GET THE APP

Replication of Identified Inflammatory Bowel Diseases Genetic Ass
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Research Article - (2013) Volume 0, Issue 0

Replication of Identified Inflammatory Bowel Diseases Genetic Associations: A Case-Control Study in the Tunisian Population

Bouzid D1*, Fourati H1, Amouri A2, Marques I3, Abida O1, Tahri N2, Penha-Gonçalves C3 and Masmoudi H1
1Immunology Department, Medicine School and Habib Bourguiba Hospital, Sfax, Tunisia
2Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
3Instituto Gulbenkian de Ciência, Oeras, Portugal
*Corresponding Author: Bouzid D, Immunology Department, Habib Bourguiba Hospital, 3029 Sfax, Tunisia, Tel: +216.74 45 16 60 Email:

Abstract

Inflammatory bowel diseases (IBD) — Crohn’s disease (CD) and ulcerative colitis (UC) — are chronic gastrointestinal inflammatory disorders with a complex genetic background. A genome wide association scan by the Welcome Trust Case Control Consortium (WTCCC) recently identified several novel susceptibility loci. We performed a replication study in 107 IBD patients (39 CD and 68 UC) and 162 controls. In total, 19 single nucleotide polymorphisms (SNPs) from previously identified susceptibility genes PTPN11, TNF α, IL23R, PTPN2, PTPN22, IL2 and IL10 were studied. In UC, we confirmed the association with PTPN2 (rs254215, GG, P=0.013, Pcorr=0.039; OR= 6.23 (1.18; 32.95)). In CD, we confirmed a marginal association with ((rs11066320, GG, P=0.018, Pcorr=0.054, OR= 0.4 (0.19; 0.82)). No significant association was found at the allele and genotype levels of SNPs in TNF α, IL23R, PTPN22, IL2, and IL10. However, on the haplotype analysis the AG haplotype of TNFα was more frequent in CD patients compared to controls (23.1% vs. 13.7%; P = 0.039; OR= 1.89 (1.02; 3.5)). The PTPN11ATG haplotype was also more frequent in CD patients compared to controls (32.1% vs 21.3%; P = 0.04; OR= 1.75 (1.01; 3.01). These results reveal limited replication in Tunisian population and indicate differences in genetic architecture between populations.

Introduction

Inflammatory bowel diseases (IBD) are disabling inflammatory disorders of the gastrointestinal tract, in which patients frequently suffer from relapsing bouts of intense abdominal pain, typically associated with diarrhoea and rectal bleeding [1]. While Crohn’s disease (CD) shows transmural and often discontinuous patches of inflammation, ulcerative colitis (UC) characteristically involves inflammation associated with only the superficial mucosal and submucosal layers of the intestinal wall [1]. Both conditions may have extra-intestinal manifestations, and may be associated with other chronic inflammatory diseases such as arthritis, ankylosing spondylitis or psoriasis [1].

The prevalence of IBD varies depending on ethnicity or race. Ashkenazi Jews have a much higher risk of developing IBD with a 2–4 times higher incidence when compared to non-Jewish Caucasians [2]. However, there is evidence to suggest that the prevalence of IBD increases in populations and regions with industrialization. The highest incidences are reported in northern Europe and North America where they range from 12 to 19/100,000/year and from 5 to 29/100,000/year, respectively and the lower incidences are reported in Asia and Africa, which probably reflects both genetic and environmental factors [1,3].

Currently, environmental risk factors and dysregulated mucosal immune responses are thought to cause chronic relapsing and remitting inflammation in genetically predisposed individuals, although the etiology of IBD remains unclear [4]. Family studies have provided overwhelming evidence that genetic factors play a significant role in determining an individual’s susceptibility to inflammatory bowel disease (IBD) [5-8]. The most compelling evidence comes from twin studies conducted in Sweden, Denmark, and the United Kingdom [5-7]. These studies reported Crohn’s disease (CD) and ulcerative colitis (UC) concordance rates of 58% and 6-19% for monozygotic twins, and 0.5-7% for dizygotic twin pairs [5-7]. The significantly higher concordance rates observed among monozygotic twin pairs across these studies indicate that there is a strong genetic component to IBD susceptibility.

Genome-wide association studies (GWASs) applying high-density SNP array technology have greatly expanded the number of genetic factors implicated in IBD pathogenesis to include over seventy genes and loci associated with CD and UC, spanning pathways involved in adaptive (IL23R, IL10, STAT3, PTPN2, PTPN22, TNFSF15, IL12B, CCR6) and innate (CARD15, ATG16L1, IRGM) immunity [9-14]. The TNFα, IL2 and PTPN11genes may also be considered as a plausible candidate for a genetic association with IBD [14-18].

Although IBD occurs worldwide, its epidemiologic and clinical characteristics vary depending upon the geographic location and the ethnicity of the population. Identifying the susceptibility genes of IBD in populations living in different geographical locations and with different ethnicities may provide significant clues about its etiology and pathophysiology [19].

However, all the published GWAS for IBD studies have been performed in populations of European descent. It will be extremely informative to test whether these new risk alleles associate with disease susceptibility in other ethnic groups.

We undertook a replication study in a cohort of IBD patients from Tunisia. We tested markers involved in adaptive immunity that showed moderate or strong association in previous case-control study.

Patients And Methods

Patients

Blood samples were obtained from 107 IBD patients (68 with UC, 39 with CD) diagnosed at the Department of Gastroenterlogy of Hedi Chaker University Hospital (Sfax, Tunisia) from March 2004 to May 2008. All patients were from Tunisian descent (North Africa). The diagnosis was based on standard clinical, radiographic, endoscopic, and histopathological criteria [20]. Table 1 summarizes clinical data: Patients with CD were assessed according to the Montreal classification [21] based on the age at diagnosis, localization, and behavior of disease. In patients with UC, anatomic localization was also determined according Montreal classification, using the criteria ulcerative proctitis, left-sided, and extensive. As unaffected controls (HC), 162 healthy Tunisian subjects with no history of digestive system disease, unrelated to each other or to the patients were recruited in this study.

  Crohn’s disease Ulcerative colitis Total (IBD)
Patients (n) 39 68 107
Sex (male/female) 24/15 33/35 57/50
Age (years),mean ± SD 39.4 ± 13.8 40.5 ± 12.4 40.1 ± 13.2
Pathology location
Ileum
Colon
Ileocolon
Pancolitis
Left-sided
Proctitis
14
11
14
35
26
7
 
Extra intestinal
manifestations
18 13 31

Table 1: Clinical description of Crohn’s disease and ulcerative colitis patients.

The study was approved by the local ethics committee and all enrolled patients gave their informed consent to participate.

Methods

Genotyping methods: Genomic DNA was extracted from whole blood samples using a standard proteinase K digestion and phenol/ chloroform extraction procedure. Genotyping was performed using primer extension chemistry and mass spectrometric analysis (iPlex assay, Sequenom, San Diego, CA) on the Sequenom MassArray at the Instituto Gulbenkian de Ciência, Oeiras, Portugal. We genotyped 3 SNPs in PTPN11 (rs12423190, rs11066323, rs11066320), 2 SNPs in TNFα (rs1800629, rs361525), 1 SNP in IL23R (rs11805303), 1 SNP in PTPN2 (rs2542151), 3 SNPs in PTPN22 (rs1310182, rs2476601, rs2488457), 4 SNPs in IL2 (rs2069763, rs1157812, rs2069762, rs6822844), and 5 SNPs in IL10 (rs3024498, rs3024495, rs1800871, rs1800896, rs1800872). SNP details are given in Table 2. All information about the selected SNPs was extracted from dbSNP public database, built 126 and gene information from the genome browser Ensembl, release 61. For quality control purposes these SNPs were also typed using a panel of HapMap samples.

locus Chr number Gene gene name   Position on Chr (bp) Allele HW
rs12423190 12 PTN11 protein tyrosine phosphatase, non-receptor type 11 intron 112909340 T/C 6,86E-01
rs11066323 12 PTN11 protein tyrosine phosphatase, non-receptor type 11 intron 112923361 G/A 9,97E-01
rs11066320 12 PTN11 protein tyrosine phosphatase, non-receptor type 11 intron 112906415 G/A 4.56E-03
rs1800629 6 TNF α tumor necrosis factor alpha 5'near gene prom 31543031 G/A 1,61E-01
rs361525 6 TNF α tumor necrosis factor alpha 5'near gene 31543101 G/A 1,94E-01
rs11805303 1 IL23R Interleukine 23 recepteur intron 67675516 C/T 2,99E-01
rs2542151 18 PTPN2 protein tyrosine phosphatase, non-receptor type 2 3'near gene 12779947 G/T 8,38E-01
rs1310182 1 PTPN22 Protein tyrosine phosphatase, non-receptor type 22   114373503 A/G 6,18E-01
rs2476601 1 PTPN22 Protein tyrosine phosphatase, non-receptor type 22 exon 114377568 A/G 8,72E-01
rs2488457 1 PTPN22 Protein tyrosine phosphatase, non-receptor type 22 promoter 114415368 G/C (G/C/A) 6,71E-01
rs2069763 4 IL2 Interleukine2 exon 123377482 C/A 1,62E-01
rs11575812 4 IL2 Interleukine2 3'near gene 123371049 A/G 7,72E-01
rs2069762 4 IL2 Interleukine2   123377980 A/C 2,72E-01
rs6822844 4 IL2 Interleukine 2 3'near gene 123509421 G/T 4,98E-01
rs3024498 1 IL10 Interleukine 10 3'UTR 206941529 T/C 7,51E-01
rs3024495 1 IL10 Interleukine 10 intron 206942413 C/T 9,52E-01
rs1800871 1 IL10 Interleukine 10 5'near gene 206946634 A/G 6,86E-02
rs1800896 1 IL10 Interleukine 10 5'near gene 206946897 T/C 9,97E-01
rs1800872 1 IL10 Interleukine 10 5'near gene 206946407 T/G 2,02E-01

Table 2: Selected polymorphisms in the candidate genes.

Assay design was performed according to manufacturer’s instructions, whereby the genomic sequence containing the SNP is amplified by multiplex PCR reactions. The amplified product was cleaned using shrimp alkaline phosphatase and used for allele specific primer extension reaction according the MassEXTEND protocol. The reaction mixture was then spotted onto a SpectroCHIP microarray and subjected to the MALDI-TOF mass spectrometry. SpectroTYPER software identifies the SNP-specific peaks and automatically assigns the genotype calls.

Data analysis: Quality control criteria excluded SNPs with call rate lower than 90% in cases and controls. Genotype frequencies were in Hardy-Weinberg equilibrium for all the typed SNPs (P>0.05) of the Tunisian control population. Case-control association analysis was performed for each SNP. Calculations of allelic and genotypic associations of SNPs with susceptibility to IBD were performed using a home-made program written in R language (www.r-project.org). This is a simple program which computes classical allelic and genotypic chi-square tests in an automated manner to avoid computing tests for each SNP one by one. A result is considered statistically significant when P<0.05. Bonferroni’s correction was carried out when single SNP analysis yield significant results. Relative risks were calculated as odds ratio (OR) using 2×2 contingency tables.

Results

Using the iPlex assay, we genotyped 19 SNPs in PTPN11, TNFα, IL23R, PTPN22, PTPN2, IL2 and IL10 genes from 107 IBD patients and 162 healthy subjects, all of South Tunisian origin. All SNPs tested except rs11066320 of PTPN11 were in Hardy-Weinberg equilibrium in the control group.

PTPN11

For the rs11066320 polymorphism of PTPN11, a significant increase in the frequency of the A allele was observed in IBD patients (29.7% vs. 20.5% for case and controls, respectively, OR=1.64, 95% CI=1.08-2.49, P=0.02, Pcorr=0.04) (Table 4). The AG genotype of the SNP rs11066320 was significantly more frequent in IBD patients than controls p=0.019, OR=1.93, but Bonferroni’s correction revealed marginal association (Pcorr=0.057). When stratified by disease type, only CD patients showed a significantly increase frequency of AG genotype (p=0.022) of the SNP rs11066320 compared with controls, but this result was not confirmed after Bonferroni’s correction (Pcorr=0.066) (Table 4). For rs11066323 polymorphism of PTPN11, CD patients showed a significantly increase frequency of AA genotype (P=0.027) compared with controls (Table 4). However, statistical significance was lost after Bonferroni’s correction (Pcorr=0.0.81). No significant differences were found in the allelic and genotypic frequencies of the rs12423190 polymorphism among CD and UC patients.

Using the genotyping data, we inferred 4 haplotypes. The haplotype ATG was statistically significantly associated with IBD (P=0.026). When stratified by disease type, only CD patients showed a significantly increase frequency of ATG haplotype (p=0.04) as compared with controls (Table 5).

TNF α

For each of the polymorphisms studied, the risk of carrying the variant was compared between CD, UC and control groups, as shown in Table 4. No associations were found at the allele and genotype levels between the polymorphism rs1800629 studied and risk of CD or UC. When combining UC and CD data to increase the statistical power there were still no associations between the polymorphism rs1800629 and risk of IBD. The rs361525 showed no statistically significant effect on patterns of UC disease risk (Table 4). However, CD patients showed a significant difference in the AA genotype frequency (p=0.038; OR=13.36 (1.18; 151.24)) of the SNP rs361525 compared with controls, but this result was not confirmed after Bonferroni’s correction (Pcorr=0.11) (Table 4). Haplotype analysis of two-SNPs was performed using Haploview. The results are summarized in Table 5. Haplotype frequencies were estimated, and association analyses were performed with respect to CD and UC and controls.

The results showed that there was no significant association between these haplotypes for UC patients as compared with control subjects (Table 5). However, we found that the AG haplotype was significantly associated (P=0.039) with CD as compared with control subjects.

IL23 R

By screening IBD patients and controls for the IL23R polymorphism (rs11805303), significant difference in the TT genotype frequency was detected in IBD patients (p=0.028, OR=2.37 (1.11; 5.07)), but this result was not confirmed after Bonferroni’s correction (Pcorr=0.069). When stratified by disease type, only UC patients showed a significant difference in the TT genotype frequency (p=0.031; OR=2.5(1.08; 5.81)) of the SNP rs11805303 compared with controls. However, statistical significance was lost after Bonferroni’s correction (P=0.0.87). No association was found between the rs11805303 variant and gender or other phenotypes (Table 4).

PTPN2

For rs2542151 polymorphism of PTPN2, a significant genotypic association was observed (GG: P=0.039, OR=4.76 (0.94; 24.04)) in IBD patients when compared with controls, but this result was not confirmed after Bonferroni’s correction (P=0.11). When stratified by disease type, only UC patients showed a significant difference in the GG genotype frequency (P=0.013; P=0.039; OR=6.23 (1.18; 32.95)) of the SNP rs2542151 compared with controls (Table 4).

PTPN22

For PTPN22, no significant differences in the frequencies of alleles and genotypes were detected between IBD patients and healthy controls.

The results of haplotype analysis of the three SNPs of PTPN22, showed that there was no significant association between these haplotypes for UC and CD patients as compared with control subjects (Table 5).

IL2

For IL2, no significant differences in the frequencies of alleles and genotypes were detected between IBD patients and healthy controls. Whereas, for the SNP rs11575812, UC patients showed a lower frequency (30.2%) of the AG genotype compared with controls (43.6%) (Table 3) without reaching significance level (p=0.0675, OR=0.59 (0.32; 1.09)).

Gene/SNP Genotypes CD UC IBD Controls
(Reference Allele) (%) (%) (%) (%)
PTPN11          
Rs11066320 AA 10.6 10.4 10.5 8.2
AG 44.7 35.8 39 24.6
GG 44.7 53.8 50.5 67.2
A 32.9 28.4 30 20.5
G 67.1 71.6 70 79.5
Rs12423190 CC 0 0 0 0
CT 13.2 19.4 17.1 23.4
TT 86.8 80.6 82.9 76.6
C 6.6 9.7 8.6 12.8
T 93.4 90.3 91.4 87.2
Rs11066323 AA 10.8 2.9 5.8 2.6
AG 24.3 25.4 25 26.9
GG 64.9 70.2 69.2 70.5
A 20.3 15.9 18.3 16
G 79.7 84.1 81.7 84
TNFα          
Rs1800629 AA 5.1 1.5 65.1 3.9
AG 33.3 31.3 32.1 23.2
GG 61.6 67.2 2.8 72.9
A 21.8 17.2 18.8 15.5
G 78.2 82.8 81.2 84.5
Rs361525 AA 2.7 0 0 0
AG 8.1 17.9 14.6 18.5
GG 89.2 82.1 85.4 81.5
A 6.8 9 8.2 9.3
G 93.2 91 91.8 90.7
IL23R          
Rs11805303 TT 15.8 17.9 17.2 8.2
TC 42.1 40.3 40.9 46.8
CC 42.1 41.8 41.9 45
T 36.8 38.1 37.6 31.6
C 36.2 61.9 62.4 68.4
PTPN2          
Rs2542151 GG 2.7 7.4 5.7 1.2
GT 16.2 13.2 14.3 21.4
TT 81.1 79.4 80 77.4
G 10.8 14 12.9 11.9
T 89.2 86 87.1 88.1
PTPN22          
Rs1310182 AA 18.4 17.9 18.1 13.2
AG 42.1 41.8 41.9 43.7
GG 39.5 40.3 40 43.1
A 39.5 38.8 39 36.3
G 60.5 61.2 61 63.7
Rs2476601 AA 0 0 0 0
AG 2.7 4.5 3.9 2.5
GG 97.3 95.5 96.1 97.5
A 1.4 2.3 1.9 1.3
G 98.6 97.7 98.1 98.7
Rs2488457 CC 75 66.7 69.8 67.3
CG 19.4 30 26 30.1
GG 5.6 3.3 4.2 2.6
C 84.7 81.7 82.8 82.4
G 15.3 18.3 17.2 17.6
IL2          
Rs2069763 AA 0 4.9 3.1 2.5
AC 22.2 21.3 21.6 18.5
CC 77.8 73.8 75.3 79
A 11.1 8.6 13.9 11.8
C 88.9 84.4 86.1 88.2
Rs11575812 AA 52.8 57.1 56 47.4
AG 41.7 30.2 34 43.6
GG 5.5 12.7 10 9
A 73.6 72.2 73 69.2
G 26.4 27.8 27 30.8
Rs2069762 AA 32.4 35.8 34.6 32.3
CA 54.1 41.8 46.2 45.2
CC 13.5 22.4 19.2 22.5
A 59.5 56.7 57.7 54.8
C 40.5 43.3 42.3 45.2
Rs6822844 GG 91.2 82.5 85.6 89.7
GT 0.8 17.5 14.4 10.3
TT 0 0 0 0
G 95.6 91.3 92.8 94.8
T 0.4 8.7 7.2 5.2
IL10          
Rs1800872 GG 55.6 47 50 43.6
GT 41.7 45.5 44.1 51.9
TT 2.7 7.5 5.9 4.5
G 76.4 69.7 72.1 69.6
T 23.6 30.3 27.9 30.4
Rs1800871 AA 5.4 8.9 7.7 5.9
AG 37.8 49.3 45.2 48.3
GG 56.8 41.8 47.1 45.8
A 24.3 33.6 30.3 30.1
Rs1800896 G 75.7 66.4 69.7 69.9
CC 18.4 7.6 11.6 14.9
CT 36.8 48.5 44.2 47.4
TT 44.8 43.9 44.2 37.7
C 36.8 31.8 33.7 38.6
T 63.2 68.2 66.3 61.4
Rs3024498 CC 2.7 0 0.9 2
CT 25 29.2 26.2 26.5
TT 72.3 70.8 72.9 71.5
C 15.3 14.6 14 15.2
T 84.7 85.4 86 84.8
Rs3024495 CC 72.3 76.5 75 78.2
CT 25 22.1 23.1 20.5
TT 2.7 1.4 1.9 1.3
C 84.7 87.5 86.5 84.8
T 15.3 12.5 13.5 15.2

Table 3: Genotype and allele frequencies of PTPN11, TNFα, IL23R, PTPN2, PTPN22, IL2 and IL10 in patients with CD, UC, IBD and unaffected controls.

GENE/SNP Genotype/ CD UC IBD
Allele P valuea (Pcorr)b OR (95 % CI) P valuea (Pcorr)b OR (95 % CI) P valuea (Pcorr)b OR (95 % CI)
PTPN11              
Rs11066320 AA 0.69   0.6   0.56  
  AG 0.022 (0.066) 2.48 (1.17; 5.25) 0.09 1.67(0.89;3.15) 0.019 (0.057) 1.93(1.11; 3.36)
  GG 0.018 (0.054) 0.4 (0.19; 0.82) 0.06 0.58 (0.32; 1.06) 0.011 (0.033) 0.51 (0.3; 0.86)
  A 0.034 (0.068) 1.90(1.08; 3.33) 0.079 1.50(0.93; 2.42) 0.02 (0.04) 1.64 (1.08; 2.49)
  G            
Rs12423190 CC 0.48   0.35   0.24  
  CT 0.16   0.54   0.23  
  TT 0.12   0.42   0.15  
  C 0.11   0.35   0.12  
  T            
Rs11066323 AA 0.027 (0.081) 4.61(1.1; 19.37) 0.84 1.17 (0.21; 6.54) 0.19 2.33 (0.64; 8.46)
  AG 0.69   0.85   0.73  
  GG 0.58   0.92   0.83  
  A 0.19   0.97   0.5  
  G            
TNFα              
Rs1800629 AA 0.75   0.35   0.64  
  AG 0.13   0.2   0.09  
  GG 0.11   0.39   0.15  
  A 0.14   0.67   0.27  
  G            
Rs361525 AA 0.038 (0.11) 13.36(1.18;151.24) -   0.21  
  AG 0.238   0.74   0.39  
  GG 0.44   0.74   0.52  
  A            
  G 0.71   0.75   0.67  
IL23R              
Rs11805303 TT 0.18   0.029 (0.087) 2.50(1.08; 5.81) 0.023 (0.069) 2.37 (1.11; 5.07)
  TC 0.72   0.31   0.35  
  CC 0.66   0.73   0.63  
  T            
  C 0.35   0.2   0.16  
PTPN2              
RS2542151 GG 0.53   0.013 (0.039) 6.23(1.18;32.95) 0.038 (0.11) 4.76 (0.94; 24.04)
  GT 0.44   0.16   0.15  
  TT 0.58   0.77   0.61  
  G 0.73   0.51   0.76  
  T            

CI: Confidence Interval; OR: Odds Ratio
Significant associations are highlighted in bold. a Two-tailed P values were calculated by home-made program written in R language. b Corrected P values Pcorr — after Bonferroni correction— are shown in brackets.

Table 4: Genotypic and allelic association tests of PTPN11, TNFα, IL23R and PTPN2 SNP polymorphisms with IBD (CD and UC).

The results of haplotype analysis of the four SNPs of IL2, showed that there was no significant association between these haplotypes for UC and CD patients as compared with control subjects (Table 5).

haplotype controls (n = 162) UC patients (n = 67) CD paients (n = 39) IBD paients (n = 107)
PTPN11        
GTG 0,498 0,461 0,397 0,437
ATG 0,213 0,284 0,321* 0,297**
GTA 0,16 0,159 0,218 0,181
GCG 0,129 0,097 0,064 0,085
TNFα        
GG 0,763 0,746 0,691 0,725
AG 0,137 0,172 0,231*** 0,193
GA 0,101 0,083 0,078 0,081
PTPN22        
GGC 0,645 0,606 0,618 0,611
AGC 0,175 0,206 0,227 0,214
AGG 0,163 0,156 0,142 0,156
AAG 0,013 0,023 0,013 0,019
IL2        
ACCG 0,442 0,432 0,419 0,427
GCAG 0,254 0,197 0,236 0,21
ACAG 0,131 0,149 0,189 0,155
AAAG 0,119 0,136 0,108 0,134
GCAT 0,052 0,087 0,047 0,074
IL10        
ACC 0,294 0,333 0,38 0,35
GCC 0,39 0,326 0,368 0,341
ATA 0,302 0,294 0,224 0,268
ACA 0,01 0 0,014 0,01
ATC 0 0,039 0,013 0,03

*P=0.04 OR=1.75 (1.01; 3.01); **P=0.026 OR=1.56 (1.05; 2.32); ***P=0.039, OR= 1,89 (1,02; 3,50)

Table 5: PTPN11, TNFα, PTPN22, IL2 and IL10 haplotypes frequencies in Crohn’s disease (CD), ulcerative colitis (UC), inflammatory bowel disease (IBD) patients and controls.

IL10

No associations were found at the allele and genotype levels between the five IL-10 polymorphisms studied and risk of CD or UC. When combining UC and CD data to increase the statistical power there were still no associations between the five IL-10 polymorphisms and risk of IBD.

We performed the haplotype analysis of the three SNPs in the 5’ flanking region of IL-10 (rs1800871, rs1800872 and rs180096). The results are summarized in Table 5. We showed that there was no significant association between these haplotypes for UC and CD patients as compared with control subjects.

Discussion

In recent years, GWA studies have provided a broad view of the relative contributions of various genomic loci and have substantially improved the understanding of the molecular pathways leading to IBD. GWA have stressed an association between IBD and specific chromosomal loci and thereby have led to the identification of candidate genes which are involved in the development of inflammatory processes [9,14]. As far as we know, no study has been yet performed in North African populations. In this work we evaluated the genetic contribution to IBD susceptibility of polymorphisms in some genes involved in the regulation of immune function.

SHP-2, coded by PTPN11, is a key downstream molecule that downregulates T- and B-cell activation through interaction with CTLA4 and PD-1 [22-25]. SHP-2 is recruited to the phosphorylated tyrosine residue in the immunoreceptor tyrosine-based switch motif of the cytoplasmic tail of PD-1 and in the immunoreceptor tyrosine-based inhibitory motif of the cytoplasmic tail of CTLA4 [26,27]. Although the mechanisms of the PTPN11 polymorphisms identified in this study are unknown, PTPN11 polymorphisms may affect the expression, affinity to immunoreceptors, and inhibitory activities of SHP-2, leading to the reduction in the SHP-2 down regulatory effect on T- and B-cell activation.

The present study is the first report to demonstrate an association between PTPN11polymorphisms and susceptibility to IBD in the Tunisian population. The presence of the allele A and the heterozygous AG genotype for SNP rs11066320 conferred susceptibility to IBD. When stratified by disease type, the AG genotype conferred susceptibility to CD. The AA genotype for rs11066323 conferred susceptibility to CD.

The association of the haplotype ATG with IBD, was statistically significant. These findings imply that PTPN11is a genetic determinant for predisposition to the onset of IBD in Tunisian individuals. However, the current population was relatively small, and further study on a larger number of the Tunisian population and on other ethnic populations is necessary to confirm the association between PTPN11and IBD.

TNFα is thought to be an important mediator of CD. Increased levels of TNFα messenger RNA and protein have been seen in inflamed and normal intestinal mucosa of patients with CD as well as in the serum [28,29]. As reviewed by Najarian and Gottlieb [30], TNFα may contribute to CD by causing apoptosis or inhibition of T-helper type 1 downregulatory cells in the lamina propria, thus stimulating mononuclear cells to release interferon gamma, interleukin-2, and TNFα. Other authors postulate that TNFα is involved in increased intestinal permeability, formation of granulomas, upregulation of endothelial cell adhesion molecules and coagulation pathways [30]. For the SNP rs361525, in the promoter region of TNFα, we found that AA genotype was associated with the CD. Furthermore, among the three TNFα promoter haplotypes resolved, AG haplotype was over represented in our CD cohort as compared with control subjects (OR=1,89; CI=1,02; 3,50). According to our data, selected studies of SNPs in the TNF promoter region have been found to be associated with IBD [31-33]. A Japanese study reported a significantly increased incidence of polymorphisms in the promoter region of the TNFα at positions -1031, -863, and -857 in patients with CD, versus ethnically matched controls [34,35] in a study on Korean patients observed a significantly higher frequency of -238A allele of TNF-α in CD patients with perianal lesions compared with patients without perianal disease. However, other studies that have confirmed evidence for linkage for an IBD susceptibility locus on 6p23 have failed to demonstrate an association to TNFα [36]. Santana et al. [37] also showed that TNF-α -308 polymorphism does not appear to be important for the susceptibility in the development of CD but may affect the severity of the disease.

It has been suggested that common IL23R gene variations on chromosome1p31 are likely to have a major impact on the differentiation of T helper cells into Th17 cells [38] and may thus contribute to autoimmunity [39]. The identification of susceptibility loci for IBD in IL23R and in other genes, whose gene products play a role in the IL23 ⁄ Th17 pathway, strongly suggests that proper IL23 ⁄ Th17 pathway regulation is crucial for intestinal immune homoeostasis and that its dysregulation plays a key role in IBD development [40,41]. Recent studies demonstrated that IL-23 is important for the survival and expansion of Th17 lymphocytes [42]. As a distinct subset of T helper cells, Th17 lymphocytes produce mostly IL17 and to a lesser extent also IL6 and TNFα [43]. IL17 acts in vitro and in vivo as a potent inflammatory cytokine. The IL23⁄ IL17cytokine axis is thus a key pathogenic mechanism that mediates the development and progress of inflammation via Th17 cells [42].

We showed a significant increase of the genotypic frequency of SNP rs11805303 of the IL23R gene in our UC patients but not in CD patients. This SNP was already reported to be associated with the susceptibility for CD and to be associated with ileal location in New Zealand population [44]. Multiple SNPs showing an association with both CD and UC have been identified in the IL23R gene by recent GWA studies. These findings were meanwhile replicated in a number of adult [13,45] and paediatric [46-50] cohorts. However, no associations were seen in a Japanese cohort [51].

PTPN2 (located on chromosome 18) encodes T cell PTP 2, an anti-inflammatory molecule. Dysregulated PTPN2 is known to be associated with abnormal JAK-STAT signalling, and evidence from mouse knockouts suggests that lack of dephosphorylation within the signalling cascade leads to marked elevation of several proinflammatory cytokines, including IFN-γ, TNF-α and IL12 [52]. The present study demonstrated an association between PTPN2 polymorphisms and susceptibility to UC in the Tunisian population. PTPN2 was first published as a susceptibility gene for CD in the GWAS by the WTCCC [53] in which strong associations for CD at four novel loci was reported. PTPN2 was one of these novel loci and rs2542151 was highly significantly associated with CD. Following the WTCCC results, there was a replication study [54] which showed modest evidence of replication for rs2542151 (P= 0.048) (Pcomb = 3.2 × 10−8). In addition, a very recent Italian study found no association of PTPN2 SNP with UC but a weak association of the PTPN2 SNP rs2542151 with susceptibility to CD (P=0.015; OR=1.34) [55].

PTPN22 (located on chromosome 1) encodes a lymphoid specific phosphatase known as Lyp, a strong negative regulator of T cell activation, either independently or through binding to a variety of adaptor molecules. In our study, there was no evidence for an association with PTPN22 and we recommend that further replication studies are undertaken to determine if this is a true association. Our results are in agreement with previous studies of Criswell et al. [56], Martin et al. [57] in Spanish subjects, Prescott et al. [58] in British subjects, and van Oene et al. [59] in Canadian patients. However, in the meta-analysis the SNP rs2476601 of PTPN22 gene was significant with a P value of 1.81 × 10−5 (replication P=1.01 × 10−4; combined P=1.46 × 10−8) [9]. In addition, the PTPN22 263Q loss-of-function variant showed initial evidence of association with UC in the Spanish cohort (P=0.026, OR=0.61), which was confirmed in the meta-analysis (P=0.013 pooled, OR=0.69) [60]. The lack of replication with PTPN22 may be falsely negative, possibly for the reason of the inconsistencies across studies regarding case or control exclusion criteria, such that phenotypic and genetic heterogeneity may exist. It is also possible that PTPN22 is significant only in the context of a particular environmental factor that is not present in the South Tunisian population. Much work, at both the population and molecular levels, remains before dismissing PTPN22 and its possible association with CD.

Genes encoding for immunoregulatory molecules clearly constitute important candidate susceptibility loci for IBD, and a number of recent studies have highlighted the central roles played by IL2 and IL-10 in orchestrating the immune response.

IL2, together with IFNγ, is produced by activated Th1 lymphocytes and stimulates the magrophages, natural killer cells and cytotoxic T cells of the cell-mediated immune response. The presence of large numbers of activated T cells in the involved mucosa of IBD patients suggests that IL2 is likely to be playing at least some role in stimulating inflammation [61,62]. This study revealed no association of the IL2 polymorphism with IBD in our patient population. Although recent non-synonymous SNP scan [12] did not find evidence for association of IL2 with UC, previous study in Spanish population did report evidence for associations with both phenotypes, either UC or CD [63]. Although the precise reasons for the lack of replication of the unclear, phenotype and genotype heterogeneity may contribute and some of the earlier studies were based on small sample sizes. IL10 is produced by Th2 lymphocytes and acts to downregulate macrophages and other antigen-presenting cells (APC). It also inhibits production of cytokines by Th1 lymphocytes in response to these APC, but does not block synthesis of cytokines by Th2 cells [64]. In this study, we did not find evidence for an association with the five polymorphisms studied. Several candidate gene studies that examined the association of the 3 IL-10 promoter polymorphisms with IBD susceptibility have had inconsistent results. In the UC GWAS that identified rs3024505, Franke et al. [65] did not find an association between the 3 promoter polymorphisms and IBD susceptibility. Another very recent study from New Zealand found an association with CD for SNPs rs3024505 and rs1800896, and phenotypic analysis indicated an association of rs3024505 with an early age at first diagnosis, stricturing CD behavior, and requirement for bowel resection [66].

Conclusion

Our replication study clearly reveals an apparent genetic/ allelic heterogeneity at UC/CD loci. It is likely that some SNPs that did not pass correction may be associated with IBD in south Tunisia nonetheless the study did not have sufficient power to detect these associations. In conclusion, the observed disparity in the allele frequency of genetic association study hits in our cohort confirms differences in genetic architecture between populations. However, further studies involving larger number of IBD patients should be performed before arriving at a definitive conclusion regarding the implications of the analyzed genes in IBD susceptibility.

Acknowledgements

This work was supported by Ministère de la recherche Scientifique et de la Technologie (MRST) (Tunisia). Genotyping was supported by the Instituto Gulbenkian de Ciência, Oeiras, Portugal. We are grateful to João Costa for providing technical support in genotyping.

References

  1. Loftus EV Jr (2004) Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology 126: 1504-1517.
  2. Nguyen GC, Torres EA, Regueiro M, Bromfield G, Bitton A, et al. (2006) Inflammatory bowel disease characteristics among African Americans, Hispanics, and non-Hispanic Whites: characterization of a large North American cohort. Am J Gastroenterol 101: 1012-1023.
  3. Ogunbi SO, Ransom JA, Sullivan K, Schoen BT, Gold BD (1998) Inflammatory bowel disease in African-American children living in Georgia. J Pediatr 133: 103-107.
  4. Hanauer SB (2006) Inflammatory bowel disease: epidemiology, pathogenesis, and therapeutic opportunities. Inflamm Bowel Dis 12 Suppl 1: S3-9.
  5. Orholm M, Binder V, Sørensen TI, Rasmussen LP, Kyvik KO (2000) Concordance of inflammatory bowel disease among Danish twins. Results of a nationwide study. Scand J Gastroenterol 35: 1075-1081.
  6. Tysk C, Lindberg E, Järnerot G, Flodérus-Myrhed B (1988) Ulcerative colitis and Crohn's disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut 29: 990-996.
  7. Thompson NP, Driscoll R, Pounder RE, Wakefield AJ (1996) Genetics versus environment in inflammatory bowel disease: results of a British twin study. BMJ 312: 95-96.
  8. Russell RK, Satsangi J (2004) IBD: a family affair. Best Pract Res Clin Gastroenterol 18: 525-539.
  9. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, et al. (2008) Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet 40: 955-962.
  10. Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, et al. (2008) Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat Genet 40: 1319-1323.
  11. Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, et al. (2010) Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet 42: 1118-1125.
  12. Fisher SA, Tremelling M, Anderson CA, Gwilliam R, Bumpstead S, et al. (2008) Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn's disease. Nat Genet 40: 710-712.
  13. Silverberg MS, Cho JH, Rioux JD, McGovern DP, Wu J, et al. (2009) Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study. Nat Genet 41: 216-220.
  14. UK IBD Genetics Consortium, Barrett JC, Lee JC, Lees CW, Prescott NJ, et al. (2009) Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region. Nat Genet 41: 1330-1334.
  15. Narumi Y, Isomoto H, Shiota M, Sato K, Kondo S, et al. (2009) Polymorphisms of PTPN11 coding SHP-2 as biomarkers for ulcerative colitis susceptibility in the Japanese population. J Clin Immunol 29: 303-310.
  16. Sanchez R, Levy E, Costea F, Sinnett D (2009) IL-10 and TNF-alpha promoter haplotypes are associated with childhood Crohn's disease location. World J Gastroenterol 15: 3776-3782.
  17. Hradsky O, Dusatkova P, Lenicek M, Bronsky J, Nevoral J, et al. (2010) The CTLA4 variants may interact with the IL23R- and NOD2-conferred risk in development of Crohn's disease. BMC Med Genet 11: 91.
  18. Morgan AR, Han DY, Huebner C, Lam WJ, Fraser AG, et al. (2010) PTPN2 but not PTPN22 is associated with Crohn's disease in a New Zealand population. Tissue Antigens 76: 119-125.
  19. Kim ES, Kim WH (2010) Inflammatory bowel disease in Korea: epidemiological, genomic, clinical, and therapeutic characteristics. Gut Liver 4: 1-14.
  20. Podolsky DK (1991) Inflammatory bowel disease (1). N Engl J Med 325: 928-937.
  21. Silverberg MS, Satsangi J, Ahmad T, Arnott ID, Bernstein CN, et al. (2005) Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 19 Suppl A: 5-36.
  22. Chuang E, Lee KM, Robbins MD, Duerr JM, Alegre ML, et al. (1999) Regulation of cytotoxic T lymphocyte-associated molecule-4 by Src kinases. J Immunol 162: 1270-1277.
  23. Schneider H, Rudd CE (2000) Tyrosine phosphatase SHP-2 binding to CTLA-4: absence of direct YVKM/YFIP motif recognition. Biochem Biophys Res Commun 269: 279-283.
  24. Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T (2001) PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A 98: 13866-13871.
  25. Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL (2004) SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol 173: 945-954.
  26. Greenwald RJ, Freeman GJ, Sharpe AH (2005) The B7 family revisited. Annu Rev Immunol 23: 515-548.
  27. Okazaki T, Honjo T (2007) PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol 19: 813-824.
  28. MacDonald TT, Hutchings P, Choy MY, Murch S, Cooke A (1990) Tumour necrosis factor-alpha and interferon-gamma production measured at the single cell level in normal and inflamed human intestine. Clin Exp Immunol 81: 301-305.
  29. Plevy SE, Landers CJ, Prehn J, Carramanzana NM, Deem RL, et al. (1997) A role for TNF-alpha and mucosal T helper-1 cytokines in the pathogenesis of Crohn's disease. J Immunol 159: 6276-6282.
  30. Najarian DJ, Gottlieb AB (2003) Connections between psoriasis and Crohn's disease. J Am Acad Dermatol 48: 805-821.
  31. Bouma G, Xia B, Crusius JB, Bioque G, Koutroubakis I, et al. (1996) Distribution of four polymorphisms in the tumour necrosis factor (TNF) genes in patients with inflammatory bowel disease (IBD). Clin Exp Immunol 103: 391-396.
  32. González S, Rodrigo L, Martínez-Borra J, López-Vázquez A, Fuentes D, et al. (2003) TNF-alpha -308A promoter polymorphism is associated with enhanced TNF-alpha production and inflammatory activity in Crohn's patients with fistulizing disease. Am J Gastroenterol 98: 1101-1106.
  33. Koss K, Satsangi J, Fanning GC, Welsh KI, Jewell DP (2000) Cytokine (TNF alpha, LT alpha and IL-10) polymorphisms in inflammatory bowel diseases and normal controls: differential effects on production and allele frequencies. Genes Immun 1: 185-190.
  34. Negoro K, Kinouchi Y, Hiwatashi N, Takahashi S, Takagi S, et al. (1999) Crohn's disease is associated with novel polymorphisms in the 5'-flanking region of the tumor necrosis factor gene. Gastroenterology 117: 1062-1068.
  35. Kim TH, Kim BG, Shin HD, Kim JW, Kim CG et al. (2003) Tumor necrosis factor-alpha and interleukin-10 gene polymorphisms in Korean patients with inflammatory bowel disease Korean J Gastroenterol 42: 377-386.
  36. Hampe J, Shaw SH, Saiz R, Leysens N, Lantermann A, et al. (1999) Linkage of inflammatory bowel disease to human chromosome 6p. Am J Hum Genet 65: 1647-1655.
  37. Santana G, Bendicho MT, Santana TC, Reis LB, Lemaire D, et al. (2011) The TNF-α -308 polymorphism may affect the severity of Crohn's disease. Clinics (Sao Paulo) 66: 1373-1378.
  38. Bettelli E, Oukka M, Kuchroo VK (2007) T(H)-17 cells in the circle of immunity and autoimmunity. Nat Immunol 8: 345-350.
  39. Afzali B, Lombardi G, Lechler RI, Lord GM (2007) The role of T helper 17 (Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune disease. Clin Exp Immunol 148: 32-46.
  40. Cho JH (2008) The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol 8: 458-466.
  41. Abraham C, Cho J (2009) Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflamm Bowel Dis 15: 1090-1100.
  42. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B (2006) TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 24: 179-189.
  43. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, et al. (2005) IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201: 233-240.
  44. Ferguson LR, Han DY, Fraser AG, Huebner C, Lam WJ, et al. (2010) IL23R and IL12B SNPs and Haplotypes Strongly Associate with Crohn's Disease Risk in a New Zealand Population. Gastroenterol Res Pract 2010: 539461.
  45. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, et al. (2006) A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314: 1461-1463.
  46. Dubinsky MC, Wang D, Picornell Y, Wrobel I, Katzir L, et al. (2007) IL-23 receptor (IL-23R) gene protects against pediatric Crohn's disease. Inflamm Bowel Dis 13: 511-515.
  47. Baldassano RN, Bradfield JP, Monos DS, Kim CE, Glessner JT, et al. (2007) Association of variants of the interleukin-23 receptor gene with susceptibility to pediatric Crohn's disease. Clin Gastroenterol Hepatol 5: 972-976.
  48. Van Limbergen J, Russell RK, Nimmo ER, Drummond HE, Smith L, et al. (2007) IL23R Arg381Gln is associated with childhood onset inflammatory bowel disease in Scotland. Gut 56: 1173-1174.
  49. Amre DK, Mack D, Israel D, Morgan K, Lambrette P, et al. (2008) Association between genetic variants in the IL-23R gene and early-onset Crohn's disease: results from a case-control and family-based study among Canadian children. Am J Gastroenterol 103: 615-620.
  50. Latiano A, Palmieri O, Valvano MR, D'Incà R, Cucchiara S, et al. (2008) Replication of interleukin 23 receptor and autophagy-related 16-like 1 association in adult- and pediatric-onset inflammatory bowel disease in Italy. World J Gastroenterol 14: 4643-4651.
  51. Yamazaki K, Onouchi Y, Takazoe M, Kubo M, Nakamura Y, et al. (2007) Association analysis of genetic variants in IL23R, ATG16L1 and 5p13.1 loci with Crohn's disease in Japanese patients. J Hum Genet 52: 575-583.
  52. Heinonen KM, Nestel FP, Newell EW, Charette G, Seemayer TA, et al. (2004) T-cell protein tyrosine phosphatase deletion results in progressive systemic inflammatory disease. Blood 103: 3457-3464.
  53. Wellcome Trust Case Control Consortium (2007) Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447: 661-678.
  54. Parkes M, Barrett JC, Prescott NJ, Tremelling M, Anderson CA, et al. (2007) Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn's disease susceptibility. Nat Genet 39: 830-832.
  55. Latiano A, Palmieri O, Latiano T, Corritore G, Bossa F, et al. (2011) Investigation of multiple susceptibility loci for inflammatory bowel disease in an Italian cohort of patients. PLoS One 6: e22688.
  56. Criswell LA, Pfeiffer KA, Lum RF, Gonzales B, Novitzke J, et al. (2005) Analysis of families in the multiple autoimmune disease genetics consortium (MADGC) collection: the PTPN22 620W allele associates with multiple autoimmune phenotypes. Am J Hum Genet 76: 561-571.
  57. Martín MC, Oliver J, Urcelay E, Orozco G, Gómez-Garcia M, et al. (2005) The functional genetic variation in the PTPN22 gene has a negligible effect on the susceptibility to develop inflammatory bowel disease. Tissue Antigens 66: 314-317.
  58. Prescott NJ, Fisher SA, Onnie C, Pattni R, Steer S, et al. (2005) A general autoimmunity gene (PTPN22) is not associated with inflammatory bowel disease in a British population. Tissue Antigens 66: 318-320.
  59. van Oene M, Wintle RF, Liu X, Yazdanpanah M, Gu X, et al. (2005) Association of the lymphoid tyrosine phosphatase R620W variant with rheumatoid arthritis, but not Crohn's disease, in Canadian populations. Arthritis Rheum 52: 1993-1998.
  60. Diaz-Gallo LM, Espino-Paisán L, Fransen K, Gómez-García M, van Sommeren S, et al. (2011) Differential association of two PTPN22 coding variants with Crohn's disease and ulcerative colitis. Inflamm Bowel Dis 17: 2287-2294.
  61. Mullin GE, Lazenby AJ, Harris ML, Bayless TM, James SP (1992) Increased interleukin-2 messenger RNA in the intestinal mucosal lesions of Crohn's disease but not ulcerative colitis. Gastroenterology 102: 1620-1627.
  62. Niessner M, Volk BA (1995) Altered Th1/Th2 cytokine profiles in the intestinal mucosa of patients with inflammatory bowel disease as assessed by quantitative reversed transcribed polymerase chain reaction (RT-PCR). Clin Exp Immunol 101: 428-435.
  63. Márquez A, Orozco G, Martínez A, Palomino-Morales R, Fernández-Arquero M, et al. (2009) Novel association of the interleukin 2-interleukin 21 region with inflammatory bowel disease. Am J Gastroenterol 104: 1968-1975.
  64. Radford-Smith G, Jewell DP (1997). Cytokines in Inflammatory Bowel Disease. In: Allan RN, Rhodes, JM, Hanauer SB (Eds) Inflammatory Bowel Diseases. (3rdedn) Churchill Livingstone, London pp:95-100.
  65. Franke A, Balschun T, Karlsen TH, Hedderich J, May S, et al. (2008) Replication of signals from recent studies of Crohn's disease identifies previously unknown disease loci for ulcerative colitis. Nat Genet 40: 713-715.
  66. Wang AH, Lam WJ, Han DY, Ding Y, Hu R, et al. (2011) The effect of IL-10 genetic variation and interleukin 10 serum levels on Crohn's disease susceptibility in a New Zealand population. Hum Immunol 72: 431-435.
Citation: Bouzid D, Fourati H, Amouri A, Marques I, Abida O, et al. (2012) Replication of Identified Inflammatory Bowel Diseases Genetic Associations: A Case–Control Study in the Tunisian Population. J Clin Cell Immunol S10:001.

Copyright: © 2012 Bouzid D, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top