006 4.08 1.52-10.94 0.005

*02-*07-*07-*15-*0602 0.40 0.15-1.06 0.07

*02-*08-*07-*03-*02 2.54 0.69-9.27 0.15

*02-*13-*06-*07-*02 8.11 1.96-33.52 0.0039 8.82 2.12-36.61 0.003

*02-*44-*05-*04-*0301 0.66 0.25-1.74 0.39

*02-*57-*06-*07-*0303 10.71 2.11-54.29 0.004 10.44 2.08-52.47 0.004

*03-*35-*04-*01-*0501 0.44 0.12-1.67 0.22

*29-*44-*16-*07-*02 0.48 0.17-1.32 0.15

Page 96: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

84

5.4.9. Linkage Disequilibrium of HLA-B and –C alleles

The MHC region contains many genes in strong LD. The magnitude of LD between HLA-C

and HLA-B alleles was extremely high with D’>0.90 for most combinations, which allowed

inferring the haplotypes for most individuals with high certainty. The LD values for the

haplotypes of interest are presented in Table 5.19.

The magnitude of LD for several of the HLA-C and –B alleles that were found to be

associated with PsA was high including: HLA-C*06/B*57 (D’=0.97), HLA-C*12/B*38

(D’=0.97), HLA-C*07/B*08 (D’=0.98), HLA-C*01/B*27 (D’=0.60) and HLA-C*02/B*27

(D’=0.61).

5.4.10. The association between HLA-B and –C haplotypes and PsA compared to Psoriasis

Based on the results of the single locus analysis that showed that most of the significant

associations are within the HLA-B and –C region, the haplotype analysis included only these

loci. A total of 107 possible haplotypes were estimated, however, due to low frequencies

only 23 common haplotypes were included in the analyses (Table 5.20). The following

haplotypes remained significantly associated with PsA compared to psoriasis after

multivariate analysis: HLA-B*18-C*07 (OR 10.1, p=0.004), HLA-B*27-C*01 (OR 41.1,

p<0.0001). HLA-B*27-C*02 (OR 19.9, p<0.0001), HLA-B*38-C*12 (OR 2.9, p=0.01),

HLA-B*08-C*07 (OR 2.6, p=0.004) and HLA-B*57-C*06 (OR 0.5, p=0.03). All of these

haplotypes include the risk alleles that were identified in the single locus analysis, the only

exception is HLA-B*18. For most of the haplotypes, the LD was high and ranged from 0.6 to

0.98, which illustrates the difficulty of investigating the independent effect of each of the

HLA alleles.

Analysis of the association of extended haplotypes across the MHC region with PsA revealed

only marginal associations with haplotypes that did not include the HLA risk alleles that

were identified before. Therefore, these associations may be explained by type I error.

Page 97: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

85

Table 5.19 - Linkage Disequilibrium of selected HLA-B and –C alleles

HLA-B/HLA-C Frequency

Corr. Coeff.

Lowenstein D’

*18/*07 1.7% 0.06

0.19

*27/*01 2.4% 0.48 0.60

*27/*02 2.8% 0.53 0.61

*37/*06

1.6%

0.30 0.97

*38/*12

4.2% 0.68 0.97

*57/*06 6.5% 0.62 0.93

*08/*07 11% 0.56 0.98

Page 98: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

Table 5.20 - Odds ratios comparing HLA-B/HLA-C haplotype frequencies in PsA (N=710) to psoriasis (N=334)

using logistic regression analysis

HLA-B/

HLA-C

Frequency LD

Univariate analysis Multivariate analysis

(Reduced model)

PsA

Psoriasis

D’ OR 95% CI P value OR 95% CI P

value

*18-*07 2.0% 0.6% 0.15 7.8 1.1-55.9 0.04 10.1 1.4-74.3 0.004 *27-*01 3.9% 0.7% 0.65 31.4 4.8-203.6 0.0003 41.1 6.4-266.1 <0.001

*27-*02 4.9% 1.3% 0.69 15.5 3.7-64.5 0.0002 19.9 4.7-84.1 <0.001

*38-*12 7.1% 4.8% 0.99 2.2 0.99-4.9 0.052 2.9 1.3-6.6 0.01

*08-*07 11.7% 8.5% 0.97 2.0 1.1-3.8 0.03 2.6 1.4-5.0 0.004

*57-*06 6.7% 11.5% 0.92 0.3 0.2-0.6 0.0004 0.5 0.3-0.9 0.03

*50-*06 1.4% 1.5% 1.00 0.9 0.2-4.1 0.87

*35-*04 6.4% 7.5% 0.87 1.0 0.5-2.0 0.91

*41-*17 0.9% 1.2% 0.91 0.6 0.1-3.4 0.54

*44-*16 2.6% 3.9% 0.91 0.4 0.2-1.2 0.11

*13-*06 4.1% 6.9% 1.00 0.3 0.2-0.8 0.009

*18-*12 1.0% 1.4% 0.23 0.4 0.1-2.4 0.32

*37-*06 1.3% 2.2% 1.00 0.4 0.1-1.4 0.14

*39-*07 1.3% 1.0% 0.37 2.0 0.3-12.6 0.47

*18-*05 1.1% 1.0% 0.24 2.3 0.3-16.2 0.42

*39-*12 1.7% 1.4% 0.50 1.3 0.3-6.2 0.71

*14-*08 3.7% 5.5% 0.98 0.5 0.2-1.1 0.08

*62-*03 3.1% 3.6% 0.85 0.7 0.3-2.0 0.55

*60-*03 3.2% 2.5% 1.00 1.6 0.5-4.7 0.43

*44-*05 6.3% 6.3% 0.78 1.0 0.5-2.2 0.95

*55-*03 1.2% 1.2% 0.80 1.0 0.2-5.5 0.99

*35-*04 6.4% 7.5% 0.87 0.7 0.4-1.5 0.37

Page 99: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

87

5.4.11. Family based association study

To validate our results, the HLA alleles that were identified in the population based study

were further assessed in a family based association study. Altogether, 178 PsA probands, 30

psoriasis probands and 561 first degree family members belonging to 208 families were

analyzed. Of those, 4 families (2 PsA probands, 2 psoriasis probands and their 13 family

members) were excluded due to Mendelian errors. The study population included 48

complete trios (2 parents and a proband), and 156 families with affected-unaffected sib-pairs

with or without their parents. The detailed structure of the study population is presented in

Table 5.21. One hundred of 201 (49.7%) probands had a positive family history of psoriasis

and 42 out of 201 (20.8%) probands had a family history of PsA

There was over-transmission and a significant association between HLA-C*12 and PsA

patients (Table 5.22, p=0.005). However, no significant association with PsD (Table 5.23,

p=0.14) was found. HLA-B*38, which is in strong LD with HLA-C*12, was over-

transmitted to the PsA patients when compared to the sibs with psoriasis alone, however the

strength of the association was weaker (Table 5.22, p=0.04) and there was no significant

association with PsD (Table 5.23, p=0.14). HLA-B*39, that was associated with axial PsA

compared to psoriasis in the single locus analysis, was over-transmitted to patients with PsA

(Table 5.22, p=0.045) compared to psoriasis alone. There was over-transmission of HLA-

B*27 among PsA and PsD compared to psoriasis alone (Table 5.22, p=0.002) and healthy

controls (Table 5.23, p=0.04), respectively. There was no significant association between

HLA-B*08, C*01, C*02, and C*07 and either PsA and PsD. The direction of association for

HLA-C*02 and C*01 was in accordance with what was found in the population-based

association study, while for HLA-C*07 and B*08, the direction of association was the

opposite. Interestingly, HLA-C*06, the strongest risk allele for psoriasis, was not

significantly associated with either PsA or PsD. However, the test statistics (Z) indicates

over-transmission of HLA-C*06 to PsD probands compared to healthy siblings, while under-

transmission of the allele to PsA compared to siblings with psoriasis, corresponding to the

direction of association of HLA-C*06 in the population based study.

Page 100: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

88

NA- Not applicable

Table 5.21 - Detailed family structure of the study population

Genotyping

data

available

No

siblings

1 or more healthy

sibling: no PsA or

psoriasis

1 or more

affected sibling:

PsA or psoriasis

Two or more

siblings: at least

1 healthy and at

least 1 affected

Total

PsA

proband

2 parents 40 35 14 5 94

1 parent NA 11 8 9 28

0 parents NA 32 15 7 54

Psoriasis

proband

2 parents 8 2 1 0 11

1 parent NA 4 0 0 4

0 parents NA 9 4 0 13

Page 101: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

89

*Frequency of the allele in the study population **Number of informative families

Table 5.22 - Family based association test Affected (PsA probands) - Unaffected (psoriasis) sib-pairs and Trios (N families= 131, N Pedigrees= 165, N=462)

Allele Allele frequency* No. Families** Z P value

HLA-B*27 8.5% 33 3.0 0.002

HLA-B*08 13.6% 36 -1.4 0.15

HLA-B*38 4.7% 15 2.0 0.04

HLA-B*39 3.5% 13 2.0 0.045

HLA-C*06 12.7% 35 -0.6 0.57

HLA-C*12 8.5% 28 2.8 0.005

HLA-C*07 27.7% 69 -1.0 0.32

HLA-C*01 4.7% 19 1.5 0.14

HLA-C*02 5.7% 22 1.1 0.27

Page 102: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

90

PsD – Psoriatic Disease (including PsA and psoriasis probands).

*Frequency of the allele in the study population **Number of informative families

Table 5.23 - Family based association test Affected (PsD)-Unaffected (Healthy controls) sib-pairs and Trios (N families= 204, N pedigrees=234, N=756)

Allele Allele frequency* No. Families** Z P value

HLA-B*27 6.9% 36 2.1 0.04

HLA-B*08 12.9% 49 -0.3 0.77

HLA-B*38 4.9% 24 1.4 0.14

HLA-B*39 2.4% 15 1.4 0.15

HLA-C*06 14.3% 55 1.3 0.18

HLA-C*12 8.9% 37 1.5 0.14

HLA-C*07 27.5% 87 -1.7 0.09

HLA-C*01 4.9% 26 1.3 0.21

HLA-C*02 4% 21 0.02 0.99

Page 103: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

91

5.5. Discussion

The MHC region on chromosome 6 and in particular, the HLA class I region has been

consistently associated with both psoriasis and PsA [41, 188-192]. In this large case-control

study, I aimed to identify PsA specific genetic markers among HLA alleles. The previously

reported association between HLA-B*27 and PsA was confirmed. Furthermore, HLA-C*06

that was associated with PsD and PsA compared to healthy controls was significantly less

frequent in PsA patients compared to those with psoriasis alone. Two additional HLA alleles,

HLA-B*08 and HLA-B*38 were identified as potential genetic markers for PsA in patients

with psoriasis. HLA-B*39 may be a potential marker for axial PsA.

In both population and family based analysis we have shown that HLA-B*27 is a strong

genetic marker for PsA compared to psoriasis. The absence of a detectable association

between HLA-B*27 and the psoriasis group compared to healthy individuals suggests that

B*27 is not a marker for skin disease. HLA-B*27 was also part of two haplotypes, along

with HLA-C*02 and HLA-C*01, that showed a strong association with PsA versus psoriasis.

Previous studies in different ethnic groups have consistently shown that HLA-B*27 is an

independent risk allele for PsA that is unrelated to the skin disease [41, 285-287]. However,

the prevalence of HLA-B*27 among PsA patients in our study was 19.2% and ranged from

20 to 35% in previous studies, clearly much lower than its prevalence in AS (80-95% of the

patients) [288]. Therefore, this allele can only account for a small proportion of the total

genetic risk of PsA. Several studies have shown an association between HLA-B*27 and axial

involvement in PsA [285, 289, 290]. It has also been suggested that HLA-B*27 is associated

with earlier onset of joint manifestations in patients with psoriasis, compared to non-carriers

[290]. It may be argued that the HLA-B*27 allele is merely a risk allele for AS that co-exists

with psoriasis, given the relatively high prevalence of psoriasis in the general population.

However, since the frequency of HLA-B*27 is also increased among PsA patients with

peripheral joint involvement, it suggests that HLA-B*27 is an independent risk allele for

PsA. Our group have shown that there is an association between HLA-B*27 in the presence

of DRB1*07 and more severe peripheral involvement and accumulation of joint damage,

features that are different from those of AS [291]. Therefore, HLA-B*27 can be considered

Page 104: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

92

as the strongest HLA risk allele for PsA among patients with psoriasis, and does differentiate

the two conditions.

Interpretation of the association between HLA-C*06 and PsA is more challenging since this

allele confers strong risk for psoriasis [180, 201, 202]. In the present study, the frequency of

HLA-C*06 was significantly higher in both psoriasis and PsA patients compared to controls.

However, the frequency of HLA-C*06 was lower in PsA patients compared to psoriasis. The

association remained significant in the subgroup of patients with early onset psoriasis,

however not among those with late onset disease. This appears to be inconsistent with the

report of Ho et al. who analyzed the association between HLA-C*06 and PsA. They

concluded that HLA-C*06 was associated only with early onset psoriasis and does not confer

any additional susceptibility to PsA. However, no direct comparison between PsA and

psoriasis was performed in that study. Analyzing their data shows that actually HLA-C*06 is

significantly decreased among PsA patients with early onset psoriasis compared to those with

psoriasis alone (169 (50.4%) vs. 354 (58.4%), p=0.02) [193]. Other studies have found a

lower prevalence of HLA-C*06 among PsA compared to psoriasis patients [287, 292]. In

addition, psoriatic nail involvement, a clinical marker for an increased risk of PsA, is more

common among HLA-C*06 negative psoriasis patients [178]. Although HLA-C*06 has not

been associated with a particular clinical subtype of PsA it has been found to increase the

psoriasis-arthritis latency period [293]. HLA-C*06 was also associated with a milder form of

arthritis among PsA patients. Ho et al. reported that PsA patients who carried HLA-C*06 had

fewer damaged joints and lower active joint counts [294]. It is challenging to explain the

strong association of HLA-C*06 with psoriasis while its lower frequency in PsA compared to

psoriasis. HLA-C*06 may be involved in different mechanisms that lead to the joint and the

skin disease. For example, it may be: antigen presentation and activation of the adaptive

immune system for the skin and inhibition of the innate immune system through interaction

with Killer cell Immunoglobulin like Receptors (KIRs) on NK cells in the joints. The

recently reported association between ERAP1 gene polymorphisms and psoriasis only among

patients carrying the HLA-C*0602 risk allele, suggests that antigen presentation may play a

role in the pathogenesis of psoriasis, as ERAP1 is involved in MHC class I peptide

processing [252]. Another explanation may be related to genetic heterogeneity of psoriasis. It

is possible that different HLA alleles are associated with a sub-type of psoriasis that is more

Page 105: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

93

likely to develop PsA. Therefore, although the phenotype of psoriasis is similar, one HLA

allele is associated only with skin disease (HLA-C*06) while the other is associated with

both skin and joint involvement. A potential candidate is HLA-C*12, that is associated with

both PsA and psoriasis compared to healthy controls, and was reported in the past to be

associated with psoriasis among Caucasians [170].

Several of the alleles (HLA-C*02, HLA-C*01 and HLA-DRB1*07) that were found to be

associated with PsA in the univariate analysis were no longer significant after adjusting for

the effect of the other alleles. Several studies have suggested that HLA-DRB1*07 is

associated with PsA [192, 286]. In the present study, the frequency of HLA-DRB1*07 was

significantly lower in the PsA group compared to the psoriasis group. However, HLA-

DRB1*07 is in strong LD with HLA-C*06 among Caucasians and it is part of the ancestral

haplotypes AH.13 and AH.57 that contain this strong risk allele for psoriasis in addition to

HLA-B*13 or –B*57 [145]. In our study, these extended haplotypes were associated with

PsD compared to healthy controls. The association between HLA-DRB1*07 and PsA was no

longer significant after adjusting for the effect of HLA-C*06 suggesting that the risk allele is

HLA-C*06. Similarly, HLA-C*02 and HLA-C*01 showed an association with PsA

compared to psoriasis, that was lost after accounting for the effect of HLA-B*27 in the

multivariate analysis. There was a strong LD between HLA-C*02/C*01 and HLA-B*27

(HLA-B*27-C*01 D’=0.60, HLA-B*27-C*02 D’=0.61). These alleles are part of common

haplotypes found in 1-2.9% of the Caucasians [284] and in this study both haplotypes were

significantly associated with PsA compared to psoriasis. Therefore the association between

C*02/C*01 and PsA was not independent and was related to their LD with HLA-B*27.

These findings illustrate the difficulties in investigating the independent association of genes

in the MHC region that contains numerous potential risk loci in close proximity.

Two additional alleles were significantly associated with PsA compared to psoriasis in

multivariate analysis: HLA-B*08 and HLA-B*38. HLA-B*38 was strongly associated with

PsA compared to both psoriasis (multivariate model) and controls (univariate model). HLA-

B*38 has been reported to be more frequent among non-Ashkenazi Jews with PsA compared

to those with psoriasis alone and healthy controls in a small study from Israel [295]. Another

small study from Maryland also reported higher frequencies of HLA-B*38 in PsA compared

Page 106: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

94

to psoriasis [292]. Additionally, HLA-B*38 has been associated with more peripheral joint

involvement [296]. However, in the subgroup analysis of our study, an association of HLA-

B*38 was detected in the group with axial PsA but not in the group with arthritis restricted to

the peripheral joints. HLA-B*38 is part of a common haplotype along with HLA-C*12. In

our study, this haplotype was associated with PsA compared to psoriasis. This haplotype has

been reported to be present in up to 8% of the Jewish population and in 1.3-3.4% of non-

Jewish Caucasians [284]. A recent GWAS suggested that HLA-C*12 is a risk allele for

psoriasis among Caucasians but not among Chinese [170]. The family based data in this

study supports the association between HLA-B*38 and C*12 and PsA, however in contrast to

the population based data, a larger test statistic was observed with HLA-C*12 compared to

HLA-B*38 (p=0.005 vs. p=0.04, respectively). Given the very high LD (D’=0.97) between

alleles in our sample, it is very difficult to dissect the independent effect of each one of them.

Larger samples sizes may be required for that purpose.

HLA-B*08 has not been previously reported to be associated with PsA. In our study, HLA-

B*08 frequencies were higher among PsA compared to psoriasis, but not different from

healthy controls. However, these results were not confirmed in our family based association

study. In the literature, the extended haplotypes across HLA class I and II genes, A*01-

C*07-B*08-DRB1*03-DQA1*05-DQB1*02-DPA1*01-DPB1*04, has been associated with

increased TNF production following rubella vaccination [297]. TNF is a major pro-

inflammatory cytokine in PsA and rubella vaccine was suggested to be a risk factor for

development of PsA among psoriasis patients [76]. HLA-B*08 is part of an ancestral

haplotype (8.1 AH) that also includes HLA-A*01-C*07-DRB1*03 and TNF-308A [284].

There have been conflicting results with regard to the association between PsA and the TNF-

308A polymorphism, which is part of the 8.1 AH [210-212]. Previous analysis of our cohort

combined with PsA patients from Newfoundland did not find an association with TNF-308A

polymorphism [223], however, these patients were compared to healthy controls and not to

psoriasis patients. In our study, HLA-B*08 was also included in the HLA-B*08-C*07

haplotype that was increased in patients with PsA compared to those with psoriasis alone.

HLA-C*07 was included in another haplotype that was associated with PsA (HLA-B*18-

C*07). In summary, HLA-B*08 may be an independent genetic marker for PsA among

Page 107: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

95

psoriasis patients. However, its strong LD with other HLA alleles and relevant genes in the

MHC region precludes a more confident conclusion.

In summary, in this study the independent association between HLA-B*27 and PsA was

confirmed. HLA-C*06 was more frequent in patients with PsD compared to healthy controls,

however it was less frequent in patients with PsA compared to those with psoriasis. This

association was particularly strong among patients with early onset psoriasis. Whether HLA-

C*06 has a “dual” effect, acting as a risk factor for psoriasis and a protective factor for PsA

among patients with psoriasis, needs further investigation using a longitudinal study. Several

additional alleles, B*38 or C*12 and B*08, are potential genetic markers for PsA in patients

with psoriasis, and warrant further investigation.

Page 108: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

96

Chapter 6. The effect of HLA risk alleles on the rate of development of PsA among psoriasis patients

Page 109: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

97

6.1. Background

Approximately 85% of patients with psoriasis develop PsA concurrently or thereafter [31]. In

a previous chapter the incidence of PsA among psoriasis patients was found to be higher than

previously reported. In contrast to what has been thought in the past, the risk of developing

PsA was found to be constant over time. However this observation was based on a small

number of incident cases of PsA. There is limited information in the literature about factors

that affect the interval of time from the onset of psoriasis to PsA. The importance of this

issue is related to the challenge in investigating genetic risk factors for PsA among psoriasis

patients. When a case-control study design is used for the investigation of risk factors, one of

the assumptions is that the outcome of interest has a low frequency in the control group. This

assumption is usually true for investigation of a relatively rare disease in the general healthy

population. However, this assumption does not hold when psoriasis patients are used as the

control group for PsA. In this situation a significant proportion of the controls may become

cases. Therefore, significant associations may be related to differences in age at onset and

duration of psoriasis between the cases and the controls. It has been reported that early onset

psoriasis is associated with a prolonged interval between the onset of psoriasis to the

development of PsA [298]. Another study has found that HLA-C*06 also increases that

interval [293]. Therefore, there is an interest in investigating the association between genetic

risk factors and the rate of PsA among psoriasis patients.

The first aim of this chapter is to validate the results from Chapter 4 by using data pooled

from two large cohorts of psoriasis and PsA patients to further assess whether the risk of

developing PsA remains constant over time. The second aim is to investigate clinical and

genetic risk factors that affect the rate of development of PsA among psoriasis patients.

Concerns associated with combining the two cohorts were addressed by statistical modeling.

Page 110: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

98

6.2. Methods

The present study is a retrospective cohort analysis of data pooled from 2 cohorts:

1. The University of Toronto PsA cohort.

2. The Toronto Psoriasis Cohort.

These cohorts were described in the Methods chapter (Groups 1 and 2).

Only PsA patients that developed their arthritis after the diagnosis of psoriasis were included.

6.2.1. Data collection and statistical analysis

The year of onset of psoriasis was ascertained from each patient at their initial visit to the

clinic. This was defined as the year the disease was diagnosed by a dermatologist. The onset

of PsA was defined as the year the disease was diagnosed by a rheumatologist, in most of the

patients this occurred shortly prior or at their first visit to the PsA clinic.

For each participant the cumulative person-years at risk were calculated. For the PsA cohort

person-years at risk were calculated from the diagnosis of psoriasis to the diagnosis of PsA,

while for the psoriasis patients it was the time interval from the diagnosis of psoriasis until

either the onset of PsA or the last assessment date, whichever came first. Patients with

psoriasis alone were censored at their last visit to the clinic. This duration of time was used to

assess the rate of PsA among psoriasis patients.

As discussed previously, some patients in the psoriasis cohort did not have any follow-up

visit after their initial screening visit and therefore did not contribute to follow-up time.

Analyses were performed twice, with and without these subjects. Two parametric models

were used to evaluate whether the risk of developing PsA, as modeled by the hazard

function, changes over time. A time homogeneous (exponential) model, that assumes a

constant hazard function, was used to estimate the rate of PsA among psoriasis patients. This

model was compared to a Weibull model that assumes a trend in hazard function. The lack of

Page 111: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

99

a significant difference between the two models, as assessed by the LR test, was taken as lack

of evidence for time-dependent rate.

Performing survival analysis on data pooled from two different cohorts may lead to a

sampling bias due to truncation. Truncation of survival data occurs when only those

individuals whose event time lies within a certain observational window are included in a

sample. An individual whose event time is not in this window is not included in the sample

and hence no information on this subject is available [299]. Right truncation occurs when

only individuals that experienced the event of interest are observable. Such individuals are

the patients from the PsA cohort that have already developed the outcome of interest (PsA).

Left truncation occurs when patients that developed the outcome of interest before the

observational window are excluded. In the present study, they include the patients that

developed PsA before the onset of psoriasis.

The main impact on the analysis when data are truncated is that we must use the conditional

distribution of the event time given it fell in the truncation window when constructing the

likelihood. Parametric survival analysis for right truncated data (PsA) and for left truncated

& right censored data (psoriasis) was used to model the risk of developing PsA after the

onset of psoriasis. Parametric proportional hazard regression model was used to assess the

association between clinical and genetic variables to the interval of time from psoriasis to

PsA. The following HLA alleles that were identified as potential genetic markers for PsA

among psoriasis patients were included in the analysis: HLA-C*06, HLA-C*01, HLA-C*02,

HLA-C*12, HLA-C*07, HLA-B*27, HLA-B*38 and HLA-B*08. Two tailed p value < 0.05

was considered significant.

Page 112: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

100

6.3. Results

447 patients with psoriasis and 772 patients with PsA were included in the analysis. Their

demographic and clinical characteristics are presented in Table 6.1. The mean age at onset of

psoriasis and PsA were 26.8±14.7 and 37.3±13.3 years. The sex ratio was 1.3:1 (male:

female). The proportion of early onset psoriasis was 82.8%. The mean time from the onset

of psoriasis to development of PsA was 12.2±10.7 years.

6.3.1. The rate of PsA among psoriasis patients over time

The distribution of the time to the development of PsA was fit with an exponential model,

which has a constant hazard rate. Tests for trend did not suggest a departure from the

constant hazard (Figure 6.1). These results concur with the findings among the prospective

psoriasis cohort (Chapter 4) that showed no trend in risk of PsA. We are able to confirm

these findings in a much larger cohort.

6.3.2. The effect of HLA risk alleles on rate of PsA among psoriasis

patients

Parametric proportional hazard regression model was used to assess the effect of HLA risk

alleles on the rate of development of PsA among psoriasis patients. The effect was assessed

from the onset of psoriasis. The results are presented in Table 6.2. Patients that had at least

one copy of HLA-C*06 had longer time intervals from the onset of psoriasis to PsA (mean of

13.8±10.4 vs. 11±10.9 years, respectively). This finding was confirmed by a regression

analysis that showed that psoriasis patients who are HLA-C*06 positive had a lower rate of

development of PsA over time compared to HLA-C*06 negative. This finding was

independent of the age at onset of psoriasis, sex and other HLA risk alleles with a Relative

Risk (RR) of 0.44 (95% CI 0.36- 0.54, p<0.001). None of the remaining HLA risk alleles had

any association with the rate of PsA. There were only 102 patients who were HLA-B*38

positive and only 73 of them had information about the age at onset of PsA. These numbers

Page 113: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

101

were small and did not fit well with the truncation model and therefore they were excluded. It

is unlikely that the exclusion of HLA-B*38 from the full regression model has created a bias

that affected the association with HLA-C*06, since these HLA alleles do not usually appear

on the same haplotype.

Since HLA B*27 is the strongest genetic risk factor for PsA among psoriasis patients, an

additional analysis was performed to determine whether this allele has any effect on the time

to onset of PsA. The time to event was calculated from birth to the onset of PsA, and the

association between HLA risk alleles was assessed using a similar multivariate regression

model. The results (Table 6.3) provide no evidence that any of the alleles was associated with

the duration of time from birth to onset of PsA.

Page 114: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

102

Table 6.1 - Characteristics of the study population

PsA cohort

(N=772)

Psoriasis cohort

(N=447)

Age at first clinic visit (Mean) 42.8±12.7

46.5±13.1

Age at onset of psoriasis (Mean) 24.9±13.5

30.1±16.3

Age at onset of PsA (Mean)* 37 ±13.1 54.8±12.1

Duration of time from onset of psoriasis to PsA (Mean)*

12.1±10.5

18.7±18.9

Sex: Female (%) 339 (43.9%) 192 (43%)

Early onset psoriasis (%) 671 (86.9%) 334 (74.7%)

HLA-C*06 (%) 167 (31.5%) 174 (41.6%)

HLA-B*27 (%)

87 (16.3%) 17 (4.1%)

HLA-B*38 (%) 74 (13.7%) 40 (9.5%)

HLA-B*08 (%) 106 (19.7%) 56 (13.3%)

HLA-C*01 (%) 64 (11.9%) 31 (7.4%)

HLA-C*02 (%) 56 (10.4%) 20 (4.8%)

HLA-C*07 (%) 232 (43.3%) 167 (39.8%)

HLA-C*12 (%) 124 (23.1%) 71 (16.9%)

*15 patients from the psoriasis cohort developed PsA during the follow-up.

Page 115: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

Figure 6.1- The probability of developing PsA among psoriasis patients from the onset of psoriasis (Ps) assuming an exponential model.(prospective cohort) with and assuming they did not develop PsA. Similarly, the black lines denote the data pooled from the PsA and the psoriasis cohorts up visit.

The probability of developing PsA among psoriasis patients from the onset of psoriasis (Ps) assuming an exponential model. The red lines denote the psoriasis cohort alone

and without the patients that did not have any followassuming they did not develop PsA. Similarly, the black lines denote the data pooled from

horts with and without the patients that did not have any follow

103

The probability of developing PsA among psoriasis patients from the onset of The red lines denote the psoriasis cohort alone

the patients that did not have any follow-up visit assuming they did not develop PsA. Similarly, the black lines denote the data pooled from

the patients that did not have any follow-

Page 116: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

104

PsA - Psoriatic Arthritis, CI - Confidence Interval, RR - Relative Risk

*Problem of monotone likelihood due to sparse data

Table 6.2 - The association between HLA alleles and the risk of PsA in analysis of time from

onset of psoriasis by parametric proportional hazard model (N=939)

Variable Univariate Full model Reduced model

RR 95% CI p RR 95% CI p RR 95% CI p

Sex: Male 1.32 0.42-4.15 0.64 1.25 0.44-3.62 0.68

Late onset Psoriasis

6.46 0.53-79.3 0.14 6.05 0.18-202.7 0.32

HLA-C*01 1.78 0.28-11.2 0.54 0.96 0.19-4.89 0.96

HLA-C*02 0.49 0.28-0.86 0.01 0.56 0.03-10.1 0.69

HLA-C*06 0.44 0.36-0.54 <0.001 0.57 0.11-2.86 0.49 0.44 0.36-0.54 <0.001

HLA-C*07 1.34 0.44-4.13 0.61 1.16 0.36-3.71 0.80

HLA-C*12 1.29 0.34-4.85 0.71 1.37 0.32-5.71 0.66

HLA-B*08 1.16 0.34-3.98 0.81 1.00 0.27-3.69 0.99

HLA-B*27 1.32 0.23-7.84 0.75 2.82 0.07-123.8 0.59

HLA-B*38* NA

Page 117: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

105

PsA - Psoriatic Arthritis, CI - Confidence Interval, RR - Relative Risk

*Problem of monotone likelihood due to sparse data

Table 6.3 - The association between HLA alleles and risk of PsA in analysis of time from birth

by parametric proportional hazard model (N=939)

Variable Univariate Full model Reduced model

RR 95% CI p RR 95% CI p RR 95% CI p

Sex: Male 1.19 0.40-3.54 0.74 1.19 0.44-3.16 0.73

Late onset Psoriasis

2.72 0.45-16.45 0.27 2.63 0.35-19.81 0.34

HLA-C*01 1.41 0.27-7.22 0.68 1.49 0.41-5.41 0.54

HLA-C*02 0.54 0.25-1.16 0.11 0.91 0.08-10.0 0.93

HLA-C*06 0.61 0.35-1.09 0.09 0.68 0.17-2.76 0.60

HLA-C*07 1.15 0.42-3.18 0.78 0.93 0.30-2.91 0.91

HLA-C*12 0.91 0.33-4.44 0.85 0.75 0.21-2.64 0.66

HLA-B*08 1.09 0.33-3.59 0.88 1.07 0.29-3.93 0.92

HLA-B*27 0.73 0.27-1.97 0.53 0.61 0.24-1.58 0.31

HLA-B*38* NA

Page 118: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

106

6.4. Discussion

This study combined data from two cohorts to assess the rate of PsA among psoriasis

patients. It confirmed that the rate of PsA remains constant following the onset of psoriasis

and in contrast to the previous notion; it does not decrease over time. In addition, the only

HLA allele that was associated with the time interval from psoriasis to PsA was HLA-C*06.

Carriers of HLA-C*06 had prolonged intervals from the onset to psoriasis to development of

PsA.

There are limited data about the incidence of PsA among psoriasis patients. The mean

duration of time from psoriasis to PsA is approximately 7 years [300]. Therefore it was

assumed by experts in the field that the risk of developing PsA is highest in the first years

following the onset of psoriasis. However, there are no actual data in the literature to support

that notion. In a recent retrospective study from Germany, the cumulative incidence of PsA

among psoriasis patients was 20.5% after 30 years and the rate of PsA was constant over

time [32]. In the present study, the distribution of the time to the development of PsA was fit

with an exponential model. The lack of a significant difference between the Weibull and the

exponential model suggests that the risk of developing PsA among psoriasis patients does not

change over time. These results counter the previous notion that the risk of developing PsA

decreases over time following the onset of psoriasis.

HLA-C*06 is the strongest genetic risk factor for psoriasis, however, in Chapter 5 we have

found that its frequency was significantly lower in PsA patients compared to those with

psoriasis alone. In the present study, HLA-C*06 allele was also associated with a prolonged

interval of time from psoriasis to PsA that supports its “protective” effect. HLA-C*06

carriers had more than double the interval of time from the onset of psoriasis to PsA. These

results are in accordance with a previous study of Queiro et al. that reported that HLA-C*06

positive PsA patients have a longer psoriasis-arthritis latency periods (9 vs. 5 years, p=0.03)

[293]. The explanation for this observation is unclear. HLA-C*06 is associated with more

severe psoriasis and early onset of the disease [178], while there is no difference in the age at

onset of PsA between HLA-C*06 positive or negative patients. It is possible that the

increased time interval among HLA-C*06 positive patients is merely related to the early

Page 119: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

107

onset of psoriasis and that the expression of that allele does not affect the onset of PsA.

However, the observation that HLA-C*06 is less frequent among PsA patients and is also

related to a milder form of joint disease [294] suggests that this allele may have a

“protective” effect on PsA among patients with psoriasis that may be mediated through

different mechanisms than those of psoriasis.

None of the other HLA alleles was significantly associated with the duration of time from

psoriasis to PsA including the strongest risk allele for PsA, HLA-B*27. There are several

potential explanations for these findings. First, these alleles may only be associated with joint

disease and not with skin disease. However, the lack of association with interval from birth to

onset of PsA does not support this explanation. Secondly, although often the presence of risk

genes is associated with earlier onset of a disease, these genes can affect the susceptibility for

the disease without affecting its age at onset. Lastly, the power to detect an association is

dependent on the prevalence of these alleles. As suggested by the large confidence intervals

of the RR, for some of the HLA alleles the sample size was small and the study was

underpowered to detect an association.

A limitation of the present study is the reliance on self-reported age at onset of psoriasis,

although we do not assume that there is a difference in recall between patients with PsA and

those with psoriasis. The accuracy of reporting age at onset of psoriasis by patients with

psoriasis has been previously assessed by evaluating the age of onset distribution curve

[301]. It has been concluded that before the age of 20 years patients were accurate in

identifying the onset of psoriasis within two years, while later in life there was a tendency to

round off the age at onset to a multiple of 5 years. The duration of psoriasis prior to the

presentation to clinic also affected the accuracy of report. Despite this limitation,

dermatologists have noted that patients are able to detect the presence of psoriasis with fair

degree of accuracy [302].

In summary, in this study the rate of PsA among psoriasis patients was assessed. The study

confirmed previous observations that the rate of PsA among psoriasis patients remains

constant over time. Additionally, HLA-C*06 was found to be associated with a longer

interval of time from onset of psoriasis to development of PsA.

Page 120: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

108

108

Chapter 7. The association of KIRs and their HLA ligands with PsA

Page 121: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

109

7.1. Background

Natural Killer (NK) cells are thought to play an important role in the pathogenesis of

psoriasis and PsA [67]. Their activity is determined by a balance between activating and

inhibitory signals transmitted by a range of receptors including Killer Immunoglobulin-like

Receptors (KIRs) [63].

The KIR gene cluster spans approximately 150 kb in the leukocyte receptor complex on

chromosome 19q13. The KIR region is very diverse, and includes different combinations of

KIR genes as well as allelic polymorphisms. Each NK cell has a combination of inhibitory

and activating receptors that interact with certain HLA alleles to result in an immune

response. KIRs are classified into activating and inhibitory receptors based on the length of

their cytoplasmic tail. Activating receptors have a short (S) tail, while inhibitory receptors

have a long (L) tail. KIRs are also categorized into 2 groups on the basis of their external

immunoglobulin-like domain (2D or 3D) (Figure 7.1) [225]. The specificity of the different

KIRs for HLA class I molecules is determined by their extracellular structure. Studies have

shown that binding of inhibitory KIRs for specific HLA molecules correlates well with their

ability to inhibit NK cytolysis of target cells bearing those HLA allotypes [68, 226]. The

specificities of the different KIRs for HLA alleles are presented in Table 7.1. Overall, this

system provides a mechanism by which an individual’s HLA repertoire may directly

influence the type and extent of the immune response in the context of a particular KIR

haplotype.

Page 122: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

Figure 7.1 - Illustration of the struct

Table 7.1

KIR2DL1 and 2DS1

HLA-C group 2

C*02 C*04 C*05 C*06 C*15 C*17 C*18

Illustration of the structure of the different type of KIRs[303]

Table 7.1 - KIR ligand specificities

KIR2DL2/3 and 2DS2 KIR3DL1 and 3DS1

HLA-C group 1

C*01 C*03 C*07 C*08 C*12 C*14 C*16

HLA-

B*2702B*3801

110

KIR3DL1 and 3DS1

-Bw4 – Ile80

B*77 B*63 B*62

B*2702 B*3801

B*49 B*51 B*52 B*53 B*57 B*58

Page 123: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

111

Several studies have shown that polymorphisms of genes encoding activating KIRs are

associated with PsA. It was found that the frequencies of KIR2DS1 and KIR2DS2genes were

higher in PsA patients compared to healthy controls. HLA-C group homozygosity is also

associated with an increased risk of developing PsA [229, 230]. These studies, however,

compared PsA patients to healthy controls and since KIR genes have also been associated

with psoriasis, it is unclear whether these findings represent an association that is

independent of the skin disease.

KIR3DL1 binds the HLA-Bα1-helix around residues 76-80 with specificity for all HLA-Bw4

alleles containing isoleucine at heavy chain residues 80 (Bw4-80I) [232], whereas the ligand

for KIR3DS1 has not yet been determined. However, it was suggested that KIR3DS1 also

binds HLA-Bw4-80I since it shares 97% sequence homology in its extracellular domain with

KIR3DL1 [233]. Several studies reported higher frequencies of the activating KIR3DS1 gene

in Ankylosing Spondylitis (AS) patients compared to HLA-B*27 positive healthy controls,

suggesting that the interaction between the activating receptor and its putative ligand may

increase susceptibility to the disease [234]. However, other studies in AS were unable to

replicate these findings [235]. Ankylosing spondylitis belongs to the family of SpA as PsA

and in both diseases HLA-B*27 is an important susceptibility allele. To our knowledge, the

association between KIR3DS1 and PsA has not been tested previously.

In summary, several studies have shown an association between activating KIRs and both

psoriasis and PsA. However, it is unclear whether the KIRs; alone or in combination with

their HLA ligands confer an independent risk for PsA among psoriasis patients.

Page 124: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

112

7.2. Methods

7.2.1. Study population

In this population based association study three groups of individuals from the same ethnic

group and geographic region were compared.

The PsA group included 710 adult Caucasian PsA patients who were recruited from the

University of Toronto PsA cohort. All patients satisfied the CASPAR criteria for

classification of PsA [22]. 333 psoriasis patients were recruited from the Toronto Psoriasis

cohort. These patients had the diagnosis of psoriasis confirmed by a dermatologist and were

assessed by a rheumatologist to rule out inflammatory arthritis. Therefore, at the time of

analysis, all psoriasis patients were free of arthritis. Control DNA was obtained from 707

healthy Caucasian volunteers, cadaveric organ donors from laboratories at the University

Health Network and from a commercial biobank.

The groups of PsA and psoriasis patients were combined to create a Psoriatic Disease group

(PsD) that was compared to the healthy control group.

7.2.2. KIR typing

Extracted genomic DNA was amplified by PCR using locus specific primers for each of the

following KIR genes: 2DL1, 2DL2, 2DL3, 2DL4, 2DL5, 2DS1, 2DS2, 2DS3, 2DS4, 2DS5,

3DL1, 3DL2, 3DL3, 3DS1. The frequency of each KIR gene was expressed as the inferred

phenotype frequency in the sample population.

7.2.3. Sample size calculation

Power calculations indicate that a study comparing 710 PsA patients to 333 psoriasis patients

has >80% power to detect an odds ratio of 2.1, 1.8, 1.6 and 1.5, for the risk gene frequencies

of 0.05, 0.1, 0.2 and 0.4, respectively at 5% alpha (type I error level). Power calculations

were performed using PS Power and Sample Size Calculations version 3.0 [266].

Page 125: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

113

7.2.4. Statistical analysis

The phenotypic frequencies of the KIR genes (the presence of at least one copy of the gene)

were compared between the PsD group vs. healthy controls and between PsA vs. psoriasis by

a LR test. Because of the known association of HLA-C*06 and B*27 with PsA, the study

focused on seven KIR genes: five genes that have HLA-C as their recognized ligand (i.e.,

KIR2DL1, -2DL2, -2DL3, -2DS1, and-2DS2) and two additional genes that have HLA-Bw4

group as their ligand (i.e. KIR3DS1 and KIR3DL1). The significance level was set at 0.05

for a 2-sided test.

Since the interaction between KIR genes and HLA class I molecules has a biologic basis, I

investigated the association between combinations of activating KIRs and their HLA ligands

with PsA. The following biological interactions were investigated:

1. KIR 3DS1 gene and HLA-Bw4-80I alleles.

2. KIR 2DS1 genes and HLA-C group 2 alleles.

3. KIR 2DS2 genes and HLA-C group 1 alleles.

These combinations were selected based on the inclusion of several susceptibility alleles for

PsA (HLA-B*27, HLA-B*38, HLA-C*12 and C*06) in these HLA groups. These alleles

have been found to be independently associated with PsA compared to psoriasis. HLA B*27

and HLA-B*38 are included in the HLA-Bw4 group, HLA-C*12 is included in the HLA-C

group 1 and HLA–C*06 is included in HLA-C group 2. Data from the literature also support

the association between these KIR genes and increased susceptibility to PsA [229, 230].

Page 126: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

114

7.2.5. The combined effect of KIR 3DS1 and HLA-Bw4 alleles

HLA-B alleles were categorized according to the protein sequence alignment into HLA-Bw4

and HLA-Bw6. The HLA-Bw4 group was further classified into HLA-Bw4-80I and 80T

based on the presence of isoleucine or aspargine in position 80, respectively [232]. Due to the

high sequence homology between KIR3DS1 and 3DL1, it is assumed that they share HLA-

Bw4-80I group as a ligand [233]. Therefore, the interaction between KIR3DS1 and HLA-

Bw4-80I alleles was investigated. Assuming that the effect of KIR3DS1 is dependent on the

presence of HLA-Bw4-80I, a higher frequency of that combination in the PsA group

compared to the psoriasis group was anticipated.

The distribution of HLA-Bw4-80I and KIR3DS1 was compared in PsA vs. psoriasis and PsD

vs. healthy controls by a LR test. Due to apparent effects of multiple variables at the HLA

and KIR loci on the risk of developing PsA, a multivariate logistic regression analysis was

used to assess the association between various genotypes and PsA. The model included HLA

alleles that were previously found to be associated with PsA (HLA-B*27, HLA-C*06), the

activating KIR gene (KIR3DS1) and its inhibitory KIR homolog (KIR3DL1). The activating

KIR3DS1 gene in the presence of its HLA putative ligand, HLA-Bw4-80I was also used as a

covariate in the model. Stepwise logistic regression analysis with backward selection was

used to identify key factors that differentiate between PsA and psoriasis alone. Covariate

effects were considered statistically significant if the p-value from the 2-sided Wald-test was

less than 0.05.

7.2.6. The combined effect of KIR 2DS1/2DS2 and HLA-C1/C2 groups

HLA-C assignments were categorized, according to current protein sequence alignments, into

two C groups as follows: C group 1 - HLA-C*01, 03, 07, 08, 12, 14, and 16; C group 2 -

HLA-C*02, 04, 05, 06, 15, 17, and 18.

Based on data from the literature that suggested that the interaction between the activating

KIR2DS and the ligands for the inhibitory KIR homologs can lead to activation of NK cells

[304, 305], the following gene-gene interactions were investigated:

Page 127: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

115

� KIR 2DS1 interacts with HLA-C*group 2

� KIR 2DS2 interacts with HLA-C*group 1

The combined effect of KIR2DS1 or KIR2DS2 and their corresponding HLA-C ligands was

assessed. The distribution of KIR2DS1 or KIR2DS2 and their corresponding HLA-C ligands

was compared in PsA vs. psoriasis and PsD vs. healthy controls by LR test. Assuming that

the effect of these activating KIRs is dependent on the presence of their corresponding HLA

ligands, higher frequencies of these combinations in the PsA group compared to the psoriasis

group were anticipated.

Due to apparent effects of multiple variables at the HLA and KIR loci on the risk of

developing PsA, a multivariate logistic regression analysis was used to assess the association

between various genotypes and PsA vs. psoriasis alone and PsD vs. healthy controls. The

model included HLA alleles that were previously found to be associated with PsA (HLA-

B*27), the activating KIR genes (KIR2DS1, KIR2DS2) and their inhibitory KIR homologues

(KIR2DL1, KIR2DL2, KIR2DL3). Additionally, the two activating KIR genes in the

presence of their HLA ligands (i.e., KIR2DS1 with group 2 HLA-C alleles, KIR2DS2 with

group 1 HLA-C alleles) were also used as covariates in the model. Stepwise logistic

regression analysis with backward selection was used to identify key factors that differentiate

between PsA and psoriasis alone and between PsD and healthy controls. Covariate effects

were considered statistically significant if the p-value from the 2-sided Wald-test was less

than 0.05.

Page 128: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

116

7.3. Results

7.3.1. The association of KIR genes and PsA – single locus analysis

The phenotypic distributions of the different KIR genes in the PsD group vs. healthy controls

and in the patients with PsA vs. those with psoriasis are presented in Tables 7.2-7.3.

The framework genes KIR2DL4, KIR3DL2 and KIR3DL3 were present in almost all

subjects. Several individuals, however, did not carry these genes. These results were

confirmed by sequencing of the KIR region and by analyzing data from other family

members of these individuals. They represent a rare deletion of these framework genes. The

frequencies of inhibitory KIRs 2DL2 and 2DL3 were higher in the PsD group compared to

the healthy controls; however these gene frequencies were similar in PsA compared to

psoriasis alone. The frequency of KIR2DL1 was lower in the PsA group compared to

psoriasis alone (97% vs. 99.4%, p=0.02); however, its frequency was similar among the PsD

and the healthy control group. The frequency of KIR2DS2 was significantly higher in the

PsA group compared to healthy controls (54.7% vs. 48.9%, OR 1.3, p=0.03). The frequency

of KIR2DS2 also tended to be higher in the PsA group compared to psoriasis alone however,

it did not reach statistical significance (54.7% vs. 49.3%, p=0.09). The frequencies of the

remaining KIR genes were similar across the three groups.

7.3.2. The association between KIR genotypes and PsA

The most common genotype in all three groups was that corresponding to the homozygous

“A” haplotype (2DL1, 2DL3, 2DL4, 3DL1, 3DL2, 3DL3, and 2DS4), which was found in

approximately one third of the study population (Figure 7.2). More than 80% of the subjects

were represented by the 10 most frequent genotypes. There was no difference in the

distribution of the genotypes in the three groups.

Page 129: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

117

7.3.3. KIR/HLA interaction

To investigate the biological interaction of KIRs and HLA genes, the frequencies of the

combined genotypes of KIRs and their corresponding HLA ligands were compared between

the groups based on a suggested model of effect that is elicited by the combination of

activating or inhibitory KIRs and their corresponding HLA ligands as described in the

Methods section.

7.3.4. The interaction of KIR 2DS1/2DS2 with HLA-C group 1/C group 2

There were significant differences in the frequencies of HLA-C group 1 and group 2 among

the PsA, psoriasis and controls (Table 7.4). The HLA-C group 2 alleles were more common

in patients with PsA compared to the healthy controls (p=0.01), but were less common

compared to psoriasis alone (p=0.03). While the frequency of HLA-C group 1 alleles was

higher among the PsA patients compared to those with psoriasis alone (p=0.02), it was

similar among the PsA group and the healthy controls.

The results of the multivariate analysis that investigated the association between PsA and the

activating KIRs, 2DS1 and 2DS2, and their corresponding HLA ligands compared to

psoriasis are presented in Table 7.5. In the presence of its corresponding ligand, HLA-C

group 1, KIR2DS2 was associated with PsA compared to psoriasis alone, suggesting that

their combination may increase the susceptibility to PsA among psoriasis patients. The

presence of KIR2DS2 in the absence of HLA-C group 1 alleles was also associated with PsA

compared to psoriasis. The level of significance and the OR for the KIR2DS2/HLA-C group

1 increased after inclusion of the corresponding inhibitory KIR2DL1 in the multivariate

regression model, suggesting that the combined effect of KIR2DS2/HLA-C group 1 is

stronger in the absence of the corresponding inhibitory KIR. In order to elucidate the

combined effect of KIR2DL2 and KIR2DS2, their frequencies were compared in PsA

patients vs. those with psoriasis alone. The results in Table 7.6 show that KIR2DL2 and

KIR2DS2 are highly concordant and appear separately only in 21 out of 1043 patients (2%).

However, the frequency of the activating KIR2DS2 without its inhibitory homologue,

KIR2DL2, was higher in the PsA group compared to psoriasis (13(1.8%) vs. 0), while the

Page 130: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

118

frequency of the inhibitory KIR2DL2 without its activating homologue, KIR2DS2, was

higher in the psoriasis group compared to the PsA group (6 (1.8%) vs. 2 (0.3%)). These

figures explain the reason these two genes were not significantly associated with PsA in the

univariate, single locus analysis, however showed a strong association and an opposite

direction with PsA compared to psoriasis in the multivariate analysis. The small number of

patients in each discordant group (patients having a copy of either KIR2DS2 or KIR2DL2)

accounts for the broad confidence intervals that were observed in the multivariate analysis

for these co-variates.

With regards to KIR2DS1, in the presence of its ligand HLA-C group 2, KIR2DS1 had a

similar frequency in PsA as in psoriasis alone. However, the presence of KIR2DS1 with its

HLA ligand was associated with PsD compared to healthy controls, while KIR2DS2 and its

ligand were not associated with PsD (Table 7.7).

7.3.5. The interaction of KIR3DS1 with HLA-Bw4-80I group

The frequencies of KIR3DS1 were similar in the three groups (37.5% in the PsA group,

41.1% in the psoriasis group and 36.8% in the controls, p=0.16). The frequencies of carriers

of HLA-Bw4-80I were higher in the PsA and the psoriasis groups compared to the controls

(38.7% in the PsA, 39.4% in the psoriasis and 28% in the controls), but there was no

difference between the psoriasis and the PsA groups (p=0.24). The frequencies of KIR3DS1

were also similar among HLA-Bw4-80I positive individuals, suggesting that the interaction

between HLA-Bw4-80I alleles and KIR3DS1 is not associated with the susceptibility to PsA

(Table 7.8). These findings were confirmed in a regression analysis (Table 7.9). The

combination of HLA-Bw4-80I and KIR3DS1 was not significantly associated with PsA

compared to psoriasis in either the univariate or the multivariate regression model that

included HLA-B*27, HLA-C*06 and KIR3DL1. The presence of KIR3DS1 without its

corresponding HLA ligand showed an inverse association of borderline significance with

PsA.

Page 131: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

119

Table 7.2 - KIR gene distribution – Psoriatic disease vs. Healthy controls

KIR PsD (N=1043) Controls (N=707) OR 95% CI P value

2DL1 1020

(97.8%) 684 (96.8%) 1.49 0.83-2.67 0.18 2DL2 548 (52.5%) 336 (47.5%) 1.22 1.00-1.48 0.04

2DL3

963 (92.3%) 634 (89.7%) 1.38 1.00-1.93 0.05

2DL4 1040 (99.8%) 706 (99.9%) 0.74 0.07-8.13 0.80

2DL5 524 (50.3%) 344 (48.7%) 1.06 0.88-1.29 0.49

2DS1 433 (41.5%) 271 (38.3%) 1.14 0.93-1.38 0.18

2DS2 553 (53.0%) 346 (48.9%) 1.17 0.97-1.42 0.09

2DS3 293 (28.1%) 183 (25.9%) 1.11 0.90-1.39 0.30

2DS4 987 (94.6%) 674 (95.3%) 0.86 0.56-1.34 0.51

2DS5 328 (31.3%) 213 (30.2%) 0.94 0.86-1.31 0.62

3DL1 988 (94.8%) 674 (95.3%) 0.89 0.57-1.39 0.62

3DL2 1042 (100%) 707 (100%) 3DL3 1040 (99.9%) 707 (100%) 1.00

3DS1 403 (38.6%) 260 (36.8%) 1.08 0.89-1.32 0.43

Table 7.3 - KIR gene distribution – PsA vs. Psoriasis

KIR

PsA (N=710) Psoriasis (N=333)

OR 95% CI P value

2DL1 689

(97%) 331 (99.4%) 0.19 0.04-0.85 0.02 2DL2 378 (53.2%) 170 (51.1%) 1.09 0.84-1.41 0.51

2DL3

649 (91.4%) 314 (94.3%) 0.64 0.38-1.09 0.10

2DL4 708 (99.9%) 332 (99.7%) 2.13 0.13-31.2 0.58

2DL5 353 (49.8%) 171 (51.5%) 0.93 0.72-1.21 0.60

2DS1 294 (41.1%) 139 (41.9%) 0.98 0.75-1.28 0.92

2DS2 389 (54.8%) 164 (49.3%) 1.25 0.96-1.62 0.09

2DS3 197 (27.8%) 96 (28.8%) 0.94 0.71-1.25 0.72

2DS4 671 (94.5%) 316 (94.9%) 0.92 0.51-1.66 0.79

2DS5 218 (30.7%) 109 (32.7%) 0.90 0.69-1.18 0.46

3DL1 671 (94.5%) 316 (95.5%) 0.81 0.44-1.79 0.51

3DL2 709 (100%) 333 (100%) 3DL3 709 (100%) 331 (99.7%) 0.31

3DS1 266 (37.5%) 137 (41.1%) 0.85 0.66-1.12 0.25

Page 132: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

120

Figure 7.2 - The 10 most frequent KIR genotypes in PsA, psoriasis and healthy controls. A black box denotes the presence of at least one copy of the gene.

Table 7.4 - The frequency of HLA-C group 1/HLA-C group 2 alleles in PsA, psoriasis and

healthy controls

Group PsA (N=710)

Psoriasis (N=331)

P value PsA vs. Psoriasis

Controls (N=707)

P value PsA vs. Controls

HLA-C group 1

alleles

600 (84.5%)

261 (78.9%)

0.02 619

(87.6%) 0.11

HLA-C group 2

alleles

457 (64.4%)

236 (71.3%)

0.03 409

(57.9%) 0.01

2DL 2DS 3DL

1 2 3 4 5 1 2 3 4 5 1 2 3 3DS1

230 (32.5%) 203 (28.6%) 103 (30.9%)

104 (14.7%) 118 (16.6%) 48 (14.4%)

95 (13.4%) 70 (9.9%) 43 (12.9%)

52 (7.4%) 55 (7.6%) 25 (7.5%)

32 (4.5%) 34 (4.8%) 17 (5.1%)

26 (3.7%) 33 (4.7%) 22 (6.6%)

22 (3.1%) 30 (4.2%) 16 (4.8%)

17 (2.4%) 14 (2%) 4 (1.2%)

10 (1.4%) 11 (1.6%) 5 (1.5%)

9 (1.3%) 9 (1.3%) 6 (1.8%)

110 (15.5%) 134 (18.8%) 44 (13.2%)Remaining genotypes

Psoriasis

(n=707) (n=710) (n=333)

Control PsA

Page 133: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

121

Table 7.5 – The association between combinations of KIR2D/HLA based on activation

model and PsA (N=710) vs. Psoriasis (N=331)

KIR/HLA Univariate analysis Multivariate Reduced

Model

OR 95% CI P value OR 95% CI P value

KIR2DL1 0.2 0.04-0.9 0.03 0.2 0.05-0.9 0.03

KIR2DL2 0.5 0.8-1.4 0.51 0.08 0.02-0.4 0.001

KIR2DL3 0.6 0.4-1.1 0.11

KIR2DS1 with HLA-C group 2

KIR2DS1 without HLA-C group 2

KIR2DS2 without HLA-C group

0.9 0.7-1.3

0.75

1.1

1.0

0.8-1.6

0.6-1.6

0.58

0.95

11.5

2.4-55.0

0.002

KIR2DS2 with HLA-C group 1 1.4 1.1-1.8 0.02 16.3 3.6-74.1 0.0003

HLA-B*27 5.1 2.9-8.7 <0.0001 5.2 3.0-9.1 <0.0001

Table 7.6 - The frequencies of KIR2DS2 and KIR2DL2 in PsA and psoriasis

KIR2DS2 KIR2DL2 PsA

(N=710)

Psoriasis

(N=333)

+ + 376 (53%) 164 (49.2%)

+ - 13 (1.8%) 0 (0%)

- + 2 (0.3%) 6 (1.8%)

- - 319 (44.9%) 163 (49%)

+ The patient carries at least one copy of the gene. - The patient does not carry the gene

Page 134: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

122

Table 7.7 - The association between combinations of KIR2D/HLA based on activation model

and PsD (N=1041) vs. Controls (N=707)

KIR/HLA Univariate analysis Multivariate Reduced

Model OR 95% CI P value OR 95% CI P value

KIR2DL1 1.5 0.8-2.3 0.18

KIR2DL2 1.2 1.01-1.5 0.04 1.3 1.1-1.6 0.006

KIR2DL3 1.4 0.99-1.9 0.05 1.7 1.2-2.4 0.003 KIR2DS1 with HLA-C group2

KIR2DS1 without HLA-C group2

KIR2DS2 without HLA-C group 1

0.003

1.4

0.8

1.3

1.1-1.8

0.6-0.99

0.9-1.9

0.002

0.04

0.15

1.4

1.1-1.8

KIR2DS2 with HLA-C group1 1.1 0.9-1.3 0.38

HLA-B*27 2.2 1.6-3.0 <0.000 2.1 1.5-3.0 <0.0001

Table 7.8 - The frequency of KIR3DS1 in PsA and psoriasis

Group PsA

(N=709)

Psoriasis

(N=331)

Healthy Controls

(N=707)

P value

(trend)

Total 266 (37.5%) 137 (41.1%) 260 (36.8%) 0.17

HLA-Bw4-I80 (positive) 104 (38.1%) 54 (41.2%) 74 (37.6%) 0.78

HLA-Bw4-I80 (negative) 162 (37.1%) 83 (41.1%) 186 (36.5%) 0.5

Page 135: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

123

Table 7.9 - The association between combinations of KIR3D/HLA based on activation model

and PsA (N=709) vs. Psoriasis (N=331)

KIR/HLA Univariate analysis Multivariate Reduced Model

OR 95% CI P value OR 95% CI P value

KIR3DL1 0.8 0.4-1.5 0.51

KIR3DS1 without HLA-Bw4-80I

0.8 0.6-1.1 0.12 0.7 0.5-0.96 0.03

KIR3DS1 with HLA-Bw4-80I

HLA-C*06

HLA-B*27

1.1 0.7-1.6 0.73

0.5

5.1

0.4-0.6

2.9-8.8

<0.0001

<0.0001

0.5

4.7

0.4-0.7

2.7-8.2

<0.0001

<0.0001

Page 136: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

124

7.4. Discussion

In this study, the association between the KIR genes and PsA was investigated. In addition,

the gene-gene interaction between selected activating KIRs and their HLA ligands in the

susceptibility to PsA was assessed. Several “functional” models that represent the likely

outcomes of HLA /KIR interactions were tested.

HLA class I genes are strongly associated with susceptibility to PsA and psoriasis [153].

HLA molecules are expressed on target cells and play an important role in the activation of

NK cells through their interaction with KIR molecules. The activation is dependent on the

combination of variable KIRs and HLA-class I molecules [226]. It has been suggested that

certain combinations of receptor-ligand may result in altered NK cell mediated immunity

against pathogens which may in turn lead to autoimmunity [303, 306].

Our results show that the frequency of the inhibitory KIR2DL1 was lower in PsA patients

compared to those with psoriasis alone. However, this gene was observed in nearly all

individuals, therefore, this difference can only account for a small fraction of the risk.

KIR2DS2 was found to be associated with PsA compared to healthy controls. However,

although it was more frequent in the PsA group compared to the psoriasis patients, the

association was not significant. This may be due to the existence of a weak association since

the study was not powered to detect an association with an OR of less than 1.5. The direction

of the association was in accordance with our expectation: the activating KIR2DS2 was more

frequent in PsA while the inhibitory KIRs were less frequent in PsA compared to psoriasis.

In the literature, KIR2DS1 and KIR2DS2 have been associated with susceptibility to

psoriasis and PsA. It has been reported that KIR2DS1 is associated with psoriasis compared

to healthy controls among Japanese [229] and Caucasians from Brazil, Sweden and Poland

[230, 307, 308]. These results concur with our findings in the multivariate analysis that

showed that KIR2DS1 in the presence of its HLA-C group 2 ligand is associated with

psoriatic disease, suggesting that the strongest association of this gene is with the skin and

not the joint disease.

Page 137: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

125

Two models of gene-gene interaction were investigated in this study. The combination of

HLA-Bw4-80I alleles and KIR3DS1 gene was found not to be associated with PsA or PsD.

This interaction has not been investigated in PsA previously. However, it was suggested to be

important in the susceptibility to AS. Two studies, one of Caucasians from Spain and the

other, Chinese people in China, have found a higher frequency of KIR3DS1 with a lower

frequency of the inhibitory KIR3DL1 among AS patients [234, 309, 310]. However, a large

study from the UK reported a lack of an association between KIR3DS1 and AS [235]. In our

study, KIR3DS1 by itself was not associated with PsA or with PsD. Possible explanations for

the lack of association with KIR3DS1 are that only a small proportion (less than 20%) of our

PsA patients are HLA-B*27 positive, as opposed to almost all of the AS patients in these

studies. Additional possible reasons include: an effect of different alleles encoded at other

KIR loci, expression of different combinations of KIRs encoded within the genome,

differences in populations and type I error.

Another interaction that was tested was between KIR2DS2 and 2DS1 and their

corresponding HLA-C ligands. It was hypothesized that the association with the activating

KIRs would be greatest in the presence of their HLA ligands. A significant association was

found between KIR2DS2 in the presence of its ligand HLA-C group 1, in which higher

frequencies were found in the PsA group, compared to psoriasis alone. However, that gene

combination was not associated with PsD, while KIR2DS1 in the presence of its ligand

HLA-C group 2 was associated with PsD. These associations may indicate that KIR2DS2 is a

susceptibility gene for PsA among psoriasis patients, while KIR2DS1 is associated only with

the skin disease. This model is supported by the “dual” associations that were found for

HLA-C*06 and PsA: a risk factor for psoriasis alone but an inverse association (“protective”

effect) for PsA compared to psoriasis. Since HLA-C*06 is included in HLA-C group 2 that is

the ligand for KIR2DS1, its interaction with KIR2DS1 may be important in the susceptibility

to psoriasis. However, since psoriasis patients who carry HLA-C*06 are less likely to have

HLA-C group 1, simply because one of their HLA-C alleles already belongs to HLA-C group

2, they are less likely to have a compatible ligand for the KIR2DS2 that may be a risk gene

for PsA among psoriasis patients.

Page 138: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

126

In the previous literature, several models of interaction between KIR2DS and their HLA

ligands were assessed for susceptibility to PsA. Martin et al. suggested a different model

where these activating KIRs conferred risk for PsA when HLA ligands for the corresponding

inhibitory KIRs were missing [227]. This model was found to be associated with PsA

compared to healthy controls. They hypothesized that activating KIR molecules bind poorly

to HLA molecules compared with that observed for inhibitory KIR and that the ligands for

the KIR2DS molecules are not the same HLA molecules recognized by homologous

KIR2DL receptors. In our model we hypothesized that the activating KIRs bind the same

ligands as their corresponding inhibitory KIRs. Although the association between the

activating KIRs and the HLA class I molecules is weaker than that for the inhibitory KIRs, it

has been suggested that a high level of HLA class I expression, such as occurs under

conditions of IFN-γ production associated with infections, may allow binding of activating

KIRs. Furthermore, it has been shown that under these conditions, KIR2DS1 binds to the

HLA-C allele group of its corresponding inhibitory KIRs and leads to activation of NK cells

[304, 305].

The activating KIR2DS are attractive candidates for exerting a direct effect on pathogenesis,

as they interact with several HLA-C alleles that are strongly associated with psoriasis and

PsA. However, to date, the activating KIRs have not been identified as susceptibility genes

for psoriasis by genome-wide scans. This might be attributable to the complex relation

between HLA and other KIRs, as very large sample sizes are required to determine a

statistical interaction. Additional studies, including functional studies, will be required to

determine the relation between KIR2DS2/2DS1 and PsA.

Activation of NK cells that is mediated through binding of activating KIRs can explain

several diseases that are associated with KIRs and HLA genes. It has been shown that the

combination of HLA-Bw4 alleles containing Ile-80 and KIR3DS1 are associated with a

slower progression of AIDS [233, 236] and type I diabetes is associated with KIR2DS2 in the

presence of HLA-C group 1 [311]. Selective tissue expression of HLA-class I genes may

explain the association of the different diseases with certain gene combinations. Another

explanation is that the peptide preferences of KIR–HLA interactions may define which

combinations are important in particular tissues. In addition, the mechanism of activation of

Page 139: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

127

NK cells is complex and is not mediated only through the interaction of KIR/HLA. NK cells

carry additional receptors including Leukocyte Immunoglobulin Like Receptors (LILR) and

CD94/NKG2 that mediate signal transmission that lead to activation of NK cells [312].

Furthermore, the complexity of KIR is compounded by allelic diversity and each KIR gene

has between four to nineteen alleles. These polymorphisms were not assessed in our study

but can affect NK cell activation.

The KIR gene diversity is related to genetic mechanisms that have led over many generations

to different combinations of KIR genes as well as allelic polymorphism. This diversity in

different human populations results in various activation and inhibition signaling potentials

that when combined with HLA diversity may be related to neutral selection in response to

exogenous stimuli [306]. Since the activating KIRs are present in approximately half of the

population, they may have an evolutionary advantage that may be related to a better ability of

the immune system to fight infections. This advantage was demonstrated in the protection of

KIR genes against AIDS progression. On the other hand, since KIRs may increase the

susceptibility to immune-mediated diseases, such as PsA, that are not associated with

mortality at young age, they continue to exist in the population.

Page 140: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

Chapter 8. The association between environmental factors and onset of PsA in patients with psoriasis

Page 141: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

129

8.1. Background

In the majority of the patients with PsA, inflammatory arthritis develops on average 7 years

after the onset of the skin disease [35]. PsA is a complex disease with genetic and

environmental risk factors playing a major role in disease susceptibility [73]. Genetic factors

alone cannot account for all cases of PsA. One of the suggested pathogenic models for PsA is

that psoriasis patients who carry susceptibility genes for arthritis develop PsA after exposure

to triggering environmental factors [74]. Suggested factors include physical and emotional

trauma, infections and hormonal changes[75]. A Few studies have systematically assessed

environmental risk factors for PsA among psoriasis patients and these showed conflicting

results [76, 77]. The objective of this case-control study is to investigate the association

between several putative environmental risk factors and the occurrence of PsA among

psoriasis patients.

Page 142: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

130

8.2. Methods

8.2.1. Study population

Cases – This group included adults with recent onset PsA that satisfied the CASPAR

classification criteria [22] selected from the University of Toronto PsA cohort. For the

purpose of this study, I identified all patients with PsA of less than 7 years duration since

diagnosis. I attempted to contact all patients in the database that satisfied the study criteria,

either during their follow-up visit in the clinic, or by telephone or mail.

Controls: This group included psoriasis patients without arthritis that were recruited from the

Toronto Psoriasis Cohort, which enrolls psoriasis patients uncomplicated by arthritis and

aims to study risk factors for PsA. These patients are assessed by a rheumatologist initially to

rule out the presence of inflammatory arthritis, and then annually for symptoms or signs of

inflammatory arthritis. If inflammatory arthritis is diagnosed, the subject is considered to

have developed the outcome of interest and is censored. This process ensures that all of the

psoriasis cohort subjects are free of arthritis. For this study, the controls were recruited

consecutively upon their visit to the clinic for their annual follow-up visit.

8.2.2. Data collection

Demographic and clinical data were available from the cohorts’ computerized databases.

Severity of psoriasis was assessed by the highest PASI in the first 3 years of follow-up.

Severe psoriasis was defined as PASI≥10.

Based on literature that has suggested that different environmental exposures are associated

with PsA, a broad range of potential triggering factors was investigated. A questionnaire for

the assessment of these exposures was designed. The study population was asked to report

whether they have had any of the environmental exposure events under investigation in the

previous 10 years. Patients were asked to specify for each event, the year of occurrence,

whether they required medical consultation or if they were admitted to hospital (See

Appendix 1). The following exposures were recorded:

Page 143: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

131

1. Physical trauma that included injuries, fractures and road traffic accidents.

2. Infections that required antibiotic treatment and infectious diarrhea.

3. Vaccinations to hepatitis A and B, influenza, pneumococcus, rubella and tetanus.

4. Emotional stress, including death of a close family member, divorce or separation,

moving house, change of job, becoming unemployed and treatment for anxiety or

depression.

5. Female hormonal exposures – ages of first and last menstrual periods, use of

hormonal contraception and hormone replacement therapy, fertility treatment and

pregnancies.

6. Occupational exposures – I hypothesized that recurrent micro-trauma that are related

to certain occupations may be associated with PsA. The literature was searched for

studies that assessed the association between several occupational tasks and

musculoskeletal problems. The cut-off levels that were found to increase the risk of

these problems were chosen [313]. Only subjects that have been employed in the last

10 years were asked to report exposures to the following: prolonged standing of more

than 30 minutes/hour, squatting for more than 5 minutes/hour, lifting cumulative

heavy loads of more than 100 pounds/ hour, pushing cumulative loads of more than

200 pounds/hour, using vibrating tools for more than 4 hours/day, frequent repetitive

hand movements of more than 45 minutes/hour, forceful gripping of more than 4

hours/day and bending of the wrist for more than 4 hours/day .

7. Smoking status – If a person smoked at least one cigarette a day for at least one year

before the diagnosis of PsA or before the date of assessment for the psoriasis only

group, they were defined as “ever smoked”.

8. Alcohol – Alcohol consumption was categorized as social, daily, or non-drinker

before the diagnosis of PsA or before the date of assessment for patients in the

psoriasis group.

Page 144: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

132

Recall bias is a potential problem in this type of study. I expected patients with arthritis to

recall more environmental events, since patients may link these events to the onset of

arthritis. In order to minimize this bias, the environmental events were not related explicitlyto

the onset of the arthritis, but the participants were simply asked to report about events that

occurred within the 10 years prior to the assessment date and thus a linkage between the

onset of arthritis and exposure was avoided. Only exposures that occurred before the onset of

arthritis were considered. In order to ensure that cases and controls had equal duration of

time for potential exposures, a reference date was assigned for each control that was derived

from the date of onset of arthritis for the corresponding matched case having a similar

duration of psoriasis. Thus the matching ensured only a similar time period at risk of

exposure and therefore potential confounding variables were controlled for through

regression analysis.

8.2.3. Statistical analysis

Baseline descriptive statistics were computed with continuous variables summarized by their

means and standard deviations and categorical variables summarized by proportions. The

proportions of patients that were exposed to the different types of environmental factors

before the reference date were calculated. For several of the exposures, mostly in the

vaccination section, a significant proportion of the responses were “don’t know” or missing.

Missing variables were assigned a distinct code. This process allowed us to avoid a selection

bias by excluding those individuals from the full regression model. Since there were too

many environmental factors for inclusion as co-variates in the full regression model I

performed a “screening” step. This step aimed to detect the most significant exposures (p

value <0.1) after adjusting for age, sex, level of education, severity and duration of psoriasis,

and included only those in the final logistic regression model. I then constructed a logistic

regression model that included all of the exposures that were found to be significant in the

“screening” step adjusting for the same confounders. Logistic regression analysis was used to

identify key factors predictive of PsA. Covariate effects were considered statistically

significant if the p-value from the 2-sided Wald-test was less than 0.05. As this study was

considered to be explorative, no adjustment for multiple testing was applied.

Page 145: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

133

8.3. Results

8.3.1. Study population characteristics

One hundred and ninety PsA patients that satisfied the study criteria were identified through

a search in the computerized PsA database. I attempted to contact all of them, however, I was

unable to contact 29 (6 deceased) and 2 were excluded due to poor English language skills.

Overall, 159 patients from each group were included in the study (83.6% for the PsA group).

I then aimed to recruit a similar number of psoriasis patients. A total of 196 psoriasis patients

was approached for enrollment of whom 159 (81.1%) completed the questionnaire.

The demographic and clinical characteristics of the study population are presented in Table

8.1. The cases and controls had a similar mean duration of psoriasis and male to female ratio.

Level of education, often associated with socioeconomic status, was also similar in the two

groups.

The psoriasis group was slightly older than the PsA group (by a mean of 3.5 years). The

mean duration of PsA at the time of assessment was relatively short, 3.1 ± 2.2 years, which

indicates the recent onset of the disease and minimizes problems due to recall. A mean time

window of 6.9±2.2 years prior to the onset of arthritis and a comparable time period for the

controls were captured (range 2 to 10 years).

Surprisingly there was not a significant difference in psoriasis severity between the PsA and

psoriasis patients. This may be explained by the fact that many of the psoriasis patients were

reviewed after several phototherapy treatments which may have improved their skin scores,

while many of the PsA patients were reviewed prior to starting therapy at a time when their

PASI scores were higher.

8.3.2. Proportion of exposure to the different factors

The proportions of patients that were exposed to different environmental factors are

presented in Table 8.2.

Page 146: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

134

8.3.3. Smoking and alcohol consumption

The proportion of smokers (current and past) was lower in the PsA group compared to the

psoriasis group (40.2% vs. 56%, respectively), leading to a significant inverse association

between smoking and PsA (OR 0.6, 95% CI 0.36-0.88). The ORs for past smokers (OR 0.52,

95% CI 0.31-0.88, p=0.015) and currents smokers (OR 0.54, 95% CI 0.31-0.96, p=0.038)

compared to those who never smoked were similar.

The proportions of social, daily and non-drinkers were found to be similar in the PsA and

psoriasis groups.

8.3.4. Occupational exposures

Only the 148 PsA and 146 psoriasis patients who were employed during the 10 years prior to

the assessment date were evaluated. Lifting cumulative heavy loads of at least 100

pound/hour was more common in the PsA group (30.1% vs. 13%, OR 2.8, 95% CI 1.48-

4.98). The following occupational tasks were more common in the PsA group: squatting for

at least 5 minutes/hour (30.8% vs. 21.9%, 95% CI 0.92-2.66, p=0.1) and pushing cumulative

heavy weights of more than 200 pounds/hour (19.8% vs. 12.3%, 95% CI 0.83-3.06, p=0.16).

However, as noted by the p values, none of these differences reached statistical significance

at the 5% level. No particular pattern of joint involvement was associated with carrying

heavy loads.

8.3.5. Infections

A history of infectious diarrhea in the exposure time window under study was more common

in the PsA group however the difference did not reach statistical significance (12.7% vs. 7%,

p=0.19). Other infections that required antibiotic treatment were significantly associated with

PsA (OR 1.7, 95% CI 1.01-2.78, p=0.19). In addition, the proportion of severe infections that

required hospitalization was significantly higher in the PsA group during the exposure time

window under study (7.2% vs. none in the psoriasis group, p<0.001). The types of infections

Page 147: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

135

reported in the PsA group were lower respiratory tract infection (11%), upper respiratory

tract infection (22.2%), urinary tract infection (8.3%), skin and soft tissue infections (25%),

sinus infection (8.3%), sepsis (2.8%), hepatitis C or B (8.3%) and other type of infections

(13.9%). There was no significant difference in the types of infections between the psoriasis

and the PsA group.

8.3.6. Injuries

There was no difference between the cases and control groups in the proportions of subjects

that had road traffic accidents and fractures. However, more PsA patients reported having

other types of injuries (21.4% vs. 10.7%, OR 2.1, 95% CI 1.13-4.01, p=0.02). The types of

injuries in the PsA group were very variable and included cuts, falls, burns, blunt and

penetrating trauma, and affected all body areas, often several body parts at once. Therefore, it

was difficult to analyze the correlation between the site of injury and the joint of onset of the

arthritis. Polyarticular pattern was more common among patients with a history of infection

(74%) compared to those without (58.6%). There was a trend for an association between a

history of infection and a polyarticular pattern at presentation (OR 2, 95%CI 0.95-4.25,

p=0.06). Therefore, it is unlikely that there was a significant misclassification with reactive

arthritis, as the typical pattern of this type of arthritis is oligoarticular.

8.3.7. Vaccination

Since vaccinations were reported to be associated with PsA in a previous study, I included

this exposure in the analysis although up to 30% of the information was missing. The

proportion of patients with missing information was similar in the cases and the controls. I

analyzed the data twice, first by excluding the patients with the missing information from the

analysis and again by considering the missing data as “no exposure” to the particular

vaccination. There were no significant changes in the OR of the two analyses. The following

vaccinations were assessed: hepatitis A and B, pneumococcus, rubella and tetanus. There

were no statistically significant differences in the proportions of exposures to the different

Page 148: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

136

types of vaccinations in the two groups during the exposure time window under study after

adjusting for potential confounders.

8.3.8. Emotional stress

Exposures to the following potential emotional stressful events were assessed: death of a

close family member, divorce or separation, moving house, changing job, becoming

unemployed and treatment for depression or anxiety. None of these factors was found to be

associated with PsA in the univariate or multivariate analyses.

8.3.9. Female hormonal exposures

Overall 73 psoriasis and 68 PsA female patients completed the female hormonal

questionnaire. The following events were investigated: age at first menstrual period,

becoming menopausal, use of oral contraceptive pills, use of hormone replacement therapy

and pregnancy. The results are presented in Table 8.3. None of these exposures were

significantly associated with PsA in the univariate or multivariate analyses.

8.3.10. Multivariable logistic regression analysis

The exposures that were found to be associated with PsA after adjustment for age, sex, level

of education, severity and duration of psoriasis were included in a full logistic regression

model to test their independent effect. The results of the analysis are presented in Table

8.4.The following variables remained significantly associated with PsA: smoking (OR 0.47,

95% CI 0.29-0.77), occupations that required lifting heavy weights (OR 2.92, 95% CI 1.56-

5.46) and infections that required antibiotic treatment (OR 1.72, 95% CI 1.01-2.95). Having a

history of an injury was of borderline significance after inclusion in the full logistic

regression model (OR 1.98, p=0.054).

Page 149: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

137

Table 8.1 - Clinical characteristics of the study population

Variable PsA (N=159) Psoriasis (N=159) P value

Age* (Years ± SD) Sex: Male (%) Ethnicity - Caucasian Family history of psoriasis (%) Family history of PsA (%) Education status (%)

- College/University Duration of psoriasis (Years ± SD) Duration of PsA (Years ± SD) Pattern of arthritis at diagnosis (%)

- Polyarthritis - Oligoarthritis - Spine+polyarthritis - Spine+oligoarthritis - Distal (DIP,PIP) only

Maximal PASI (in the first 3 years of follow-up) Severe Psoriasis (%)

44.9±13.1 89 (55.9%)

124 (81.6%) 70 (44.6%) 13 (8.2%)

122 (77.7%)

17.2±13 3.1±2.2

57 (42.7%) 40 (29.8%) 22 (16.4%) 12 (8.9%) 3 (2.2%) 7.3±9.6

33 (20.9%)

48.4±13.3 86 (54.1%)

137 (86.2%) 71 (44.9%)

5 (3.1%)

127 (79.9%) 18.6±14.5

NA

NA

7.1±7.2

34 (21.4%)

0.02

0.08 0.91 0.09

0.58 0.36

0.83

0.89

* At first visit to the clinic

Page 150: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

138

*Adjusted for age, sex, duration and severity of psoriasis, level of education. OR- Odds ratio, CI – Confidence Interval,

highlighted rows indicate covariates achieving statistical significance.

Table 8.2 – The association between environmental exposures and PsA (N=318)

Exposure Frequency

PsA

Frequency

Psoriasis

OR

95% CI

p

value

Adjusted

p value*

Car accidents that required medical treatment 6 (3.8%) 5 (3.2%)

1.1 0.32-3.74 0.79 0.89

Fractures 16 (10.1%)

14 (8.9%) 1.2 0.54-2.51 0.72 0.69

Injuries that required medical attention

30 (18.9%) 17 (10.7%) 1.8 0.95-3.49 0.04 0.07

Any other injury 34 (21.4%) 17 (10.7%) 2.1 1.13-4.01 0.01 0.02

Infectious diarrhea 20 (12.7%) 11 (7%) 1.7 0.76-3.79 0.09 0.19

Infections that required Antibiotic

treatment*

53 (34.4%) 37 (23.7%) 1.8 1.01-2.78 0.04 0.05

Infections that required hospitalization 11 (7.2%) 0 (0%) <0.001

Vaccination –Hepatitis A 3 (2.5%) 4 (3.6%) 0.8 0.14-3.07 0.63 0.59

Vaccination – Hepatitis B

29 (22.6%) 19 (16.2%) 1.4 0.75-2.79 0.21 0.27

Vaccination – Pneumococcus 30 (23.6%) 20 (17.1%) 1.4 0.74-2.71 0.51 0.29

Vaccination - Flu 59 (41.5%) 60 (44.7%) 1.0 0.60-1.61 0.58 0.94

Vaccination - Rubella 4 (2.9%) 5 (3.9%) 0.9 0.22-3.46 0.68 0.84

Vaccination - Tetanus

6 (4.8%) 4 (3.7%) 1.0 0.26-3.84 0.66 0.99

Death in the family 41 (26.8%) 44 (28.9%) 1.1 0.63-1.79 0.67 0.82

Divorce/Separation 17 (10.9%) 12 (7.7%) 1.5 0.69-3.35 0.33 0.29

Move house 74 (50%) 64 (43%) 1.1 0.68-1.82 0.24 0.68

Changed job 70 (46.7%) 56 (40%) 1.2 0.73-2.06 0.25 0.44

Becoming unemployed 34 (22.7%) 35 (22.8%) 0.9 0.54-1.66 0.96 0.85

Treated for anxiety/depression 21 (13.9%) 26 (16.8%) 0.8 0.39-1.46 0.47 0.41

Occupational tasks-

- Prolonged standing (>30 min/Hr)

95 (65.7%) 85 (58.2%) 1.3 0.77-2.05 0.22 0.34

- Squatting (>5 min/Hr) 45 (30.8%) 32 (21.9%) 1.7 0.92-2.66 0.09 0.10

- Lifting heavy weights (>100 pound/Hr) 44 (30.1%) 19 (13%) 2.8 1.48-4.98 <0.001 0.001

- Pushing heavy weights (>200 pound/Hr) 29 (19.8%) 18 (12.3%) 1.7 0.83-3.06 0.08 0.16

- Using vibrating tools (>4 Hr/Day) 7 (4.8%) 8 (5.5%) 0.8 0.27-2.32 0.79 0.67

- Repetitive hand movement (>45 min/Hr) 88 (6%) 85 (5.8%) 1.1 0.65-1.69 0.72 0.83

- Forceful gripping (>4 Hr/Day) 28 (19.2%) 21 (14.4%) 1.0 0.64-2.27 0.27 0.56

- Bending the wrist (>4 Hr/Day) 65 (44.5%) 65 (44.5%) 0.9 0.59-1.53 1.00 0.84

Smoking –

- Ever vs. Never Alcohol consumption

- Social vs. None - Daily vs. None

64

88 13

(40.2%)

(28.2%) (4.2%)

89

83 11

(56.0%)

(26.6%) (3.5%)

0.6

0.8 0.8

0.36-0.88

0.49-1.31 0.34-2.03

0.005

0.45 0.52

0.01

0.39 0.68

Page 151: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

139

Table 8.3 – The Association between female hormonal exposures and PsA

Exposure

PsA (N=68)

Psoriasis (N=73)

OR

95% CI

p value

Adjusted p value*

Age at first period 12.5±1.6

12.7±1.6

0.85

Becoming post-menopausal 13/19 (68.4%)

12/20 (60%)

1.8 0.31-11.49 0.58

0.50

Using oral OCT 24 (35.3%)

26 (35.6%)

1.6

0.71-4.24

0.96

0.23

Using HRT 9 (13.2%)

10 (13.7%)

0.7 0.26-2.33

0.93 0.66

Pregnancy Fertility treatment

13 (19.1%)

5 (7.5%)

13 (17.8%)

7 (9.6%)

1.2

0.9

0.47-2.90

0.26-2.98

0.84

0.63

0.73

0.83

OR – Odds ratio, HRT – Hormone Replacement Therapy OCT - Oral Contraceptive Therapy, CI – Confidence Interval *Adjusted for age, duration and severity of psoriasis and education level.

Page 152: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

140

OR – Odds ratio, CI- Confidence Interval, highlighted rows indicate covariates achieving statistical significance

Table 8.4 - Full regression model adjusted for age, sex, education level, duration and severity

of psoriasis

Exposure

OR

95% CI

P value

Smoking status – Ever vs. Never 0.47 0.29-0.77 0.003

Any injury 1.98 0.99-3.96 0.054

Occupational task- Lifting heavy weight 2.92 1.56-5.46 0.0008

Infection that required antibiotic treatment 1.72 1.01-2.95 0.046

Duration of psoriasis (1 year increase) 0.99 0.98-1.01 0.63

Age (1 year increase) 0.99 0.97-1.01 0.29

Sex (Female) 0.86 0.53-1.42 0.57

Severe Psoriasis (PASI>10) 0.89 0.49-1.61 0.70

Level of education (College/University vs. else) 1.18 0.66-2.13 0.56

Page 153: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

141

8.4. Discussion

In this study the association between a number of a priori chosen environmental factors and

PsA among patients with psoriasis was investigated. The results suggest that there is an

association between PsA and infections, physical trauma including physically demanding

occupational tasks, and smoking.

Only a limited number of studies have investigated environmental risk factors for PsA.

Pattison et al. compared the prevalence of environmental exposures among 98 British PsA

and 163 psoriasis patients over a window of exposure that ranged from 5 to 10 years prior to

the onset of arthritis. Information about environmental exposures was collected through

questionnaires. In their study, physical trauma, rubella vaccination, oral ulcers and moving

house were found to be associated with PsA [76]. Another study by Thumboo et al. used an

administrative database from Rochester, Minnesota and retrospectively evaluated exposure to

several environmental risk factors among 60 PsA and 120 psoriasis patients [77]. They did

not define a specific window of exposure. In that study, pregnancy was found to be

protective of PsA, while steroid use was associated with higher risk of the disease. These

studies had several limitations; first, their small sample size precluded identifying important

associations due to reduced power. The use of an administrative database as a source of

information may have led to underestimation of events that are often not coded or are coded

incorrectly. Pattison’s study included controls from a dermatology clinic while the PsA

patients were recruited from their rheumatologists or by advertisement. The controls were not

evaluated to exclude the presence of PsA. It has been shown that PsA is underestimated

among psoriasis patients, as approximately 18% of the psoriasis patients that were found to

have inflammatory arthritis were unaware of their condition and had never been seen by a

rheumatologist [119]. The problem of misclassification of patients and controls may

significantly affect the results in case-control studies where the outcome (PsA in this case) is

not rare among the control group. Misclassification can decrease the power of the study to

detect significant associations. In the present study, I aimed to minimize the misclassification

of cases and controls by having a rheumatologist evaluate all of the controls to rule out

clinical inflammatory arthritis. The cases and controls were recruited from the same program,

and were evaluated according to the same protocol.

Page 154: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

142

Several case reports and case series have highlighted the potential role of local trauma in the

pathogenesis of PsA [80, 314, 315]. However, only a few studies have systematically

assessed its role in PsA. Scarpa et al.[118] reported more acute medical events in the PsA

group, including injuries that occurred less than 10 days before the onset of the arthritis,

compared to the group with rheumatoid arthritis. Pattison et al.[76] found that the

occurrence of an injury was more common in patients with PsA, compared to psoriasis

patients. However, Thumboo et al. did not find an association between trauma and PsA[77].

In our study, patients with PsA had double the risk of experiencing injuries in the time

window under study compared to psoriasis patients. However, in the full regression model,

that association became borderline and there was a small reduction in effect size after the

inclusion of injuries in the full regression model, suggesting that injuries were related to one

of the other risk factors in the model. The most likely candidate is physically demanding

occupations that involved lifting heavy weights, because when that covariate was excluded

from the model in order to include those individuals that did not work, injuries were found to

be associated with PsA (data not shown).

Work related physical activity can lead to musculoskeletal disorders and specific

occupational tasks can lead to osteoarthritis [316, 317]. The role of occupational related

physical activities in the pathogenesis of PsA has not been thoroughly investigated. In

ankylosing spondylitis, occupations that require recurrent bending, twisting and stretching

are associated with more functional limitation and higher radiographic spinal scores [318]. In

the present study occupations that were associated with lifting heavy weights were more

common in the PsA group, and recurrent squatting and pushing heavy weights also tended to

be more common in the PsA group. Recurrent microtrauma that are associated with these

physically demanding occupations may lead to greater susceptibility to PsA.

It has been suggested that infections play a role in the pathogenesis of PsA. The role of

infection as a triggering event for arthritis is well established in reactive arthritis that has

some common features with PsA [87]. Infections of the gastrointestinal and urogenital tract

are most commonly associated with reactive arthritis. While the frequency of urogenital tract

Page 155: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

143

infection was similar in the two groups, infectious diarrhea tended to be more common in the

PsA group. HIV infection that has been linked to severe psoriasis and PsA [95] was found

only in the psoriasis group.

An interesting finding was the decreased prevalence of smoking among the PsA patients.

While smoking is a well-established risk factor for psoriasis [109, 319],in the present study it

was found to be significantly less common among the PsA cases. A similar trend was

reported by Pattison et al. who found a trend to fewer smokers among the PsA patients [76].

In their study the OR (0.68) was similar to the OR in our study (0.5) but was not significant

possibly due to a smaller sample size. Rakkhit et al. reported that the time to development of

PsA decreases with smoking prior to psoriasis onset and increases with smoking after

psoriasis onset [114]. Smoking was also found to have a protective effect in the pathogenesis

of ulcerative colitis, a disorder that bears some clinical and genetic similarities to PsA [320-

322]. The mechanisms underlying these observations are unknown, but suggested

explanations include decreased expression of IL-1β, IL-8 and altered response of the toll-like

receptor pathway to infectious agents among smokers [323, 324]. Another mechanism that

may explain the protective effect of smoking is through the activation of the nicotinic

receptor. Nicotine can activate the alpha7 nicotinic acetylcholine receptor that inhibits

intracellular pro-inflammatory pathways that are associated with development of arthritis

[325, 326]. This pathway is a target for novel therapeutic agents for treatment of arthritis.

There are several limitations to this study. Its retrospective nature may have led to a recall

bias that stems from the tendency of the cases to recall more events that preceded the onset of

arthritis than the controls. I have tried to minimize that problem in several ways. By

approaching patients with a recent onset of arthritis, the study population had a short interval

from the onset of PsA of approximately 3 years. I also avoided linking environmental

exposures and arthritis in the questionnaire, and requested information about events that

occurred in the past 10 years. I have also assessed recall bias by comparing the reported

information about exposure to infections and injuries with available data from a

computerized database that stores medical records from 6 medical centers and outpatient

clinics in Toronto. I aimed to assess whether there has been under-reporting of events by

psoriasis patients. Overall, three out of 3 psoriasis patients reported a previous injury and 0

Page 156: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

144

out of 2 reported an infection. In the PsA group, only 1 out of 3 reported an injury and 1 out

of 6 reported an infection. Therefore, although these figures are small, it seems that recall

bias is not a threat to the validity of the study as the rates of reporting were not lower in the

psoriasis group.

In summary, in this study the association between several environmental exposures and PsA

was investigated. Infections that required antibiotic treatment and occupations that involved

lifting heavy weights were associated with PsA, while there was an inverse association with

the disease. These results need to be confirmed by a prospective study among psoriasis

patients.

Page 157: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

145

Chapter 9. Smoking is inversely associated with development of psoriatic arthritis among psoriasis patients

Page 158: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

146

9.1. Background

Several models have been suggested to describe the genetic and clinical association between

psoriasis and PsA. Since the majority of the patients with PsA develop inflammatory arthritis

following the onset of the psoriasis, PsA may be considered as a “disease within a disease”

with psoriasis as the parent disease. PsA may be considered a more severe phenotype of the

psoriatic disease that occurs due to a greater number of susceptibility genes or exposure to

environmental factors. Additional susceptibility genes or environmental risk factors on the

background of psoriasis are postulated to lead to development of PsA [73].

Genetic studies have shown a strong association of the MHC region on chromosome 6p with

psoriasis and PsA. In psoriasis, the strongest and most consistently reported association is

with HLA-C*06 [327]. While great effort has been expended in the investigation of genetic

risk factors of these diseases, the role of environmental factors has been less defined,

particularly in PsA. In psoriasis, smoking, alcohol consumption, obesity, stressful life events

and infections have been considered to be potential risk factors [75, 328], while physical

trauma, vaccinations and pregnancies have been suggested as risk factors for PsA [75].

Smoking was associated with psoriasis in several case-control studies [105, 106, 329].

Recently, a prospective cohort study has established the association between smoking and

psoriasis [109]. It has been suggested that the effect of smoking may be modified in the

presence of HLA-C*06.Their co-existence was associated with a significantly higher risk of

developing psoriasis compared to the presence of each factor individually and led to a

significantly higher risk of developing psoriasis [330]. In the Chapter 8 the prevalence of

smoking was found to be decreased among PsA patients compared to psoriasis patients

without arthritis. The aim of this study was to validate these results by analyzing the

information available for two large cohorts of PsA and psoriasis patients and to further

investigate the association between smoking and PsA and its interaction with HLA-C*06

allele.

Page 159: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

147

9.2. Methods

9.2.1. Study population In this case-control study, two groups of patients were compared:

Cases – This group included 791 Adult prevalent PsA patients that were all part of the

University of Toronto Psoriatic Arthritis cohort. These patients satisfied the CASPAR criteria

for classification of PsA [22].

Controls – This group included 404 psoriasis patients without arthritis that were recruited

from the University of Toronto Psoriasis Cohort. The diagnosis of psoriasis was confirmed

by a dermatologist. All patients were evaluated by a rheumatologist to exclude inflammatory

arthritis. For the purpose of this study psoriasis patients that developed PsA during the follow

up were removed.

The cohorts were described in details in the methods section (Chapter 4).

Reference population -For comparison, smoking rates among the general population of

Ontario were obtained from the National Population Health Survey (NPHS), a longitudinal

survey conducted by Statistics Canada since 1994. The survey was designed to measure the

health status and determinants of health among Canadians and provides periodic cross-

sectional information. For our study we used the 2005 NPHS survey. The survey collected

information at the provincial level and included 17,276 respondents. The sample was

weighted and the results extrapolated to the population of Ontario. Available summary tables

that were stratified by age groups and gender were used for analysis [331].

9.2.2. Patients selection

PsA patients included in the study were registered in the PsA clinic between 1978 and 2010.

Psoriasis patients included in the study were registered in the Psoriasis cohort between 2006

and 2010. PsA and psoriasis patients within the database with available information about

their smoking status at the time of the diagnosis of PsA and at their first visit, respectively,

were included in the study.

Page 160: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

148

9.2.3. Determination of smoking status

Smoking status was categorized as current smoker, past smoker and life-time non-smoker. A

smoker was defined as a person who smoked ≥ 1 cigarette per day for at least 1 year.

At enrolment, all study participants were asked to report their current smoking status and

what was their smoking status at the time of the diagnosis of PsA (for PsA patients) and at

the time of the diagnosis of psoriasis (for psoriasis patients). Patients were also asked to

report the number of cigarettes and smoking years at the above defined time point.

The reference dates for determination of smoking status were: the time of diagnosis of PsA

(for PsA patients) and the first visit (for psoriasis patients). These dates were chosen because

information about potential confounders was available only from the first assessment date.

Information about potential confounders was retrieved from the database including: age, sex,

level of education, duration of psoriasis, severity of psoriasis and alcohol consumption.

Similar to smoking status, the reference dates used to determine alcohol consumption were:

prior to the diagnosis of PsA (for PsA patients) and at first visit (for psoriasis patients).

Alcohol consumption was categorized as daily (≥ 1 alcoholic beverage a day), social (≥ 1

alcoholic beverage a week) and non-drinker. Since smoking is known to be associated with

the severity of psoriasis; patients were categorized as those with severe psoriasis (Psoriasis

Area Severity Index (PASI) ≥ 10 or mild to moderate psoriasis (PASI< 10) based on the

maximal PASI score in the first 3 years of the follow-up.

9.2.4. HLA-C*06 Allele typing

HLA-C*06 is the strongest known risk factor for psoriasis [327]. Recently, it was suggested

that the interaction between HLA-C*06 and smoking may play a role in the susceptibility to

psoriasis [330]. We therefore investigated whether the effect of smoking is modified by this

allele. For HLA typing, extracted genomic DNA was amplified by PCR using locus specific

primers for the HLA-C locus. PCR amplicons were identified by Sequence Specific

Page 161: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

149

Oligonucleotide (SSO) probes. Ambiguous results were resolved using Sequence Specific

Primers (PCR-SSP).

9.2.5. Statistical analysis

Baseline descriptive statistics were computed with continuous variables summarized by their

means and standard deviations and categorical variables summarized by proportions. The

proportions with exposure to smoking were compared between the PsA and psoriasis patients

through calculation of the Odds Ratio (OR) and the corresponding 95% Confidence Interval

(CI).

In addition, the prevalence of smokers in the PsA and psoriasis groups was assessed within

the same gender and age groups as in the NPHS survey. The prevalence of smoking status in

each group was then compared with data on smoking rates from the NPHS through

Standardized Prevalence Ratios (SPRs). In this analysis only PsA patients that were

diagnosed between 1999 and 2010 were included to avoid bias due to changing trends in

cigarette smoking over the years.

In order to avoid bias due to ecological trends in smoking, an additional analysis was

performed in a subset of the study population that included 404 PsA and psoriasis patients

matched by age (within 5 years) and gender. Each psoriasis patient was assigned a reference

year that was derived from the year of onset of PsA for the corresponding case. From the

available information we were able to infer the years of exposure to smoking for 381 of the

matched psoriasis patients. I have then performed a conditional logistic regression for

matched pairs data to determine the association between smoking status and PsA compared

to psoriasis.

In order to adjust for potential confounders, a logistic regression model that included all

observations was performed. The model included as a key predictor smoking status and the

following potential confounders as covariates: age, sex, duration of psoriasis, level of

education, alcohol consumption and severity of psoriasis. Multivariate logistic regression

analysis was used to assess the independent association between smoking and PsA. A

sensitivity analysis was performed to assess whether the exclusion of patients with missing

Page 162: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

150

information of psoriasis severity and alcohol consumption from the multivariate analysis

affected the results by the removal of these variables from the regression model.

Only Caucasians were included in the analysis of the interaction between HLA-C*06 and

smoking. The proportions with smoking exposure were compared between PsA and psoriasis

after stratification by HLA-C*06 status using a LR test. A logistic regression analysis was

performed to assess the statistical interaction between HLA-C*06 and smoking.

The significance level was set at 0.05 for a 2-sided test. Statistical analysis was performed

using SAS 9.2© statistical software.

Page 163: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

151

9.3. Results

Overall 791 PsA and 404 psoriasis patients with complete information were included in the

study. Most of the PsA patients enrolled the cohort over the last decade (20.6% from 1978 to

1989, 27% from 1990 to 1999 and 52.4% from 2000 to 2010). Overall, 257 PsA patients

were not included in the analysis due to missing information about their smoking status. The

majority of these patients were evaluated at the establishment of the PsA cohort, at that time

such information was not routinely collected. Comparison of the excluded PsA patients to

those included in the study revealed no significant differences in demographic and clinical

characteristics apart from gender. The proportion of females was slightly higher in the PsA

patients with missing information (49.8% vs. 43.8%). The median time interval from the

onset of PsA until the first assessment in the clinic was 3 years.

The PsA patients were on average 10 years younger and had a shorter duration of psoriasis

(Table 9.1). These differences are related to the use of different reference dates for

determination of smoking status: year at onset of PsA (ranged from 1951 to 2010) for the

PsA group vs. first assessment date for the psoriasis group (ranged from 2006 to 2010). PsA

patients had more severe psoriasis as reflected by maximal PASI scores. There were no

differences in the male to female ratio and age at onset of psoriasis. Among the psoriasis

patients, 7 (1.7%) were using biologic agents and 23 (5.7%) were using immunosuppressive

agents.

9.3.1. Comparison of smoking status among PsA and psoriasis to the general population

The smoking rates among psoriasis and PsA patients were compared to the general

population in Ontario through SPRs. The SPRs for smokers among males with psoriasis and

PsA were: 1.12 (95% CI 1.00-1.26) and 1.27 (95% CI 1.09-1.48), respectively. The SPRs for

smokers among females with psoriasis and PsA were: 1.29 (95% CI 1.12-1.47) and PsA 1.28

(95% CI 1.14-1.43), respectively.

Page 164: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

152

9.3.2. Comparison of smoking status between PsA and psoriasis

The distribution of smoking status among patients with PsA and psoriasis alone is presented

in Table 9.2. The proportions of current smokers and past smokers were higher in the

psoriasis patients than the PsA patients (current smokers 26.7% vs. 21.9% and past smokers

30.2% vs. 23.4%, p=0.0006, respectively). The psoriasis group also smoked, on average, for

more years than the PsA group; however, these results may be attributed to the mean older

age of psoriasis patients. There was no difference in the mean number of cigarettes smoked

per day between the PsA and the psoriasis group; however, this information was available

only for a much smaller sample, particularly in the PsA group (N=226).

9.3.3. Analysis for matched pairs data

An analysis for matched pairs was performed to determine whether the association between

smoking and PsA was due to ecological trends in smoking. Of the 404 matched pairs of PsA

and psoriasis patients I was able to infer the smoking status at the reference year for 381

psoriasis patients. The results confirmed the inverse association between current smoking and

PsA compared to psoriasis patients (Table 9.3). In the conditional regression analysis

“current smoking” was less common in PsA compared to psoriasis patients (OR 0.57, 95%

CI 0.39-0.82, p=0.002). In contrast, the prevalence of past smokers was higher in patients

with PsA compared to those with psoriasis (OR 1.92, 95% CI 1.2-3.07, p=0.007). This

change in the direction of the association for the “past smoking” group may be related to the

different reference dates considered for the determination of the status of smoking in the

analysis presented in Table 9.2.

9.3.4. Multivariate regression analysis

A multivariable logistic regression analysis was performed to adjust for potential

confounders; the results are presented in Table 9.4. After adjustment, current smoker

compared to life-time non-smoker remained inversely associated with PsA (OR 0.54, 95% CI

0.38-0.77), while past smoker vs. life-time non-smoker was no longer significant (OR 0.87,

Page 165: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

153

95% CI 0.62-1.21). The increase in OR in the “past-smoker” group suggests a confounding

effect of one or several of the co-variates included in the model. These results were not

appreciably changed after removal of alcohol consumption and psoriasis severity from the

regression model to allow the inclusion of patients with missing data in the analysis.

9.3.5. The interaction between HLA-C*06 and smoking

In the previous sections HLA-C*06 was found to be decreased in patients with PsA

compared to those with psoriasis alone. I have assessed whether the effect of smoking is

modified by HLA-C*06 status. Overall 572 PsA and 376 psoriasis Caucasians were included

in the analysis. In a subgroup analysis (Figure 9.1), the inverse association between smoking

and PsA compared to psoriasis was found only among those who were HLA-C*06 negative

(current smoking vs. non-smoker: OR 0.46, 95% CI 0.3-0.7, p=0.0004 and past smoker vs.

non-smoker: OR 0.46, 95% CI 0.3-0.7, p=0.0003). Among those who were HLA-C*06

carriers, there was no significant difference in smoking status between the two groups

(current smoker vs. non-smoker OR 0.97, 95% CI 0.6-1.7, p=0.91 and past smoker vs. non-

smoker OR 0.96, 95% CI 0.6-1.6, p=0.87). The interaction between HLA-C*06 and smoking

status was statistically significant in a logistic regression analysis (HLA-C*06-current

smoking OR 2.11, 95% CI 1.04-4.32, p=0.039, HLA-C*06-past smoking OR 2.07, 95% CI

1.05-4.11, p= 0.035) suggesting a departure from a multiplicative model.

Page 166: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

154

*Determined at the time of the diagnosis of PsA (for PsA patients) and at first assessment (for psoriasis patients).

Table 9.1 – Demographic and clinical characteristics of the study population

Variable Psoriasis (N=404) PsA (N=791) P value

Age (Years ± SD)* 46.3 ±13.1 36.7 ±13.0 <0.001 Sex: Female (%) 177 (43.8%) 335 (41.6%) 0.66 Age at Onset Ps 30.2(16.0) 28.6 (14.6) 0.08 Age at Onset PsA --- 36.7 (13.0) Duration of Psoriasis* 16.1 ±14.1 8.5 ±10.2 <0.001 Level of Education (%)

- College/University

311(77.2%)

499 (66.3%)

<0.001

PASI (Max. in 3 years)* Severe Psoriasis

6.4 ±6.1 64 (16.1%)

8.7 ±10.1 220 (28.8%)

<0.001 <0.001

Psoriatic nail lesions

Alcohol consumption - Daily

- Social

288 (72.2%)

38 (9.4%) 252 (62.5%)

511 (92.6%)

85 (11.6%) 428 (58.3%)

<0.001

0.53 0.64

Skin lesions prior to arthritis --- 560 (72.3%)

Arthritis pattern at diagnosis - Distal joints only - Oligoarthritis

- Polyarthritis

- Axial only

- Axial+Distal joints

- Axial+Oligoarthritis

- Axial+Polyarthritis

--- 58 (8.1%)

253 (35.2%) 256 (35.6%)

28 (3.9%) 6 (0.8%)

41 (5.7%) 77 (10.7%)

Total Damaged Joints --- 2.9 ±7.5

Page 167: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

155

* Psoriasis (N=209) and PsA (N= 226)

Table 9.3 – Smoking status at the time of the diagnosis by matched pairs (N=381 pairs)

PsA group

Never smoked Current smoker Past smoker

Psoriasis

group

Never smoked 138 (32.6%) 37 (9.7%) 38 (10%)

Current smoker 72 (18.9%) 27 (7.1%) 26 (6.8%)

Past smoker 16 (4.2%) 11 (2.9%) 16 (4.2%)

Grey box – concordant pairs. White box – discordant pairs.

Table 9.2 – Smoking characteristics among psoriasis and PsA patients

Psoriasis (N=404) PsA (N=791) P value

Smoking status

- Never smoked

- Current

- Past

174 (43%)

108 (26.7%)

122 (30.2%)

433 (54.7%)

173 (21.9%)

185 (23.4%)

0.0006

No. of Cigarettes Smoked (per day)*

- 1-9 cigarettes

- 10-19 cigarettes

- More than 20 cigarettes

59 (33.3%)

53 (29.9%)

65 (36.7%)

73 (31.1%)

54 (23.2%)

106 (45.5%)

0.15

No. of Years Smoked* 19.4 (12.6) 16.3 (11.4) 0.0064

Page 168: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

156

*Determined at the time of the diagnosis for PsA and at first visit for psoriasis.

Table 9.4 – The association between smoking status and PsA (N=701) compared to psoriasis alone (N=400) by logistic regression analysis

Univariate Model Multivariate Model

Covariate OR 95% CI P-value OR 95% CI P-value

Smoking Status:

- Current vs. Life-time Non- Smokers

0.65

0.48-0.89

0.007

0.54

0.38-0.77

0.0007

- Past vs. Life-time Non- smokers 0.66 0.49- 0.88 0.006 0.87

0.62-1.21

0.41

Age -1 yr increase* 0.95 0.94 - 0.96 <0.0001 0.95 0.94-0.96 <0.0001

Sex: Male vs. Female 1.06 0.82 - 1.36 0.67 1.04 0.78-1.39 0.78

Duration of Psoriasis -1 yr increase* 0.95 0.94- 0.96 <0.0001 0.96 0.95-0.97 <0.0001

Alcohol Consumption:

- Social vs. None

0.94

0.69-1.21

0.53

0.98

0.71-1.35

0.91

- Daily vs. None 1.11 0.71-1.75 0.64 1.57 0.94-2.63 0.08

Severe psoriasis: Yes vs. No

2.06 1.51-2.82

<0.0001

1.83 1.29-2.60

0.0007

Level of education - College/University

0.58

0.44-0.77

0.0001

0.58

0.42-0.80

0.0009

Page 169: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

157

Page 170: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

158

9.4. Discussion

In this study the association between smoking and PsA was analyzed. I have shown that

smoking is inversely associated with PsA compared to psoriasis patients without arthritis.

The prevalence of smoking among patients with PsA was higher than in the general

population. I have also found an interaction between smoking and HLA-C*06, as the inverse

association between smoking and PsA was present only among patients who were HLA-

C*06 negative.

The role of smoking as a risk factor for psoriasis is well established. The first studies drew

attention to the linkage between smoking and palmo-plantar pustular psoriasis [104]. Since

then several case-control studies have established the association between smoking and

psoriasis vulgaris [105-108], the most common type of psoriasis. The most conclusive

epidemiological evidence for the causative role of smoking in the pathogenesis of psoriasis

was provided by the Nurses’ Health Study II that showed, for the first time, a strong

association between smoking and incident psoriasis in a large population-based prospective

cohort study [109]. In this study the relative risk of psoriasis was 1.8 for current smokers and

1.4 for past smokers. The dose-effect relation between smoking intensity and psoriasis risk

also supports its etiological role. Smoking has also been associated with more severe

psoriasis and poor response to treatment [110, 111]. The independent association between

smoking and psoriasis remained significant even after adjustment for another potential

confounder, excessive alcohol consumption, that is also increased among psoriasis patients

[112, 113]. In the present study the SPRs of smoking were higher in men and women with

psoriasis and PsA compared to the general population.

One of the challenges in investigating risk factors for PsA is selecting the appropriate control

group. When comparing PsA to healthy controls, it is uncertain whether any significant

association is related to the skin or the joint disease. Therefore, psoriasis patients without

arthritis comprise a more informative control group. There are few studies that have

investigated environmental risk factors for PsA among psoriasis patients. In a study by

Pattison et al., smoking was less common among patients with PsA compared to those with

Page 171: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

159

psoriasis alone [76]; however it did not reach statistical significance, possibly due to the

small sample size. Rakkhit et al. found a temporal association between psoriasis, PsA and

smoking [114]. They reported that the duration of time from the onset of psoriasis to

development of PsA decreases with a history of smoking prior to psoriasis onset and

increases with smoking after psoriasis onset. On the other hand, a recent study from

Singapore has found similar proportions of smokers among patients with psoriasis and PsA

patients [38]. In the present study, current smoking was associated with PsA even after

adjustment for potential confounders. Previous studies were also limited by a potential

misclassification of cases and controls since the exclusion of PsA among psoriasis patients

was based upon self-report. Misclassification may decrease the power to detect significant

association. One of the strengths of this study is the careful phenotyping of both cases and

controls by experienced rheumatologists.

The interaction between genetic and environmental risk factors has received only minimal

attention in genetic studies of psoriasis. It has been suggested that HLA-C*06 may modify

the effect of smoking and lead to an increased risk for psoriasis of about 11-fold over that for

non-smokers without HLA-C*06 [330], much higher than the combined effect of both risk

factors. Statistical interaction in epidemiology refers to a model dependent concept and is

considered to be present on a multiplicative scale when the joint effect of risk factors differs

from the product of the effect of the individual factors [332]. In other words, the effect of one

risk factor depends on the presence of the other risk factor. Biological interaction, on the

other hand, measures the joint effect of two risk factors that act together in the same causal

mechanism of a disease. In this study I analyzed the statistical interaction between HLA-

C*06 and smoking. The inverse association between smoking and PsA was present only

among patients who were HLA-C*06 negative. These findings were explained by identifying

a statistically significant interaction term between HLA*06 and smoking status in a logistic

regression analysis. The interaction between HLA-C*06 and smoking may explain the lack

of association between smoking and PsA in studies that recruited the entire study population

from dermatology clinics, as opposed to our study that enrolled cases and controls from two

different sources though within the same centre [38]. By sampling only patients with severe

psoriasis, it is likely that the proportion of HLA-C*06 carriers was high, and the studies were

thus underpowered to detect an association between smoking and PsA.

Page 172: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

160

The inverse association between smoking and PsA is intriguing as smoking is a known risk

factor for numerous diseases. Smoking is associated with increased risk of psoriasis as well

as rheumatoid arthritis [333]. On the other hand, smoking has a protective effect against

ulcerative colitis that bears some clinical and genetic similarities to PsA [320]. Current

smokers have a 40% lower risk of developing ulcerative colitis [321]. Smoking also reduces

the rate of flare-ups, hospitalizations and use of steroids in ulcerative colitis [322]. The

mechanisms underlying these observations are unknown, but suggested explanations include

decreased expression of IL-1β, IL-8 and altered response of the toll-like receptor pathway to

infectious agents among smokers [323, 324]. Another mechanism that may explain the

protective effect of smoking is through the activation of the nicotinic receptor. Nicotine can

activate the alpha7 nicotinic acetylcholine receptor that inhibits intracellular pro-

inflammatory pathways that are associated with development of arthritis [325, 326]. This

pathway is a target for novel therapeutic agents for the treatment of arthritis.

There are several limitations to the study that are inherent to a case-control study. First, recall

bias is one of the limitations of a retrospective study. In this study, PsA patients had to report

their smoking status at the time of the diagnosis, while the psoriasis patients had to report

their current smoking status, therefore it is possible that the information about smoking status

was less accurate for the PsA patients. However, the information has been collected

systematically in all patients upon their first visit to the clinic which was usually close to the

time of the diagnosis of PsA and in that way, minimized recall bias. The reliability of a

retrospective report of smoking status at a specific time-point has been extensively

investigated. It has been shown that people can reliably recall aspects of first tobacco use

occurring on average 30 years before the time of assessment [334]. Other types of biases in

report of smoking habits are more difficult to control. They may depend on factors such as:

socio-cultural aspects associated with the acceptability of smoking, prior knowledge about

the association between smoking and the disease and on the amount of emphasis that is laid

by the investigator on the smoking question. These sources of potential bias are associated

with any case-control study and are acknowledged as one of the limitation of this type of

study.

Another potential threat to the validity of the study is related to ecological trends in smoking

habits over time, since different reference dates were used for determination of smoking

Page 173: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

161

status. However, given the trend of decline in smoking over the past decades [335], the true

difference in smokers between the groups may be underestimated. Furthermore, I have

confirmed the inverse association between current smoking at the time of the diagnosis and

PsA compared to psoriasis in a subset of patients in a matched-pair analysis.

One of the difficulties in designing a case-control study is the selection of an appropriate

control group that would reflect the source population from which the cases have originated.

In this study, all of the controls had psoriasis and came from the same geographic region as

the cases. Since most of the patients came from dermatology clinics they reflect a moderate

to severe spectrum of the disease. It has been reported that patients with severe psoriasis are

at higher risk of developing PsA [24], therefore this group of psoriasis patients serves as an

appropriate control group, although the generalizability of the results may be limited. The

study population was well phenotyped and I was able to adjust for differences between the

groups in the statistical analysis. However, I am aware that there may be other confounders

that were not considered and may explain the differences in smoking prevalence between the

two groups.

Finally, I was unable to analyze the exact temporal relation between smoking and the onset

of psoriasis and PsA. There were conflicting results with regards to the association between

past smoking and PsA compared to psoriasis patients: in the univariate analysis past smoking

was less frequent among patients with PsA, however in the multivariate analysis the

association between past smoking and PsA was not statistically significant while in the

matched-pair analysis past smoking was increased among PsA patients compared to psoriasis

patients. These differences in the direction of association may be explained by the lag

between reference dates used to determine smoking status. I cannot determine the effect of

smoking cession on the risk of developing PsA in patients with psoriasis. This question

would require a prospective cohort study design with an appropriate statistical analysis.

In summary, current smoking was less frequent in patients with PsA compared to those with

psoriasis alone. Furthermore, the findings support a possible interaction between HLA-C*06

and smoking. Among patients who are HLA-C*06 positive, the association between PsA and

smoking was smaller than in HLA-C*06 negative and not statistically significant. Additional

Page 174: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

162

studies that investigate environmental risk factors for PsA and gene-environment interaction

are required to confirm these results.

Page 175: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

163

Chapter 10. General Discussion

The nature of the relationship between PsA and psoriasis is not completely clear. PsA can be

viewed as a “disease within a disease” and psoriasis patients can be considered as a

population at risk for developing PsA. There has been limited information in the literature

about the clinical, environmental and genetic risk factors for PsA among patients with

psoriasis. The present study aimed to fill this gap by directly comparing two well

characterized cohorts of PsA and psoriasis patients from the same geographic area.

The incidence of PsA among patients with psoriasis was investigated, for the first time, in a

prospective cohort of psoriasis patients. The estimated annual incidence of PsA of 1.87 cases

per 100 psoriasis patients was higher than previously reported. Although several

epidemiologic studies have assessed the incidence of PsA in the general population, there is

limited information about the incidence of PsA among psoriasis patients. In a population

based retrospective study that used medical records to confirm the diagnosis of PsA, Wilson

et al., from Rochester, estimated that the cumulative incidence of PsA was 3.1% after 10

years from the onset of psoriasis and 5.1% after 20 years [33]. Several reasons, other than

geographic and ethnic differences, may explain the differences in the cumulative incidence

between our study and those of the group from Rochester. First, Wilson et al. identified cases

based on a retrospective review of computerized medical records, while in our study each

subject was examined prospectively by a rheumatologist. Underestimation and

misclassification of cases are major threats in epidemiological studies that rely on

computerized data registries, particularly since widely accepted classification criteria for PsA

were unavailable until recently. Furthermore, underestimation of PsA among psoriasis

patients may be even more pronounced. In a German study, approximately 18% of the

psoriasis patients that were found to have inflammatory arthritis were unaware of their

condition and had never seen by a rheumatologist [119]. A referral bias may also account

for the difference, since by relying on medical records some of the mild cases of arthritis may

not come to medical attention. The prospective design of this study and careful assessment

of each subject allowed us to diagnose early and milder cases that potentially could have

escaped medical attention. Finally, the differences in the source populations of psoriasis

Page 176: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

164

patients may explain some of the differences. The present study included mainly patients

with moderate to severe psoriasis from dermatology clinics, while the Rochester study

assessed patients with a wider range of psoriasis severities. Several studies in the past

suggested that the severity of psoriasis is associated with higher risk of developing PsA [24,

262]. A recently published study from Europe that assessed the presence of PsA among

psoriasis patients attending dermatology clinics reported a cumulative incidence of PsA that

was closer to that found in our study (20.5% of patients developed PsA after 30 years from

the diagnosis of psoriasis) [32]. In summary, our results of 4 years of follow-up of a newly

established cohort of psoriasis patients without arthritis suggest that the incidence rate of PsA

may be higher than previously reported, particularly among patients with moderate to severe

psoriasis.

In contrast to the prevailing notion that the greatest risk of developing PsA is during the first

years following the onset of psoriasis, the retrospective analysis of data pooled from both the

PsA and psoriasis cohorts demonstrated that the risk of developing PsA among patients with

psoriasis does not change over time [336]. There are limited data about the incidence of PsA

among psoriasis patients. The mean duration of time from psoriasis to PsA is approximately

7 years [300]. Therefore it was assumed by experts in the field that the risk of developing

PsA is highest in the first years following the onset of psoriasis. However, there are no actual

data in the literature to support that notion. A recent retrospective study from Germany

supports our findings. The authors have found that the rate of PsA was constant over time

[32].

A primary aim of this thesis was to identify PsA specific genetic markers among HLA

alleles. The previously reported association between HLA-B*27 and PsA was confirmed.

Furthermore, HLA-C*06 that was associated with PsD and PsA compared to healthy controls

was significantly less frequent in PsA patients compared to those with psoriasis alone. Two

additional HLA alleles, HLA-B*08 and HLA-B*38 were identified as potential genetic

markers for PsA in patients with psoriasis. HLA-B*39 may be a potential marker for axial

PsA.

Page 177: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

165

Our group has previously assessed the association between HLA antigens and PsA compared

to patients with psoriasis alone using serological methods [32]. In that study, HLA-B27 and

B7 antigens conferred an increased risk for PsA, while HLA-B17 (split to B*57 and B*58),

Cw6 and DR7 occurred with a lower frequency among PsA than in those with psoriasis

alone. HLA-B38 and B39 were associated with polyarthritis while HLA-B27, Cw2 and

DRw52 were associated with axial involvement. In the present study I was able to replicate

several of these associations using an independent sample, this time using molecular

techniques to detect HLA alleles.

In both population and family based analysis I have found that HLA-B*27 is a strong genetic

marker for PsA among psoriasis patients. The absence of a detectable association between

HLA-B*27 and the psoriasis group compared to healthy individuals suggests that B*27 is not

a marker for skin disease. Previous studies in different ethnic groups have consistently shown

that HLA-B*27 is an independent risk allele for PsA that is unrelated to the skin disease [41,

285-287]. However, the prevalence of HLA-B*27 among PsA patients in our study was

19.2% and ranged from 20 to 35% in previous studies, clearly much lower than its

prevalence in AS (80-95% of the patients) [288]. Therefore, this allele can only account for a

small proportion of the total genetic risk of PsA.

HLA-C*06 allele confers a strong risk for psoriasis [180, 201, 202]. In the present study, the

frequency of HLA-C*06 was significantly higher in both psoriasis and PsA patients

compared to controls. However, the frequency of HLA-C*06 was lower in PsA patients

compared to psoriasis. Other studies have found a lower prevalence of HLA-C*06 among

PsA compared to psoriasis patients [287, 292]. In addition, psoriatic nail involvement, a

clinical marker for an increased risk of PsA, is more common among HLA-C*06 negative

psoriasis patients [178]. Furthermore, HLA-C*06 has been found to increase the psoriasis-

arthritis latency period [293]. In this thesis I have also found that HLA-C*06 allele was

associated with a prolonged interval of time from psoriasis to PsA that supports its

“protective” effect. HLA-C*06 carriers had more than double the interval of time from the

onset of psoriasis to PsA. It is challenging to explain the strong association of HLA-C*06

with psoriasis while its lower frequency in PsA compared to psoriasis. HLA-C*06 may be

involved in different immunological mechanisms that lead to the joint and the skin disease.

Page 178: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

166

For example, it may be: antigen presentation and activation of the adaptive immune system

for the skin and inhibition of the innate immune system through interaction with Killer cell

Immunoglobulin like Receptors (KIRs) on NK cells in the joints. The recently reported

association between ERAP1 gene polymorphisms and psoriasis only among patients carrying

the HLA-C*0602 risk allele, suggests that antigen presentation may play a role in the

pathogenesis of psoriasis, as ERAP1 is involved in MHC class I peptide processing [252].

Another explanation may be related to genetic heterogeneity of psoriasis. It is possible that

different HLA alleles are associated with a sub-type of psoriasis that is more likely to

develop PsA. Therefore, although the phenotype of psoriasis is similar, one HLA allele is

associated only with skin disease (HLA-C*06) while the other is associated with both skin

and joint involvement. A potential candidate is HLA-C*12, that is associated with both PsA

and psoriasis compared to healthy controls, and was reported in the past to be associated with

psoriasis among Caucasians [170].

Two additional alleles were significantly associated with PsA compared to psoriasis in

multivariate analysis: HLA-B*08 and HLA-B*38. HLA-B*38 was strongly associated with

PsA compared to both psoriasis and controls. HLA-B*38 has been reported to be more

frequent among PsA patients compared to those with psoriasis alone in several small studies

[292, 295]. Additionally, HLA-B*38 has been associated with more peripheral joint

involvement [296]. However, in the subgroup analysis of our study, an association of HLA-

B*38 was detected in the group with axial PsA but not in the group with arthritis restricted to

the peripheral joints. HLA-B*38 is part of a common haplotype along with HLA-C*12. In

our study, this haplotype was associated with PsA compared to psoriasis. A recent GWAS

suggested that HLA-C*12 is a risk allele for psoriasis among Caucasians [170]. The family

based data in our study supports the association between HLA-B*38 and C*12 and PsA,

however in contrast to the population based data, a stronger association was observed with

HLA-C*12 compared to HLA-B*38. Given the very high LD between alleles in our sample,

it is very difficult to dissect the independent effect of each one of them. Larger samples sizes

may be required for that purpose.

HLA-B*08 has not been previously reported to be associated with PsA. In our study, HLA-

B*08 frequencies were higher among PsA compared to psoriasis, but not different from

Page 179: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

167

healthy controls. However, these results were not confirmed in our family based association

study. In the literature, the extended haplotypes across HLA class I and II genes, A*01-

C*07-B*08-DRB1*03-DQA1*05-DQB1*02-DPA1*01-DPB1*04, has been associated with

increased TNF production following rubella vaccination [297]. TNF is a major pro-

inflammatory cytokine in PsA and rubella vaccine was suggested to be a risk factor for

development of PsA among psoriasis patients [76]. HLA-B*08 is part of an ancestral

haplotype (8.1 AH) that also includes HLA-A*01-C*07-DRB1*03 and TNF-308A [284].

There have been conflicting results with regard to the association between PsA and the TNF-

308A polymorphism, which is part of the 8.1 AH [210-212]. Previous analysis of our cohort

combined with PsA patients from Newfoundland did not find an association with TNF-308A

polymorphism [223], however, these patients were compared to healthy controls and not to

psoriasis patients. In summary, HLA-B*08 may be an independent genetic marker for PsA

among psoriasis patients. However, its strong LD with other HLA alleles and relevant genes

in the MHC region precludes a more confident conclusion.

HLA molecules are expressed on target cells and play an important role in the activation of

NK cells through their interaction with KIR molecules. Since I have found that the HLA

class I genes are strongly associated with susceptibility to PsA and psoriasis [153], the

biological interaction between selected activating KIR genes and their HLA ligands in the

susceptibility to PsA was assessed. Several “functional” models that represent the likely

outcomes of HLA /KIR interactions were tested.

Overall, KIR genes showed only weak associations with both PsA and psoriasis. The

frequency of the inhibitory KIR2DL1 gene was lower in PsA patients compared to those with

psoriasis alone. However, this gene was observed in nearly all individuals, therefore, this

difference can only account for a small fraction of the risk. The activating KIR2DS2 gene

was found to be associated with PsA compared to healthy controls. However, although it was

more frequent in the PsA group compared to the psoriasis patients, the association was not

significant. This may be due to the existence of a weak association since the study was not

powered to detect an association with an OR of less than 1.5. The direction of the association

was in accordance with our expectation: the activating KIR2DS2 gene was more frequent in

PsA while the inhibitory KIRs were less frequent in PsA compared to psoriasis. In the

Page 180: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

168

literature, KIR2DS1 and KIR2DS2 have been associated with susceptibility to psoriasis and

PsA. It has been reported that KIR2DS1 is associated with psoriasis compared to healthy

controls among Japanese [229] and Caucasians from Brazil, Sweden and Poland [230, 307,

308]. These results concur with our findings in the multivariate analysis that showed that

KIR2DS1 in the presence of its HLA-C group 2 ligand is associated with psoriatic disease,

suggesting that the strongest association of this gene is with the skin and not the joint disease.

Two models of KIR-HLA interaction were investigated in this study. The combination of

HLA-Bw4-80I alleles and KIR3DS1 gene was found not to be associated with PsA or PsD.

This interaction has not been investigated in PsA previously. However, it was suggested to be

important in the susceptibility to AS. Two studies have found a higher frequency of

KIR3DS1 with a lower frequency of the inhibitory KIR3DL1 among AS patients [234, 309,

310]. However, a large study from the UK reported a lack of an association between

KIR3DS1 and AS [235]. The second interaction that was tested was between the activating

genes KIR2DS2 and KIR2DS1 and their corresponding HLA-C ligands. It was hypothesized

that the association with the activating KIRs would be greatest in the presence of their HLA

ligands. A significant association was found between KIR2DS2 in the presence of its ligand

HLA-C group 1, in which higher frequencies were found in the PsA group, compared to

psoriasis alone. However, that gene combination was not associated with PsD, while

KIR2DS1 in the presence of its ligand HLA-C group 2 was associated with PsD. These

associations may indicate that KIR2DS2 is a susceptibility gene for PsA among psoriasis

patients, while KIR2DS1 is associated only with the skin disease. This model is supported by

the “dual” associations that were found for HLA-C*06 and PsA: a risk factor for psoriasis

alone but an inverse association (“protective” effect) for PsA compared to psoriasis. Since

HLA-C*06 is included in HLA-C group 2 that is the ligand for KIR2DS1, its interaction with

KIR2DS1 may be important in the susceptibility to psoriasis. However, since psoriasis

patients who carry HLA-C*06 are less likely to have HLA-C group 1, simply because one of

their HLA-C alleles already belongs to HLA-C group 2, they are less likely to have a

compatible ligand for the KIR2DS2 that may be a risk gene for PsA among psoriasis patients.

Genetic factors alone cannot account for all cases of PsA. I hypothesized that psoriasis

patients who carry susceptibility genes for arthritis develop PsA after exposure to triggering

Page 181: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

169

environmental factors [74]. In this thesis the association between several putative

environmental risk factors and the occurrence of PsA among psoriasis patients was

investigated. The results suggest that there is an association between PsA and infections,

physical trauma including physically demanding occupational tasks, and smoking [337].

Only a limited number of studies have investigated environmental risk factors for PsA.

Pattison et al. have found that physical trauma, rubella vaccination, oral ulcers and moving

house were associated with PsA compared to psoriasis alone [76]. Another study by

Thumboo et al. [77] have found that pregnancy was protective of PsA, while steroid use was

associated with higher risk of the disease. These studies had several limitations including:

their small sample size, the use of an administrative database as a source of information that

may have led to underestimation of events and potential misclassification of cases and

controls as psoriasis patients were not evaluated to exclude the presence of PsA. In the

present study, I aimed to minimize the misclassification of cases and controls by having a

rheumatologist evaluate all of the controls to rule out clinical inflammatory arthritis. The

cases and controls were recruited from the same program, and were evaluated according to

the same protocol.

Several case reports and case series have highlighted the potential role of local trauma in the

pathogenesis of PsA [80, 314, 315]. However, only a few studies have systematically

assessed its role in PsA. Scarpa et al. [118] reported more acute medical events in the PsA

group, including injuries that occurred less than 10 days before the onset of the arthritis,

compared to the group with rheumatoid arthritis. Pattison et al. [76] found that the

occurrence of an injury was more common in patients with PsA, compared to psoriasis

patients. However, Thumboo et al. did not find an association between trauma and PsA [77].

In our study, patients with PsA had double the risk of experiencing injuries in the time

window under study compared to psoriasis patients. The role of occupational related physical

activities in the pathogenesis of PsA has not been thoroughly investigated. In ankylosing

spondylitis, occupations that require recurrent bending, twisting and stretching are associated

with more functional limitation and higher radiographic spinal scores [318]. In the present

study occupations that were associated with lifting heavy weights were more common in the

PsA group, and recurrent squatting and pushing heavy weights also tended to be more

Page 182: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

170

common in the PsA group. Recurrent microtrauma that are associated with these physically

demanding occupations may lead to greater susceptibility to PsA.

It has been suggested that infections play a role in the pathogenesis of PsA. The role of

infection as a triggering event for arthritis is well established in reactive arthritis that has

some common features with PsA [87]. Infections of the gastrointestinal and urogenital tract

are most commonly associated with reactive arthritis. While the frequency of urogenital tract

infection was similar in the two groups, infectious diarrhea tended to be more common in the

PsA group.

I have found that smoking is inversely associated with PsA compared to psoriasis patients

without arthritis. The prevalence of smoking among patients with PsA was higher than in the

general population. I have also found an interaction between smoking and HLA-C*06, as the

inverse association between smoking and PsA was present only among patients who were

HLA-C*06 negative. Pattison et al. have found that smoking was less common among

patients with PsA compared to those with psoriasis alone [76]; however it did not reach

statistical significance, possibly due to the small sample size. Rakkhit et al. founda temporal

association between psoriasis, PsA and smoking [114]. They reported that the duration of

time from the onset of psoriasis to development of PsA decreases with a history of smoking

prior to psoriasis onset and increases with smoking after psoriasis onset. On the other hand, a

recent study from Singapore has found similar proportions of smokers among patients with

psoriasis and PsA patients [38]. In the present study, current smoking was associated with

PsA even after adjustment for potential confounders.

The interaction between genetic and environmental risk factors has received only minimal

attention in genetic studies of psoriasis. It has been suggested that HLA-C*06 may modify

the effect of smoking and lead to an increased risk for psoriasis of about 11-fold over that for

non-smokers without HLA-C*06 [330], much higher than the combined effect of both risk

factors. In this study I analyzed the statistical interaction between HLA-C*06 and smoking.

The inverse association between smoking and PsA was present only among patients who

were HLA-C*06 negative. The interaction between HLA-C*06 and smoking may explain the

lack of association between smoking and PsA in studies that recruited the entire study

Page 183: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

171

population from dermatology clinics, as opposed to our study that enrolled cases and controls

from two different sources [38]. By sampling only patients with severe psoriasis, it is likely

that the proportion of HLA-C*06 carriers was high, and the studies were thus underpowered

to detect an association between smoking and PsA.

The inverse association between smoking and PsA is intriguing as smoking is a known risk

factor for numerous diseases. Smoking is associated with increased risk of psoriasis as well

as rheumatoid arthritis [333]. On the other hand, smoking has a protective effect against

ulcerative colitis that bears some clinical and genetic similarities to PsA [320]. Current

smokers have a 40% lower risk of developing ulcerative colitis [321]. Smoking also reduces

the rate of flare-ups, hospitalizations and use of steroids in ulcerative colitis [322]. The

mechanisms underlying these observations are unknown, but suggested explanations include

decreased expression of IL-1β, IL-8 and altered response of the toll-like receptor pathway to

infectious agents among smokers [323, 324]. Another mechanism that may explain the

protective effect of smoking is through the activation of the nicotinic receptor. Nicotine can

activate the alpha7 nicotinic acetylcholine receptor that inhibits intracellular pro-

inflammatory pathways that are associated with development of arthritis [325, 326]. This

pathway is a target for novel therapeutic agents for the treatment of arthritis.

10.1. Conclusions

This work provides valuable information about the epidemiology of PsA. I was able to

estimate for the first time the incidence of PsA in a prospective cohort study of patients with

psoriasis. The results contradict the commonly held notion that the risk of developing PsA

decreases over time following the onset of psoriasis. Furthermore, the incidence of PsA

among psoriasis patients as estimated in this thesis is significantly higher than previously

reported in the literature suggesting that PsA is underestimated in patients with psoriasis.

The results of the thesis help to better understand the pathogenesis of PsA. I have shown that

genetic and environmental factors are associated with the development of PsA among

patients with psoriasis, and that there may be interactions between these factors. In

Page 184: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

172

accordance with the previous literature, the role of trauma is suggested as a triggering event

for PsA and additional novel potential environmental risk factors were identified:

occupations that involve heavy lifting and infections. Furthermore, I have shown for the first

time that active smoking is inversely associated with PsA among psoriasis patients and that

its effect is modified by the HLA-C*06 allele. This association, if replicated in future studies,

may contribute to the understanding of the pathogenesis of the disease and may have

implications related to treatment, as medications that target signaling pathway involving the

nicotinic receptor may be effective in PsA.

The association between HLA alleles and PsA was investigated. I was able to replicate the

previously reported associations of PsA with HLA-B*27 and B*38 and to identify novel

associations with HLA-B*08, HLA-C*06 and HLA-B*39. The KIR genes, 2DS2 and 2DL1

were also associated with PsA among psoriasis patients. These genetic markers differentiate

patients with PsA from those with psoriasis alone and may serve as markers for higher risk of

PsA among psoriasis patients. Early treatment in these patients may prevent joint damage and

disability that result from a delayed diagnosis and therapy of PsA.

10.2. Future directions

Future studies related to HLA alleles and their association with PsA can focus on grouping

these alleles based on antigen binding motifs. The high levels of polymorphism in the HLA

molecules are required for presentation of a diverse repertoire of peptides to immune cells.

Most of the variation in protein sequences is located in the HLA antigen binding pockets.

HLA associations can be analyzed by incorporating structural and functional information

about HLA molecules. This process may identify associations that may be related to the

pathogenic mechanism of the disease based on variation in functional sequences.

We also aim to further investigate the “protective” effect of smoking in the susceptibility to

PsA that has been identified in the present study. One of the potential mechanisms that may

explain the “protective” effect of smoking is through the activation of alpha7 nicotinic

acetylcholine receptor that inhibits intracellular pro-inflammatory pathways. We aim to

Page 185: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

173

investigate the association of polymorphisms within the alpha7 nicotinic acetylcholine

receptor gene and their interaction with smoking to PsA.

Finally, this study has used the information collected from a recently established psoriasis

cohort. This cohort was established to investigate genetic and clinical risk factors for PsA. A

prospective cohort is considered the preferred design to evaluate risk factors in epidemiologic

studies. Since at the present time there are not sufficient incident cases of PsA to evaluate

possible risk factors, we had to use case-control and retrospective study designs which are

subjected to more bias. In the future we will be able to validate these results by analyzing the

data that has been collected prospectively, by continuing to follow the Toronto Psoriasis

Cohort.

Page 186: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

174

References

1. Gelfand JM, Weinstein R, Porter SB, Neimann AL, Berlin JA, Margolis DJ: Prevalence and treatment of psoriasis in the United Kingdom: a population-based study. Arch Dermatol 2005, 141(12):1537-1541.

2. Brandrup F, Green A: The prevalence of psoriasis in Denmark. Acta Derm

Venereol 1981, 61(4):344-346. 3. Ferrandiz C, Bordas X, Garcia-Patos V, Puig S, Pujol R, Smandia A: Prevalence of

psoriasis in Spain (Epiderma Project: phase I). J Eur Acad Dermatol Venereol

2001, 15(1):20-23. 4. Shai A, Vardy D, Zvulunov A: [Psoriasis, biblical afflictions and patients' dignity].

Harefuah 2002, 141(5):479-482, 496. 5. Willan R (ed.): On cutaneous diseases. London; 1798. 6. Langley RG, Krueger GG, Griffiths CE: Psoriasis: epidemiology, clinical features,

and quality of life. Ann Rheum Dis 2005, 64 Suppl 2:ii18-23; discussion ii24-15. 7. Henseler T, Christophers E: Psoriasis of early and late onset: characterization of

two types of psoriasis vulgaris.J Am Acad Dermatol 1985, 13:450-456. 8. Griffiths CE, Barker JN: Pathogenesis and clinical features of psoriasis.Lancet

2007, 370:263-271. 9. Farber EM, Nall LM: The natural history of psoriasis in 5600

patients.Dermatologica 1974, 148:1–18. 10. Finlay AY, Kelly SE: Psoriasis—an index of disability.Clin Exp Dermatol 1987,

12:8–11. 11. Finlay AY, Coles EC: The effect of severe psoriasis on the quality of life of 369

patients.Br J Dermatol 1995, 132:236–244. 12. Christophers E: Comorbidities in psoriasis. Clin Dermatol 2007, 25:529-534. 13. Gottlieb AB, Dann F, Menter A: Psoriasis and the metabolic syndrome.J Drugs

Dermatol 2008, 7:563-572. 14. Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB: Risk of

myocardial infarction in patients with psoriasis. . JAMA 2006, 296:1735-1741. 15. Gladman DD: Psoriatic arthritis: epidemiology, clinical features, course, and

outcome. Annals of the Rheumatic Diseases 2005, 64(suppl_2):ii14-ii17. 16. Fredriksson T, Pettersson U: Severe psoriasis--oral therapy with a new retinoid.

Dermatologica 1978, 157(4):238-244. 17. Feldman SR: Psoriasis assessment tools in clinical trials. Annals of the Rheumatic

Diseases 2005, 64(suppl_2):ii65-ii68. 18. Menter A, Korman NJ, Elmets CA, Feldman SR, Gelfand JM, Gordon KB, Gottlieb

A, Koo JY, Lebwohl M, Lim HW et al: Guidelines of care for the management of

psoriasis and psoriatic arthritis: Section 5. Guidelines of care for the treatment of psoriasis with phototherapy and photochemotherapy. J Am Acad Dermatol

2010, 62(1):114-135. 19. Wright V, Moll JMH (eds.): Psoriatic arthritis. Amsterdam: North Holland

Publishing Co; 1976.

Page 187: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

175

20. Alibert JL (ed.): Précis théorique et pratique sur les maladies de la peau. Paris: Caille et Ravier; 1818.

21. BLUMBERG BS, BUNIM JJ, CALKINS E, PIRANI CL, ZVAIFLER NJ: ARA

Nomenclature and Classification of Arthritis and Rheumatism (Tentative). Arthritis Rheum 1964, 7:93-97.

22. Taylor W, Gladman D, Helliwell P, Marchesoni A, Mease P, Mielants H: Classification criteria for psoriatic arthritis: development of new criteria from a large international study. Arthritis Rheum 2006, 54(8):2665-2673.

23. Healy PJ, Helliwell PS: Classification of the spondyloarthropathies. Curr Opin

Rheumatol 2005, 17(4):395-399. 24. Gelfand JM, Gladman DD, Mease PJ, Smith N, Margolis DJ, Nijsten T, Stern RS,

Feldman SR, Rolstad T: Epidemiology of psoriatic arthritis in the population of

the United States. J Am Acad Dermatol 2005, 53(4):573. 25. Alamanos Y, Voulgari PV, Drosos AA: Incidence and prevalence of psoriatic

arthritis: a systematic review. J Rheumatol 2008, 35(7):1354-1358. 26. Kaipiainen-Seppanen O: Incidence of psoriatic arthritis in Finland. Br J

Rheumatol 1996, 35(12):1289-1291. 27. Shbeeb M, Uramoto KM, Gibson LE, O'Fallon WM, Gabriel SE: The epidemiology

of psoriatic arthritis in Olmsted County, Minnesota, USA, 1982-1991. J

Rheumatol 2000, 27(5):1247-1250. 28. Soriano ER, Rosa J, Velozo E, Schpilberg M, Imamura PM, Diaz J, Catoggio LJ:

Incidence and prevalence of psoriatic arthritis in Buenos Aires, Argentina: a 6-year health management organization-based study. Rheumatology (Oxford) 2010.

29. Zachariae H: Prevalence of joint disease in patients with psoriasis: implications

for therapy. Am J Clin Dermatol 2003, 4(7):441-447. 30. Scarpa R, Oriente P, Pucino A, Torella M, Vignone L, Riccio A, Biondi Oriente C:

Psoriatic arthritis in psoriatic patients. Br J Rheumatol 1984, 23(4):246-250. 31. Gladman D (ed.): Psoriatic arthritis. Berlin: Springer-Verlag; 2005. 32. Christophers E, Barker JN, Griffiths CE, Dauden E, Milligan G, Molta C, Sato R,

Boggs R: The risk of psoriatic arthritis remains constant following initial

diagnosis of psoriasis among patients seen in European dermatology clinics. J

Eur Acad Dermatol Venereol 2010, 24(5):548-554. 33. Wilson FC, Icen M, Crowson CS, McEvoy MT, Gabriel SE, Kremers HM: Incidence

and clinical predictors of psoriatic arthritis in patients with psoriasis: a population-based study. Arthritis Rheum 2009, 61(2):233-239.

34. Helliwell PS, Porter G, Taylor WJ: Polyarticular psoriatic arthritis is more like

oligoarticular psoriatic arthritis, than rheumatoid arthritis. Ann Rheum Dis 2007, 66(1):113-117.

35. Gladman DD, Shuckett R, Russell ML, Thorne JC, Schachter RK: Psoriatic arthritis

(PSA)--an analysis of 220 patients. Q J Med 1987, 62(238):127-141. 36. McGonagle D, Khan MA, Marzo-Ortega H, O'Connor P, Gibbon W, Emery P:

Enthesitis in spondyloarthropathy. Curr Opin Rheumatol 1999, 11(4):244-250. 37. Brockbank JE, Stein M, Schentag CT, Gladman DD: Dactylitis in psoriatic

arthritis: a marker for disease severity?Ann Rheum Dis 2005, 64(2):188-190.

Page 188: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

176

38. Tey HL, Ee HL, Tan AS, Theng TS, Wong SN, Khoo SW: Risk factors associated

with having psoriatic arthritis in patients with cutaneous psoriasis. J Dermatol

2010, 37(5):426-430. 39. Elkayam O, Ophir J, Yaron M, Caspi D: Psoriatic arthritis: interrelationships

between skin and joint manifestations related to onset, course and distribution. Clin Rheumatol 2000, 19(4):301-305.

40. Cohen MR, Reda DJ, Clegg DO: Baseline relationships between psoriasis and

psoriatic arthritis: analysis of 221 patients with active psoriatic arthritis.

Department of Veterans Affairs Cooperative Study Group on Seronegative Spondyloarthropathies. J Rheumatol 1999, 26(8):1752-1756.

41. Gladman DD, Anhorn KA, Schachter RK, Mervart H: HLA antigens in psoriatic

arthritis. J Rheumatol 1986, 13(3):586-592. 42. Griffiths CE, Christophers E, Barker JN, Chalmers RJ, Chimenti S, Krueger GG,

Leonardi C, Menter A, Ortonne JP, Fry L: A classification of psoriasis vulgaris

according to phenotype. Br J Dermatol 2007, 156(2):258-262. 43. Gladman DD, Hing EN, Schentag CT, Cook RJ: Remission in psoriatic arthritis. J

Rheumatol 2001, 28(5):1045-1048. 44. Gladman DD: Natural history of psoriatic arthritis. Baillieres Clin Rheumatol

1994, 8(2):379-394. 45. Torre Alonso JC, Rodriguez Perez A, Arribas Castrillo JM, Ballina Garcia J, Riestra

Noriega JL, Lopez Larrea C: Psoriatic arthritis (PA): a clinical, immunological

and radiological study of 180 patients. Br J Rheumatol 1991, 30(4):245-250. 46. Rahman P, Nguyen E, Cheung C, Schentag CT, Gladman DD: Comparison of

radiological severity in psoriatic arthritis and rheumatoid arthritis. J Rheumatol

2001, 28(5):1041-1044. 47. Siannis F, Farewell VT, Cook RJ, Schentag CT, Gladman DD: Clinical and

radiological damage in psoriatic arthritis. Ann Rheum Dis 2006, 65(4):478-481. 48. Bond SJ, Farewell VT, Schentag CT, Gladman DD: Predictors for radiological

damage in psoriatic arthritis: results from a single centre. Ann Rheum Dis 2007, 66(3):370-376.

49. Wong K, Gladman DD, Husted J, Long JA, Farewell VT: Mortality studies in

psoriatic arthritis: results from a single outpatient clinic. I. Causes and risk of death. Arthritis Rheum 1997, 40(10):1868-1872.

50. Ali Y, Tom BD, Schentag CT, Farewell VT, Gladman DD: Improved survival in

psoriatic arthritis with calendar time. Arthritis Rheum 2007, 56(8):2708-2714. 51. Gladman DD, Ang M, Su L, Tom BD, Schentag CT, Farewell VT: Cardiovascular

morbidity in psoriatic arthritis. Ann Rheum Dis 2009, 68(7):1131-1135. 52. Ritchlin CT, Kavanaugh A, Gladman DD, Mease PJ, Helliwell P, Boehncke WH, de

Vlam K, Fiorentino D, Fitzgerald O, Gottlieb AB et al: Treatment

recommendations for psoriatic arthritis. Ann Rheum Dis 2009, 68(9):1387-1394. 53. Gladman DD: Toward unraveling the mystery of psoriatic arthritis. Arthritis

Rheum 1993, 36(7):881-884. 54. Tassiulas I, Duncan SR, Centola M, Theofilopoulos AN, Boumpas DT: Clonal

characteristics of T cell infiltrates in skin and synovium of patients with psoriatic arthritis. Hum Immunol 1999, 60(6):479-491.

Page 189: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

177

55. Prinz JC, Vollmer S, Boehncke WH, Menssen A, Laisney I, Trommler P: Selection

of conserved TCR VDJ rearrangements in chronic psoriatic plaques indicates a common antigen in psoriasis vulgaris. Eur J Immunol 1999, 29(10):3360-3368.

56. Chang JC, Smith LR, Froning KJ, Schwabe BJ, Laxer JA, Caralli LL, Kurland HH, Karasek MA, Wilkinson DI, Carlo DJ et al: CD8+ T cells in psoriatic lesions

preferentially use T-cell receptor V beta 3 and/or V beta 13.1 genes. Proc Natl

Acad Sci U S A 1994, 91(20):9282-9286. 57. van Kuijk AW, Reinders-Blankert P, Smeets TJ, Dijkmans BA, Tak PP: Detailed

analysis of the cell infiltrate and the expression of mediators of synovial

inflammation and joint destruction in the synovium of patients with psoriatic arthritis: implications for treatment. Ann Rheum Dis 2006, 65(12):1551-1557.

58. Szodoray P, Alex P, Chappell-Woodward CM, Madland TM, Knowlton N, Dozmorov I, Zeher M, Jarvis JN, Nakken B, Brun JG et al: Circulating cytokines in

Norwegian patients with psoriatic arthritis determined by a multiplex cytokine array system. Rheumatology (Oxford) 2007, 46(3):417-425.

59. Partsch G, Steiner G, Leeb BF, Dunky A, Broll H, Smolen JS: Highly increased

levels of tumor necrosis factor-alpha and other proinflammatory cytokines in psoriatic arthritis synovial fluid. J Rheumatol 1997, 24(3):518-523.

60. Gaspari AA: Innate and adaptive immunity and the pathophysiology of psoriasis. J Am Acad Dermatol 2006, 54(3 Suppl 2):S67-80.

61. Ritchlin CT, Fitzgerald O: Pathogenesis of Psoriatic Arthritis. In. USA: MOSBY Elsevier; 2007: 54-57.

62. Liu Y, Krueger JG, Bowcock AM: Psoriasis: genetic associations and immune

system changes. Genes Immun 2007, 8(1):1-12. 63. McQueen FM, Skinner MA, Krissansen GW, Robinson E, Tan PL: Natural killer

cell function and expression of beta 7 integrin in psoriatic arthritis. J Rheumatol

1994, 21(12):2266-2273. 64. Spadaro A, Scrivo R, Moretti T, Bernardini G, Riccieri V, Taccari E, Strom R,

Valesini G: Natural killer cells and gamma/delta T cells in synovial fluid and in

peripheral blood of patients with psoriatic arthritis. Clin Exp Rheumatol 2004, 22(4):389-394.

65. Zhang AL, Colmenero P, Purath U, Teixeira de Matos C, Hueber W, Klareskog L, Tarner IH, Engleman EG, Soderstrom K: Natural killer cells trigger differentiation

of monocytes into dendritic cells. Blood 2007, 110(7):2484-2493. 66. Cameron AL, Kirby B, Fei W, Griffiths CE: Natural killer and natural killer-T

cells in psoriasis. Arch Dermatol Res 2002, 294(8):363-369. 67. Zucchini N, Crozat K, Baranek T, Robbins SH, Altfeld M, Dalod M: Natural killer

cells in immunodefense against infective agents. Expert Review of Anti-infective

Therapy 2008, 6(6):867-885. 68. Diaz-Pena R, Blanco-Gelaz MA, Lopez-Larrea C: KIR genes and their role in

spondyloarthropathies. Adv Exp Med Biol 2009, 649:286-299. 69. Williams F, Meenagh A, Sleator C, Cook D, Fernandezvina M, Bowcock A,

Middleton D: Activating Killer Cell Immunoglobulin-Like Receptor Gene

KIR2DS1 Is Associated With Psoriatic Arthritis. Human Immunology 2005, 66(7):836-841.

Page 190: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

178

70. McGonagle D, Lories RJ, Tan AL, Benjamin M: The concept of a "synovio-

entheseal complex" and its implications for understanding joint inflammation and damage in psoriatic arthritis and beyond. Arthritis Rheum 2007, 56(8):2482-2491.

71. McGonagle D: Enthesitis: an autoinflammatory lesion linking nail and joint

involvement in psoriatic disease. J Eur Acad Dermatol Venereol 2009, 23 Suppl

1:9-13. 72. Tan AL, Benjamin M, Toumi H, Grainger AJ, Tanner SF, Emery P, McGonagle D:

The relationship between the extensor tendon enthesis and the nail in distal

interphalangeal joint disease in psoriatic arthritis--a high-resolution MRI and histological study. Rheumatology (Oxford) 2007, 46(2):253-256.

73. Ritchlin CT: Pathogenesis of psoriatic arthritis. Curr Opin Rheumatol 2005, 17(4):406-412.

74. Bruce IN, Silman AJ: The aetiology of psoriatic arthritis. Rheumatology (Oxford)

2001, 40(4):363-366. 75. Chandran V, Raychaudhuri SP: Geoepidemiology and environmental factors of

psoriasis and psoriatic arthritis. J Autoimmun 2010, 34(3):J314-321. 76. Pattison E, Harrison BJ, Griffiths CE, Silman AJ, Bruce IN: Environmental risk

factors for the development of psoriatic arthritis: results from a case-control study. Ann Rheum Dis 2008, 67(5):672-676.

77. Thumboo J, Uramoto K, Shbeeb MI, O'Fallon WM, Crowson CS, Gibson LE, Michet CJ, Jr., Gabriel SE: Risk factors for the development of psoriatic arthritis: a

population based nested case control study. J Rheumatol 2002, 29(4):757-762. 78. Doury P: Psoriatic arthritis with physical trauma. J Rheumatol 1993, 20(9):1629. 79. Goupille P, Soutif D, Valat JP: Psoriatic arthritis precipitated by physical trauma.

J Rheumatol 1991, 18(4):633. 80. Langevitz P, Buskila D, Gladman DD: Psoriatic arthritis precipitated by physical

trauma. J Rheumatol 1990, 17(5):695-697. 81. Mohla G, Brodell RT: Koebner phenomenon in psoriasis. A common response to

skin trauma. Postgrad Med 1999, 106(3):39-40. 82. Kalayciyan A, Aydemir EH, Kotogyan A: Experimental Koebner phenomenon in

patients with psoriasis. Dermatology 2007, 215(2):114-117. 83. Eyre RW, Krueger GG: Response to injury of skin involved and uninvolved with

psoriasis, and its relation to disease activity: Koebner and 'reverse' Koebner reactions. Br J Dermatol 1982, 106(2):153-159.

84. Dika E, Bardazzi F, Balestri R, Maibach HI: Environmental factors and psoriasis. Curr Probl Dermatol 2007, 35:118-135.

85. Olivieri I, Padula A, D'Angelo S, Scarpa R: Role of trauma in psoriatic arthritis. J

Rheumatol 2008, 35(11):2085-2087. 86. Sieper J, Braun J, Kingsley GH: Report on the Fourth International Workshop on

Reactive Arthritis. Arthritis Rheum 2000, 43(4):720-734. 87. Rihl M, Klos A, Kohler L, Kuipers JG: Infection and musculoskeletal conditions:

Reactive arthritis. Best Pract Res Clin Rheumatol 2006, 20(6):1119-1137. 88. Rasmussen JE: The relationship between infection with group A beta hemolytic

streptococci and the development of psoriasis. Pediatr Infect Dis J 2000, 19(2):153-154.

Page 191: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

179

89. Prinz JC: Psoriasis vulgaris--a sterile antibacterial skin reaction mediated by

cross-reactive T cells? An immunological view of the pathophysiology of psoriasis. Clin Exp Dermatol 2001, 26(4):326-332.

90. Gudjonsson JE, Thorarinsson AM, Sigurgeirsson B, Kristinsson KG, Valdimarsson H: Streptococcal throat infections and exacerbation of chronic plaque psoriasis:

a prospective study. Br J Dermatol 2003, 149(3):530-534. 91. Valdimarsson H, Thorleifsdottir RH, Sigurdardottir SL, Gudjonsson JE, Johnston A:

Psoriasis--as an autoimmune disease caused by molecular mimicry. Trends

Immunol 2009, 30(10):494-501. 92. Vasey FB, Deitz C, Fenske NA, Germain BF, Espinoza LR: Possible involvement of

group A streptococci in the pathogenesis of psoriatic arthritis. J Rheumatol 1982, 9(5):719-722.

93. Rahman MU, Ahmed S, Schumacher HR, Zeiger AR: High levels of

antipeptidoglycan antibodies in psoriatic and other seronegative arthritides. J

Rheumatol 1990, 17(5):621-625. 94. Mody GM, Parke FA, Reveille JD: Articular manifestations of human

immunodeficiency virus infection. Best Pract Res Clin Rheumatol 2003, 17(2):265-287.

95. Menon K, Van Voorhees AS, Bebo BF, Jr., Gladman DD, Hsu S, Kalb RE, Lebwohl MG, Strober BE: Psoriasis in patients with HIV infection: from the medical

board of the National Psoriasis Foundation. J Am Acad Dermatol 2010, 62(2):291-299.

96. Mijiyawa M, Oniankitan O, Khan MA: Spondyloarthropathies in sub-Saharan

Africa. Curr Opin Rheumatol 2000, 12(4):281-286. 97. Bremmer S, Van Voorhees AS, Hsu S, Korman NJ, Lebwohl MG, Young M, Bebo

BF, Jr., Blauvelt A: Obesity and psoriasis: from the Medical Board of the

National Psoriasis Foundation. J Am Acad Dermatol 2010, 63(6):1058-1069. 98. Lindegard B: Diseases associated with psoriasis in a general population of

159,200 middle-aged, urban, native Swedes. Dermatologica 1986, 172(6):298-304. 99. Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB, Gelfand JM: Prevalence

of cardiovascular risk factors in patients with psoriasis. J Am Acad Dermatol

2006, 55(5):829-835. 100. Setty AR, Curhan G, Choi HK: Obesity, waist circumference, weight change, and

the risk of psoriasis in women: Nurses' Health Study II. Arch Intern Med 2007, 167(15):1670-1675.

101. Wolk K, Mallbris L, Larsson P, Rosenblad A, Vingard E, Stahle M: Excessive body

weight and smoking associates with a high risk of onset of plaque psoriasis. Acta

Derm Venereol 2009, 89(5):492-497. 102. Puig L: Obesity and psoriasis: body weight and body mass index influence the

response to biological treatment. J Eur Acad Dermatol Venereol 2011. 103. Soltani-Arabshahi R, Wong B, Feng BJ, Goldgar DE, Duffin KC, Krueger GG:

Obesity in early adulthood as a risk factor for psoriatic arthritis. Arch Dermatol

2010, 146(7):721-726. 104. O'Doherty CJ, MacIntyre C: Palmoplantar pustulosis and smoking. Br Med J 1985,

291:861-864.

Page 192: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

180

105. Naldi L, Chatenoud L, Linder D, Belloni Fortina A, Peserico A, Virgili AR, al. e: Cigarette smoking, body mass index, and stressful life events as risk factors for psoriasis: results from an Italian case-control study. J Invest Dermatol 2005, 125:61-67.

106. Wolk K, Mallbris L, Larsson P, Rosenblad A, Vingård E, Ståhle M: Excessive body

weight and smoking associates with a high risk of onset of plaque psoriasis. Acta

Derm Venereol 2009, 89:492-497. 107. Herron MD, Hinckley M, Hoffman MS, Papenfuss J, Hansen CB, Callis KP, al. e:

Impact of obesity and smoking on psoriasis presentation and management. Arch

Dermatol 2005, 141:1527-1534. 108. Poikolainen K, Reunala T, Karvonen J: Smoking, alcohol and life events related to

psoriasis among women. Br J Dermatol 1994, 130:473-477. 109. Setty AR, Curhan G, Choi HK: Smoking and the risk of psoriasis in women:

Nurses' Health Study II.Am J Med 2007, 120:953-959. 110. Fortes C, Mastroeni S, Leffondré K, Sampogna F, Melchi F, Mazzotti E, al. e:

Relationship between smoking and the clinical severity of psoriasis. Arch

Dermatol 2005, 141:1580-1584. 111. Serwin AB, Sokolowska M, Chodynicka B: Tumor necrosis factor-alpha-

converting enzyme as a potential mediator of the influence of smoking on the response to treatment with narrowband ultraviolet B in psoriasis patients. Photodermatol Photoimmunol Photomed 2010, 26:36-40.

112. Jankovic S, Raznatovic M, Marinkovic J, Jankovic J, Maksimovic N: . Risk factors

for psoriasis: A case-control study. J Dermatol 2009, 36:328-334. 113. Gerdes S, Zahl VA, Weichenthal M, Mrowietz U: Smoking and alcohol intake in

severely affected patients with psoriasis in Germany. Dermatol 2010, 220:38-43. 114. Rakkhit T, Wong B, Nelson TS, Hansen CB, Papenfuss JS, Panko JM, al. E: Time to

development of psoriatic arthritis decreases with smoking prior to psoriasis onset and increases with smoking after psoriasis onset. . J invest Dermatol 2007, 127 (Suppl. 1):S52.

115. Al'Abadie MS, Kent GG, Gawkrodger DJ: The relationship between stress and the

onset and exacerbation of psoriasis and other skin conditions. Br J Dermatol

1994, 130(2):199-203. 116. Miller AH: Neuroendocrine and immune system interactions in stress and

depression. Psychiatr Clin North Am 1998, 21(2):443-463. 117. Maes M, Goossens F, Lin A, De Meester I, Van Gastel A, Scharpe S: Effects of

psychological stress on serum prolyl endopeptidase and dipeptidyl peptidase IV

activity in humans: higher serum prolyl endopeptidase activity is related to stress-induced anxiety. Psychoneuroendocrinology 1998, 23(5):485-495.

118. Scarpa R, Del Puente A, di Girolamo C, della Valle G, Lubrano E, Oriente P: Interplay between environmental factors, articular involvement, and HLA-B27 in patients with psoriatic arthritis. Ann Rheum Dis 1992, 51(1):78-79.

119. Radtke MA, Reich K, Blome C, Rustenbach S, Augustin M: Prevalence and clinical

features of psoriatic arthritis and joint complaints in 2009 patients with psoriasis: results of a German national survey. J Eur Acad Dermatol Venereol

2009, 23(6):683-691.

Page 193: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

181

120. King MC, Lee GM, Spinner NB, Thomson G, Wrensch MR: Genetic epidemiology. Annu Rev Public Health 1984, 5:1-52.

121. Elston RC: Segregation analysis. Adv Hum Genet 1981, 11:63-120, 372-123. 122. Allison AK (ed.): Determining Genetic Component of a Disease, 1st edn. Hoboken,

New Jersey: John Wiley & Sons; 2006. 123. Dawnteare M, Barrett J: Genetic linkage studies. The Lancet 2005, 366(9490):1036-

1044. 124. Terwilliger JD (ed.): Linkage analysis model based. Chichester: Wiley; 2002. 125. Martin ER (ed.): Linkage Disequilibrium and Association Analysis. Hoboken:

Wiley-Liss; 2006. 126. Cordell H, Clayton D: Genetic association studies. The Lancet 2005,

366(9491):1121-1131. 127. Risch N, Merikangas K: The future of genetic studies of complex human diseases.

Science 1996, 273(5281):1516-1517. 128. Hirschhorn JN, Daly MJ: Genome-wide association studies for common diseases

and complex traits. Nat Rev Genet 2005, 6(2):95-108. 129. Risch N: Linkage strategies for genetically complex traits. III. The effect of

marker polymorphism on analysis of affected relative pairs. Am J Hum Genet

1990, 46(2):242-253. 130. Risch N: Linkage strategies for genetically complex traits. II. The power of

affected relative pairs. Am J Hum Genet 1990, 46(2):229-241. 131. Risch N: Linkage strategies for genetically complex traits. I. Multilocus models.

Am J Hum Genet 1990, 46(2):222-228. 132. Hellgren L (ed.): Psoriasis. Stockholm, Sweden: Almqvist & Wiksell; 1967. 133. Lomholt G (ed.): Psoriasis: Prevalence, Spontaneous Course, and Genetics.

Copenhagen, Denmark: GEC GAD; 1963. 134. Elder JT, Nair RP, Guo SW, Henseler T, Christophers E, Voorhees JJ: The genetics

of psoriasis. Arch Dermatol 1994, 130(2):216-224. 135. Brandrup F, Holm N, Grunnet N, Henningsen K, Hansen HE: Psoriasis in

monozygotic twins: variations in expression in individuals with identical genetic constitution. Acta Derm Venereol 1982, 62(3):229-236.

136. Duffy DL, Spelman LS, Martin NG: Psoriasis in Australian twins. J Am Acad

Dermatol 1993, 29(3):428-434. 137. Farber EM, Nall ML, Watson W: Natural history of psoriasis in 61 twin pairs.

Arch Dermatol 1974, 109(2):207-211. 138. Bhalerao J, Bowcock AM: The genetics of psoriasis: a complex disorder of the

skin and immune system. Hum Mol Genet 1998, 7(10):1537-1545. 139. Pietrzyk JJ, Turowski G, Kapinska-Mrowka M, Rozanski B: Family studies in

psoriasis. I. Complex segregation analysis. Arch Dermatol Res 1982, 273(3-4):287-294.

140. Moll JM, Wright V: Familial occurrence of psoriatic arthritis. Ann Rheum Dis

1973, 32(3):181-201. 141. Myers A: Recurrence risk for psoriasis and psoriatic arthritis within sibships.

Rheumatology 2005, 44(6):773-776.

Page 194: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

182

142. Karason A, Love TJ, Gudbjornsson B: A strong heritability of psoriatic arthritis

over four generations--the Reykjavik Psoriatic Arthritis Study. Rheumatology

(Oxford) 2009, 48(11):1424-1428. 143. Chandran V, Schentag CT, Brockbank JE, Pellett FJ, Shanmugarajah S, Toloza SMA,

Rahman P, Gladman DD: Familial aggregation of psoriatic arthritis. Annals of the

Rheumatic Diseases 2009, 68(5):664-667. 144. Pedersen OB, Svendsen AJ, Ejstrup L, Skytthe A, Junker P: On the heritability of

psoriatic arthritis. Disease concordance among monozygotic and dizygotic twins. Annals of the Rheumatic Diseases 2008, 67(10):1417-1421.

145. Elder JT, Nair RP, Henseler T, Jenisch S, Stuart P, Chia N, Christophers E, Voorhees JJ: The genetics of psoriasis 2001: the odyssey continues. Arch Dermatol 2001, 137(11):1447-1454.

146. Abele DC, Dobson RL, Graham JB: Heredity and Psoriasis. Study of a Large

Family. Arch Dermatol 1963, 88:38-47. 147. Swanbeck G, Inerot A, Martinsson T, Enerback C, Enlund F, Samuelsson L, Yhr M,

Wahlstrom J: Genetic counselling in psoriasis: empirical data on psoriasis among

first-degree relatives of 3095 psoriatic probands. Br J Dermatol 1997, 137(6):939-942.

148. Rahman P, Gladman DD, Schentag CT, Petronis A: Excessive paternal

transmission in psoriatic arthritis. Arthritis Rheum 1999, 42(6):1228-1231. 149. Traupe H, van Gurp PJ, Happle R, Boezeman J, van de Kerkhof PC: Psoriasis

vulgaris, fetal growth, and genomic imprinting. Am J Med Genet 1992, 42(5):649-654.

150. Reik W: Genomic imprinting and genetic disorders in man. Trends Genet 1989, 5(10):331-336.

151. Burden AD, Javed S, Bailey M, Hodgins M, Connor M, Tillman D: Genetics of

psoriasis: paternal inheritance and a locus on chromosome 6p. J Invest Dermatol

1998, 110(6):958-960. 152. Karason A, Gudjonsson JE, Upmanyu R, Antonsdottir AA, Hauksson VB,

Runasdottir EH, Jonsson HH, Gudbjartsson DF, Frigge ML, Kong A et al: A

susceptibility gene for psoriatic arthritis maps to chromosome 16q: evidence for imprinting. Am J Hum Genet 2003, 72(1):125-131.

153. Winchester R (ed.): Genetics of Psoriatic Arthritis, 1st edn. Philadelphia: Mosby; 2007.

154. Hauser ER, Haines J, Goldgar EG (eds.): Nonparametric Linkage Analysis, 1st edn. Hoboken: Wiley-Liss; 2006.

155. Enlund F, Samuelsson L, Enerback C, Inerot A, Wahlstrom J, Yhr M, Torinsson A, Martinsson T, Swanbeck G: Analysis of three suggested 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(1):2-8.

156. 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.

Page 195: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

183

157. Matthews D, Fry L, Powles A, Weber J, McCarthy M, Fisher E, Davies K, Williamson R: Evidence that a locus for familial psoriasis maps to chromosome

4q. Nat Genet 1996, 14(2):231-233. 158. Capon F, Semprini S, Chimenti S, Fabrizi G, Zambruno G, Murgia S, Carcassi C,

Fazio M, Mingarelli R, Dallapiccola B et al: Fine mapping of the PSORS4

psoriasis susceptibility region on chromosome 1q21. J Invest Dermatol 2001, 116(5):728-730.

159. Enlund F, Samuelsson L, Enerback C, Inerot A, Wahlstrom J, Yhr M, Torinsson A, Riley J, Swanbeck G, Martinsson T: Psoriasis susceptibility locus in chromosome

region 3q21 identified in patients from southwest Sweden. Eur J Hum Genet 1999, 7(7):783-790.

160. Hensen P, Windemuth C, Huffmeier U, Ruschendorf F, Stadelmann A, Hoppe V, Fenneker D, Stander M, Schmitt-Egenolf M, Wienker TF et al: Association scan of

the novel psoriasis susceptibility region on chromosome 19: evidence for both susceptible and protective loci. Exp Dermatol 2003, 12(4):490-496.

161. Veal CD, Clough RL, Barber RC, Mason S, Tillman D, Ferry B, Jones AB, Ameen M, Balendran N, Powis SH et al: Identification of a novel psoriasis susceptibility

locus at 1p and evidence of epistasis between PSORS1 and candidate loci. J Med

Genet 2001, 38(1):7-13. 162. The International Psoriasis Genetics Study: assessing linkage to 14 candidate

susceptibility loci in a cohort of 942 affected sib pairs. Am J Hum Genet 2003, 73(2):430-437.

163. Sagoo GS, Tazi-Ahnini R, Barker JW, Elder JT, Nair RP, Samuelsson L, Traupe H, Trembath RC, Robinson DA, Iles MM: Meta-analysis of genome-wide studies of

psoriasis susceptibility reveals linkage to chromosomes 6p21 and 4q28-q31 in Caucasian and Chinese Hans population. J Invest Dermatol 2004, 122(6):1401-1405.

164. Asumalahti K, Laitinen T, Lahermo P, Suomela S, Itkonen-Vatjus R, Jansen C, Karvonen J, Karvonen SL, Reunala T, Snellman E et al: Psoriasis susceptibility

locus on 18p revealed by genome scan in Finnish families not associated with PSORS1. J Invest Dermatol 2003, 121(4):735-740.

165. Nair RP, Henseler T, Jenisch S, Stuart P, Bichakjian CK, Lenk W, Westphal E, Guo SW, Christophers E, Voorhees JJ 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(8):1349-1356.

166. Henseler T, Christophers E: Psoriasis of early and late onset: characterization of

two types of psoriasis vulgaris. J Am Acad Dermatol 1985, 13(3):450-456. 167. Russell TJ, Schultes LM, Kuban DJ: Histocompatibility (HL-A) antigens

associated with psoriasis. N Engl J Med 1972, 287(15):738-740. 168. Tiilikainen A, Lassus A, Karvonen J, Vartiainen P, Julin M: Psoriasis and HLA-

Cw6. Br J Dermatol 1980, 102(2):179-184. 169. Balendran N, Clough RL, Arguello JR, Barber R, Veal C, Jones AB, Rosbotham JL,

Little AM, Madrigal A, Barker JN et al: Characterization of the major

susceptibility region for psoriasis at chromosome 6p21.3. J Invest Dermatol 1999, 113(3):322-328.

Page 196: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

184

170. Feng BJ, Sun LD, Soltani-Arabshahi R, Bowcock AM, Nair RP, Stuart P, Elder JT, Schrodi SJ, Begovich AB, Abecasis GR et al: Multiple Loci within the major

histocompatibility complex confer risk of psoriasis. PLoS Genet 2009, 5(8):e1000606.

171. Fan X, Yang S, Huang W, Wang ZM, Sun LD, Liang YH, Gao M, Ren YQ, Zhang KY, Du WH et al: Fine mapping of the psoriasis susceptibility locus PSORS1

supports HLA-C as the susceptibility gene in the Han Chinese population. PLoS

Genet 2008, 4(3):e1000038. 172. Elder JT: PSORS1: linking genetics and immunology. J Invest Dermatol 2006,

126(6):1205-1206. 173. Marcusson JA, Johannesson A, Moller E: HLA-A,B,C and DR antigens in

psoriasis. Tissue Antigens 1981, 17(5):525-529. 174. Jenisch S, Westphal E, Nair RP, Stuart P, Voorhees JJ, Christophers E, Kronke M,

Elder JT, Henseler T: Linkage disequilibrium analysis of familial psoriasis:

identification of multiple disease-associated MHC haplotypes. Tissue Antigens

1999, 53(2):135-146. 175. Yang S, Ge HS, Zhang AP, Wei SC, Gao M, Wang HY, Chen JJ, Li M, Liang YH,

He PP et al: Haplotype associations of the MHC with psoriasis vulgaris in

Chinese Hans. Clin Exp Dermatol 2004, 29(4):399-405. 176. Gazit E, Brenner S, Efter T, Orgad S, Mizrachi Y, Krakowski A: HLA antigens in

patients with psoriasis. Tissue Antigens 1978, 12(3):195-199. 177. Tiwari JL, Lowe NJ, Abramovits W, Hawkins BR, Park MS: Association of

psoriasis with HLA-DR7. Br J Dermatol 1982, 106(2):227-230. 178. Gudjonsson JE, Karason A, Antonsdottir AA, Runarsdottir EH, Gulcher JR,

Stefansson K, Valdimarsson H: HLA-Cw6-positive and HLA-Cw6-negative

patients with Psoriasis vulgaris have distinct clinical features. J Invest Dermatol

2002, 118(2):362-365. 179. Enerback C, Martinsson T, Inerot A, Wahlstrom J, Enlund F, Yhr M, Swanbeck G:

Evidence that HLA-Cw6 determines early onset of psoriasis, obtained using sequence-specific primers (PCR-SSP). Acta Derm Venereol 1997, 77(4):273-276.

180. Nair RP, Stuart PE, Nistor I, Hiremagalore R, Chia NV, Jenisch S, Weichenthal M, Abecasis GR, Lim HW, Christophers E et al: Sequence and haplotype analysis

supports HLA-C as the psoriasis susceptibility 1 gene. Am J Hum Genet 2006, 78(5):827-851.

181. Schmitt-Egenolf M, Eiermann TH, Boehncke WH, Stander M, Sterry W: Familial

juvenile onset psoriasis is associated with the human leukocyte antigen (HLA)

class I side of the extended haplotype Cw6-B57-DRB1*0701-DQA1*0201-DQB1*0303: a population- and family-based study. J Invest Dermatol 1996, 106(4):711-714.

182. Jenisch S, Henseler T, Nair RP, Guo SW, Westphal E, Stuart P, Kronke M, Voorhees JJ, Christophers E, Elder JT: Linkage analysis of human leukocyte antigen (HLA)

markers in familial psoriasis: strong disequilibrium effects provide evidence for a major determinant in the HLA-B/-C region. Am J Hum Genet 1998, 63(1):191-199.

183. Asumalahti K, Laitinen T, Itkonen-Vatjus R, Lokki ML, Suomela S, Snellman E, Saarialho-Kere U, Kere J: A candidate gene for psoriasis near HLA-C, HCR

Page 197: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

185

(Pg8), is highly polymorphic with a disease-associated susceptibility allele. Hum

Mol Genet 2000, 9(10):1533-1542. 184. Tazi Ahnini R, Camp NJ, Cork MJ, Mee JB, Keohane SG, Duff GW, di Giovine FS:

Novel genetic association between the corneodesmosin (MHC S) gene and susceptibility to psoriasis. Hum Mol Genet 1999, 8(6):1135-1140.

185. Chia NV, Stuart P, Nair RP, Henseler T, Jenisch S, Lim HW, Christophers E, Voorhees JJ, Elder JT: Variations in the HCR (Pg8) gene are unlikely to be causal

for familial psoriasis. J Invest Dermatol 2001, 116(5):823-824. 186. Enerback C, Nilsson S, Enlund F, Inerot A, Samuelsson L, Wahlstrom J, Swanbeck

G, Martinsson T: Stronger association with HLA-Cw6 than with corneodesmosin

(S-gene) polymorphisms in Swedish psoriasis patients. Arch Dermatol Res 2000, 292(11):525-530.

187. Zhang XJ, Huang W, Yang S, Sun LD, Zhang FY, Zhu QX, Zhang FR, Zhang C, Du WH, Pu XM et al: Psoriasis genome-wide association study identifies

susceptibility variants within LCE gene cluster at 1q21. Nat Genet 2009, 41(2):205-210.

188. Gerber LH, Murray CL, Perlman SG, Barth WF, Decker JL, Nigra TA, Mann DL: Human lymphocyte antigens characterizing psoriatic arthritis and its subtypes. J

Rheumatol 1982, 9(5):703-707. 189. Espinoza LR, Vasey FB, Gaylord SW, Dietz C, Bergen L, Bridgeford P, Germain BF:

Histocompatibility typing in the seronegative spondyloarthropathies: a survey. Semin Arthritis Rheum 1982, 11(3):375-381.

190. Eastmond CJ, Woodrow JC: The HLA system and the arthropathies associated

with psoriasis. Ann Rheum Dis 1977, 36(2):112-120. 191. Lopez-Larrea C, Torre Alonso JC, Rodriguez Perez A, Coto E: HLA antigens in

psoriatic arthritis subtypes of a Spanish population. Ann Rheum Dis 1990, 49(5):318-319.

192. McHugh NJ, Laurent MR, Treadwell BL, Tweed JM, Dagger J: Psoriatic arthritis:

clinical subgroups and histocompatibility antigens. Ann Rheum Dis 1987, 46(3):184-188.

193. 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. Ann Rheum Dis 2008, 67(5):677-682.

194. Gudjonsson JE, Elder JT: Psoriasis: epidemiology. Clin Dermatol 2007, 25(6):535-546.

195. Kantor SM, Hsu SH, Bias WB, Arnett FC: Clinical and immunogenetic subsets of

psoriatic arthritis. Clin Exp Rheumatol 1984, 2(2):105-109. 196. Beaulieu AD, Roy R, Mathon G, Morissette J, Latulippe L, Lang JY, Mathieu JP,

Brunet D, Hebert J, Archambault H et al: Psoriatic arthritis: risk factors for

patients with psoriasis - a study based on histocompatibility antigen frequencies. J Rheumatol 1983, 10(4):633-636.

197. McKendry RJ, Sengar DP, DesGroseilliers JP, Dunne JV: Frequency of HLA

antigens in patients with psoriasis or psoriatic arthritis. Can Med Assoc J 1984, 130(4):411-415.

Page 198: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

186

198. Gladman DD, Farewell VT, Pellett F, Schentag C, Rahman P: HLA is a candidate

region for psoriatic arthritis. evidence for excessive HLA sharing in sibling pairs. Hum Immunol 2003, 64(9):887-889.

199. Beck S, Trowsdale J: The human major histocompatability complex: lessons from

the DNA sequence. Annu Rev Genomics Hum Genet 2000, 1:117-137. 200. Leder RO, Mansbridge JN, Hallmayer J, Hodge SE: Familial psoriasis and HLA-B:

unambiguous support for linkage in 97 published families. Hum Hered 1998, 48(4):198-211.

201. Holm SJ, Sanchez F, Carlen LM, Mallbris L, Stahle M, O'Brien KP: HLA-Cw*0602

associates more strongly to psoriasis in the Swedish population than variants of the novel 6p21.3 gene PSORS1C3. Acta Derm Venereol 2005, 85(1):2-8.

202. Helms C, Saccone NL, Cao L, Daw JA, Cao K, Hsu TM, Taillon-Miller P, Duan S, Gordon D, Pierce B et al: Localization of PSORS1 to a haplotype block harboring

HLA-C and distinct from corneodesmosin and HCR. Hum Genet 2005, 118(3-4):466-476.

203. Chang YT, Chou CT, Shiao YM, Lin MW, Yu CW, Chen CC, Huang CH, Lee DD, Liu HN, Wang WJ et al: Psoriasis vulgaris in Chinese individuals is associated

with PSORS1C3 and CDSN genes. Br J Dermatol 2006, 155(4):663-669. 204. Gonzalez S, Martinez-Borra J, Del Rio JS, Santos-Juanes J, Lopez-Vazquez A,

Blanco-Gelaz M, Lopez-Larrea C: The OTF3 gene polymorphism confers

susceptibility to psoriasis independent of the association of HLA-Cw*0602. J

Invest Dermatol 2000, 115(5):824-828. 205. Asumalahti K, Veal C, Laitinen T, Suomela S, Allen M, Elomaa O, Moser M, de Cid

R, Ripatti S, Vorechovsky I et al: Coding haplotype analysis supports HCR as the

putative susceptibility gene for psoriasis at the MHC PSORS1 locus. Hum Mol

Genet 2002, 11(5):589-597. 206. Elomaa O, Majuri I, Suomela S, Asumalahti K, Jiao H, Mirzaei Z, Rozell B,

Dahlman-Wright K, Pispa J, Kere J et al: Transgenic mouse models support HCR

as an effector gene in the PSORS1 locus. Hum Mol Genet 2004, 13(15):1551-1561. 207. Holm SJ, Carlen LM, Mallbris L, Stahle-Backdahl M, O'Brien KP: Polymorphisms

in the SEEK1 and SPR1 genes on 6p21.3 associate with psoriasis in the Swedish population. Exp Dermatol 2003, 12(4):435-444.

208. Jenisch S, Koch S, Henseler T, Nair RP, Elder JT, Watts CE, Westphal E, Voorhees JJ, Christophers E, Kronke M: Corneodesmosin gene polymorphism demonstrates

strong linkage disequilibrium with HLA and association with psoriasis vulgaris. Tissue Antigens 1999, 54(5):439-449.

209. Schmitt-Egenolf M, Windemuth C, Hennies HC, Albis-Camps M, von Engelhardt B, Wienker T, Reis A, Traupe H, Blasczyk R: Comparative association analysis

reveals that corneodesmosin is more closely associated with psoriasis than HLA-Cw*0602-B*5701 in German families. Tissue Antigens 2001, 57(5):440-446.

210. Hohler T, Kruger A, Schneider PM, Schopf RE, Knop J, Rittner C, Meyer zum Buschenfelde KH, Marker-Hermann E: A TNF-alpha promoter polymorphism is

associated with juvenile onset psoriasis and psoriatic arthritis. J Invest Dermatol

1997, 109(4):562-565. 211. Reich K, Westphal G, Schulz T, Muller M, Zipprich S, Fuchs T, Hallier E, Neumann

C: Combined analysis of polymorphisms of the tumor necrosis factor-alpha and

Page 199: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

187

interleukin-10 promoter regions and polymorphic xenobiotic metabolizing enzymes in psoriasis. J Invest Dermatol 1999, 113(2):214-220.

212. Mossner R, Kingo K, Kleensang A, Kruger U, Konig IR, Silm H, Westphal GA, Reich K: Association of TNF -238 and -308 promoter polymorphisms with

psoriasis vulgaris and psoriatic arthritis but not with pustulosis palmoplantaris. J Invest Dermatol 2005, 124(1):282-284.

213. Butt C, Rahman P, Siannis F, Farewell VT, Gladman DD: Corneodesmosin

polymorphisms in psoriatic arthritis. Rheumatology (Oxford) 2005, 44(5):684-685. 214. Gonzalez S, Martinez-Borra J, Torre-Alonso JC, Gonzalez-Roces S, Sanchez del Rio

J, Rodriguez Perez A, Brautbar C, Lopez-Larrea C: The MICA-A9 triplet repeat

polymorphism in the transmembrane region confers additional susceptibility to

the development of psoriatic arthritis and is independent of the association of Cw*0602 in psoriasis. Arthritis Rheum 1999, 42(5):1010-1016.

215. Gonzalez S, Brautbar C, Martinez-Borra J, Lopez-Vazquez A, Segal R, Blanco-Gelaz MA, Enk CD, Safriman C, Lopez-Larrea C: Polymorphism in MICA rather than

HLA-B/C genes is associated with psoriatic arthritis in the Jewish population. Hum Immunol 2001, 62(6):632-638.

216. Gonzalez S, Martinez-Borra J, Lopez-Vazquez A, Garcia-Fernandez S, Torre-Alonso JC, Lopez-Larrea C: MICA rather than MICB, TNFA, or HLA-DRB1 is

associated with susceptibility to psoriatic arthritis. J Rheumatol 2002, 29(5):973-978.

217. Grubic Z, Peric P, Eeeuk-Jelicic E, Zunec R, Stingl K, Curkovic B, Kerhin-Brkljacic V: The MICA-A4 triplet repeats polymorphism in the transmembrane region

confers additional risk for development of psoriatic arthritis in the Croatian population. Eur J Immunogenet 2004, 31(2):93-98.

218. Fojtikova M, Stolfa J, Novota P, Cejkova P, Dostal C, Cerna M: HLA-Cw*06 class I

region rather than MICA is associated with psoriatic arthritis in Czech population. Rheumatol Int 2009, 29(11):1293-1299.

219. Pollock R, Chandran v, Barrett J, Eder L, Pellett F, Yao C, Lino M, Shanmugarajah S, Farewell V, Gladman DD: MICA*016 Differentiates Skin And Joint

Manifestations Of Psoriatic Disease. 220. Ritchlin C, Haas-Smith SA, Hicks D, Cappuccio J, Osterland CK, Looney RJ:

Patterns of cytokine production in psoriatic synovium. J Rheumatol 1998, 25(8):1544-1552.

221. Danning CL, Illei GG, Hitchon C, Greer MR, Boumpas DT, McInnes IB: Macrophage-derived cytokine and nuclear factor kappaB p65 expression in synovial membrane and skin of patients with psoriatic arthritis. Arthritis Rheum

2000, 43(6):1244-1256. 222. Gladman DD: Traditional and newer therapeutic options for psoriatic arthritis:

an evidence-based review. Drugs 2005, 65(9):1223-1238. 223. Rahman P, Siannis F, Butt C, Farewell V, Peddle L, Pellett F, Gladman D: TNFalpha

polymorphisms and risk of psoriatic arthritis. Ann Rheum Dis 2006, 65(7):919-923.

224. Lanier LL: NK cell recognition. Annu Rev Immunol 2005, 23:225-274.

Page 200: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

188

225. Wilson MJ, Torkar M, Haude A, Milne S, Jones T, Sheer D, Beck S, Trowsdale J: Plasticity in the organization and sequences of human KIR/ILT gene families. Proc Natl Acad Sci U S A 2000, 97(9):4778-4783.

226. Gardiner CM: Killer cell immunoglobulin-like receptors on NK cells: the how,

where and why. Int J Immunogenet 2008, 35(1):1-8. 227. Martin MP, Nelson G, Lee JH, Pellett F, Gao X, Wade J, Wilson MJ, Trowsdale J,

Gladman D, Carrington M: Cutting edge: susceptibility to psoriatic arthritis:

influence of activating killer Ig-like receptor genes in the absence of specific HLA-C alleles. J Immunol 2002, 169(6):2818-2822.

228. Nelson GW, Martin MP, Gladman D, Wade J, Trowsdale J, Carrington M: Cutting

edge: heterozygote advantage in autoimmune disease: hierarchy of

protection/susceptibility conferred by HLA and killer Ig-like receptor combinations in psoriatic arthritis. J Immunol 2004, 173(7):4273-4276.

229. Suzuki Y, Hamamoto Y, Ogasawara Y, Ishikawa K, Yoshikawa Y, Sasazuki T, Muto M: Genetic polymorphisms of killer cell immunoglobulin-like receptors are

associated with susceptibility to psoriasis vulgaris. J Invest Dermatol 2004, 122(5):1133-1136.

230. Luszczek W, Manczak M, Cislo M, Nockowski P, Wisniewski A, Jasek M, Kusnierczyk P: Gene for the activating natural killer cell receptor, KIR2DS1, is

associated with susceptibility to psoriasis vulgaris. Hum Immunol 2004, 65(7):758-766.

231. Williams F, Meenagh A, Sleator C, Cook D, Fernandez-Vina M, Bowcock AM, Middleton D: Activating killer cell immunoglobulin-like receptor gene KIR2DS1

is associated with psoriatic arthritis. Hum Immunol 2005, 66(7):836-841. 232. Cella M, Longo A, Ferrara GB, Strominger JL, Colonna M: NK3-specific natural

killer cells are selectively inhibited by Bw4-positive HLA alleles with isoleucine 80. J Exp Med 1994, 180(4):1235-1242.

233. Carr WH, Rosen DB, Arase H, Nixon DF, Michaelsson J, Lanier LL: Cutting Edge:

KIR3DS1, a gene implicated in resistance to progression to AIDS, encodes a

DAP12-associated receptor expressed on NK cells that triggers NK cell activation. J Immunol 2007, 178(2):647-651.

234. Lopez-Larrea C, Blanco-Gelaz MA, Torre-Alonso JC, Bruges Armas J, Suarez-Alvarez B, Pruneda L, Couto AR, Gonzalez S, Lopez-Vazquez A, Martinez-Borra J: Contribution of KIR3DL1/3DS1 to ankylosing spondylitis in human leukocyte antigen-B27 Caucasian populations. Arthritis Res Ther 2006, 8(4):R101.

235. Harvey D, Pointon JJ, Sleator C, Meenagh A, Farrar C, Sun JY, Senitzer D, Middleton D, Brown MA, Wordsworth BP: Analysis of killer immunoglobulin-like

receptor genes in ankylosing spondylitis. Annals of the Rheumatic Diseases 2009, 68(4):595-598.

236. Martin MP, Gao X, Lee JH, Nelson GW, Detels R, Goedert JJ, Buchbinder S, Hoots K, Vlahov D, Trowsdale J et al: Epistatic interaction between KIR3DS1 and HLA-

B delays the progression to AIDS. Nat Genet 2002, 31(4):429-434. 237. Sanchez FO, Linga Reddy MV, Mallbris L, Sakuraba K, Stahle M, Alarcon-Riquelme

ME: IFN-regulatory factor 5 gene variants interact with the class I MHC locus in

the Swedish psoriasis population. J Invest Dermatol 2008, 128(7):1704-1709.

Page 201: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

189

238. Rahman P, Bartlett S, Siannis F, Pellett FJ, Farewell VT, Peddle L, Schentag CT, Alderdice CA, Hamilton S, Khraishi M et al: CARD15: a pleiotropic autoimmune

gene that confers susceptibility to psoriatic arthritis. Am J Hum Genet 2003, 73(3):677-681.

239. Borgiani P, Vallo L, D'Apice MR, Giardina E, Pucci S, Capon F, Nistico S, Chimenti S, Pallone F, Novelli G: Exclusion of CARD15/NOD2 as a candidate susceptibility

gene to psoriasis in the Italian population. Eur J Dermatol 2002, 12(6):540-542. 240. Nair RP, Stuart P, Ogura Y, Inohara N, Chia NV, Young L, Henseler T, Jenisch S,

Christophers E, Voorhees JJ et al: Lack of association between NOD2 3020InsC

frameshift mutation and psoriasis. J Invest Dermatol 2001, 117(6):1671-1672. 241. Lascorz J, Burkhardt H, Huffmeier U, Bohm B, Schurmeyer-Horst F, Lohmann J,

Stander M, Wendler J, Kelsch R, Baumann C et al: Lack of genetic association of

the three more common polymorphisms of CARD15 with psoriatic arthritis and psoriasis in a German cohort. Ann Rheum Dis 2005, 64(6):951-954.

242. Rahman P, Sun S, Peddle L, Snelgrove T, Melay W, Greenwood C, Gladman D: Association between the interleukin-1 family gene cluster and psoriatic arthritis. Arthritis Rheum 2006, 54(7):2321-2325.

243. Chang YT, Chou CT, Yu CW, Lin MW, Shiao YM, Chen CC, Huang CH, Lee DD, Liu HN, Wang WJ et al: Cytokine gene polymorphisms in Chinese patients with

psoriasis. Br J Dermatol 2007, 156(5):899-905. 244. Rahman P, Inman RD, Maksymowych WP, Reeve JP, Peddle L, Gladman DD:

Association of interleukin 23 receptor variants with psoriatic arthritis. J

Rheumatol 2009, 36(1):137-140. 245. Cargill M, Schrodi SJ, Chang M, Garcia VE, Brandon R, Callis KP, Matsunami N,

Ardlie KG, Civello D, Catanese JJ et al: A large-scale genetic association study

confirms IL12B and leads to the identification of IL23R as psoriasis-risk genes. Am J Hum Genet 2007, 80(2):273-290.

246. Nair RP, Duffin KC, Helms C, Ding J, Stuart PE, Goldgar D, Gudjonsson JE, Li Y, Tejasvi T, Feng BJ et al: Genome-wide scan reveals association of psoriasis with

IL-23 and NF-kappaB pathways. Nat Genet 2009, 41(2):199-204. 247. Wu Y, Lu Z, Chen Y, Xue F, Chen X, Zheng J: Replication of association between

interleukin-23 receptor (IL-23R) and its ligand (IL-12B) polymorphisms and psoriasis in the Chinese Han population. Hum Immunol 2010, 71(12):1255-1258.

248. Huffmeier U, Lascorz J, Bohm B, Lohmann J, Wendler J, Mossner R, Reich K, Traupe H, Kurrat W, Burkhardt H et al: Genetic variants of the IL-23R pathway:

association with psoriatic arthritis and psoriasis vulgaris, but no specific risk factor for arthritis. J Invest Dermatol 2009, 129(2):355-358.

249. Duffin KC, Freeny IC, Schrodi SJ, Wong B, Feng BJ, Soltani-Arabshahi R, Rakkhit T, Goldgar DE, Krueger GG: Association between IL13 polymorphisms and

psoriatic arthritis is modified by smoking. J Invest Dermatol 2009, 129(12):2777-2783.

250. Eder L, Chandran V, Pellett F, Pollock R, Shanmugarajah S, Gladman D: The

association between IL-13 polymorphism with psoriatic arthritis among psoriasis patients. Artthritis Rheum 2010, 62S(10):S830.

Page 202: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

190

251. Bowes J, Ho P, Salah S, Warren RB, Korendowych E, McHugh N, Fitzgerald O, Packham J, Morgan AW, Griffiths C et al: IL13 is a risk locus for psoriatic

arthritis but not psoriasis vulgaris.Arthritis Rheum 2010, 62S(10):S348. 252. Strange A, Capon F, Spencer CC, Knight J, Weale ME, Allen MH, Barton A, Band

G, Bellenguez C, Bergboer JG et al: A genome-wide association study identifies

new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet 2010, 42(11):985-990.

253. Ellinghaus E, Ellinghaus D, Stuart PE, Nair RP, Debrus S, Raelson JV, Belouchi M, Fournier H, Reinhard C, Ding J et al: Genome-wide association study identifies a

psoriasis susceptibility locus at TRAF3IP2. Nat Genet 2010, 42(11):991-995. 254. Liu Y, Helms C, Liao W, Zaba LC, Duan S, Gardner J, Wise C, Miner A, Malloy MJ,

Pullinger CR et al: A genome-wide association study of psoriasis and psoriatic

arthritis identifies new disease loci. PLoS Genet 2008, 4(3):e1000041. 255. Huffmeier U, Uebe S, Ekici AB, Bowes J, Giardina E, Korendowych E, Juneblad K,

Apel M, McManus R, Ho P et al: Common variants at TRAF3IP2 are associated

with susceptibility to psoriatic arthritis and psoriasis. Nat Genet 2010, 42(11):996-999.

256. Gladman DD, Schentag CT, Tom BD, Chandran V, Brockbank J, Rosen C, Farewell VT: Development and initial validation of a screening questionnaire for psoriatic

arthritis: the Toronto Psoriatic Arthritis Screen (ToPAS). Ann Rheum Dis 2009, 68(4):497-501.

257. Gladman DD, Inman RD, Cook RJ, van der Heijde D, Landewe RB, Braun J, Davis JC, Mease P, Brandt J, Vargas RB et al: International spondyloarthritis

interobserver reliability exercise--the INSPIRE study: I. Assessment of spinal measures. J Rheumatol 2007, 34(8):1733-1739.

258. Cassell S, Bieber JD, Rich P, Tutuncu Z, Lee SJ, Kalunian K, Wu CW, Kavanaugh A: The modified Nail Psoriasis Severity Index: Validation of an instrument to

assess psoriatic nail involvement in patients with psoriatic arthritis. J Rheumatol

2007, 34(1):123-129. 259. Rahman P, Gladman DD, Cook RJ, Zhou Y, Young G, Salonen D: Radiological

assessment in psoriatic arthritis. Br J Rheumatol 1998, 37(7):760-765. 260. Spoorenberg A, de Vlam K, van der Linden S, Dougados M, Mielants H, van de

Tempel H, van der Heijde D: Radiological scoring methods in ankylosing

spondylitis. Reliability and change over 1 and 2 years. J Rheumatol 2004, 31(1):125-132.

261. van der Linden S, Valkenburg HA, Cats A: Evaluation of diagnostic criteria for

ankylosing spondylitis. A proposal for modification of the New York criteria. Arthritis Rheum 1984, 27(4):361-368.

262. Zachariae H, Zachariae R, Blomqvist K, Davidsson S, Molin L, Mork C, Sigurgeirsson B: Quality of life and prevalence of arthritis reported by 5,795

members of the Nordic Psoriasis Associations. Data from the Nordic Quality of Life Study. Acta Derm Venereol 2002, 82(2):108-113.

263. Kane D, Stafford L, Bresnihan B, FitzGerald O: A prospective, clinical and

radiological study of early psoriatic arthritis: an early synovitis clinic experience. Rheumatology (Oxford) 2003, 42(12):1460-1468.

Page 203: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

191

264. Rahman P: Genetic epidemiology of psoriasis and psoriatic arthritis. Annals of the

Rheumatic Diseases 2005, 64(suppl_2):ii37-ii39. 265. Begovich AB, Erlich HA: HLA typing for bone marrow transplantation. New

polymerase chain reaction-based methods. JAMA 1995, 273(7):586-591. 266. Dupont WD, Plummer WD: "Power and Sample Size Calculations: A Review and

Computer Program". Controlled Clinical Trials 1990, 11:116-128. 267. Benjamini Y, Hochberg Y: Controlling the False Discovery Rate: A practical and

powerful approach to multiple testing. J R Stat Soc B 1995, 57(1):289-300. 268. van den Oord EJCG: Controlling false discoveries in genetic studies. American

Journal of Medical Genetics Part B: Neuropsychiatric Genetics 2008, 147B(5):637-644.

269. Ghosh S, Collins FS: The geneticist's approach to complex disease. Annu Rev Med

1996, 47:333-353. 270. Deighton C, Criswell LA: Recent advances in the genetics of rheumatoid arthritis.

Curr Rheumatol Rep 2006, 8(5):394-400. 271. SAS/Genetics. Version 9.1.3. SAS Institute, Cary, NC. In.

272. Excoffier L, Slatkin M: Maximum-likelihood estimation of molecular haplotype

frequencies in a diploid population. Mol Biol Evol 1995, 12(5):921-927. 273. Hawley ME, Kidd KK: HAPLO: a program using the EM algorithm to estimate

the frequencies of multi-site haplotypes. J Hered 1995, 86(5):409-411. 274. Long JC, Williams RC, Urbanek M: An E-M algorithm and testing strategy for

multiple-locus haplotypes. Am J Hum Genet 1995, 56(3):799-810. 275. Laird NM, Horvath S, Xu X: Implementing a unified approach to family-based

tests of association. Genet Epidemiol 2000, 19 Suppl 1:S36-42. 276. Rabinowitz D, Laird N: A unified approach to adjusting association tests for

population admixture with arbitrary pedigree structure and arbitrary missing marker information. Hum Hered 2000, 50(4):211-223.

277. Spielman RS, McGinnis RE, Ewens WJ: Transmission test for linkage

disequilibrium: the insulin gene region and insulin-dependent diabetes mellitus (IDDM). Am J Hum Genet 1993, 52(3):506-516.

278. Lake SL, Blacker D, Laird NM: Family-based tests of association in the presence

of linkage. Am J Hum Genet 2000, 67(6):1515-1525. 279. Malhotra AK, Goldman D: Benefits and pitfalls encountered in psychiatric genetic

association studies.Biological Psychiatry 1999, 45:544–550. 280. Hutchison KE, Stallings M, McGeary J, Bryan A: Population Stratification in the

Candidate Gene Study: Fatal Threat or Red Herring?Psychological Bulletin

2004, 130(1):66-79. 281. Wacholder S, Rothman N, Caporaso N: Population stratification in epidemiologic

studies of common genetic variants and cancer: Quantification of bias.Journal of

the National Cancer Institute 2000, 92:1151–1158. 282. Bacanu SA, Devlin B, & Roeder B: The power of genomic control.American

Journal of Human Genetics 2000, 66:1933–1944. 283. Khlat M, Cazes MH, Ge´nin E, Guiguet M: Robustness of Case-Control Studies of

Genetic Factors to Population Stratification: Magnitude of Bias and Type I Error Cancer Epidemiol Biomarkers Prev 2004, 13(10):1660-1664.

Page 204: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

192

284. Middleton D, Menchaca L, Rood H, Komerofsky R: New allele frequency database:

http://www.allelefrequencies.net. Tissue Antigens 2003, 61(5):403-407. 285. Liao HT, Lin KC, Chang YT, Chen CH, Liang TH, Chen WS, Su KY, Tsai CY, Chou

CT: Human leukocyte antigen and clinical and demographic characteristics in

psoriatic arthritis and psoriasis in Chinese patients. J Rheumatol 2008, 35(5):891-895.

286. Armstrong RD, Panayi GS, Welsh KI: Histocompatibility antigens in psoriasis,

psoriatic arthropathy, and ankylosing spondylitis. Ann Rheum Dis 1983, 42(2):142-146.

287. Woodrow JC, Ilchysyn A: HLA antigens in psoriasis and psoriatic arthritis. J Med

Genet 1985, 22(6):492-495. 288. Brown MA: Genetics of ankylosing spondylitis. Curr Opin Rheumatol 2010,

22(2):126-132. 289. 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. Clin Rheumatol 2008, 27(6):709-712. 290. Queiro R, Sarasqueta C, Belzunegui J, Gonzalez C, Figueroa M, Torre-Alonso JC:

Psoriatic spondyloarthropathy: a comparative study between HLA-B27 positive and HLA-B27 negative disease. Semin Arthritis Rheum 2002, 31(6):413-418.

291. Gladman DD, Farewell VT, Kopciuk KA, Cook RJ: HLA markers and progression

in psoriatic arthritis. J Rheumatol 1998, 25(4):730-733. 292. Murray C, Mann DL, Gerber LN, Barth W, Perlmann S, Decker JL, Nigra TP:

Histocompatibility alloantigens in psoriasis and psoriatic arthritis. Evidence for the influence of multiple genes in the major histocompatibility complex. J Clin

Invest 1980, 66(4):670-675. 293. Queiro R, Gonzalez S, Lopez-Larrea C, Alperi M, Sarasqueta C, Riestra JL, Ballina J:

HLA-C locus alleles may modulate the clinical expression of psoriatic arthritis. Arthritis Res Ther 2006, 8(6):R185.

294. Ho PY, Barton A, Worthington J, Thomson W, Silman AJ, Bruce IN: HLA-Cw6 and

HLA-DRB1*07 together are associated with less severe joint disease in psoriatic arthritis. Ann Rheum Dis 2007, 66(6):807-811.

295. Elkayam O, Segal R, Caspi D: Human leukocyte antigen distribution in Israeli

patients with psoriatic arthritis. Rheumatol Int 2004, 24(2):93-97. 296. Espinoza LR, Vasey FB, Oh JH, Wilkinson R, Osterland CK: Association between

HLA-BW38 and peripheral psoriatic arthritis. Arthritis Rheum 1978, 21(1):72-75. 297. Ovsyannikova IG, Vierkant RA, Pankratz VS, Jacobson RM, Poland GA: Extended

LTA, TNF, LST1 and HLA gene haplotypes and their association with rubella vaccine-induced immunity. PLoS One 2010, 5(7):e11806.

298. Rahman P, Schentag CT, Gladman DD: Immunogenetic profile of patients with

psoriatic arthritis varies according to the age at onset of psoriasis. Arthritis

Rheum 1999, 42(4):822-823. 299. Klein JP, Moeschberger ML (eds.): Censoring and Truncation. USA: Springer;

2005. 300. Gladman DD, Antoni C, Mease P, Clegg DO, Nash P: Psoriatic arthritis:

epidemiology, clinical features, course, and outcome. Ann Rheum Dis 2005, 64

Suppl 2:ii14-17.

Page 205: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

193

301. Swanbeck G, Inerot A, Martinsson T, Wahlstrom J, Enerback C, Enlund F, Yhr M: Age at onset and different types of psoriasis. Br J Dermatol 1995, 133(5):768-773.

302. Fleischer AB, Jr., Rapp SR, Reboussin DM, Vanarthos JC, Feldman SR: Patient

measurement of psoriasis disease severity with a structured instrument. J Invest

Dermatol 1994, 102(6):967-969. 303. Kulkarni S, Martin MP, Carrington M: The Yin and Yang of HLA and KIR in

human disease. Semin Immunol 2008, 20(6):343-352. 304. Biassoni R, Pessino A, Malaspina A, Cantoni C, Bottino C, Sivori S, Moretta L,

Moretta A: Role of amino acid position 70 in the binding affinity of p50.1 and

p58.1 receptors for HLA-Cw4 molecules. Eur J Immunol 1997, 27(12):3095-3099. 305. Stewart CA, Laugier-Anfossi F, Vely F, Saulquin X, Riedmuller J, Tisserant A,

Gauthier L, Romagne F, Ferracci G, Arosa FA et al: Recognition of peptide-MHC

class I complexes by activating killer immunoglobulin-like receptors. Proc Natl

Acad Sci U S A 2005, 102(37):13224-13229. 306. Parham P: MHC class I molecules and KIRs in human history, health and

survival. Nat Rev Immunol 2005, 5(3):201-214. 307. Holm SJ, Sakuraba K, Mallbris L, Wolk K, Stahle M, Sanchez FO: Distinct HLA-

C/KIR genotype profile associates with guttate psoriasis. J Invest Dermatol 2005, 125(4):721-730.

308. Jobim M, Jobim LF, Salim PH, Cestari TF, Toresan R, Gil BC, Jobim MR, Wilson TJ, Kruger M, Schlottfeldt J et al: A study of the killer cell immunoglobulin-like

receptor gene KIR2DS1 in a Caucasoid Brazilian population with psoriasis vulgaris. Tissue Antigens 2008, 72(4):392-396.

309. Diaz-Pena R, Vidal-Castineira JR, Alonso-Arias R, Suarez-Alvarez B, Vicario JL, Solana R, Collantes E, Lopez-Vazquez A, Martinez-Borra J, Lopez-Larrea C: Association of the KIR3DS1*013 and KIR3DL1*004 alleles with susceptibility to ankylosing spondylitis. Arthritis Rheum 2010, 62(4):1000-1006.

310. Diaz-Pena R, Blanco-Gelaz MA, Suarez-Alvarez B, Martinez-Borra J, Lopez-Vazquez A, Alonso-Arias R, Bruges-Armas J, Vidal-Castineira JR, Lopez-Larrea C: Activating KIR genes are associated with ankylosing spondylitis in Asian populations. Hum Immunol 2008, 69(7):437-442.

311. van der Slik AR, Koeleman BP, Verduijn W, Bruining GJ, Roep BO, Giphart MJ: KIR in type 1 diabetes: disparate distribution of activating and inhibitory natural killer cell receptors in patients versus HLA-matched control subjects. Diabetes 2003, 52(10):2639-2642.

312. Cheent K, Khakoo SI: Natural killer cells: integrating diversity with function. Immunology 2009, 126(4):449-457.

313. Andersen JH, Haahr JP, Frost P: Risk factors for more severe regional

musculoskeletal symptoms: a two-year prospective study of a general working population. Arthritis Rheum 2007, 56:1355-1364.

314. Padula A, Belsito F, Barozzi L, Cantini F, Salvarani C, Pavlica P, Olivieri I: Isolated

tenosynovitis associated with psoriasis triggered by physical injury. Clin Exp

Rheumatol 1999, 17(103-4). 315. Thomachot B, Lafforgue P, Acquaviva PC: Post-traumatic psoriatic arthritis. 2

cases. Presse Med 1996(25):21-24.

Page 206: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

194

316. D'Souza JC, Franzblau A, Werner RA: Review of epidemiologic studies on

occupational factors and lower extremity musculoskeletal and vascular disorders and symptoms. J Occup Rehabil 2005, 15:129-165.

317. Natvig B, Picavet HS: The epidemiology of soft tissue rheumatism.Best Pract Res

Clin Rheumatol 2002, 16:777-793. 318. Ward MM, Reveille JD, Learch TJ, Davis JCJ, Weisman MH: Occupational

physical activities and long-term functional and radiographic outcomes in patients with ankylosing spondylitis. Arthritis Rheum 2008, 59:822-832.

319. Jankovic S, Raznatovic M, Marinkovic J, Jankovic J, Maksimovic N: Risk factors

for psoriasis: A case-control study. J Dermatol 2009, 36:328-334. 320. Lakatos PL: Environmental factors affecting inflammatory bowel disease: have

we made progress?Dig Dis 2009, 27:215-225. 321. Calkins BM: A meta-analysis of the role of smoking in inflammatory bowel

disease. Dig Dis Sci 1989, 34:1841-1854. 322. Boyko EJ, Perera DR, Koepsell TD, Keane EM, Inui TS: Effects of cigarette

smoking on the clinical course of ulcerative colitis. Scand J Gastroenterol 1988, 23:1147-1152.

323. Cosnes J: Tobacco and IBD: relevance in the understanding of disease

mechanisms and clinical practice. Best Pract Res Clin Gastroenterol 2004, 18:481-496.

324. Savoye G, Lerebours E: Toll-like receptor-4 signaling: a possible candidate

pathway to support tobacco smoking effects in ulcerative colitis. Am J

Gastroenterol 2008, 103:2947-2948. 325. van Maanen MA, Stoof SP, Larosa GJ, Vervoordeldonk MJ, Tak PP: Role of the

cholinergic nervous system in rheumatoid arthritis: aggravation of arthritis in nicotinic acetylcholine receptor alpha7 subunit gene knockout mice. Ann Rheum

Dis 2010, 69:1717-1723. 326. Bencherif M, Lippiello PM, Lucas R, Marrero MB: Alpha7 nicotinic receptors as

novel therapeutic targets for inflammation-based diseases.Cell Mol Life Sci 2010, Oct 15. [Epub ahead of print].

327. Duffin KC, Chandran V, Gladman DD, Krueger GG, Elder JT, Rahman P: Genetics

of psoriasis and psoriatic arthritis: update and future direction. J Rheumatol

2008, 35:1449-1453. 328. Dika E, Bardazzi F, Balestri R, Maibach HI: Environmental factors and psoriasis.

Curr Probl Dermatol 2007, 35:118-135. 329. Naldi L, Mercuri SR: Smoking and psoriasis: from epidemiology to

pathomechanisms. J Invest Dermatol 2009, 129:2741-2743. 330. Jin Y, Yang S, Zhang F, Kong Y, Xiao F, Hou Y, al e: Combined effects of HLA-

Cw6 and cigarette smoking in psoriasis vulgaris: a hospital-based case-control study in China. J Eur Acad Dermatol Venereol 2009, 23:132-137.

331. Statistics Canada. National Population Health Survey. [http://www.statcan.gc.ca] 332. Ahlbom A, Alfredsson L: Interaction: A word with two meanings creates

confusion. Eur J Epidemiol 2005, 20:563-564. 333. Liao KP, Alfredsson L, Karlson EW: Environmental influences on risk for

rheumatoid arthritis. Curr Opin Rheumatol 2009, 21:279-283.

Page 207: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

195

334. Brigham J, Lessov-Schlaggar CN, Javitz HS, McElroy M, Krasnow R, Swan GE: Reliability of adult retrospective recall of lifetime tobacco use. Nicotine Tob Res

2008, 10(2):287-299. 335. Shiffman S: How many cigarettes did you smoke? Assessing cigarette

consumption by global report, Time-Line Follow-Back, and ecological momentary assessment. Health Psychol 2009, 28(5):519-526.

336. Eder L, Chandran V, Shen H, Cook RJ, Shanmugarajah S, Rosen CF, Gladman DD: Incidence of arthritis in a prospective cohort of psoriasis patients. Arthritis Care

Res (Hoboken) 2011, 63(4):619-622. 337. Eder L, Loo T, Chandran V, Kalman-Lamb G, Shanmugarajah S, Shen H, Rosen CF,

Cook RJ, Gladman DD: The association between environmental factors and onset

of psoriatic arthritis in patients with psoriasis. Arthritis Care Res (Hoboken) 2011.

Page 208: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

196

Appendices

Appendix 1 - Questionnaire for the assessment of exposure to environmental factors

1 Employment NO YES

1a. In the last 10 years, have you ever had paid employment?

� �

go to 2a

� �

complete sections below

current

or

past

c p

part-time

or

full-time

p f

# of Years Worked

i. Job #1 (specify): �� ��

ii. Job #2 (specify): �� ��

iii. Job #3 (specify): �� ��

iv. Job #4 (specify): �� ��

v. Job #5 (specify): �� ��

vi. Job #6 (specify): �� ��

In the last 10 years, did you ever have a job that involved:

NO YES

1b. …standing on your feet for more than 30 minutes per hour?

� �

1c. …squatting for more than 5 minutes per hour? � �

1d. …lifting cumulative loads of more than 100 pounds per hour?

� �

1e. …pushing cumulative loads of more than 200 pounds per hour?

� �

1f. …using a vibrating tool for more than 4 hours a day?

� �

1g. …frequent repetitive hand movements for more than 45 minutes per hour?

� �

1h. …forceful gripping for more than 4 hours per day?

� �

1i. …bending your wrist for more than 4 hours per day?

� �

2 Life Events

In the last 10 years, have you experienced any of the following:

NO YES Year of Event #1

Year of Event #2

2a. Death of a close family member (e.g. parent,

sibling, child or partner)?

� ��

Page 209: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

197

2b. Divorce or separation � ��

2c. Moved house � ��

2d. Changed job � ��

2e. Become unemployed � ��

2f. Treated for anxiety or depression � ��

3 Infections and Immunizations

3a. In the last 10 years have you had any of the following immunizations?

Don’t Know

NO YES Year specify year(s) immunized

i. BCG (for tuberculosis) � � ��

ii. Hepatitis A � � ��

iii. Hepatitis B � � ��

iv. Influenza (the flu shot) � � ��

v. Pneumovax (for pneumonia) � � ��

vi. Rubella � � ��

vii. Tetanus � � ��

viii. Other (specify) � � ��

ix. Other (specify) � � ��

3b. In the last 10 years, have you had any serious infections?

(i.e. requiring antibiotic treatment or admission to hospital)

NO YES

Year

Were you treated with antibiotics?

Were you admitted to hospital?

� �

go to 3c

� �

complete sections below

Don’t Know

NO YES NO YES

i. Type of infection #1 (specify): ____ � � � � �

ii. Type of infection #2 (specify): ____ � � � � �

iii. Type of infection #3 (specify): ____ � � � � �

iv. Type of infection #4 (specify): ____ � � � � �

v. Type of infection #5 (specify): ____ � � � � �

3c. In the last 10 years: have you had infective diarrhea?

(e.g. food poisoning or ‘traveler’s diarrhea’)

Don’t Know

NO YES

Year

Were you treated with antibiotics?

� �

go to 4

� �

go to 4

� �

complete sections below

Don’t Know

NO YES

i. Incident #1 (specify diagnosis): ____ � � �

ii. Incident #2 (specify diagnosis): ____ � � �

iii. Incident #3 (specify diagnosis): ____ � � �

4 Physical Trauma

4a. In the last 10 years, have you been involved in a road traffic accident?

NO YES

Year

Were you seen in

emergency?

Were you admitted to hospital?

� �

go to 4b

� �

complete sections below NO YES NO YES

i. Accident #1 (describe): ____ � � � �

ii. Accident #2 (describe): ____ � � � �

Page 210: Genetic and Environmental Risk Factors for Psoriatic ... · Genetic and Environmental Risk Factors for Psoriatic Arthritis among Patients with Psoriasis Lihi Eder PhD Institute of

198

4b. In the last 10 years, Have you had any fractures (broken bones)?

NO YES

Year

Were you seen in

emergency?

Were you admitted to hospital?

� �

go to 4c

� �

complete sections below NO YES NO YES

i. Fracture #1 (which bone?): ____ � � � �

ii. Fracture #2 (which bone?): ____ � � � �

iii. Fracture #3 (which bone?): ____ � � � �

4c. In the last 10 years, have you had any other injury (e.g. fall, cut, accident etc.) for which you consulted your family doctor, walk-in clinic or emergency room?

NO YES

Year

Were you seen in

emergency?

Did you consult a family doctor?

� �

complete sections below NO YES NO YES

i. Other injury #1 (describe): ____ � � � �

ii. Other injury #2 (describe): ____ � � � �

iii. Other injury #3 (describe): ____ � � � �