You are on page 1of 7

Clin Exp Med

DOI 10.1007/s10238-017-0469-y

ORIGINAL ARTICLE

Nonassociation of homocysteine gene polymorphisms


with treatment outcome in South Indian Tamil Rheumatoid
Arthritis patients
Niveditha Muralidharan1 • Reena Gulati2 • Durga Prasanna Misra1 •

Vir S. Negi1

Received: 12 January 2017 / Accepted: 3 August 2017


Ó Springer International Publishing AG 2017

Abstract The aim of the study was to look for any asso- Abbreviations
ciation of MTR 2756A[G and MTRR 66A[G gene poly- ACPA Anticitrullinated peptide antibody
morphisms with clinical phenotype, methotrexate (MTX) ACR American College of Rheumatology
treatment response, and MTX-induced adverse events in CEU Utah residents with Northern and Western
South Indian Tamil patients with rheumatoid arthritis (RA). European ancestry
A total of 335 patients with RA were investigated. MTR CHD Chinese in Metropolitan Denver
2756A[G gene polymorphism was analyzed by PCR– DAS28 (ESR) Disease Activity Score based on 28 joints
RFLP, and MTRR 66A[G SNP was analyzed by TaqMan score and Erythrocyte Sedimentation Rate
50 nuclease assay. The allele frequencies were compared DMARD Disease-modifying antirheumatic drug
with HapMap groups. MTR 2756G allele was found to be EULAR European League Against Rheumatism
associated with risk of developing RA. The allele fre- GIH Gujarat Indians in Houston
quencies of MTR 2756A[G and MTRR 66A[G SNPs in HWE Hardy Weinberg equilibrium
controls differed significantly when compared with Hap- JPT Japanese in Tokyo Japan
Map groups. Neither of the SNPs influenced the MTX LWK Luhya in Webuye, Kenya
treatment outcome and adverse effects. Neither of the SNPs MKK Maasai in Kinyawa, Kenya
seems to be associated with MTX treatment outcome and MTHFR Methylene tetrahydrofolate reductase
adverse events in South Indian Tamil patients with RA. MTX Methotrexate
PCR Polymerase chain reaction
Keywords Rheumatoid arthritis  MTR  MTRR  RA Rheumatoid arthritis
Methotrexate  Treatment response  Adverse events  RF Rheumatoid factor
South Indian Tamils RFLP Restriction fragment length
polymorphism
MTR 5-Methyltetrahydrofolate-homocysteine
methyltransferase
Electronic supplementary material The online version of this MTRR Methionine synthase reductase
article (doi:10.1007/s10238-017-0469-y) contains supplementary SNP Single nucleotide polymorphism
material, which is available to authorized users.
YRI Yoruba in Ibadan, Nigeria
& Vir S. Negi
vsnegi22@yahoo.co.in
1
Introduction
Department of Clinical Immunology, Jawaharlal Institute of
Postgraduate Medical Education and Research (JIPMER),
Puducherry 605 006, India Rheumatoid arthritis (RA) is the most prevalent chronic
2 systemic inflammatory autoimmune disease [1–3] charac-
Genetic Services Unit, Department of Paediatrics, Jawaharlal
Institute of Postgraduate Medical Education and Research terized by synovial inflammation and joint destruction. If
(JIPMER), Puducherry, India left untreated, the disease leads to severe disability,

123
Clin Exp Med

systemic complications, and early death [4–6]. on disease activity and patient tolerance, to a maximum
Methotrexate (MTX) is the most commonly used drug for of 25 mg/week at the end of 3 months. The patients were
treatment of RA which is effective in suppressing symp- followed up every 2 weeks during the course of MTX
toms and reducing disability [7]. dose escalation and once a month after a stable dose was
A significant inter-patient variability is known to occur reached. During each hospital visit, disease activity was
in clinical response to MTX [8] as 30–40% of the patients assessed by EULAR DAS28 criteria [17]. Response to
on MTX therapy fail to attain remission [9]. The lack of therapy was assessed at 16 weeks by EULAR response
reliable biomarkers necessitates the identification of criteria. The response was classified as ‘Good Response,’
genetic polymorphisms in MTX metabolic pathway to ‘Partial Response,’ and ‘No Response.’ Nonresponders
predict treatment response [10]. were offered combination DMARDs consisting of
Folate metabolism is an important therapeutic target of methotrexate, sulfasalazine/leflunomide, and hydroxy-
MTX, and numerous studies were conducted in diverse chloroquine. Partial responders were followed up till
ethnic populations exploring the associations of genetic 24 weeks and offered additional immunomodulator ther-
variants in folate pathway [11]. Genetic polymorphisms in apy if they failed to improve further. Complete responders
MTHFR gene have been thoroughly investigated [12, 13]. continued with methotrexate.
The 5-methyltetrahydrofolate-homocysteine methyltrans- Patients were monitored for MTX-induced toxicity by
ferase (MTR) and 5-methyltetrahydrofolate-homocysteine clinical assessment (oral ulcers, nausea, vomiting, diarrhea,
methyltransferase reductase (MTRR) genes play pivotal methotrexate flu, methotrexate nodulosis, and methotrex-
roles in homocysteine metabolism. There are fewer studies ate-induced interstitial lung disease) and laboratory inves-
conducted in the Caucasian group [14, 15] which investi- tigations including complete blood count (CBC) and liver
gated the association of MTR and MTRR gene polymor- function tests [to look for cytopenias (anemia, leucopenia,
phisms with methotrexate treatment outcome; however, thrombocytopenia or pancytopenia), rise in aminotrans-
there are no studies in the south Indian cohort of patients ferases or serum bilirubin, and rise of serum creatinine
with RA. Hence the aim of this study was to establish allele greater than 30% from baseline]. Patients were also mon-
frequencies of MTR 2756A[G and MTRR 66A[G SNPs in itored for development of infections, classifying those as
348 South Indian Tamil healthy controls and compare the serious if they required in-hospital treatment.
allele frequencies with HapMap groups and to identify any
associations of these SNPs with clinical phenotype, treat- Data collection
ment response, and MTX-induced adverse events in 335
patients with RA. Identification of genetic variants may Variables which could possibly influence the treatment
help in stratifying patients who are more likely to respond outcome and the development of MTX-induced adverse
or develop adverse events to MTX which will in turn aid in events were recorded. These variables included age, gen-
formulating better treatment options and management of der, disease duration, age at onset (young onset/late onset),
patients with RA. autoantibody status (RF and ACPA), and disease activity.

Genotyping
Patients and methods
Five milliliters of venous blood was collected, and DNA
Subjects was extracted by the salting-out procedure [18]. MTR
2756A[G genotyping (rs1805087) was performed by PCR
The study was conducted at the Department of Clinical RFLP method as per the published protocol [19]. The PCR
Immunology, Jawaharlal Institute of Postgraduate Medical product was digested with Hae III enzyme (New England
Education and Research (JIPMER), Puducherry, a major BioLabs) and electrophoresed in 3% agarose gel. Wild
tertiary referral center for South India. Newly diagnosed, types (2756AA) produced a single band at 211 bp.
DMARD naı̈ve patients fulfilling the modified ACR cri- Heterozygotes (2756AG) produced 211-, 131-, and 80-bp
teria for RA [16] were included in the study. A total of fragments. Homozygous mutants (2756GG) produced two
348 age- and gender-matched healthy individuals of fragments of 131 and 80 bp.
Tamil ethnicity, without any family history of autoim- MTRR 66A[G (rs1801394) genotyping was performed
mune disease were enrolled as healthy controls (HC). by TaqMan 50 nuclease assay consisting of allele-specific
Informed consent was taken from all participants, and the fluorogenic oligonucleotide probes and primers specific for
study was approved by the Institute Ethics Committee. MTRR 66A[G designed by Applied Biosystems, USA. The
Treatment was initiated with an initial MTX dose of sequences allowed discrimination of genotypes of each
10 mg/week and escalated by 5 mg every 2 weeks based studied pair of alleles.

123
Clin Exp Med

Laboratory measurement Table 1 Demographic and clinical profile of study subjects


Patients/controls (no.) 335/348
IgM rheumatoid factor (RF) in sera was measured at Sex (female/male) (%) 93/7
baseline by nephelometry (BN Prospec, Dade Behring,
Mean age (years) ± SEM 42.72 ± 0.55 years
Germany). Serum anticyclic citrullinated peptide antibody
Mean disease duration (years) ± SEM 3.75 ± 0.23 years
(ACPA) levels were examined by second-generation
Young onset disease (YORA) 314 (93.73%)
commercial ELISA kits (Biosystems, Barcelona, Spain).
Late onset disease (LORA) 21 (6.26%)
Antibody levels [10 and [20 IU/ml were regarded as RF
Erosive, deforming disease 237 (70.74%)
positive and ACPA positive, respectively.
Extra-articular manifestations 87 (25.97%)
Levels of homocysteine in plasma were measured at
Autoantibody phenotype
baseline by nephelometry (BN Prospec, Dade Behring,
RF positive 262 (78.20%)
Germany). Homocysteine levels of [15 lmol/L were
ACPA positive 250 (74.62%)
considered to be elevated.
Disease activity

Statistical analysis High disease activity 248 (74.02%)


Moderate disease activity 81 (24.17%)
Statistical analysis was carried out using GraphPad Instat Low disease activity 6 (1.79%)
version 3.0 and SPSS version 16.0. Differences in genotype Baseline DAS28 (mean ± SEM) 5.77 ± 0.05
and allele frequencies of MTR 2756A[G and MTRR Treatment response
66A[G between responders and nonresponders were ana- Mean dose of MTX (mg/week) 16.79 ± 0.21
lyzed by Chi-square test with Yate’s continuity correction. Good responders 121 (36.11%)
Odds ratio and 95% confidence interval were calculated for Partial responders 107 (31.94%)
each allele and genotype. p \ 0.05 was considered to be Nonresponders 107 (31.94%)
statistically significant. Sample size and power of the study YORA—young onset RA (onset \55 years); LORA—late onset RA
(80%) were calculated using CaTS software (Power cal- (onset 55 years or above); high disease activity (DAS28 score [5.1),
culator for genetic studies, compiled and published by moderate disease activity (DAS28 3.2 B 5.1); good response—(an
improvement of[1.2 in DAS28 score and a DAS of B2.6 (remission)
Centre for Statistical Genetics, Michigan University, on follow-up); no response—(B0.6 change in DAS score or a change
USA). between 0.6 and 1.2 with DAS score [3.7 on follow-up); moderate
Univariate logistic regression analysis for the associa- response—(an improvement 0.6–1.2 in DAS score with overall DAS
tion between methotrexate nonresponse and development between 2.6 and 3.7)
of MTX-induced adverse events with respective SNPs,
gender, and other clinical characteristics were tested, and
those factors that were significant in the univariate analysis gastrointestinal (GI) side effects like oral ulcers, nausea,
were included in a second multivariate logistic analysis. vomiting, or diarrhea. Thirteen (19.11%) had hematologi-
The binary outcome variables were ‘responders’ (good and cal manifestations in the form of anemia, leucopenia, or
partial responders) and ‘nonresponders’ for treatment out- pancytopenia. Five (7.35%) developed hepatotoxicity as
come and ‘with adverse events’ and ‘without adverse evidenced by an increase in amino transferase levels more
events’ for development of adverse events. than two times the upper limit of normal range which
improved on discontinuation of MTX. Another nine
(13.23%) developed infections such as herpes zoster, oral
Results candidiasis, cellulitis, or urinary tract infection. Three
patients (4.41%) had pulmonary toxicity, and one (1.47%)
Characteristics of study participants developed MTX-induced nodulosis.

The study included 335 patients with rheumatoid arthritis Clinical variables influencing the treatment outcome
treated with methotrexate, of which 93% were females. and development of adverse events
The baseline demographic details of the patients are sum-
marized in Table 1. At the time of enrollment all patients Univariate analysis showed that patients who were positive
were DMARD naı̈ve with active disease. for RF [p = 0.01, OR 2.37, 95% CI (1.23–4.57)] and
All patients received 5 mg two times per week folic acid ACPA [p = 0.03, OR 1.86, 95% CI (1.04–3.31)] showed
supplementation. Sixty-eight patients (20.29%) developed poor response to treatment. However, the statistical sig-
adverse events (AEs) related to MTX administration during nificance was retained only for RF [p = 0.04, OR 2.03,
the course of treatment. Thirty-seven of them (54.41%) had 95% CI (1.01–4.09)] after multivariate analysis

123
Clin Exp Med

(Supplementary Table 1). None of the variables influenced concordant with CEU and statistically different from JPT,
the development of MTX-induced adverse events. YRI, and CHD ethnic groups (Supplementary Table 3).
The major and minor allele frequencies of MTR
Relationship between homocysteine levels and gene 2756A[G and MTRR 66A[G SNPs remained same in
polymorphisms good and nonresponders groups, and hence neither of the
SNPs was found be associated with MTX treatment
The mean homocysteine levels were 10.29 ± 0.35 lmol/L response (Tables 3, 4). Similarly, no association was seen
(n = 108). Eleven patients (10.18%) had hyperhomocys- for both the SNPs with development of MTX adverse
teinemia. No association was found between levels of events (Supplementary Tables 4, 5).
homocysteine and any of the gene polymorphisms studied.

MTR 2756A>G and MTRR 66A>G gene Discussion


polymorphisms
The present study on South Indian Tamil patients with RA
The MTR 2756A[G genotype distribution in cases were found no significant association of MTR 2756A[G and
found to be AA 166 (50.30%), AG 154 (46.66%) and GG MTRR 66A[G SNPs with treatment outcome and adverse
10 (3.03%), and in controls it was found to be AA 227 events. Also neither of the SNPs influenced the plasma
(65.22%), AG 106 (30.45%) and GG 15 (4.31%). The homocysteine levels. However, MTR 2756G allele was
ancestral A allele frequency was 73.63% in cases and found to be associated with susceptibility to RA.
80.45% in controls, and mutant G allele frequency was The folate metabolism is critical in regulating the
26.36% in cases and 19.54% in controls. The genotype intracellular folate pool needed for synthesis and methy-
frequencies of MTR 2756A[G SNP among controls were lation of DNA [20]. MTR located at 1q 43 is essential for
in HWE, and in cases there was a minor deviation. maintaining adequate intracellular methionine, intracellular
The genotype and allele frequency of MTR 2756A[G folate, and normal homocysteine concentrations [21–23]. A
SNP in South Indians were compared with HapMap pop- common polymorphism in MTR gene involves a base
ulations, and it was found that the allele frequency were transition near the crucial vitamin B12-binding site
significantly different from YRI, GIH, LWK, and MKK resulting in a change of aspartic acid to glycine (2756A[G,
ethnic groups (Supplementary Table 2).The mutant MTR D919G) [21, 24, 25]. This SNP influence the enzyme’s
2756 G allele was associated with risk of developing RA secondary structure [24, 25]. The enzyme is maintained in
[p = 0.003, OR 1.47, 95% CI (1.14–1.90)] (Table 2). an active state by MTRR [26]. MTRR located on chromo-
The MTRR 66A[G genotype distribution in cases were some 5q15.3–p15.2 is a dual flavo protein that catalyzes the
found to be AA 87 (25.97%), AG 178 (53.13%), and GG reductive activation of cobalamin(II) to cobalamin(I), thus
70 (20.89%), and in controls it was found to be AA 82 making it available for remethylation of homocysteine to
(25.94%), AG 149 (47.15%), and GG 85 (26.89%). The methionine [26–29]. The common polymorphism in MTRR
ancestral A allele frequency was 52.53% in cases and gene is A[G substitution, leading to a change of isoleucine
49.52% in controls, and mutant G allele frequency was to methionine at amino acid 22 [30]. This substitution is
47.46% in cases and 50.47% in controls. The cases and thought to disrupt the binding of MTRR to the MTR-cob (I)-
controls of MTRR 66A[G SNP were in HWE. The geno- alanine complex which could decrease the rate of homo-
type and allele frequency of MTRR 66A[G SNP in South cysteine remethylation [27].
Indians were compared with HapMap populations, and it MTR 2756A[G SNP result in increased homocysteine
was found that the MTRR 66A[G allele frequencies were levels, decreased folate levels, and decreased cobalamin

Table 2 Genotype and allele frequencies of MTR 2756A[G in cases and controls
MTR 2756A[G genotype Cases (n = 330) Controls (n = 348) PC (Yates corrected) OR (95% CI)

AA 166 (50.30%) 227 (65.22%) 0.0001 0.53 (0.39–0.73)


AG 154 (46.66%) 106 (30.45%) \0.0001 1.99 (1.45–2.73)
GG 10 (3.03%) 15 (4.31%) 0.49 0.69 (0.30–1.56)
Allele
A 486 (73.63%) 560 (80.45%) 0.003 0.67 (0.52–0.87)
G 174 (26.36%) 136 (19.54%) 0.003 1.47 (1.14–1.90)

123
Clin Exp Med

Table 3 Genotype and allele frequencies of MTR 2756A[G in good, partial and nonresponders
MTR 2756A[G genotype Good responders (n = 121) Partial responders (n = 103) Nonresponders (n = 106)
n (%) n (%) PC OR (95% CI) n (%) PC OR (95% CI)

AA 55 (45.45%) 58 (56.31%) 0.13 0.64 (0.38–1.09) 53 (50%) 0.58 0.83 (0.49–1.40)


AG 64 (52.89%) 42 (40.77%) 0.09 1.63 (0.95–2.77) 48 (45.28%) 0.31 1.35 (0.80–2.29)
GG 2 (1.65%) 3 (2.91%) 0.85 0.56 (0.09–3.42) 5 (4.71%) 0.34 0.33 (0.06–1.78)
Allele
A 174 (71.90%) 158 (76.69%) 0.29 0.77 (0.50—1.19) 154 (72.64%) 0.94 0.96 (0.63–1.45)
G 68 (28.09%) 48 (23.30%) 0.29 1.28 (0.83—1.97) 58 (27.35%) 0.94 1.03 (0.68–1.56)

Table 4 Genotype and allele frequencies of MTRR 66A[G in good, partial and nonresponders
MTRR 66A[G genotype Good responders (n = 121) Partial responders (n = 107) Nonresponders (n = 107)
n (%) n (%) PC OR (95% CI) n (%) PC OR (95% CI)

AA 27 (22.31%) 27 (25.23%) 0.71 0.85 (0.46–1.56) 33 (30.84%) 0.19 0.64 (0.35–1.16)


AG 69 (57.02%) 59 (55.14%) 0.87 1.08 (0.63–1.82) 50 (46.72%) 0.15 1.51 (0.89–2.55)
GG 25 (20.66%) 21 (19.62%) 0.97 1.06 (0.55–2.04) 24 (22.42%) 0.87 0.90 (0.47–1.69)
Allele
A 123 (50.82%) 113 (52.80%) 0.74 0.92 (0.63–1.33) 116 (54.20%) 0.53 0.87 (0.60–1.26)
G 119 (49.17%) 101 (47.19%) 0.74 1.08 (0.74–1.56) 98 (45.79%) 0.53 1.14 (0.79–1.65)

levels in patient carriers of the 2756A variant versus those (1.14–1.90)]. Similar to our results, MTR 2756 GG geno-
with the 2756G variant. On the other hand, an 66A[G type and MTR 2756 G allele were associated with sus-
polymorphism in MTRR was associated with decreased ceptibility to RA [34]. Also in 106 Polish group of patients
homocysteine levels, increased folate levels, and increased with SLE, MTR 2756G allele was associated with SLE
cobalamin levels in those with the 66A variant versus those susceptibility [37]. The present finding of how MTR
with the 66G variant [31, 32]. Thus, both variants seem to 2756A[G gene polymorphism confers susceptibility to RA
have opposite contributions to homocysteine remethylation could be interpreted by the shared chromosomal location of
activity [33]. MTR with PTPN22, IL23R, and TGFb genes at chromo-
MTR 2756A allele frequency was 80.45%, and mutant G some 1. Any of these immune-related genes could be in
allele frequency was 19.54% in controls. In 324 healthy linkage disequilibrium with MTR 2756A[G SNP which in
subjects from the Jewish cohort [34], the MTR 2756A and turn could be responsible for the observed susceptibility in
G allele frequency was 86 and 14%, respectively. In 144 RA [38–40].
Western Indian healthy subjects [35], MTR 2756 G allele In the present study on South Indian Tamils, MTR
frequency was 34%. MTR 2756 allele frequencies were 2756A[G and MTRR 66A[G gene polymorphisms did not
significantly different from YRI, GIH, LWK, and MKK influence the MTX treatment response and adverse events.
ethnic groups. Our results are concordant to reports from Chaabane et al.
MTRR 66G allele frequency was 50% in this study and [41] and Wessels et al. [15]. Chaabane et al. [41] in a group
58% in volunteers of Uttar Pradesh [27]. In 144 Western of 141 Tunisian patients with RA reported no significant
Indian healthy subjects [35], MTRR G allele frequency was association of these SNPs with development of toxicities.
50%. In 294 South Indian healthy volunteers [36], MTRR Wessels et al. in 205 patients with active RA [15] found
66 A and G allele frequency was 33 and 67%, respectively. that neither of the SNPs had any association with clinical
MTRR 66A[G allele frequencies were concordant with response or with the development of toxicities. Grabar
CEU and statistically different from JPT, YRI, and CHD et al. [14] in 213 Caucasians reported that MTR 2756A[G
ethnic groups. and MTRR 66A[G SNPs had no role in predicting the
The mutant MTR 2756 G allele was associated with risk treatment outcome, MTX adverse events, and total homo-
of developing RA [p = 0.003, OR 1.47, 95% CI cysteine concentrations.

123
Clin Exp Med

On the contrary, James et al. [33] observed that Ethical approval The study was carried out in compliance with
patients with MTR 2756 A allele were more likely to international, national and institutional regulations. Institute Ethics
committee approved the study.
respond to methotrexate [OR 2.6, 95% CI (1.1–6.1)].
Similarly, in a cross-sectional study involving 86 patients Informed consent All persons gave informed consent prior to the
treated with MTX [34], it was found that MTR 2756GG inclusion in the study.
genotype was associated with MIARN (p = 0.013). In
213 patients with RA [14], MTRR 66A[G gene poly-
morphism was associated with lower risk of developing
dermatologic complaints (p = 0.007, OR 0.191, 95% CI References
0.057–637). Stamp et al. [42] found that MTRR 66A[G
1. Holers VM. Autoimmunity to citrullinated proteins and the ini-
gene polymorphism (p \ 0.0001) influenced RBC folate tiation of rheumatoid arthritis. Curr Opin Immunol.
concentrations. 2013;25:728–35.
Differences in the heterogeneity of results could be due 2. Deane KD. Can rheumatoid arthritis be prevented? Best Pract Res
to differences in demographic features, variations in the Clin Rheumatol. 2013;27:467–85.
3. Tobón GJ, Youinou P, Saraux A. The environment, geo-epi-
measures of treatment outcome, interethnic variations, and demiology, and autoimmune disease: rheumatoid arthritis.
lack of adequate sample size. Establishing pharmacoge- Autoimmun Rev. 2010;9:A288–92.
netic models [43] could provide better insights into the role 4. Tanaka Y. Current concepts in the management of rheumatoid
of folate metabolizing gene polymorphisms with MTX arthritis. Korean J Intern Med. 2016;31:210–8.
5. Vivar N, Van Vollenhoven RF. Advances in the treatment of
treatment outcome in RA. Measuring the intracellular rheumatoid arthritis. F1000prime Rep. 2014;6:31.
folate levels and analyzing other functional genetic variants 6. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis.
in homocysteine metabolism may provide better insights N Engl J Med. 2011;365:2205–19.
into the role of genetic variants with MTX treatment 7. Colmegna I, Ohata BR, Menard HA. Current understanding of
rheumatoid arthritis therapy. Clin Pharmacol Ther.
outcome. 2012;91:607–20.
The strength of the present study is that this is a 8. Wesoly J, Wessels JAM, Guchelaar H-J, Huizinga TWJ. Genetic
prospective genetic study conducted in South Indian markers of treatment response in rheumatoid arthritis. Curr
Tamils with adequate sample size (statistical power of Rheumatol Rep. 2006;8:369–77.
9. Ranganathan P, McLeod HL. Methotrexate pharmacogenetics:
[80%). However, the limitation of the study is that not all the first step toward individualized therapy in rheumatoid
functionally relevant genetic variants in MTR and MTRR arthritis. Arthritis Rheum. 2006;54:1366–77.
genes were analyzed. 10. Burgos PI, Danila MI, Kelley JM, Hughes LB, Bridges SL.
Understanding personalized medicine in rheumatoid arthritis: a
clinician’s guide to the future. Ther Adv Musculoskelet Dis.
2009;1:97–105.
Conclusion 11. Hider SL, Bruce IN, Thomson W. The pharmacogenetics of
methotrexate. Rheumatol Oxf Engl. 2007;46:1520–4.
MTR 2756G allele confers susceptibility to RA in South 12. Spyridopoulou KP, Dimou NL, Hamodrakas SJ, Bagos PG.
Methylene tetrahydrofolate reductase gene polymorphisms and
Indians. MTR 2756A[G and MTRR 66A[G gene poly- their association with methotrexate toxicity: a meta-analysis.
morphisms had no role with MTX treatment outcome and Pharmacogenet Genomics. 2012;22:117–33.
adverse events. 13. Owen SA, Lunt M, Bowes J, Hider SL, Bruce IN, Thomson W,
et al. MTHFR gene polymorphisms and outcome of methotrexate
Funding The study was supported by an intramural research grant by treatment in patients with rheumatoid arthritis: analysis of key
JIPMER, Puducherry, India. In addition, the infrastructure and other polymorphisms and meta-analysis of C677T and A1298C poly-
support provided by DST-FIST and ICMR-INSERM programs are morphisms. Pharmacogenomics J. 2013;13:137–47.
also acknowledged. 14. Bohanec Grabar P, Logar D, Lestan B, Dolzan V. Genetic
determinants of methotrexate toxicity in rheumatoid arthritis
patients: a study of polymorphisms affecting methotrexate
Author contributions VSN and RG conceived and planned the
transport and folate metabolism. Eur J Clin Pharmacol.
study. VSN recruited the patients and collected clinical data. NM
2008;64:1057–68.
performed laboratory evaluation and molecular analysis on patient
15. Wessels JAM, Kooloos WM, De Jonge R, De Vries-Bouwstra JK,
samples. NM and DPM carried out the statistical analysis and wrote
Allaart CF, Linssen A, et al. Relationship between genetic vari-
the manuscript. RG critically evaluated and gave useful suggestions to
ants in the adenosine pathway and outcome of methotrexate
improve the manuscript. VSN is the lead author and takes primary
treatment in patients with recent-onset rheumatoid arthritis.
responsibility for the data and the study.
Arthritis Rheum. 2006;54:2830–9.
16. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham
Compliance with ethical standards CO, et al. 2010 Rheumatoid arthritis classification criteria: an
American College of Rheumatology/European League Against
Conflict of interest All authors declare that there are no conflicts of Rheumatism collaborative initiative. Arthritis Rheum.
interest. 2010;62:2569–81.

123
Clin Exp Med

17. Van Gestel AM, Prevoo ML, van’t Hof MA, van’t Hof MA, van 31. Miriuka SG, Langman LJ, Evrovski J, Miner SES, D’Mello N,
Rijswijk MH, van de Putte LB, van Riel PL. Development and Delgado DH, et al. Genetic polymorphisms predisposing to
validation of the European League Against Rheumatism response hyperhomocysteinemia in cardiac transplant patients. Transpl Int
criteria for rheumatoid arthritis. Comparison with the preliminary Off J Eur Soc Organ Transplant. 2005;18:29–35.
American College of Rheumatology and the World Health 32. Silaste ML, Rantala M, Sämpi M, Alfthan G, Aro A, Kesäniemi
Organization/International League Against Rheumatism Criteria. YA. Polymorphisms of key enzymes in homocysteine metabo-
Arthritis Rheum. 1996;39:34–40. lism affect diet responsiveness of plasma homocysteine in healthy
18. Miller SA, Dykes DD, Polesky HF. A simple salting out proce- women. J Nutr. 2001;131:2643–7.
dure for extracting DNA from human nucleated cells. Nucleic 33. James HM, Gillis D, Hissaria P, Lester S, Somogyi AA, Cleland
Acids Res. 1988;16:1215. LG, et al. Common polymorphisms in the folate pathway predict
19. Skibola CF, Smith MT, Hubbard A, Shane B, Roberts AC, Law efficacy of combination regimens containing methotrexate and
GR, et al. Polymorphisms in the thymidylate synthase and serine sulfasalazine in early rheumatoid arthritis. J Rheumatol.
hydroxymethyltransferase genes and risk of adult acute lym- 2008;35:562–71.
phocytic leukemia. Blood. 2002;99:3786–91. 34. Berkun Y, Abou Atta I, Rubinow A, Orbach H, Levartovsky D,
20. Waseem M, Hussain SR, Kumar S, Serajuddin M, Mahdi F, Aamar S, et al. 2756GG genotype of methionine synthase
Sonkar SK, et al. Association of MTHFR (C677T) gene poly- reductase gene is more prevalent in rheumatoid arthritis patients
morphism with breast cancer in North India. Biomark Cancer. treated with methotrexate and is associated with methotrexate-
2016;8:111–7. induced nodulosis. J Rheumatol. 2007;34:1664–9.
21. Nikbakht M, MalekZadeh K, Kumar Jha A, Askari M, Marwaha 35. Ghodke Y, Chopra A, Shintre P, Puranik A, Joshi K, Patwardhan
RK, Kaul D, et al. Polymorphisms of MTHFR and MTR genes B. Profiling single nucleotide polymorphisms (SNPs) across
are not related to susceptibility to childhood ALL in North India. intracellular folate metabolic pathway in healthy Indians. Indian J
Exp Oncol. 2012;34:43–8. Med Res. 2011;133:274–9.
22. Zhang Z, Shi Q, Liu Z, Sturgis EM, Spitz MR, Wei Q. Poly- 36. Rai PS, Murali TS, Vasudevan TG, Prasada SK, Bhagavath AK,
morphisms of methionine synthase and methionine synthase Pai P, et al. Genetic variation in genes involved in folate and drug
reductase and risk of squamous cell carcinoma of the head and metabolism in a south Indian population. Indian J Hum Genet.
neck: a case-control analysis. Cancer Epidemiol Biomark Prev. 2011;17(Suppl 1):S48–53.
2005;14:1188–93. 37. Burzynski M, Duriagin S, Mostowska M, Wudarski M,
23. Yu K, Zhang J, Zhang J, Dou C, Gu S, Xie Y, et al. Methionine Chwalinska-Sadowska H, Jagodzinski PP. MTR 2756 A[G
synthase A2756G polymorphism and cancer risk: a meta-analy- polymorphism is associated with the risk of systemic lupus ery-
sis. Eur J Hum Genet EJHG. 2010;18:370–8. thematosus in the Polish population. Lupus. 2007;16:450–4.
24. Van der Put NM, van der Molen EF, Kluijtmans LA, Heil SG, 38. Rincón JFM, Cano DL, Morales SJ, Jiménez MLR, Cobos REB,
Trijbels JM, Eskes TK, et al. Sequence analysis of the coding Bello JR. The functional PTPN22 C1858T polymorphism confers
region of human methionine synthase: relevance to hyperhomo- risk for rheumatoid arthritis in patients from Central Mexico. Clin
cysteinaemia in neural-tube defects and vascular disease. QJM Rheumatol. 2016;35:1457–62.
Mon J Assoc Physicians. 1997;90:511–7. 39. Raslan HM, Attia HR, Salama I, Ibrahim MH, Hassan EM, El
25. Yates Z, Lucock M. Methionine synthase polymorphism A2756G Hussieny MS, et al. Association of PTPN22 1858C ? T poly-
is associated with susceptibility for thromboembolic events and morphism, HLA-DRB1 shared epitope and autoantibodies with
altered B vitamin/thiol metabolism. Haematologica. rheumatoid arthritis. Rheumatol Int. 2016;36:1167–75.
2002;87:751–6 (discussion 756). 40. Chen Y, Dawes PT, Packham JC, Mattey DL. Interaction
26. Mir MM, Dar JA, Dar NA, Dar MS, Salam I, Lone MM, et al. between smoking and functional polymorphism in the TGFB1
Combined impact of polymorphism of folate metabolism genes; gene is associated with ischaemic heart disease and myocardial
glutamate carboxypeptidase, methylene tetrahydrofolate reduc- infarction in patients with rheumatoid arthritis: a cross-sectional
tase and methionine synthase reductase on breast cancer sus- study. Arthritis Res Ther. 2012;14:R81.
ceptibility in kashmiri women. Int J Health Sci. 2008;2:3–14. 41. Chaabane S, Marzouk S, Akrout R, Ben Hamad M, Achour Y,
27. Rai V, Yadav U, Kumar P. MTRR A66G polymorphism among Rebai A, et al. Genetic determinants of methotrexate toxicity in
two caste groups of Uttar Pradesh (India). Indian J Med Sci. tunisian patients with rheumatoid arthritis: a study of polymor-
2012;66:136–40. phisms involved in the MTX metabolic pathway. Eur J Drug
28. Kim DJ, Park BL, Koh J-M, Kim GS, Kim LH, Cheong HS, et al. Metab Pharmacokinet. 2016;41:385–93.
Methionine synthase reductase polymorphisms are associated 42. Stamp LK, Chapman PT, O’Donnell JL, Zhang M, James J,
with serum osteocalcin levels in postmenopausal women. Exp Frampton C, et al. Polymorphisms within the folate pathway
Mol Med. 2006;38:519–24. predict folate concentrations but are not associated with disease
29. Naushad SM, Jain Jamal MN, Prasad CK, Rama Devi AR. activity in rheumatoid arthritis patients on methotrexate. Phar-
Relationship between methionine synthase, methionine synthase macogenet Genomics. 2010;20:367–76.
reductase genetic polymorphisms and deep vein thrombosis 43. Wessels JAM, van der Kooij SM, le Cessie S, Kievit W, Barerra
among South Indians. Clin Chem Lab Med. 2008;46:73–9. P, Allaart CF, et al. A clinical pharmacogenetic model to predict
30. Hu S, Liu H-C, Xi S-M. Methionine synthase reductase A66G the efficacy of methotrexate monotherapy in recent-onset
polymorphism is not associated with breast cancer susceptibil- rheumatoid arthritis. Arthritis Rheum. 2007;56:1765–75.
ity—a meta-analysis. Asian Pac J Cancer Prev APJCP.
2014;15:3267–71.

123

You might also like