Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

ARTHRITIS & RHEUMATISM

Vol. 50, No. 10, October 2004, pp 3093–3103


DOI 10.1002/art.20555
© 2004, American College of Rheumatology

Interaction Between RANKL and HLA–DRB1 Genotypes


May Contribute to Younger Age at Onset of
Seropositive Rheumatoid Arthritis in an Inception Cohort

Hui Wu,1 Dinesh Khanna,2 Grace Park,2 Vivian Gersuk,3 Gerald T. Nepom,3
Weng Kee Wong,2 Harold E. Paulus,2 and Betty P. Tsao,2
for the Western Consortium of Practicing Rheumatologists

Objective. To determine whether the RANKL and SNP1 and fluorescence-based PCR for the presence or
HLA–DRB1 “shared epitope” (SE) genotypes contribute absence of the TAAA insertion.
to the development of rheumatoid arthritis (RA). Results. The presence of SE-containing DRB1*04
Methods. We studied 237 patients with early RA alleles was associated with an earlier age at RA onset
(within 15 months of symptom onset) who were seropos- (mean ⴞ SD 47 ⴞ 12.7 years versus 53 ⴞ 12.5 years in
itive for rheumatoid factor. HLA–DRB1 genotyping was SE– patients; P ⴝ 0.0004). The 2 novel RANKL polymor-
performed using the polymerase chain reaction (PCR)– phisms were in strong linkage disequilibrium (P <
based oligonucleotide probe assay. RANKL polymor- 0.0001) and were associated with earlier ages at disease
phisms were analyzed using PCR pyrosequencing for onset (e.g., for the CC versus CT/TT genotypes, 44 ⴞ
13.5 years versus 51 ⴞ 12.7 years; P ⴝ 0.0080). The
mean age at disease onset in SEⴙ patients with the
Supported in part by grants from the NIH (P60-AR-36834),
the Southern California Chapter of the Arthritis Foundation, and the RANKL-CC genotype (35 ⴞ 7.2 years) was a mean of 18
Paxson Family Foundation. Dr. Khanna’s work was supported by a years younger than in SE– patients with RANKL-CT/TT
Centocor Health Outcomes in Rheumatic Diseases Fellowship.
1 (53 ⴞ 12.5 years; P < 0.0001) and was 17 years younger
Hui Wu, MD: University of California, Los Angeles, and
Ren Ji Hospital, Shanghai Second Medical University, Shanghai, than in SE– patients with RANKL-CC (52 ⴞ 13.2 years;
China; 2Dinesh Khanna, MD, MS (current address: University of P ⴝ 0.0005). The proportion of patients with both the
Cincinnati, Cincinnati, Ohio), Grace Park, MPH, Weng Kee Wong,
PhD, Harold E. Paulus, MD, Betty P. Tsao, PhD: University of
SE and RANKL risk alleles was highest (23%) in those
California, Los Angeles; 3Vivian Gersuk, PhD, Gerald T. Nepom, MD, who developed RA during their third decade of life (ages
PhD: Benaroya Research Institute at Virginia Mason, Seattle, Wash- 20–30 years), with a declining trend among those who
ington.
Members of the Western Consortium of Practicing Rheuma- developed RA during their fourth (16%), fifth (5%), and
tologists are as follows: Drs. Robert Shapiro, Maria W. Greenwald, H. sixth or later (0%) decades. Interestingly, 92% of the
Walter Emori, Fredrica E. Smith, Craig W. Wiesenhutter, Charles patients diagnosed as having RA between ages 20 and 30
Boniske, Max Lundberg, Anne MacGuire, Jeffry Carlin, Robert
Ettlinger, Michael H. Weisman, Elizabeth Tindall, Karen Kolba, years carried at least 1 of the RA-associated DRB1*04
George Krick, Melvin Britton, Rudy Greene, Ghislaine Bernard alleles, suggesting a strong influence of the SE in the
Medina, Raymond T. Mirise, Daniel E. Furst, Kenneth B. Wiesner, early onset of RA. The majority of patients who devel-
Robert F. Willkens, Kenneth Wilske, Karen Basin, Robert Gerber,
Gerald Schoepflin, Marcia J. Sparling, George Young, Philip J. Mease, oped RA symptoms in their third to fifth decades (74 of
Ina Oppliger, Douglas Roberts, J. Javier Orozco Alcala, John Seaman, 119 [62%]) carried at least 1 copy of the DRB1*04
Martin Berry, Ken J. Bulpitt, Grant Cannon, Gregory Gardner, Allen
Sawitzke, Andrew Lun Wong, Daniel O. Clegg, Timothy Spiegel,
alleles; in contrast, fewer than half of the patients who
Wayne Jack Wallis, Mark Wener, Robert Fox. developed RA in their sixth decade or later (50 of 118
Drs. Wu and Khanna contributed equally to this work. [42%]) had DRB1*04 alleles. RANKL genotypes were
Address correspondence and reprint requests to Betty P.
Tsao, PhD, Division of Rheumatology, Department of Medicine, not associated with erosive disease at baseline or with
Rehabilitation Center, Room 32-59, 1000 Veteran Avenue, UCLA the yearly progression rate of radiographic joint dam-
School of Medicine, Los Angeles, CA 90095-1670. E-mail: age.
Btsao@mednet.ucla.edu.
Submitted for publication February 26, 2004; accepted in Conclusion. This study provides the first evidence
revised form June 22, 2004. that novel RANKL polymorphisms were associated with
3093
3094 WU ET AL

an earlier age at RA onset in SEⴙ, but not SE–, patients ulation of RANK (23). Levels of both OPG and RANKL
and that an interaction between SE-containing HLA– are elevated in the sera of RA patients, but are normal-
DRB1 and RANKL polymorphisms increased the disease ized without influencing the ratio of OPG to RANKL
penetrance, resulting in a mean age at RA onset that after treatment with anti-TNF (25). The interaction
was 18–20 years younger. Our results also suggested between RANK and RANKL appears to mediate bone
genetic differences between patients with early-onset and cartilage destruction in animal models of arthritis,
and those with late-onset RA. and OPG treatment is effective in preventing bone
erosion and has partially beneficial effects on cartilage
Rheumatoid arthritis (RA) is a systemic auto- destruction (26–28). Taken together, the functional
immune disease characterized by chronic inflammation properties of RANKL make it a logical candidate gene
of the joints, which may lead to structural damage of the for RA.
cartilage and bone. Genetic influences in RA have been To our knowledge, no studies published thus far
supported by cumulative studies (for review, see refs. 1 have described an association between genetic polymor-
and 2). The best known and the most dominant genetic phisms of RANKL and RA. Single-nucleotide polymor-
risk factor for RA is the HLA class II region containing phisms (SNPs) that affect the binding of transcription
the DRB1 locus. This has been a consistent finding of 4 factors have recently been associated with susceptibility
independent genome scans using family collections from to multiple autoimmune diseases including RA (29–32).
Europe, the US (2 studies), and the UK (3–6). The This provided a rationale for selecting 3 intronic SNPs of
RA-associated DRB1 alleles (DRB1*0101, *0102, *0401, RANKL from the National Center for Biotechnology
*0404, *0405, *0408, *0409, *0410, *0413, *0416, *0421, Information SNP database that had a predictive alter-
*1001, *1402, and *1406) share a conserved linear
ation in transcription factor affinity between the 2
sequence of amino acids between positions 70 and 74 in
alleles, using the TFSearch computer program (33).
the DR␤1 chain (QKRAA, QRRAA, or RRRAA) of
Among these 3 SNPs (dbSNP nos. rs7334187, rs387856,
the HLA–DR␣/␤ heterodimer, which has led to the
and rs922996) the former 2 were not polymorphic in our
“shared epitope” (SE) hypothesis (7).
samples, leaving 1 SNP (SNP1) that was appropriate for
The risk of RA conferred by different SE-
association study. Since SNP1 is located in intron 7
containing alleles may vary. In addition, there are ethnic
toward the 3⬘ end of the gene, we selected another
variations in the allele frequency of SE-containing al-
polymorphism (rs5803141; the insertion of TAAA or
leles associated with RA. In Caucasians, DRB1*04 al-
none) lying 37.5 kb upstream of the entire 45.3-kb
leles (especially *0401) are the major SE-containing
alleles associated with a higher risk of RA (1,8). The RANKL for this initial study.
presence and the dose effect of these DRB1 alleles have In the present study, we assessed novel RANKL
also been associated with early disease onset (8–10), polymorphisms and HLA–DRB1*04 alleles for their
radiologic erosions (11–18), and extraarticular manifes- potential association with RA in an early (⬍15 months
tations (19,20). Most estimates of the HLA component after symptom onset) observational cohort of patients
of the overall genetic risk of RA are ⬍50% (21), which with seropositive active RA. Since an inception cohort
makes it an important challenge to identify non-HLA tends to have a more accurately documented date of RA
genes that may contribute to the pathogenesis of RA. onset, we examined whether the age at disease onset
RANKL (also known as osteoprotegerin ligand, correlated with the DRB1*04 and RANKL genotypes. In
TRANCE, and osteoclast differentiation factor), a addition, we studied whether RANKL genotypes were
member of the tumor necrosis factor (TNF) ligand associated with the presence of erosive disease at base-
family, has been shown to play a pivotal role in inflam- line and with the rate of radiographic progression of
matory joint diseases (for review, see ref. 22). Cell joint damage.
membrane–anchored or secreted RANKL binds to
PATIENTS AND METHODS
RANK that is expressed on osteoclasts, dendritic cells,
and B and T lymphocytes, resulting in osteoclastogen- Patients. The patients included in this study were a
esis, osteoclast activation, dendritic cell survival, lym- subset of a group of RA patients who were participating in a
phocyte development, and lymph node organogenesis long-term observational study involving the Western Consor-
tium of Practicing Rheumatologists, which is a regional con-
(23,24). A natural, soluble decoy receptor, osteoprote- sortium of 29 rheumatology practices in the western United
gerin (OPG), binds both the secreted and cell-bound States and Mexico, as described previously (34–36). The
forms of RANKL, preventing the interaction and stim- inclusion criteria for this study were a diagnosis of RA
RANKL AND HLA–DRB1 GENOTYPES AND AGE AT ONSET OF RA 3095

according to the American College of Rheumatology (for- was assessed by the intraclass correlation coefficient and the
merly, the American Rheumatism Association) criteria (37) smallest detectable difference. The intraclass correlation coef-
within 15 months of symptom onset, no previous treatment ficient and the smallest detectable difference for the average of
with disease-modifying antirheumatic drugs, seropositivity for the 2 readers’ scores was 0.97 and 3.07 for the erosion score,
rheumatoid factor (a titer ⱖ1:80 or a level ⱖ40 IU/ml), ⱖ6 0.93 and 7.52 for the joint space narrowing score, and 0.90 and
swollen joints, and ⱖ9 tender joints, and availability of 12.71 for the total Sharp score (36).
genomic DNA. Symptom onset was defined as the date when Because the time interval between pairs of radiographs
musculoskeletal symptoms began, provided that these symp- varied among patients, radiographic progression between ob-
toms persisted and led to the diagnosis of RA. This study was servations was determined by dividing the difference between
approved by appropriate institutional review boards. the scores of each pair of radiographs by the number of months
Clinical and laboratory assessments. The consortium that had elapsed between them. The progression rate was
rheumatologists assessed the patients’ disease status at study expressed as the change in score (total, erosion, or joint space
entry (baseline), 6 months, 1 year, and yearly thereafter. Using narrowing) per month; this was annualized when necessary to
standard methods, the detailed physician’s assessment in- express the progression rate per year (36). For each patient
cluded all of the core set of outcome measures required to with ⱖ2 radiographic observations, the slope of the least-
calculate the Disease Activity Score (DAS), including com- squares linear regression line was calculated to estimate the
plete tender and swollen joint counts (of 68 tender and 66 annualized progression rates of the erosion score, joint space
swollen joints) and measures of acute-phase reactants, as well narrowing score, and total Sharp score, assuming that progres-
as assessments of global disease activity and pain on 0–100-mm sion remains relatively constant over time.
visual analog scales. The DAS was calculated according to the DNA preparations and HLA–DRB1 genotyping.
published algorithm, using the Ritchie Articular Index, swollen Genomic DNA was isolated by standard protocol from blood
joint count of 44 joints, and erythrocyte sedimentation rate (in mononuclear cells obtained from RA patients. Patients were
mm/hour) (38). In addition, assays for rheumatoid factor and genotyped using a DNA-based sequence-specific oligonucleo-
self-report measurements, such as the disability index of the tide probe assay for HLA–DRB1*04 alleles (including *0401,
Health Assessment Questionnaire (HAQ) and the Center for *0404, *0408, *0405, and *0410) as described elsewhere (42).
Epidemiologic Studies Depression Scale (39), were performed Confirmatory sequencing or allele-specific quantitative poly-
at each visit. At each scheduled physician visit, blood speci- merase chain reaction (PCR) was used to verify the gene dose
mens were collected for measurement of C-reactive protein in homozygotes. Distinction between the *0404 and *0408
levels; Westergren erythrocyte sedimentation rates were deter- DRB1*04 alleles was not made because of their structural
mined, when clinically indicated, in the rheumatologist’s office similarity and the rarity of allele type *0408. This was also true
or local laboratory. for DRB1*04 alleles *0405 and *0410, since *0410 was infre-
Radiographic assessments. Radiographs of the hands, quent.
wrists, and forefeet were obtained at entry, at 6 months, 1 year, RANKL polymorphism genotyping. The SNP of
and yearly thereafter. Standard posteroanterior radiographs RANKL, SNP1 (reference no. rs922996), was determined by
that included both hands and wrists and anteroposterior PCR pyrosequencing. The PCR was performed in a 25-␮l
radiographs that included both forefeet were obtained in the mixture containing 20–40 ng of genomic DNA template,
rheumatologists’ offices or at the radiology facility they usually standard PCR buffer, 200 ␮M dNTPs, 2 mM MgCl2, 200 nM
used. All available radiographs were scored by 2 experienced primers, and 1 unit of AmpliTaq Gold Polymerase (Applied
readers for the presence of erosions (0–5 scale) and joint space Biosystems, Foster City, CA). The PCR began with a denatur-
narrowing (0–4 scale), and a total score (sum of the erosion ation step at 95°C for 5 minutes, followed by 50 cycles of
and joint space narrowing scores) was obtained. Sclerosis or denaturing at 95°C for 15 seconds, annealing at 65°C for 30
healing of erosions was not scored. seconds, plus extension at 72°C for 15 seconds, and ended with
After a brief “training” session in which the readers a final elongation step at 72°C for 5 minutes. Using the forward
discussed the scoring scale and reviewed a small group of primer 5⬘-TTT-CTG-GAC-AGA-GGG-ATT-GG-3⬘ and re-
radiographs to be sure they agreed on the features to be verse primer 5⬘-TGA-TCT-GGG-AGC-CTC-AAA-AC-3⬘, a
scored, the radiographs were scored independently using the 153-bp amplicon was generated and checked for size and
method described by Sharp et al (40,41), assessing 17 joints in purity by agarose gel electrophoresis. The forward primer was
each hand and wrist for erosions, 16 joints in each hand and purified by high-performance liquid chromatography and was
wrist for joint space narrowing, and 6 joints in each forefoot for biotinylated at the 5⬘ end to allow immobilization onto
erosions and joint space narrowing. The maximum possible streptavidin-coated Dynabeads M-280 (Dynal Biotech, Oslo,
scores for the hands and wrists were 170 for erosions and 128 Norway).
for joint space narrowing, with a total score of 298. The Pyrosequencing of 20 ␮l of the PCR product immobi-
maximum possible scores for the feet were 60 for erosions and lized onto the beads was performed using the sequencing
48 for joint space narrowing, with a total score of 108. The primer 5⬘-CAA-CGG-TGG-GGC-A-3⬘ and SNP Reagent kits
maximum possible scores for all joints assessed (hands, wrists, (Pyrosequencing, Uppsala, Sweden) according to the manufac-
and feet bilaterally) were 230 for erosions and 176 for joint turer’s instructions. SNP genotype analysis was performed
space narrowing, with a total score of 406. using the SNP software in a PSQ 96MA System (both from
Radiographs were read in patient sets, which had been Pyrosequencing, Uppsala, Sweden).
randomized and blinded for sequence. The independent scores Genotyping of the RANKL polymorphism (rs5803141;
of readers 1 and 2 were averaged for each radiograph, and this –/TAAA) was performed by fluorescence-based PCR using the
average was used for the analyses. Reliability of the readers forward primer 5⬘-TET-TGA-GCT-GAG-ATT-GCA-CCA-
3096 WU ET AL

Table 1. Major baseline characteristics of the 237 patients with early rheumatoid arthritis
Characteristic Value
Age at disease onset, mean ⫾ SD years 50.0 ⫾ 12.9
Female, % 75.5
Ethnicity, %
Caucasian 77.1
Hispanic 15.7
Asian, Pacific Islander 3.4
African American 1.3
Disease duration, mean ⫾ SD months 6.16 ⫾ 3.35
No. of comorbidities per patient, mean ⫾ SD* 1.60 ⫾ 1.66
Patient-reported total annual household income, mean ⫾ SD dollars (⫻1,000) 38.9 ⫾ 26.4
Highest year of education completed, mean ⫾ SD 12.7 ⫾ 2.9
Smoker, % 56.5
Rheumatoid nodules, % 11.5
Extraarticular manifestations other than rheumatoid nodules, %† 9.0
Rheumatoid factor, mean ⫾ SD IU/ml 385 ⫾ 526
Erythrocyte sedimentation rate, mean ⫾ SD mm/hour 42.0 ⫾ 25.4
C-reactive protein, mean ⫾ SD mg/dl 3.12 ⫾ 6.20
Disease Activity Score, mean ⫾ SD 4.71 ⫾ 1.15
Health Assessment Questionnaire disability index, mean ⫾ SD (range 0–3.0) 1.20 ⫾ 0.72
Center for Epidemiologic Studies Depression Scale, mean ⫾ SD (range 0–60) 15.7 ⫾ 11.7

* Frequent comorbidities observed in this cohort were depression, hypertension, broken bones or
fractures, coronary heart disease, gall bladder disease, and osteoporosis. Other comorbidities were cancer
of the lung, breast, uterus, cervix, and prostate, as well as diabetes, hyperthyroidism, cataracts, glaucoma,
myocardial infarction, cardiomyopathy, pneumonia, emphysema, asthma, ulcer, gastritis, endometriosis,
duodenal spasms, back problems, fibrositis, insomnia, allergies, angina, stroke, chronic bronchitis, hiatal
hernia, mitral valve prolapse, diverticulitis, etc.
† Most had self-reported symptoms of keratoconjunctivitis sicca (dry eyes, dry mouth).

CT-3⬘ and the reverse primer 5⬘-TCC-TGA-ATC-TGT-CTC- Pairwise linkage disequilibrium between 2 genetic
CAC-TTC-A-3⬘. The PCR condition and contents were similar polymorphisms was assessed using a chi-square test of associ-
to those described for SNP1 except that the reaction volume ation on phase-known haplotypes from DNA samples. Haplo-
was 5 ␮l. A 207-bp (without the TAAA insertion) or 211-bp types were inferred using the partition–ligation algorithm to
(with the TAAA insertion) amplicon generated by PCR was reconstruct individual haplotypes from population genotype
mixed with the loading buffer and GeneScan-350 TAMRA size data (Haplotyper 1.0 Linux software [43]). The extent of
standard (Applied Biosystems) and subjected to electrophor- linkage disequilibrium between the 2 RANKL polymorphisms
esis in polyacrylamide sequencing gels (Long Ranger Singel (D⬘ and r2) was assessed using the GOLD program (44).
Packs; Cambrex Bioscience, Rockland, ME) for 2 hours at 3 Statistical analyses were performed using software packages
kV using an ABI 377 DNA sequencer (Applied Biosystems). SAS release 8.2 (SAS Institute, Cary, NC) and Prism 3.02
The gel image was analyzed by GeneScan software version 3.1 (GraphPad Software, San Diego, CA). P values less than 0.05
for sizing amplified products and Genotyper software version were considered statistically significant. We considered these
2.5 for allele assignment (both from Applied Biosystems). exploratory analyses to be preliminary and hypothesis-
Allele frequencies of RANKL polymorphisms of this cohort generating; thus, no Bonferroni correction was made.
showed no significant departure from expected frequencies
according to Hardy-Weinberg equilibrium.
Statistical analysis. Comparisons between patient RESULTS
groups were made by 2-sample, 2-sided t-tests for continuous RA patient characteristics. This study cohort
variables and by chi-square test for categorical variables. The consisted of 237 patients with early, seropositive, active
nonparametric Wilcoxon rank sum test and Fisher’s exact test
were performed when necessary for continuous and categori- RA who had been enrolled within 15 months of symp-
cal variables, respectively. For comparisons of continuous tom onset, before initiation of disease-modifying anti-
variables among 3 groups, we used the Kruskal-Wallis non- rheumatic drug therapy. Table 1 shows the baseline
parametric test of one-way analysis of variance for ranks. In characteristics of the study patients. Their age at RA
addition, to explore the contribution of the DRB1*04 and onset was normally distributed, with a mean ⫾ SD of
RANKL gene polymorphisms to the age at RA onset, we used
a multiple regression model, in which age at onset was 50.0 ⫾ 12.9 years, and their disease duration at study
regressed on the presence of the shared epitope, the RANKL entry was 6.16 ⫾ 3.35 months (mean ⫾ SD). Most of the
at-risk alleles, and their interaction. patients (75.5%) were women and most had active
RANKL AND HLA–DRB1 GENOTYPES AND AGE AT ONSET OF RA 3097

Table 2. Distribution and ethnic variation of the rheumatoid arthritis–associated DRB1*04 alleles*
Total study group DRB1*04⫹ cohort

Caucasians Hispanics Caucasians Hispanics


HLA–DRB1*04 allele All patients (n ⫽ 182) (n ⫽ 37) P (n ⫽ 98) (n ⫽ 17) P
1 copy of a DRB1*04 allele 43 43.5 43.3 NS 100 100 NS
*0401 27 32.4 5.4 0.0005 60.2 11.8 0.0003
*0404 or *0408 12 9.9 29.7 0.0012 18.4 64.7 ⬍0.0001
*0405 or *0410 4 1.1 10.8 0.0082 2.0 23.5 0.0042
2 copies of a DRB1*04 allele 10 10.4 2.7 NS 19.4 5.9 NS
*0401/*0401 homozygous 6 6.0 2.7 NS 11.2 5.9 NS
*0401/*0404 or *0408 heterozygous 2 2.8 0 NS 5.1 0 NS
*0401/*0405 or *0410 heterozygous 1 1.6 0 NS 3.1 0 NS

* Values are percentages. NS ⫽ not significant.

disease (mean DAS 4.71) (45) and moderate functional tients (42.2 ⫾ 11.4 versus 51.6 ⫾ 12.9 years; P ⬍ 0.0001).
disability (mean HAQ disability index 1.20). This trend was also seen for comparisons of DRB1*04⫹
Distribution of DRB1*04 alleles. RA-associated Hispanics and Caucasians (Table 3). This association
DRB1*04 alleles (DRB1*0401, *0404, *0405, *0408, or between the SE and an earlier age at RA onset was
*0410) were present in 53% of the patients; 43% had 1 observed in Caucasians (48.5 ⫾ 12.4 years in SE⫹ versus
copy of DRB1*04 (27% had *0401, 12% had *0404 or 55.3 ⫾ 12.5 years in SE–; P ⫽ 0.003) but not in Hispanics
*0408, and 4% had *0405 or *0410) and 10% had 2 (40.4 ⫾ 13.1 years in SE⫹ versus 43.7 ⫾ 9.7 years in SE–;
copies (6% were *0401 homozygotes and 4% had *0401 P ⫽ 0.4). The sample size of the Hispanic group was
and other *04 alleles) (Table 2). Comparison of patients insufficiently powered to detect a difference between
with (n ⫽ 124) and without (n ⫽ 113) RA-associated SE⫹ and SE– subgroups.
DRB1*04 alleles revealed that the presence of the SE The presence of RA-associated DRB1*04 alleles
was associated with an earlier age at disease onset was higher in the Caucasians (54%) than in the Hispan-
(mean ⫾ SD 47.2 ⫾ 12.7 years versus 53.0 ⫾ 12.5 years ics (46%). There was a striking difference in the distri-
in the SE– group; P ⫽ 0.0004) (Table 3). There was no bution of specific RA-associated DRB1*04 alleles be-
trend toward a younger disease onset among patients tween the 2 ethnic groups (Table 2). DRB1*0401 was the
with 2 copies of DRB1*04 (50.2 ⫾ 13.6 years; n ⫽ 23) most frequent allele in the Caucasians, whereas *0404
compared with those with a single copy (46.5 ⫾ 12.4 (*0408) and *0405 (*0410) accounted for the majority of
years; n ⫽ 101) (P ⫽ 0.2). SE⫹ Hispanics. Patients who had 2 copies of SE-
Ethnic variation in age at disease onset and containing DRB1*04 represented 10.4% of the Cauca-
RA-associated DRB1*04 alleles. The major ethnic sians of this cohort, but only 2.7% of the Hispanics.
groups of the cohort were Caucasians (n ⫽ 182 [77%]) HLA–DRB1 and RANKL gene polymorphisms
and Hispanics (n ⫽ 37 [16%]). The numbers of patients and age at RA onset. Because RANK and RANKL are
of other ethnic backgrounds were too small to make a novel therapeutic targets for arthritis, RANKL located
meaningful comparison. The Hispanic patients had an on chromosome 13q14 was chosen as a non-HLA can-
earlier disease onset compared with the Caucasian pa- didate gene for RA. We observed that an intronic SNP,

Table 3. Age at onset of rheumatoid arthritis symptoms, according to the presence and absence of DRB1*04 alleles*
Total study group DRB1*04⫹ cohort DRB1*04⫺ cohort P†
All study patients 50.0 ⫾ 12.9 (n ⫽ 237) 47.2 ⫾ 12.7 (n ⫽ 124) 53.0 ⫾ 12.5 (n ⫽ 113) 0.0004
Caucasian patients 51.6 ⫾ 12.9 (n ⫽ 182) 48.5 ⫾ 12.4 (n ⫽ 98) 55.3 ⫾ 12.5 (n ⫽ 84) 0.0003
Hispanic patients 42.2 ⫾ 11.4 (n ⫽ 37) 40.4 ⫾ 13.1 (n ⫽ 17) 43.7 ⫾ 9.7 (n ⫽ 20) NS
P* ⬍0.0001 0.0150 0.0002 –

* The presence of rheumatoid arthritis–associated HLA–DRB1*04 alleles was associated with early onset of the disease. Values
are the mean ⫾ SD years of age.
* For comparisons between Hispanics and Caucasians.
† For comparisons between DRB1*04⫹ and DRB1*04⫺ cohorts. NS ⫽ not significant.
3098 WU ET AL

Table 4. Association between the presence of HLA–DRB1*04 (SE-containing) and RANKL risk alleles and early age at onset
of rheumatoid arthritis*
Total study group (n ⫽ 237) Caucasian patients (n ⫽ 182)

Age at onset, Age at onset,


Genotype mean ⫾ SD years P mean ⫾ SD years P
HLA–DRB1*04
SE⫹ 47 ⫾ 12.7 (n ⫽ 124) 0.0004 48 ⫾ 12.4 (n ⫽ 98) 0.0003
SE⫺ 53 ⫾ 12.5 (n ⫽ 113) 55 ⫾ 12.5 (n ⫽ 84)
RANKL
CC 44 ⫾ 13.5 (n ⫽ 25) 0.0080 44 ⫾ 14.0 (n ⫽ 18) 0.0110
CT plus TT 51 ⫾ 12.7 (n ⫽ 212) 52 ⫾ 12.5 (n ⫽ 164)
RANKL†
TAAA-C⫹/⫹ 44 ⫾ 14.4 (n ⫽ 21) 0.0190 44 ⫾ 14.8 (n ⫽ 16) 0.0140
TAAA-C⫹/⫺ ⫹ TAAA-C⫺/⫺ 51 ⫾ 12.6 (n ⫽ 216) 52 ⫾ 12.5 (n ⫽ 166)
RANKL/DRB1*04
CC/SE⫹ 35 ⫾ 7.2 (n ⫽ 13) 0.0003 36 ⫾ 6.1 (n ⫽ 10) 0.0005
CT plus TT/SE⫹ 49 ⫾ 12.5 (n ⫽ 111) 50 ⫾ 12.1 (n ⫽ 88)
RANKL/DRB1*04
CC/SE⫹ 35 ⫾ 7.2 (n ⫽ 13) 0.0005 36 ⫾ 6.1 (n ⫽ 10) 0.0013
CC/SE⫺ 52 ⫾ 13.2 (n ⫽ 12) 55 ⫾ 14.0 (n ⫽ 8)
RANKL/DRB1*04
CC/SE⫹ 35 ⫾ 7.2 (n ⫽ 13) ⬍0.0001 36 ⫾ 6.1 (n ⫽ 10) ⬍0.0001
CT plus TT/SE⫺ 53 ⫾ 12.5 (n ⫽ 101) 55 ⫾ 12.4 (n ⫽ 76)
RANKL/DRB1*04†
TAAA-C⫹/⫹ and SE⫹ 34 ⫾ 7.2 (n ⫽ 10) ⬍0.0001 35 ⫾ 6.3 (n ⫽ 9) ⬍0.0001
TAAA-C⫹/⫺ ⫹ TAAA-C⫺/⫺ and SE⫺ 53 ⫾ 12.4 (n ⫽ 102) 55 ⫾ 12.3 (n ⫽ 77)

* SE ⫽ shared epitope.
† Haplotype TAAA-C⫹/⫹ ⫽ TAAA-C/TAAA-C, haplotype TAAA-C⫹/⫺ ⫽ TAAA-C/absence of TAAA insertion -T, and
haplotype TAAA-C⫺/⫺ ⫽ homozygous for the absence of TAAA insertion -T.

SNP1 (rs922996; T⬎C), of RANKL was associated with who had RANKL-CT/TT (55 ⫾ 12.4 years; P ⬍ 0.0001)
age at RA symptom onset. As shown in Table 4, among (Table 4).
the 237 study patients, those with RANKL-CC genotypes A multiple regression model, in which age at RA
had earlier ages at disease onset (44 ⫾ 13.5 years) than onset was regressed on the presence of SE, RANKL
did those with RANKL-CT/TT genotypes (51 ⫾ 12.7 at-risk allele, and their interaction, showed that age at
years) (P ⫽ 0.0080). The presence of RA-associated onset was decreased an average of 18 years in the total
DRB1*04 alleles was associated with an earlier disease group of patients with both SE and RANKL-CC alleles
onset (47 ⫾ 12.7 years versus 53 ⫾ 12.5 years in SE– in comparison with patients with neither allele (age at
patients; P ⫽ 0.0004). In the presence of SE-containing onset ⫽ 53.10 – 4.6 ⫻ SE – 0.8 ⫻ RANKL-CC – 12.5
DRB1*04 alleles, RA patients with RANKL-CC geno- [SE ⫻ RANKL-CC] interaction). Individual P values
types had an earlier age at RA onset than did those with were P ⬍ 0.007 for the presence of SE, P ⫽ 0.85 for the
CT/TT genotypes (35 ⫾ 7.2 years versus 49 ⫾ 12.5 years; presence of RANKL-CC, P ⬍ 0.02 for the interaction of
P ⫽ 0.0003). In the absence of SE-containing DRB1*04 these 2 genotypes, and P ⬍ 0.0001 for the model. Similar
alleles, the RANKL-CC genotype was not associated multiple regression modeling resulted in an average 19.6
with an early age at RA onset compared with the CT/TT years younger age at RA onset in Caucasian patients
genotypes (52 ⫾ 13.2 years versus 53 ⫾ 12.5 years). with both SE and RANKL-CC alleles in comparison with
The mean age at disease onset in RA patients patients with neither allele (age at onset ⫽ 55.37 – 5.5 ⫻
harboring SE plus RANKL-CC (35 ⫾ 7.2 years) was an SE – 0.4 ⫻ RANKL-CC – 13.7 [SE ⫻ RANKL-CC]
average of 18 years younger than that in patients without interaction; P ⬍ 0.0001 for the model). This further
SE but with RANKL-CT/TT (53 ⫾ 12.5 years; P ⬍ supported the idea that an interaction between SE-
0.0001). Similarly, among the 182 Caucasian RA pa- containing DRB1*04 and RANKL-CC genotypes might
tients, those who had SE plus RANKL-CC (36 ⫾ 6.1 contribute to the younger age at RA onset. These P
years) developed RA an average of 19 years earlier than values remained significant after correction for multiple
did those without the RA-associated DRB1*04 alleles testing of 3 gene polymorphisms (P ⬍ 0.0003). The
RANKL AND HLA–DRB1 GENOTYPES AND AGE AT ONSET OF RA 3099

least 1 copy of the RA-associated DRB1*04 alleles, while


the majority of the patients who developed RA after age
50 (68 of 118 [58%]) lacked these DRB1*04 alleles
(Figure 1A).
Although only a few patients had RANKL-CC, a
higher proportion of those who had the RANKL-CC
genotype developed the disease before age 40 (Figure
1B). The co-occurrence of RA-associated DRB1*04 and
RANKL-CC genotypes was present in 23% of patients
who developed the disease during the third decade, 16%
who developed it during the fourth decade, 5% who
developed it fifth decade, and was absent in patients who
developed RA after the age of 50 years (Figure 1C). It
appeared that the co-occurrence of RA-associated
DRB1*04 and RANKL-CC genotypes was more preva-
lent in a subset of patients who developed RA at a
younger age, but the majority of our early RA patients
did not have these genotypes. Our results suggested that
1) the co-occurrence of DRB1*04 and RANKL-CC ge-
notypes increased the disease penetrance, resulting in an
earlier age at onset of RA, and 2) there might be genetic
differences between RA patients with younger age at
onset and those with older age at onset.
The tested SNP1 was predicted by the TFSearch
database to affect binding of transcription factors, which
might cause differential gene expression of RANKL.
SNP1 located in the 3⬘ end of the gene (Figure 2) might
be in linkage disequilibrium with the downstream gene.
To confirm the expression of RANKL rather than its
downstream gene, we tested another intronic polymor-
phism (the presence or absence of the 4-nucleotide
Figure 1. Proportion of patients with the rheumatoid arthritis (RA)–
TAAA insertion; –/TAAA, rs5803141) in the 5⬘ region
associated DRB1*04 and RANKL-CC alleles, according to age at RA
onset (3rd decade ⫽ ages 20–29 years; 4th decade ⫽ ages 30–39 years; of RANKL located 37.5 kb upstream of SNP1 (Figure 2).
5th decade ⫽ ages 40–49 years; 6th decade ⫽ ages 50–59 years; 7th Significant linkage disequilibrium between these 2 poly-
decade ⫽ 60–69 years; 8th decade ⫽ 70–79 years). morphisms was observed in our Caucasian patients
(D⬘ ⫽ 0.734, r2 ⫽ 0.52, P ⬍ 0.000001).

combination of both at-risk alleles lowered the age at


disease onset much more than did each individual risk
genotype, but the age effect of the RANKL-CC genotype
was not observed in the absence of SE (Table 4).
We grouped these 237 RA patients according to
their age at RA onset (by decades) to assess whether the
proportion of patients harboring the RA-associated Figure 2. The RANKL gene structure and the relative positions of
DRB1*04 alleles, the RANKL-CC genotype, and polymorphisms tested in this study. This gene located on 13q14 covers
DRB1*04 plus RANKL-CC genotypes varied according 45.28 kb, from 40,934,872 to 40,980,153 bp (National Center for
to age at disease onset (Figure 1). Strikingly, 12 of the 13 Biotechnology Information build 34; August 2003). The positions of
RA patients who had an onset between 20 and 30 years exons 1–8 are marked; exons 6 and 7 are physically close and are
indistinguishable on the scale used here. The SNP1 (T⬎C at
of age had at least 1 copy of the RA-associated DRB1*04 40,976,671 bp) and the insertion polymorphism (absence or presence
alleles (Figure 1A). The majority of the patients who of the 4-nucleotide TAAA insertion; –/TAAA, at 40,939,161 bp) are
developed RA before age 50 (74 of 119 [62%]) had at 37.5 kb apart.
3100 WU ET AL

Table 5. Lack of association between the RANKL SNP1 genotype and the yearly radiographic progression rate*
Erosions, Joint space narrowing, Total progression,
median (IQR) median (IQR) median (IQR)
Total (n ⫽ 190)
CC (n ⫽ 20) 0.73 (⫺0.0068, 3.17) 0.17 (⫺0.45, 1.69) 0.97 (⫺0.39, 4.77)
CT (n ⫽ 112) 0.48 (⫺0.041, 1.82) 0.025 (⫺0.11, 1.23) 1.01 (⫺0.12, 2.74)
TT (n ⫽ 58) 0.68 (0, 1.55) 0.33 (⫺0.075, 1.30) 1.14 (⫺0.037, 2.89)
CT ⫹ TT (n ⫽ 170) 0.49 (⫺0.023, 1.79) 0.081 (⫺0.11, 1.26) 1.06 (⫺0.10, 2.87)
P (CC vs. CT vs. TT)† 0.62 0.76 0.90
P (CC vs. [CT ⫹ TT])‡ 0.33 0.73 0.94
Caucasian patients (n ⫽ 149)
CC (n ⫽ 13) 0.55 (⫺0.027, 1.5) 0.064 (⫺1.17, 1.33) 0.82 (⫺1.30, 4.5)
CT (n ⫽ 89) 0.40 (⫺0.053, 1.79) 0.040 (⫺0.13, 1.07) 0.97 (⫺0.20, 2.48)
TT (n ⫽ 47) 0.66 (0, 1.49) 0.46 (⫺0.070, 1.25) 1.16 (⫺0.069, 2.87)
CT ⫹ TT (n ⫽ 136) 0.45 (⫺0.043, 1.62) 0.081 (⫺0.11, 1.09) 1.06 (⫺0.16, 2.59)
P (CC vs. CT vs. TT)† 0.94 0.42 0.61
P (CC vs. [CT ⫹ TT])‡ 0.73 0.32 0.47

* Values are the total Sharp score units per year. IQR ⫽ interquartile range (25th, 75th percentiles) of radiographic
progression.
† By Kruskal-Wallis nonparametric test for one-way analysis of variance.
‡ By Wilcoxon’s nonparametric rank-sum test.

These 2 polymorphisms were used to construct RANKL genotype and the erosion score at baseline was
haplotypes, which showed the absence of insertion (-)-T observed (P ⫽ 0.7). There was no statistically significant
(54.43%), TAAA-C (33.12%), TAAA-T (6.33%), and difference in the yearly radiographic progression rate
--C (6.12%) in our cohort. The frequencies of these 4 (erosion, joint space narrowing, and total Sharp scores)
haplotypes were 54.1%, 33.1%, 6.6%, and 5.8% in the in patients with the RANKL-CC genotype compared
Caucasians and 56.6%, 31.6%, 6.6%, and 5.3% in the with patients with the RANKL-CT/TT genotypes among
Hispanics, showing no significant difference between the the 149 Caucasians and the 190 total group patients in
2 ethnic groups (P ⫽ 0.98). RA patients with 2 copies of whom serial radiographs were obtained (Table 5). Sim-
the TAAA-C haplotype had a younger age at disease ilarly, no significant association between the RANKL
onset (44 ⫾ 14.4 years) compared with patients with 1 haplotype and the yearly radiographic progression rate
copy or with no copy of the TAAA-C haplotype (51 ⫾ was observed in this cohort (data not shown).
12.6 years; P ⫽ 0.019) (Table 4). The presence of
RANKL TAAA-C⫹/⫹ and SE⫹ was associated with
DISCUSSION
younger age at RA onset (34 ⫾ 7.2 years) compared with
the presence of RANKL TAAA-C⫹/– plus TAAA-C–/– This study explored one well-known genetic fac-
and SE– (53 ⫾ 12.4 years; P ⬍ 0.0001) (Table 4). These tor for RA (HLA–DRB1*0401, *0404, *0405, *0408
data supported the idea that RANKL polymorphisms alleles) and one novel candidate gene (RANKL) for their
and SE containing HLA–DRB1*04 alleles might interact potential roles in the development of RA. Using a
to predispose an individual to the early onset of RA. cohort of patients with early RA, we confirmed the
RANKL gene polymorphisms and erosive disease. report of (8–10) an association between DRB1*04 alleles
The role of RANK/RANKL in the regulation of bone and ages at RA onset (P ⫽ 0.0004) (Table 3) and the
destruction provided a rationale for assessing the asso- ethnic variation (36) in the RA-associated DRB1*04
ciation of RANKL genotypes with the presence of ero- alleles (Table 2). We observed an average of 6 years
sive disease at baseline and with the radiographic rate of younger age at RA onset among patients with SE-
progression. At study entry, 63 of 217 patients (29%) containing DRB1*04 alleles. The novel RANKL poly-
had erosive damage, as indicated by radiographic ero- morphisms (the insertion of TAAA, SNP1-C, and the
sion scores ⱖ2 (20 patients had no radiographic data at TAAA-C haplotype) were also associated with an earlier
study entry). Six of these 63 patients had the age at RA onset (⬃7 years). Most interesting was our
RANKL-CC genotype. Among the 154 patients with observation that the co-occurrence of DRB1*04 and
radiographic erosion scores ⬍2, 17 had the RANKL-CC RANKL risk alleles (haplotypes) was associated with a
genotype. No significant association between the much younger age at RA onset, an effect of ⬃18–20
RANKL AND HLA–DRB1 GENOTYPES AND AGE AT ONSET OF RA 3101

years (Table 4). The co-occurrence of DRB1*04 and ble 2), similar to previous findings by Del Rincon et al
RANKL risk alleles appeared to have a stronger impact (49). While the Hispanic patients in our study and the
on the earlier age at disease onset than did each gene study by Del Rincon were mainly of Mexican descent,
variant individually. Of note, this effect of RANKL risk Del Rincon and colleagues found no difference in the
alleles was present only among patients who had SE- age at symptom onset (or age at diagnosis) between their
containing DRB1*04 alleles. These RANKL genotypes Hispanic and Caucasian RA patients (49). We have no
appeared not to be associated with erosive disease at explanation for this difference except for the possible
baseline or with the yearly rate of progression of radio- difference in inclusion criteria: one of the inclusion criteria
graphic joint damage (Table 5). for our cohort but not theirs was rheumatoid factor sero-
We observed that the majority of the patients positivity.
who developed RA symptoms in their third to fifth We observed strong linkage disequilibrium (P ⬍
decades of life carried at least 1 copy of the DRB1*04
0.0001) between the 2 RANKL polymorphisms (the
alleles. In contrast, fewer than half of the patients who
insertion polymorphism and SNP1 shown in Figure 2)
developed RA in their sixth or later decades had these
that are 37.5 kb apart within the entire 45.3-kb gene.
alleles (Figure 1A). Interestingly, 92% of the patients
Either of the polymorphisms or the combined haplotype
who were diagnosed as having RA between the ages of
20 and 30 years carried at least 1 of the RA-associated was associated with an earlier age at RA onset, suggest-
DRB1*04 alleles, suggesting strong influences of the SE ing a role of RANKL variants in the pathogenesis of RA.
in the early onset of RA. The interaction between RANKL, RANK, and OPG is
The most prevalent DRB1*04 allele in this cohort pivotal in bone and cartilage homeostasis, as well as in
was *0401. Genotypes containing DRB1*0401, unlike the regulation of bone metabolism and immune func-
other SE-containing DRB1 alleles, have been shown to tions (23,28). The RANKL polymorphisms we tested, or
increase susceptibility or penetrance even if only 1 copy other polymorphisms in linkage disequilibrium, might
is present (8). This might explain our failure to find an affect levels of gene expression, resulting in differential
SE dose effect on the age at onset. To our knowledge, expression of RANKL protein. It is tempting to specu-
there has been no clear demonstration of an SE dose late that an abnormal level of RANKL expression might
effect on the age at RA onset (8). The proportion of RA dysregulate communication between dendritic cells and
patients harboring both DRB1*04 and RANKL risk T cells, resulting in a breakdown of the immune toler-
alleles was highest in patients who developed the disease ance to self components in the joint, while the presence
during their third decade of life (23%), exhibited a of SE-containing HLA–DRB1 alleles might shape the T
declining trend among patients diagnosed during their cell repertoire, leading to enhanced T cell reactivity
fourth (16%) and fifth (5%) decades, and was absent in toward arthritogenic antigens. The co-occurrence of
patients diagnosed after age 50 years (Figure 1C). These DRB1 and RANKL at-risk genotypes might accelerate
results suggested that genetic interactions between escape from tolerance, resulting in a much earlier devel-
HLA–DRB1*04 and RANKL genotypes enhanced dis- opment of arthritis.
ease penetrance and provided evidence for genetic In summary, the results of our study provide the
differences between early-onset and late-onset RA pa- first evidence of a strong association between an early
tients. We did not genotype other, non-DRB1*04 SE age at onset of RA and the combined presence of
alleles (*01, *10, *1402), since the presence of these HLA–DRB1 and RANKL genotypes. However, in view
alleles has not been reported to confer an increase in of the large number of genetic polymorphisms that were
either susceptibility or disease severity of seropositive not tested by us, it is possible that our findings could
RA among North American Caucasians (8,46–48). have occurred by chance and, at this time, should be
The age at onset of RA symptoms in Hispanics considered preliminary. Further studies are needed to
was earlier compared with that in Caucasians (42.2 ⫾ confirm this association and to elucidate the role attrib-
11.4 years versus 51.6 ⫾ 12.9 years; P ⬍ 0.0001) (Table uted to RANKL polymorphisms in the pathogenesis of
3). This ⬃8-year difference was also observed among RA.
SE⫹ Hispanics compared with SE⫹ Caucasians (40.4 ⫾
13.1 years versus 48.5 ⫾ 12.4 years; P ⫽ 0.015) (Table 3).
Compared with Caucasian patients, Hispanic patients ACKNOWLEDGMENTS
showed ethnic variations in the frequency and distribu- We thank all of the participating patients, and we
tion of the specific SE-containing DRB1*04 alleles (Ta- thank Elly Park for technical help in this study.
3102 WU ET AL

REFERENCES and disease severity in rheumatoid arthritis. Arthritis Rheum


1996;39:1802–7.
1. Jawaheer D, Gregersen PK. Rheumatoid arthritis: the genetic 19. Weyand CM, Hicok KC, Conn DL, Goronzy JJ. The influence of
components. Rheum Dis Clin North Am 2002;28:1–15. HLA-DRB1 genes on disease severity in rheumatoid arthritis. Ann
2. Huizinga TW. Genetics in rheumatoid arthritis. Best Pract Res Intern Med 1992;117:801–6.
Clin Rheumatol 2003;17:703–16. 20. Weyand CM, Goronzy JJ. Disease mechanisms in rheumatoid
3. Cornelis F, Faure S, Martinez M, Prud’homme JF, Fritz P, Dib C, arthritis: gene dosage effect of HLA–DR haplotypes. J Lab Clin
et al. New susceptibility locus for rheumatoid arthritis suggested by Med 1994;124:335–8.
a genome-wide linkage study. Proc Natl Acad Sci U S A 1998;95: 21. Seldin MF, Amos CI, Ward R, Gregersen PK. The genetics
10746–50. revolution and the assault on rheumatoid arthritis. Arthritis
4. Jawaheer D, Seldin MF, Amos CI, Chen WV, Shigeta R, Monteiro Rheum 1999;42:1071–9.
J, et al. A genomewide screen in multiplex rheumatoid arthritis 22. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature
families suggests genetic overlap with other autoimmune diseases. 2003;423:356–61.
Am J Hum Genet 2001;68:927–36. 23. Hofbauer LC, Heufelder AE. The role of osteoprotegerin and
5. Jawaheer D, Seldin MF, Amos CI, Chen WV, Shigeta R, Etzel C, receptor activator of nuclear factor ␬B ligand in the pathogenesis
et al. Screening the genome for rheumatoid arthritis susceptibility and treatment of rheumatoid arthritis. Arthritis Rheum 2001;44:
genes: a replication study and combined analysis of 512 multicase 253–9.
families. Arthritis Rheum 2003;48:906–16. 24. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C,
6. MacKay K, Eyre S, Myerscough A, Milicic A, Barton A, Laval S, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte
et al. Whole-genome linkage analysis of rheumatoid arthritis development and lymph-node organogenesis. Nature 1999;397:
susceptibility loci in 252 affected sibling pairs in the United 315–23.
Kingdom. Arthritis Rheum 2002;46:632–9. 25. Ziolkowska M, Kurowska M, Radzikowska A, Luszczykiewicz G,
7. Gregersen PK, Silver J, Winchester RJ. The shared epitope Wiland P, Dziewczopolski W, et al. High levels of osteoprotegerin
hypothesis: an approach to understanding the molecular genetics and soluble receptor activator of nuclear factor ␬B ligand in serum
of susceptibility to rheumatoid arthritis. Arthritis Rheum 1987;30: of rheumatoid arthritis patients and their normalization after
1205–13. anti–tumor necrosis factor ␣ treatment. Arthritis Rheum 2002;46:
8. Fries JF, Wolfe F, Apple R, Erlich H, Bugawan T, Holmes T, et al. 1744–53.
HLA–DRB1 genotype associations in 793 white patients from a 26. Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, et al.
rheumatoid arthritis inception cohort: frequency, severity, and Activated T cells regulate bone loss and joint destruction in
treatment bias. Arthritis Rheum 2002;46:2320–9. adjuvant arthritis through osteoprotegerin ligand. Nature 1999;
9. MacGregor A, Ollier W, Thomson W, Jawaheer D, Silman A. 402:304–9.
HLA-DRB1*0401/0404 genotype and rheumatoid arthritis: in- 27. Pettit AR, Ji H, von Stechow D, Muller R, Goldring SR, Choi Y,
creased association in men, young age at onset, and disease et al. TRANCE/RANKL knockout mice are protected from bone
severity. J Rheumatol 1995;22:1032–6. erosion in a serum transfer model of arthritis. Am J Pathol
10. Gonzalez-Gay MA, Garcia-Porrua C, Hajeer AH. Influence of 2001;159:1689–99.
human leukocyte antigen-DRB1 on the susceptibility and severity 28. Nakashima T, Wada T, Penninger JM. RANKL and RANK as
of rheumatoid arthritis. Semin Arthritis Rheum 2002;31:355–60. novel therapeutic targets for arthritis. Curr Opin Rheumatol
11. Massardo L, Gareca N, Cartes MA, Cervilla V, Gonzalez A, 2003;15:280–7.
Jacobelli S. The presence of the HLA-DRB1 shared epitope 29. Alarcon-Riquelme ME. A RUNX trio with a taste for autoimmu-
correlates with erosive disease in Chilean patients with rheumatoid nity. Nat Genet 2003;35:299–300.
arthritis. Rheumatology (Oxford) 2002;41:153–6. 30. Helms C, Cao L, Krueger JG, Wijsman EM, Chamian F, Gordon
12. Mattey DL, Dawes PT, Gonzalez-Gay MA, Garcia-Porrua C, D, et al. A putative RUNX1 binding site variant between
Thomson W, Hajeer AH, et al. HLA-DRB1 alleles encoding an SLC9A3R1 and NAT9 is associated with susceptibility to psoriasis.
aspartic acid at position 70 protect against development of rheu- Nat Genet 2003;35:349–56.
matoid arthritis. J Rheumatol 2001;28:232–9. 31. Prokunina L, Castillejo-Lopez C, Oberg F, Gunnarsson I, Berg L,
13. Mattey DL, Hassell AB, Dawes PT, Cheung NT, Poulton KV, Magnusson V, et al. A regulatory polymorphism in PDCD1 is
associated with susceptibility to systemic lupus erythematosus in
Thomson W, et al. Independent association of rheumatoid factor
humans. Nat Genet 2002;32:666–9.
and the HLA–DRB1 shared epitope with radiographic outcome in
32. Tokuhiro S, Yamada R, Chang X, Suzuki A, Kochi Y, Sawada T,
rheumatoid arthritis. Arthritis Rheum 2001;44:1529–33.
et al. An intronic SNP in a RUNX1 binding site of SLC22A4,
14. Plant MJ, Jones PW, Saklatvala J, Ollier WE, Dawes PT. Patterns
encoding an organic cation transporter, is associated with rheu-
of radiological progression in early rheumatoid arthritis: results of matoid arthritis. Nat Genet 2003;35:341–8.
an 8 year prospective study. J Rheumatol 1998;25:417–26. 33. Heinemeyer T, Wingender E, Reuter I, Hermjakob H, Kel
15. Wagner U, Kaltenhauser S, Sauer H, Arnold S, Seidel W, AE, Kel OV, et al. Databases on transcriptional regulation:
Hantzschel H, et al. HLA markers and prediction of clinical course TRANSFAC, TRRD and COMPEL. Nucleic Acids Res 1998;26:
and outcome in rheumatoid arthritis. Arthritis Rheum 1997;40: 362–7.
341–51. 34. Paulus HE, Bulpitt KJ, Ramos B, Park G, Wong WK. Relative
16. Gough A, Faint J, Salmon M, Hassell A, Wordsworth P, Pilling D, contributions of the components of the American College of
et al. Genetic typing of patients with inflammatory arthritis at Rheumatology 20% criteria for improvement to responder status
presentation can be used to predict outcome. Arthritis Rheum in patients with early seropositive rheumatoid arthritis. Arthritis
1994;37:1166–70. Rheum 2000;43:2743–50.
17. Emery P, Salmon M, Bradley H, Wordsworth P, Tunn E, Bacon 35. Paulus HE, Wiesner J, Bulpitt KJ, Patnaik M, Law J, Park GS, et
PA, et al. Genetically determined factors as predictors of radio- al. Autoantibodies in early seropositive rheumatoid arthritis, be-
logical change in patients with early symmetrical arthritis. BMJ fore and during disease modifying antirheumatic drug treatment.
1992;305:1387–9. J Rheumatol 2002;29:2513–20.
18. Reveille JD, Alarcon GS, Fowler SE, Pillemer SR, Neuner R, 36. Paulus HE, Oh M, Sharp JT, Gold RH, Wong WK, Park GS, et al.
Clegg DO, et al, for the MIRA Trial Group. HLA–DRB1 genes Correlation of single time-point damage scores with observed
RANKL AND HLA–DRB1 GENOTYPES AND AGE AT ONSET OF RA 3103

progression of radiographic damage during the first 6 years of multiple linked single-nucleotide polymorphisms. Am J Hum
rheumatoid arthritis. J Rheumatol 2003;30:705–13. Genet 2002;70:157–69.
37. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, 44. Abecasis GR, Cookson WO. GOLD—graphical overview of link-
Cooper NS, et al. The American Rheumatism Association 1987 age disequilibrium. Bioinformatics 2000;16:182–3.
revised criteria for the classification of rheumatoid arthritis. 45. Van Gestel AM, Prevoo ML, van ’t Hof MA, van Rijswijk MH, van
Arthritis Rheum 1988;31:315–24. de Putte LB, van Riel PL. Development and validation of the
38. Van der Heijde DM, van ’t Hof MA, van Riel PL, Theunisse LM, European League Against Rheumatism response criteria for rheu-
Lubberts EW, van Leeuwen MA, et al. Judging disease activity in matoid arthritis: comparison with the preliminary American Col-
clinical practice in rheumatoid arthritis: first step in the develop- lege of Rheumatology and the World Health Organization/Inter-
ment of a disease activity score. Ann Rheum Dis 1990;49:916–20. national League Against Rheumatism criteria. Arthritis Rheum
39. Blalock SJ, DeVellis RF, Brown GK, Wallston KA. Validity of the 1996;39:34–40.
Center for Epidemiological Studies Depression Scale in arthritis 46. Nepom GT, Gersuk V, Nepom BS. Prognostic implications of
populations. Arthritis Rheum 1989;32:991–7. HLA genotyping in the early assessment of patients with rheuma-
40. Sharp JT, Lidsky MD, Collins LC, Moreland J. Methods of scoring toid arthritis. J Rheumatol Suppl 1996;44:5–9.
the progression of radiologic changes in rheumatoid arthritis: 47. Nepom GT, Nepom BS. Prediction of susceptibility to rheumatoid
correlation of radiologic, clinical and laboratory abnormalities. arthritis by human leukocyte antigen genotyping. Rheum Dis Clin
Arthritis Rheum 1971;14:706–20. North Am 1992;18:785–92.
41. Sharp JT, Young DY, Bluhm GB, Brook A, Brower AC, Corbett 48. Nepom GT, Byers P, Seyfried C, Healey LA, Wilske KR, Stage D,
M, et al. How many joints in the hands and wrists should be et al. HLA genes associated with rheumatoid arthritis: identifica-
included in a score of radiologic abnormalities used to assess tion of susceptibility alleles using specific oligonucleotide probes.
rheumatoid arthritis? Arthritis Rheum 1985;28:1326–35. Arthritis Rheum 1989;32:15–21.
42. O’Dell JR, Nepom BS, Haire C, Gersuk VH, Gaur L, Moore GF, 49. Del Rincon ID, I, Battafarano DF, Arroyo RA, Murphy FT,
et al. HLA–DRB1 typing in rheumatoid arthritis: predicting Fischbach M, Escalante A. Ethnic variation in the clinical mani-
response to specific treatments. Ann Rheum Dis 1998;57:209–13. festations of rheumatoid arthritis: role of HLA–DRB1 alleles.
43. Niu T, Qin ZS, Xu X, Liu JS. Bayesian haplotype inference for Arthritis Rheum 2003;49:200–8.

You might also like