Skip to main content
Erschienen in: BMC Medical Genetics 1/2010

Open Access 01.12.2010 | Research article

The polymorphism rs3024505 proximal to IL-10 is associated with risk of ulcerative colitis and Crohns disease in a Danish case-control study

verfasst von: Vibeke Andersen, Anja Ernst, Jane Christensen, Mette Østergaard, Bent A Jacobsen, Anne Tjønneland, Henrik B Krarup, Ulla Vogel

Erschienen in: BMC Medical Genetics | Ausgabe 1/2010

Abstract

Background

Crohns disease (CD) and ulcerative colitis (UC) are characterized by a dysregulated inflammatory response to normal constituents of the intestinal flora in the genetically predisposed host. Heme oxygenase-1 (HO-1/HMOX1) is a powerful anti-inflammatory and anti-oxidant enzyme, whereas the pro-inflammatory interleukin 1β (IL-1β/IL1B) and anti-inflammatory interleukin 10 (IL-10/IL10) are key modulators for the initiation and maintenance of inflammation. We investigated whether single nucleotide polymorphisms (SNPs) in the IL-1β, IL-10, and HO-1 genes, together with smoking, were associated with risk of CD and UC.

Methods

Allele frequencies of the IL-1β T-31C (rs1143627), and IL-10 rs3024505, G-1082A (rs1800896), C-819T (rs1800871), and C-592A (rs1800872) and HO-1 A-413T (rs2071746) SNPs were assessed using a case-control design in a Danish cohort of 336 CD and 498 UC patients and 779 healthy controls. Odds ratio (OR) and 95% confidence interval (95% CI) were estimated by logistic regression models.

Results

Carriers of rs3024505, a marker polymorphism flanking the IL-10 gene, were at increased risk of CD (OR = 1.40, 95% CI: 1.06-1.85, P = 0.02) and UC (OR = 1.43, 95% CI: 1.12-1.82, P = 0.004) and, furthermore, with risk of a diagnosis of CD and UC at young age (OR = 1.47, 95% CI: 1.10-1.96) and OR = 1.35, 95% CI: 1.04-1.76), respectively). No association was found between the IL-1β, IL-10 G-1082A, C-819T, C-592A, and HO-1 gene polymorphisms and CD or UC. No consistent interactions between smoking status and CD or UC genotypes were demonstrated.

Conclusions

The rs3024505 marker polymorphism flanking the IL-10 gene was significantly associated with risk of UC and CD, whereas no association was found between IL-1β or HO-1 gene polymorphisms and risk of CD and UC in this Danish study, suggesting that IL-10, but not IL-1β or HO-1, has a role in IBD etiology in this population.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2350-11-82) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

UV and AE carried out the genotyping. VA, HK, AE, MØ, BAJ established the cohort and/or participated in sample preparation and collection. JC and AT performed the statistical analyses. VA and UV conceived the genotyping study, and its design and coordination and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
CD
Crohns disease
CI
confidence interval
CO
carbon monoxide
COX-2
cyclooxygenase 2
HO-1
heme oxygenase 1
IBD
inflammatory bowel disease
IL-1β
interleukin 1β
IL-10
interleukin 10
iNOS
inducible nitric oxide synthase
NO
nitric oxide
OR
odds ratio
RQ-PCR
real-time quantitative RT-PCR
SNP
single nucleotide polymorphism
UC
ulcerative colitis
PGE2
prostaglandin E2

Background

The chronic inflammatory bowel diseases (IBD), ulcerative colitis (UC) and Crohn's disease (CD), are complex diseases caused by an interplay between genetic and environmental factors [1].
The recent years have brought much progress regarding the genetics in IBD and the number of confirmed IBD associated loci and genes have risen dramatically [27]. Yet, still, only part of the genetic contribution to disease risk may be explained by the identified genes [8, 9]. Northern European populations, including the Danish, generally have low frequencies of the CD risk-associated variants of CARD15 [3, 10], and it is therefore of interest to search for more genetic determinants in these populations. Less progress has been achieved in the identification of environmental risk factors and gene-environmental interactions. Differences in environmental exposures and genetic heterogeneity between ethnic groups may have complicated the search for genetic and gene-environmental determinants.
The emerging picture of IBD pathogenesis is focused on the sequential occurrence of pivotal events leading to the initiation and subsequent perpetuation of inflammation [11, 12]. First, the initial interaction between luminal constituents and intestinal epithelial cells leads to activation of the innate immune system [11]. The recognition of highly conserved pathogen structures such as lipopolysaccharide (LPS), the main constituent of Gram-negative bacteria, by Toll-like receptors and other pattern recognition receptors on the epithelial and other immunologically active cells in the intestine, initiates the release of various cytokines and enzymes, including interleukins (IL) and heme oxygenase-1 [13, 14]. Second, the inflammation will eventually become chronic due to defective regulation of the immune response. Therefore, polymorphisms in genes encoding cytokines and other molecules involved in the innate immune system, may affect the course of the inflammatory cascade and thereby the risk of developing IBD.
Activation of the pro-inflammatory IL-1β leads to production of prostaglandin E2 (PGE2) and nitric oxide (NO) via the induction of cyclo-oxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS) among others [15]. IL-1β knock-out mice have no spontaneous abnormalities, however, on challenge with LPS, a less pronounced acute phase response is observed, suggesting that IL-1β is required for an adequate immune response [15]. In both CD and UC patients, high levels of IL-1β are found in the intestinal mucosa [16] and stimulation by IL-1β leads to a more pronounced inflammatory response in CD immune cells compared to cells from healthy controls [17]. The variant alleles of two IL-1β promoter polymorphisms, IL-1β T-31C and IL-1β C-511T, have been found to be in almost complete linkage disequilibrium [18], and the haplotypes encompassing the IL-1β T-31C variant conferred higher transcription of IL-1β compared to the wild type haplotype [18]. The role of IL-1β polymorphisms in IBD has been explored in several studies [1924]. These studies did not find any association with IL-1β, however, the studies were rather small.
IL-10 is an anti-inflammatory cytokine, which leads to dampening of the activated immune system. IL-10 knock-out mice develop colitis if they are not kept in germ-free environment [25], and the administration of IL-10 ameliorates the inflammation in animal and in vitro models [26]. In patients, an impaired IL-10 production has been fund in severe cases of CD and UC [27, 28]. Recently, a strong association between the marker rs3024505 immediately downstream of the IL-10 gene and adult UC was found in a genome-wide association study [29]. This study also found a modest association between this marker and CD risk [29]. However, no association was found for rs3024493, a linked polymorphism located in an intron in the IL-10 gene, in a case-control study of paediatric onset CD [30]. The IL-10 promoter is polymorphic and genetic variation may account for different levels of cytokine production [31]. The IL-10 promotor polymorphisms G-1082A, C-819T, and C-592A have been most extensively studied. They are in tight linkage disequilibrium [32] and the haplotype encompassing these three polymorphisms is associated with low IL-10 protein production in lymphocytes in vitro [32] and low of levels of circulating IL-10 protein in Kenyan children [33] probably because the A allele of the IL-10 promoter polymorphism C-592A leads to the formation of a binding site for the ETS family of transcription factors [34]. Studies on the IL-10 promoter polymorphisms and IBD susceptibility have been inconsistent [22, 2931, 3540].
Heme oxygenase-1 (HO-1) is involved in the degradation of heme, thereby reducing oxidative stress and protecting against acute and chronic inflammation [41]. Animal models of IBD have confirmed the anti-inflammatory effect of HO-1 [42]. Hence, blockade of HO-1 activity results in exacerbation of experimental colitis whereas increased HO-1 pathway activity ameliorates experimental murine colitis [4345]. Carbon monoxide (CO) is one of the main metabolite of the HO-1 pathway and CO administration has been shown to ameliorate chronic colitis in IL-10 deficient mice [42, 46]. In UC patients, HO-1 expression and protein levels have been reported to be increased in inflamed colon compared to normal mucosa from patients with UC [47]. Studies using luciferase reporter assays of a functional promoter polymorphism, HO-1 A-413T, indicated that the A allele promoter had significantly higher activity than the T allele promoter [48]. The AA genotype of this polymorphism has been associated with a reduced incidence of ischemic heart disease [48]. Another promoter polymorphism, the HO-1 (GT)N dinucleotide repeat polymorphism, was not associated with risk of inflammatory bowel disease [49]. Interestingly, smoking affects the risk of IBD differentially, increasing the risk of CD and reducing the risk of UC [50]. The mechanisms by which smoking affects risk of IBD is not clear and as tobacco smoke contains more than 3000 chemicals, several different mechanisms may be involved. Nicotine has been reported to modulate the immune balance in a Th1-dominant direction [51] in accordance with the beneficial effect of smoking on UC. Moreover, smoking increases the production of certain pro-inflammatory cytokines, but, on the other hand, smoking is a source of carbon monoxide (CO). Thus, another possible mechanisms may involve interactions between smoking and IL-1β, IL-10 and HO-1 activity in relation to intestinal inflammation [46].
In this study we wanted to assess the role of polymorphisms in IL-1β, IL-10, and HO-1 together with smoking in relation to risk of developing IBD in a Danish case-control study of 336 CD, 498 UC and 779 healthy controls, respectively.

Methods

Patients and controls

Patients with CD (n = 373) or UC (n = 541), and healthy controls (n = 796) were included. All information was available for 336 CD cases, 498 UC cases and 779 healthy controls. Diagnosis of CD or UC was based on clinical, radiological, endoscopic and histological examinations [52]. Infectious and other cases of inflammatory bowel diseases were excluded. IBD diagnoses were classified on basis of disease localization according to the Montreal classification; L1, L2, and L3 signifies ileal, colonic, and ileocolonic localisation in CD and E1, E2, and E3 signifies the extent of colon involvement in UC; proctitis (inflammation limited to rectum), left side (inflammation distal to the left flexure), and extensive colon (inflammation proximal to the left flexure) [53]. Patients were recruited from Viborg, Aalborg and Herning Regional Hospitals from January 2004 to March 2005. Healthy blood donors were recruited from Viborg Hospital. Subjects under the age of 18 and with ethnicity other than Caucasian were excluded from the study. Phenotypic data on age at diagnosis of disease, localisation, family disposition for IBD, and medical and surgical treatment of disease was collected. Information on smoking habits at the time of diagnosis (patients) and smoking habits at study entry (donors) was collected.

Genotyping

DNA was extracted from EDTA-stabilized peripheral blood samples from all patients and healthy controls by using either a PureGene (Gentra Systems, Minneapolis, MN, USA) or Wizard Genomic (Promega, Madison, WI, USA) DNA purification kit, according to the manufacturers' recommendations.
SNPs were chosen from literature studies. IL-1β T-31C (rs1143627), and IL-10 C-592T (rs1800872) were genotyped as previously described [54] Rs3024505, IL-10 G-1082A (rs1800896), C-819T (rs1800871) [29] were genotyped using the pre-developed allelic discrimination assays C_15983681 20, C_1747360_10 and C_1747362_10 (Applied Biosystems). The genotyping reaction was performed in a final volume of 6 μl consisting of 3 μl Universal PCR Master Mix, 0.075 μl 40 × assay-on-demand mix, 1.925 μl H2O, and 1 μl genomic DNA. HO-1 A-413T (rs2071746 was determined using the pre-developed allelic discrimination assay C_15869717_10 (Applied Biosystems). Genotyping was performed by TaqMan real-time PCR on an ABI7900HT (Applied Biosystems), using Allelic Discrimination. Twenty ng of DNA was genotyped in 5 μl containing 1 × Mastermix (Applied Biosystems, Nærum, Denmark), 100 nM probes, and 900 nM primers or as recommended by the manufacturer for predesigned assays.. Controls of known genotypes were included in each run, and repeated genotyping of a random 10% subset yielded 100% identical genotypes. Laboratory personnel were blinded to the case/control status of the study group [55].

Statistical analyses

We used logistic regression to analyse the relationship between the six polymorphisms and disease. The statistical analyses included only subjects where all information was available. Age was entered linear in the model after checking for linearity using a linear spline [56]. Subgroup analyses were done for the polymorphisms in relation to location of the disease (CD: L1, L2, L3, UC: E1, E2, E3), and age at diagnosis (above or not above 40 years of age) for the cases. The GENMOD procedure in SAS release 9.1 (SAS Institute, Inc., Cary, North Carolina, USA) was used for the statistical analyses.

Power analyses

We used Genetic Power Calculator for Case - control for discrete traits [57] for power analyses. This study has more than 80% power to detect a dominant effect with an OR of 1.5 in either CD or in UC or 1.4 if CD and UC were combined.

Ethical Considerations

All subjects received written and oral information and gave written informed consent. The study was conducted in accordance with the Declaration of Helsinki and approved by the local Scientific Ethical Committees at Viborg and Aalborg County (VN 2003/5).

Results

Characteristics of the Danish IBD patients and controls are shown in Table 1. A total of 834 Danish patients and 779 controls were included. 51% of the CD patients were current smokers at the time of diagnosis, whereas only 17% of the UC patients were current smokers. The genotype distributions among the controls did not deviate from Hardy-Weinberg equilibrium. The variant allele frequencies for IL-1β T-31C, IL-10 rs3024505, G-1082A, C-819T, C-592A and HO-1 A-413T were 0.35, 0.18, 0.45, 0.21, 0.21, and 0.42, respectively, in the control group.
Table 1
The basic descriptions of the Danish study subjects1.
 
Crohns Disease
Ulcerative Colitis
Controls
 
(n = 336)
(n = 498)
(n = 779)
Gender: n (%)
   
   male
131 (39)
241 (48)
397 (51)
   female
205 (61)
257 (52)
382 (49)
Age:
   
   Median (5%-95%)
43 (23-77)
49 (24-76)
43 (23-60)
Age at diagnosis:
   
   Median (5%-95%)
30 (15-65)
35 (17-68)
 
Smoking habits: n(%)
   
   Smokers
170 (51)
85 (17)
204 (26)
   Never smokers
120 (36)
231 (46)
391 (50)
   Former smokers
46 (14)
182 (37)
184 (24)
Location UC 2 :
   
   Proctitis (E1)
 
211 (42)
 
   Left side (E2)
 
183 (37)
 
   Extensive (E3)
 
94 (19)
 
   Data not available
 
10 (2)
 
Location CD 2 : n (%)
   
   Colonic (L2)
75 (22)
  
   Ileal (L1)
155 (46)
  
   Ileocolonic (L3)
93 (28)
  
   Data not available
13 (4)
  
Medication: n (%)
   
   Advanced3
142 (42)
104 (21)
 
   No advanced medication4
189 (56)
390 (78)
 
   Data not available
5 (1)
4 (1)
 
Operation: n (%)
   
   Yes
153 (46)
15 (3)
 
   No
176 (52)
473 (95)
 
   Data not available
7 (2)
10 (2)
 
1The analyses included subjects where all information was available,
2The location of CD and UC in accordance with the Montreal classification are shown.
3azathioprin, 6-mercaptopyrine, Tumor Necrosis Factor-inhibitors, or methrotrexate,
45-aminosalicylic acid, prednisolone

Associations between polymorphisms and disease

Carriers of the variant allele of rs3024505 flanking the IL-10 gene were at increased risk of both CD and UC. Homozygous variant allele carriers were at 2.48-fold (95% CI: 1.27-4.84) increased risk of CD and heterozygous carriers were at 1.31-fold (95% CI: 0.98-1.75) increased risk of CD after adjusting for age, gender and smoking status (Table 2). Homozygous variant allele carriers were at 2.31-fold (95% CI: 1.27-4.20) increased risk of UC and heterozygous carriers were at 1.34-fold (95% CI: 1.04-1.73) increased risk of UC after adjusting for age, gender and smoking status (Table 3). After correction for multiple testing the associations were borderline statistically significant. Minor allele frequencies of IL-10 gene polymorphisms in selected studies are shown in Table 4.
Table 2
Genotypes in Danish patients with Crohns Disease1.
 
Ncase
Ncontrol
OR (95%CI)2
OR (95%CI)3
OR (95%CI)4
P-value5
IL1 β C-31T (rs1143627)
         
   TT
165
342
1.00
-
1.00
-
1.00
-
 
   CT
139
342
0.84
(0.64-0.10)
0.85
(0.65-1.12)
0.86
(0.65-1.14)
0.29
   CC
32
95
0.70
(0.45-1.09)
0.71
(0.45-1.10)
0.69
(0.44-1.09)
0.11
   CT and CC
171
437
0.81
(0.63-1.05)
0.82
(0.63-1.07)
0.82
(0.63-1.07)
0.15
IL-10 C-592A (rs1800872)
         
   CC
214
483
1.00
-
1.00
-
1.00
-
 
   AC
114
261
0.99
(0.75-1.29)
0.98
(0.75-1.30)
1.01
(0.76-1.34)
0.97
   AA
8
35
0.52
(0.24-1.13)
0.53
(0.24-1.16)
0.54
(0.24-1.21)
0.13
   AC and AA
122
296
0.93
(0.71-1.21)
0.93
(0.71-1.22)
0.95
(0.72-1.25)
0.72
IL-10 C-819T (rs1800871)
         
   CC
216
483
1.00
-
1.00
-
1.00
-
 
   CT
111
259
0.96
(0.73-1.26)
0.95
(0.72-1.25)
0.97
(0.73-1.29)
0.83
   TT
9
37
0.54
(0.26-1.15)
0.55
(0.26-1.18)
0.56
(0.26-1.22)
0.14
   CT and TT
120
296
0.91
(0.69-1.18)
0.90
(0.69-1.18)
0.92
(0.70-1.21)
0.55
IL-10 G-1082A (rs1800896)
         
   GG
109
238
1.00
-
1.00
-
1.00
-
 
   AG
171
374
1.00
(0.75-1.33)
0.99
(0.74-1.33)
0.97
(0.72-1.32)
0.87
   AA
56
167
0.73
(0.50-1.07)
0.75
(0.51-1.10)
0.78
(0.53-1.16)
0.23
   AG and AA
227
541
0.92
(0.70-1.21)
0.92
(0.69-1.21)
0.92
(0.69-1.22)
0.56
(rs3024505)
         
   CC
203
522
1.00
-
1.00
-
1.00
-
 
   CT
114
235
1.25
(0.95-1.64)
1.27
(0.96-1.68)
1.31
(0.98-1.75)
0.07
   TT
19
22
2.22
(1.18-4.19)
2.60
(1.36-4.96)
2.48
(1.27-4.84)
0.01
   CT and TT
133
779
1.33
(1.02-1.73)
1.37
(1.05-1.80)
1.40
(1.06-1.85)
0.02
HO-1 A-413T (rs2071746)
         
   AA
110
267
1.00
-
1.00
-
1.00
-
 
   AT
165
373
1.07
(0.81-1.43)
1.06
(0.79-1.42)
1.05
(0.78-1.42)
0.75
   TT
61
139
1.07
(0.73-1.55)
1.11
(0.76-1.62)
1.04
(0.70-1.53)
0.86
   AT and TT
226
512
1.07
(0.82-1.41)
1.08
(0.82-1.42)
1.05
(0.79-1.39)
0.75
1OR = Odds Ratio, 95%CI = 95% confidence interval,
2 Crude,
3 Adjusted for age and gender,
4 Adjusted for age, gender and smoking status.
5P for the fully adjusted estimate
Table 3
Genotypes in Danish patients with ulcerative colitis1.
 
Ncase
Ncontrol
OR (95%CI)2
OR (95%CI)3
OR (95%CI)4
P-value5
IL1 β C-31T (rs1143627)
         
   TT
204
342
1.00
-
1.00
-
1.00
-
 
   CT
238
342
1.17
(0.92-1.48)
1.15
(0.90-1.47)
1.15
(0.89-1.47)
0.28
   CC
56
95
0.99
(0.68-1.43)
0.99
(0.68-1.46)
1.02
(0.69-1.49)
0.94
   CT and CC
294
437
1.13
(0.90-1.42)
1.11
(0.88-1.41)
1.12
(0.88-1.42)
0.35
IL-10 C-592A (rs1800872)
         
   CC
328
483
1.00
-
1.00
-
1.00
-
 
   AC
149
261
0.84
(0.66-1.07)
0.83
(0.65-1.07)
0.83
(0.64-1.07)
0.14
   AA
21
35
0.88
(0.51-1.55)
0.99
(0.56-1.76)
1.00
(0.56-1.77)
0.99
   AC and AA
170
296
0.85
(0.67-1.07)
0.85
(0.67-1.08)
0.85
(0.66-1.08)
0.18
IL-10 C-819T (rs1800871)
         
   CC
325
483
1.00
-
1.00
-
1.00
-
 
   CT
151
259
0.87
(0.68-1.11)
0.86
(0.67-1.11)
0.85
(0.66-1.10)
0.22
   TT
22
37
0.88
(0.51-1.53)
1.00
(0.57-1.74)
1.00
(0.57-1.75)
0.99
   CT and TT
173
296
0.87
(0.69-1.10)
0.88
(0.69-1.12)
0.87
(0.68-1.11)
0.27
IL-10 G-1082A (rs1800896)
         
   GG
169
238
1.00
-
1.00
-
1.00
-
 
   AG
239
374
0.90
(0.70-1.16)
0.91
(0.70-1.19)
0.93
(0.72-1.22)
0.61
   AA
90
167
0.76
(0.55-1.05)
0.78
(0.56-1.08)
0.76
(0.54-1.06)
0.10
   AG and AA
329
541
0.86
(0.67-1.09)
0.87
(0.68-1.12)
0.88
(0.68-1.13)
0.31
(rs3024505)
         
   CC
297
522
1.00
-
1.00
-
1.00
-
 
   CT
172
235
1.29
(1.01-1.64)
1.35
(1.05-1.73)
1.34
(1.04-1.73)
0.02
   TT
29
22
2.32
(1.31-4.11)
2.37
(1.31-4.29)
2.31
(1.27-4.20)
0.01
   CT and TT
201
779
1.37
(1.09-1.74)
1.43
(1.13-1.83)
1.43
(1.12-1.82)
0.004
HO-1 A-413T (rs2071746)
         
   AA
162
267
1.00
-
1.00
-
1.00
-
 
   AT
251
373
1.11
(0.86-1.43)
1.10
(0.85-1.42)
1.11
(0.85-1.44)
0.45
   TT
85
139
1.01
(0.72-1.41)
1.00
(0.71-1.41)
1.01
(0.71-1.42)
0.98
   AT and TT
336
512
1.08
(0.85-1.37)
1.07
(0.84-1.37)
1.08
(0.84-1.38)
0.55
1OR = Odds Ratio, 95%CI = 95% confidence interval,
2 Crude,
3 Adjusted for age and gender,
4 Adjusted for age, gender and smoking status.
5P for the fully adjusted estimate
Table 4
Odds ratios and 95% confidence intervals (OR (CI)) for associations between IL-10 gene polymorphisms and ulcerative colitis (UC) or Crohns disease (CD) in selected case-control studies3.
Ncases/control
Trs3024505C1
Grs3024493T1
C-819T2
(rs1800871)
C-592A2
(rs1800872)
G-1082A2
(rs1800896)
Crs2222202T
 
UC
       
1855/3091
1.46 (1.31-1.62)
 
Neg
Neg
neg
 
[29]
203/391
  
neg
 
1.66 (1.30-2.14)
 
[36]
CD
       
1848/1804
1.17 (1.01-1.34)
     
[29]
270/336
 
neg
0.77 (0.58-1.00)
  
1.29 (1.01-1.64)
[30]
234-6/188-231
   
neg
neg
 
[35]
1rs3024505 and rs3024493 are in complete linkage disequilibrium [30]
2 IL-10 G-1082A, C-819T, and C-592A are in linkage disequilibrium [32] C-819T, and C-592A are in complete linkage disequilibrium [34]
3Negative associations are indicated by "Neg"
No association was found between the IL-1β, IL-10 G-1082A, C-819T, C-592A,, and HO-1 polymorphisms and risk of CD or UC (Table 2 and 3). The three IL-10 promoter polymorphisms were found to be in almost complete linkage as previously described for Caucasians [32]. Therefore, no haplotype analyses were performed.
No significant difference in the genotype distribution between CD and UC was found (data not shown). When combining UC and CD data to increase the statistical power there were still no associations between the IL-1β, the three IL-10 promoter polymorphisms, and HO-1 gene polymorphisms and risk of IBD (results not shown).
Subgroup analyses showed that variant allele carriers of rs3024505 were at 1.47-fold (95% CI: 1.10-1.96) and 1.35-fold (95% CI: 1.04-1.76) higher risk of a diagnosis of CD and UC, respectively, before the age of 40 years than the homozygous wildtype carriers (results not shown). No associations between rs3024505 genotype and disease localisation, or between IL-1β, the three IL-10 promoter polymorphisms, and HO-1 polymorphisms and age at diagnosis or disease localisation were found.

Gene-smoking interaction analyses

The effect of smoking habits at diagnosis on the genotype associations was investigated for CD and UC, respectively (Additional file 1: Interaction between the studied polymorphisms and smoking status in relation to risk of Crohns Disease and Additional file 2: Interaction between the studied polymorphisms and smoking status in relation to risk of ulcerative colitis). No consistent interactions between smoking status and any of the genotypes were found.

Discussion

The present case-control study showed that the rs3024505 marker polymorphism flanking the IL-10 gene was significantly associated with risk of CD and UC, and, furthermore, with risk of a diagnosis of CD and UC at young age. None of the polymorphisms IL-1β T-31C, IL-10 G-1082A, C-819T, C-592A, or HO-1 A-413T were associated with risk of CD, UC, or UC and CD combined. No consistent interactions between smoking status and genotypes were found.
Our results replicate the findings by Franke et al. [29] (Table 4). In addition, we found that the association was carried by a stronger association in the younger age group. Franke et al found that the variant allele of rs3024505 was associated with increased risk of UC with OR of 1.46 (95% CI: 1.31-1.62) and with CD with OR of 1.17 (95% CI: 1.01-1.34). Furthermore, they found no association between the three IL-10 promoter polymorphisms and risk of UC (results regarding CD were not reported). Previous studies were unable to find association between IBD and the IL-10 promoter polymorphisms [22, 31, 35, 39, 40] whereas other studies have found associations between paediatric onset of CD and IL-10 C-819T wildtype allele [30], Crs2222202T variant allele [30] and between the IL-10 G-1082A variant allele and risk of UC [58] (Table 4).
The biological significance of rs3024505 in IBD remains unclear [29]. The polymorphism is is located in an intergenic region proximal to the 3'UTR end of the IL-10 gene. The region has a high potential for containing regulatory sequences, and may thus regulate IL-10 gene expression [29]. Furthermore, rs3024505 is in perfect linkage with other polymorphisms located within the IL-10 gene [29]. On the other hand, since no associations were found between risk of UC or CD and the IL-10 promoter polymorphisms with proven functional effects on the IL-10 gene expression, this may suggest either that the rs3024505 has a much stronger regulatory effect on IL-10 levels than the promoter polymorphisms or that the effect of the polymorphism on disease risk is unrelated to IL-10 expression.
IL-1β, IL-10 and HO-1 are key players in the homeostasis of the intestinal immune system. Due to their pro-inflammatory and anti-inflammatory effects they are of significance for the development and maintenance of chronic inflammation. IL-1β has pro-inflammatory effects, whereas IL-10 and HO-1 have anti-inflammatory effects. A substantial number of studies document the roles of the interleukins, including IL-1β and IL-10, and HO-1 in intestinal inflammation in various animal IBD models and in IBD patients [15, 25, 26, 4347]. Therefore, genetic variations in these genes may cause imbalance in intestinal homeostasis and thereby contribute to chronic inflammation. On this background, IL-1β, IL-10 and HO-1 are relevant candidates for IBD susceptibility genes.
Our results are in accordance with previous studies which were unable to find association between IBD and IL-1β T-31C [24], taqI [19, 23] or C-511T [20, 21]. The HO-1 A-413T polymorphism has not previously been studied in relation to IBD, whereas no association was found between IBD and HO-1 (GT)N [49]. However, all these studies were small, the largest studies included 500 participants, and thus with limited statistical power to exclude an association. The polymorphisms analysed in the present study, IL-1β T-31C, and HO-1 A-413T have been shown to have biological effect [18, 48, 59], and the SNPs have previously been associated to risk of various disease entities [48, 60].
We found no consistent interactions between the studied polymorphisms and smoking in relation to risk of CD or UC. Although both smoking and nicotine administration lower the exaggerated IL-1β response in IBD patients [61, 62], the present study does not indicate that smoking at the time of diagnosis influences IBD risk by pathways involving IL-1β, IL-10 or HO-1, since the polymorphisms had no effect among present smokers. Cigarette smoke has been reported to act differentially on inflammation in the small and large intestine, thus worsening small intestinal inflammation, but ameliorating colitis [63]. We were not able to perform subgroup analyses to target this question due to limited statistical power.
It is important to stress the strengths and limitations of the study. The present study included 1600 participants and power analyses showed that this study has more than 80% power to detect a dominant effect with an OR of 1.5 in relation to either CD or UC and or 1.4 if CD and UC were combined. Moreover, genetic determinants may be stronger among patients with extensive disease and ileal disease [64, 65] and disease onset at low age. The effects of the polymorphisms might thus be below the detection level of our study.

Conclusions

In conclusion, the rs3024505 marker polymorphism flanking the IL-10 gene was associated with risk of UC and CD in the present Danish case-cohort study, and, furthermore, with risk of a diagnosis of CD and UC at young age. None of the polymorphisms IL-1β T-31C, IL-10 G-1082A, C-819T, C-592A, or HO-1 A-413T were associated with risk of CD or UC. No consistent interactions between smoking status and genotypes were found. The study suggests that IL-10, but not IL-1β or HO-1, play a role in IBD etiology.

Acknowledgements

Lars Bentzen is thanked for excellent technical assistance. Patients and donors are kindly thanked for participation. We thank Dr. Andre Franke for the suggestion to include rs3024505 in this study. We thank the staff at the Library, Viborg Regional Hospital. The project has been supported by the "Familien Erichsen Mindefond", the Lundbeck Foundation, the Danish Research Council, the Western Danish Research Forum for Health Science, the County of Viborg, the Danish Colitis-Crohn Association, "John M Klein og hustrus mindelegat" and "The A.P. Møller Foundation for the Advancement of Medical Science".
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

UV and AE carried out the genotyping. VA, HK, AE, MØ, BAJ established the cohort and/or participated in sample preparation and collection. JC and AT performed the statistical analyses. VA and UV conceived the genotyping study, and its design and coordination and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Xavier RJ, Podolsky DK: Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007, 448: 427-434. 10.1038/nature06005.CrossRefPubMed Xavier RJ, Podolsky DK: Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007, 448: 427-434. 10.1038/nature06005.CrossRefPubMed
2.
Zurück zum Zitat Franke A, Balschun T, Karlsen TH, Hedderich J, May S, Lu T, et al: Replication of signals from recent studies of Crohn's disease identifies previously unknown disease loci for ulcerative colitis. Nat Genet. 2008, 40: 713-715. 10.1038/ng.148.CrossRefPubMed Franke A, Balschun T, Karlsen TH, Hedderich J, May S, Lu T, et al: Replication of signals from recent studies of Crohn's disease identifies previously unknown disease loci for ulcerative colitis. Nat Genet. 2008, 40: 713-715. 10.1038/ng.148.CrossRefPubMed
3.
Zurück zum Zitat Ernst A, Jacobsen B, Ostergaard M, Okkels H, Andersen V, Dagiliene E, et al: Mutations in CARD15 and smoking confer susceptibility to Crohn's disease in the Danish population. Scand J Gastroenterol. 2007, 42: 1445-1451. 10.1080/00365520701427102.CrossRefPubMed Ernst A, Jacobsen B, Ostergaard M, Okkels H, Andersen V, Dagiliene E, et al: Mutations in CARD15 and smoking confer susceptibility to Crohn's disease in the Danish population. Scand J Gastroenterol. 2007, 42: 1445-1451. 10.1080/00365520701427102.CrossRefPubMed
4.
5.
Zurück zum Zitat Ostergaard M, Ernst A, Labouriau R, Dagiliene E, Krarup HB, Christensen M, et al: Cyclooxygenase-2, multidrug resistance 1, and breast cancer resistance protein gene polymorphisms and inflammatory bowel disease in the Danish population. Scand J Gastroenterol. 2009, 44: 65-73. 10.1080/00365520802400826.CrossRefPubMed Ostergaard M, Ernst A, Labouriau R, Dagiliene E, Krarup HB, Christensen M, et al: Cyclooxygenase-2, multidrug resistance 1, and breast cancer resistance protein gene polymorphisms and inflammatory bowel disease in the Danish population. Scand J Gastroenterol. 2009, 44: 65-73. 10.1080/00365520802400826.CrossRefPubMed
6.
Zurück zum Zitat Fisher SA, Tremelling M, Anderson CA, Gwilliam R, Bumpstead S, Prescott NJ, et al: Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn's disease. Nat Genet. 2008, 40: 710-712. 10.1038/ng.145.CrossRefPubMedPubMedCentral Fisher SA, Tremelling M, Anderson CA, Gwilliam R, Bumpstead S, Prescott NJ, et al: Genetic determinants of ulcerative colitis include the ECM1 locus and five loci implicated in Crohn's disease. Nat Genet. 2008, 40: 710-712. 10.1038/ng.145.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al: Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 2008, 40: 955-962. 10.1038/ng.175.CrossRefPubMedPubMedCentral Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH, Rioux JD, et al: Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet. 2008, 40: 955-962. 10.1038/ng.175.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Duerr RH: Update on the genetics of inflammatory bowel disease. J Clin Gastroenterol. 2003, 37: 358-367. 10.1097/00004836-200311000-00003.CrossRefPubMed Duerr RH: Update on the genetics of inflammatory bowel disease. J Clin Gastroenterol. 2003, 37: 358-367. 10.1097/00004836-200311000-00003.CrossRefPubMed
9.
Zurück zum Zitat Brant SR, Shugart YY: Inflammatory bowel disease gene hunting by linkage analysis: rationale, methodology, and present status of the field. Inflamm Bowel Dis. 2004, 10: 300-311. 10.1097/00054725-200405000-00019.CrossRefPubMed Brant SR, Shugart YY: Inflammatory bowel disease gene hunting by linkage analysis: rationale, methodology, and present status of the field. Inflamm Bowel Dis. 2004, 10: 300-311. 10.1097/00054725-200405000-00019.CrossRefPubMed
10.
Zurück zum Zitat Hugot JP, Zaccaria I, Cavanaugh J, Yang H, Vermeire S, Lappalainen M, et al: Prevalence of CARD15/NOD2 mutations in Caucasian healthy people. Am J Gastroenterol. 2007, 102: 1259-1267. 10.1111/j.1572-0241.2007.01149.x.CrossRefPubMed Hugot JP, Zaccaria I, Cavanaugh J, Yang H, Vermeire S, Lappalainen M, et al: Prevalence of CARD15/NOD2 mutations in Caucasian healthy people. Am J Gastroenterol. 2007, 102: 1259-1267. 10.1111/j.1572-0241.2007.01149.x.CrossRefPubMed
11.
Zurück zum Zitat Baumgart DC, Carding SR: Inflammatory bowel disease: cause and immunobiology. Lancet. 2007, 369: 1627-1640. 10.1016/S0140-6736(07)60750-8.CrossRefPubMed Baumgart DC, Carding SR: Inflammatory bowel disease: cause and immunobiology. Lancet. 2007, 369: 1627-1640. 10.1016/S0140-6736(07)60750-8.CrossRefPubMed
12.
13.
Zurück zum Zitat Fukata M, Abreu MT: What Are Toll-like Receptors and what Role May They Have in IBD. Inflamm Bowel Dis. 2008, 14: S90-S91. 10.1002/ibd.20632.CrossRefPubMed Fukata M, Abreu MT: What Are Toll-like Receptors and what Role May They Have in IBD. Inflamm Bowel Dis. 2008, 14: S90-S91. 10.1002/ibd.20632.CrossRefPubMed
14.
Zurück zum Zitat Werner T, Haller D: Intestinal epithelial cell signalling and chronic inflammation: From the proteome to specific molecular mechanisms. Mutat Res. 2007, 622: 42-57.CrossRefPubMed Werner T, Haller D: Intestinal epithelial cell signalling and chronic inflammation: From the proteome to specific molecular mechanisms. Mutat Res. 2007, 622: 42-57.CrossRefPubMed
15.
Zurück zum Zitat Dinarello CA: Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009, 27 (519-50): 519-550. 10.1146/annurev.immunol.021908.132612.CrossRefPubMed Dinarello CA: Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol. 2009, 27 (519-50): 519-550. 10.1146/annurev.immunol.021908.132612.CrossRefPubMed
16.
Zurück zum Zitat Guimbaud R, Bertrand V, Chauvelot-Moachon L, Quartier G, Vidon N, Giroud JP, et al: Network of inflammatory cytokines and correlation with disease activity in ulcerative colitis. Am J Gastroenterol. 1998, 93: 2397-2404. 10.1111/j.1572-0241.1998.00694.x.CrossRefPubMed Guimbaud R, Bertrand V, Chauvelot-Moachon L, Quartier G, Vidon N, Giroud JP, et al: Network of inflammatory cytokines and correlation with disease activity in ulcerative colitis. Am J Gastroenterol. 1998, 93: 2397-2404. 10.1111/j.1572-0241.1998.00694.x.CrossRefPubMed
17.
Zurück zum Zitat Kidd M, Gustafsson BI, Drozdov I, Modlin IM: IL1beta- and LPS-induced serotonin secretion is increased in EC cells derived from Crohn's disease. Neurogastroenterol Motil. 2009, 21: 439-450. 10.1111/j.1365-2982.2008.01210.x.CrossRefPubMed Kidd M, Gustafsson BI, Drozdov I, Modlin IM: IL1beta- and LPS-induced serotonin secretion is increased in EC cells derived from Crohn's disease. Neurogastroenterol Motil. 2009, 21: 439-450. 10.1111/j.1365-2982.2008.01210.x.CrossRefPubMed
18.
Zurück zum Zitat Chen H, Wilkins LM, Aziz N, Cannings C, Wyllie DH, Bingle C, et al: Single nucleotide polymorphisms in the human interleukin-1B gene affect transcription according to haplotype context. Hum Mol Genet. 2006, 15: 519-529. 10.1093/hmg/ddi469.CrossRefPubMed Chen H, Wilkins LM, Aziz N, Cannings C, Wyllie DH, Bingle C, et al: Single nucleotide polymorphisms in the human interleukin-1B gene affect transcription according to haplotype context. Hum Mol Genet. 2006, 15: 519-529. 10.1093/hmg/ddi469.CrossRefPubMed
19.
Zurück zum Zitat Hacker UT, Bidlingmaier C, Gomolka M, Keller E, Eigler A, Hartmann G, et al: Inflammatory bowel disease: no association between allele combinations of the interleukin (IL) I beta and IL-I receptor antagonist gene polymorphisms. Eur J Clin Invest. 1998, 28: 214-219. 10.1046/j.1365-2362.1998.00277.x.CrossRefPubMed Hacker UT, Bidlingmaier C, Gomolka M, Keller E, Eigler A, Hartmann G, et al: Inflammatory bowel disease: no association between allele combinations of the interleukin (IL) I beta and IL-I receptor antagonist gene polymorphisms. Eur J Clin Invest. 1998, 28: 214-219. 10.1046/j.1365-2362.1998.00277.x.CrossRefPubMed
20.
Zurück zum Zitat Corleto VD, Pagnini C, Margagnoni G, Guagnozzi D, Torre MS, Martorelli M, et al: IL-1beta-511 and IL-1RN*2 polymorphisms in inflammatory bowel disease: An Italian population study and meta-analysis of European studies. Dig Liver Dis. 2010, 42: 179-84. 10.1016/j.dld.2009.06.016.CrossRefPubMed Corleto VD, Pagnini C, Margagnoni G, Guagnozzi D, Torre MS, Martorelli M, et al: IL-1beta-511 and IL-1RN*2 polymorphisms in inflammatory bowel disease: An Italian population study and meta-analysis of European studies. Dig Liver Dis. 2010, 42: 179-84. 10.1016/j.dld.2009.06.016.CrossRefPubMed
21.
Zurück zum Zitat Ferreira AC, Almeida S, Tavares M, Canedo P, Pereira F, Regalo G, et al: NOD2/CARD15 and TNFA, but not IL1B and IL1RN, are associated with Crohn's disease. Inflamm Bowel Dis. 2005, 11: 331-339. 10.1097/01.MIB.0000158153.71579.b4.CrossRefPubMed Ferreira AC, Almeida S, Tavares M, Canedo P, Pereira F, Regalo G, et al: NOD2/CARD15 and TNFA, but not IL1B and IL1RN, are associated with Crohn's disease. Inflamm Bowel Dis. 2005, 11: 331-339. 10.1097/01.MIB.0000158153.71579.b4.CrossRefPubMed
22.
Zurück zum Zitat Balding J, Livingstone WJ, Conroy J, Mynett-Johnson L, Weir DG, Mahmud N, et al: Inflammatory bowel disease: the role of inflammatory cytokine gene polymorphisms. Mediators Inflamm. 2004, 13: 181-187. 10.1080/09511920410001713529.CrossRefPubMedPubMedCentral Balding J, Livingstone WJ, Conroy J, Mynett-Johnson L, Weir DG, Mahmud N, et al: Inflammatory bowel disease: the role of inflammatory cytokine gene polymorphisms. Mediators Inflamm. 2004, 13: 181-187. 10.1080/09511920410001713529.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Stokkers PC, van Aken BE, Basoski N, Reitsma PH, Tytgat GN, van Deventer SJ: Five genetic markers in the interleukin 1 family in relation to inflammatory bowel disease. Gut. 1998, 43: 33-39. 10.1136/gut.43.1.33.CrossRefPubMedPubMedCentral Stokkers PC, van Aken BE, Basoski N, Reitsma PH, Tytgat GN, van Deventer SJ: Five genetic markers in the interleukin 1 family in relation to inflammatory bowel disease. Gut. 1998, 43: 33-39. 10.1136/gut.43.1.33.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Queiroz DM, Oliveira AG, Saraiva IE, Rocha GA, Rocha AM, das Gracas Pimenta SM, et al: Immune response and gene polymorphism profiles in Crohn's disease and ulcerative colitis. Inflamm Bowel Dis. 2009, 15: 353-358. 10.1002/ibd.20757.CrossRefPubMed Queiroz DM, Oliveira AG, Saraiva IE, Rocha GA, Rocha AM, das Gracas Pimenta SM, et al: Immune response and gene polymorphism profiles in Crohn's disease and ulcerative colitis. Inflamm Bowel Dis. 2009, 15: 353-358. 10.1002/ibd.20757.CrossRefPubMed
25.
Zurück zum Zitat Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W: Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993, 75: 263-274. 10.1016/0092-8674(93)80068-P.CrossRefPubMed Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W: Interleukin-10-deficient mice develop chronic enterocolitis. Cell. 1993, 75: 263-274. 10.1016/0092-8674(93)80068-P.CrossRefPubMed
26.
Zurück zum Zitat Ishizuka K, Sugimura K, Homma T, Matsuzawa J, Mochizuki T, Kobayashi M, et al: Influence of interleukin-10 on the interleukin-1 receptor antagonist/interleukin-1 beta ratio in the colonic mucosa of ulcerative colitis. Digestion. 2001, 63 (Suppl 1): 22-7-22-27.PubMed Ishizuka K, Sugimura K, Homma T, Matsuzawa J, Mochizuki T, Kobayashi M, et al: Influence of interleukin-10 on the interleukin-1 receptor antagonist/interleukin-1 beta ratio in the colonic mucosa of ulcerative colitis. Digestion. 2001, 63 (Suppl 1): 22-7-22-27.PubMed
27.
Zurück zum Zitat Correa I, Veny M, Esteller M, Pique JM, Yague J, Panes J, et al: Defective IL-10 production in severe phenotypes of Crohn's disease. J Leukoc Biol. 2009, 85: 896-903. 10.1189/jlb.1108698.CrossRefPubMed Correa I, Veny M, Esteller M, Pique JM, Yague J, Panes J, et al: Defective IL-10 production in severe phenotypes of Crohn's disease. J Leukoc Biol. 2009, 85: 896-903. 10.1189/jlb.1108698.CrossRefPubMed
28.
Zurück zum Zitat Schreiber S, Heinig T, Thiele HG, Raedler A: Immunoregulatory role of interleukin 10 in patients with inflammatory bowel disease. Gastroenterology. 1995, 108: 1434-1444. 10.1016/0016-5085(95)90692-4.CrossRefPubMed Schreiber S, Heinig T, Thiele HG, Raedler A: Immunoregulatory role of interleukin 10 in patients with inflammatory bowel disease. Gastroenterology. 1995, 108: 1434-1444. 10.1016/0016-5085(95)90692-4.CrossRefPubMed
29.
Zurück zum Zitat Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, Mayr G, et al: Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat Genet. 2008, 40: 1319-1323. 10.1038/ng.221.CrossRefPubMed Franke A, Balschun T, Karlsen TH, Sventoraityte J, Nikolaus S, Mayr G, et al: Sequence variants in IL10, ARPC2 and multiple other loci contribute to ulcerative colitis susceptibility. Nat Genet. 2008, 40: 1319-1323. 10.1038/ng.221.CrossRefPubMed
30.
Zurück zum Zitat Amre DK, Mack DR, Morgan K, Israel D, Lambrette P, Costea I, et al: Interleukin 10 (IL-10) gene variants and susceptibility for paediatric onset Crohn's disease. Aliment Pharmacol Ther. 2009, 29: 1025-1031. 10.1111/j.1365-2036.2009.03953.x.CrossRefPubMed Amre DK, Mack DR, Morgan K, Israel D, Lambrette P, Costea I, et al: Interleukin 10 (IL-10) gene variants and susceptibility for paediatric onset Crohn's disease. Aliment Pharmacol Ther. 2009, 29: 1025-1031. 10.1111/j.1365-2036.2009.03953.x.CrossRefPubMed
31.
Zurück zum Zitat Koss K, Satsangi J, Fanning GC, Welsh KI, Jewell DP: 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. 2000, 1: 185-190. 10.1038/sj.gene.6363657.CrossRefPubMed Koss K, Satsangi J, Fanning GC, Welsh KI, Jewell DP: 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. 2000, 1: 185-190. 10.1038/sj.gene.6363657.CrossRefPubMed
32.
Zurück zum Zitat Turner DM, Williams DM, Sankaran D, Lazarus M, Sinnott PJ, Hutchinson IV: An investigation of polymorphism in the interleukin-10 gene promoter. Eur J Immunogenet. 1997, 24: 1-8.CrossRefPubMed Turner DM, Williams DM, Sankaran D, Lazarus M, Sinnott PJ, Hutchinson IV: An investigation of polymorphism in the interleukin-10 gene promoter. Eur J Immunogenet. 1997, 24: 1-8.CrossRefPubMed
33.
Zurück zum Zitat Ouma C, Davenport GC, Were T, Otieno MF, Hittner JB, Vulule JM, et al: Haplotypes of IL-10 promoter variants are associated with susceptibility to severe malarial anemia and functional changes in IL-10 production. Hum Genet. 2008, 124: 515-524. 10.1007/s00439-008-0578-5.CrossRefPubMedPubMedCentral Ouma C, Davenport GC, Were T, Otieno MF, Hittner JB, Vulule JM, et al: Haplotypes of IL-10 promoter variants are associated with susceptibility to severe malarial anemia and functional changes in IL-10 production. Hum Genet. 2008, 124: 515-524. 10.1007/s00439-008-0578-5.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Shin HD, Winkler C, Stephens JC, Bream J, Young H, Goedert JJ, et al: Genetic restriction of HIV-1 pathogenesis to AIDS by promoter alleles of IL10. Proc Natl Acad Sci USA. 2000, 19 (97): 14467-14472. 10.1073/pnas.97.26.14467.CrossRef Shin HD, Winkler C, Stephens JC, Bream J, Young H, Goedert JJ, et al: Genetic restriction of HIV-1 pathogenesis to AIDS by promoter alleles of IL10. Proc Natl Acad Sci USA. 2000, 19 (97): 14467-14472. 10.1073/pnas.97.26.14467.CrossRef
35.
Zurück zum Zitat Fowler EV, Eri R, Hume G, Johnstone S, Pandeya N, Lincoln D, et al: TNFalpha and IL10 SNPs act together to predict disease behaviour in Crohn's disease. J Med Genet. 2005, 42: 523-528. 10.1136/jmg.2004.027425.CrossRefPubMedPubMedCentral Fowler EV, Eri R, Hume G, Johnstone S, Pandeya N, Lincoln D, et al: TNFalpha and IL10 SNPs act together to predict disease behaviour in Crohn's disease. J Med Genet. 2005, 42: 523-528. 10.1136/jmg.2004.027425.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Tedde A, Laura PA, Bagnoli S, Congregati C, Milla M, Sorbi S, et al: Interleukin-10 promoter polymorphisms influence susceptibility to ulcerative colitis in a gender-specific manner. Scand J Gastroenterol. 2008, 43: 712-718. 10.1080/00365520701885507.CrossRefPubMed Tedde A, Laura PA, Bagnoli S, Congregati C, Milla M, Sorbi S, et al: Interleukin-10 promoter polymorphisms influence susceptibility to ulcerative colitis in a gender-specific manner. Scand J Gastroenterol. 2008, 43: 712-718. 10.1080/00365520701885507.CrossRefPubMed
37.
Zurück zum Zitat Tagore A, Gonsalkorale WM, Pravica V, Hajeer AH, McMahon R, Whorwell PJ, et al: Interleukin-10 (IL-10) genotypes in inflammatory bowel disease. Tissue Antigens. 1999, 54: 386-390. 10.1034/j.1399-0039.1999.540408.x.CrossRefPubMed Tagore A, Gonsalkorale WM, Pravica V, Hajeer AH, McMahon R, Whorwell PJ, et al: Interleukin-10 (IL-10) genotypes in inflammatory bowel disease. Tissue Antigens. 1999, 54: 386-390. 10.1034/j.1399-0039.1999.540408.x.CrossRefPubMed
38.
Zurück zum Zitat Fernandez L, Martinez A, Mendoza JL, Urcelay E, Fernandez-Arquero M, Garcia-Paredes J, et al: Interleukin-10 polymorphisms in Spanish patients with IBD. Inflamm Bowel Dis. 2005, 11: 739-743. 10.1097/01.MIB.0000173457.64868.20.CrossRefPubMed Fernandez L, Martinez A, Mendoza JL, Urcelay E, Fernandez-Arquero M, Garcia-Paredes J, et al: Interleukin-10 polymorphisms in Spanish patients with IBD. Inflamm Bowel Dis. 2005, 11: 739-743. 10.1097/01.MIB.0000173457.64868.20.CrossRefPubMed
39.
Zurück zum Zitat Parkes M, Satsangi J, Jewell D: Contribution of the IL-2 and IL-10 genes to inflammatory bowel disease (IBD) susceptibility. Clin Exp Immunol. 1998, 113: 28-32. 10.1046/j.1365-2249.1998.00625.x.CrossRefPubMedPubMedCentral Parkes M, Satsangi J, Jewell D: Contribution of the IL-2 and IL-10 genes to inflammatory bowel disease (IBD) susceptibility. Clin Exp Immunol. 1998, 113: 28-32. 10.1046/j.1365-2249.1998.00625.x.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Klein W, Tromm A, Griga T, Fricke H, Folwaczny C, Hocke M, et al: The IL-10 gene is not involved in the predisposition to inflammatory bowel disease. Electrophoresis. 2000, 21: 3578-3582. 10.1002/1522-2683(200011)21:17<3578::AID-ELPS3578>3.0.CO;2-Z.CrossRefPubMed Klein W, Tromm A, Griga T, Fricke H, Folwaczny C, Hocke M, et al: The IL-10 gene is not involved in the predisposition to inflammatory bowel disease. Electrophoresis. 2000, 21: 3578-3582. 10.1002/1522-2683(200011)21:17<3578::AID-ELPS3578>3.0.CO;2-Z.CrossRefPubMed
41.
Zurück zum Zitat Pae HO, Kim EC, Chung HT: Integrative survival response evoked by heme oxygenase-1 and heme metabolites. J Clin Biochem Nutr. 2008, 42: 197-203. 10.3164/jcbn.2008029.CrossRefPubMedPubMedCentral Pae HO, Kim EC, Chung HT: Integrative survival response evoked by heme oxygenase-1 and heme metabolites. J Clin Biochem Nutr. 2008, 42: 197-203. 10.3164/jcbn.2008029.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Lee TS, Chau LY: Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med. 2002, 8: 240-246. 10.1038/nm0302-240.CrossRefPubMed Lee TS, Chau LY: Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med. 2002, 8: 240-246. 10.1038/nm0302-240.CrossRefPubMed
43.
Zurück zum Zitat Berberat PO, Rahim YI, Yamashita K, Warny MM, Csizmadia E, Robson SC, et al: Heme oxygenase-1-generated biliverdin ameliorates experimental murine colitis. Inflamm Bowel Dis. 2005, 11: 350-359. 10.1097/01.MIB.0000164017.06538.8a.CrossRefPubMed Berberat PO, Rahim YI, Yamashita K, Warny MM, Csizmadia E, Robson SC, et al: Heme oxygenase-1-generated biliverdin ameliorates experimental murine colitis. Inflamm Bowel Dis. 2005, 11: 350-359. 10.1097/01.MIB.0000164017.06538.8a.CrossRefPubMed
44.
Zurück zum Zitat Paul G, Bataille F, Obermeier F, Bock J, Klebl F, Strauch U, et al: Analysis of intestinal haem-oxygenase-1 (HO-1) in clinical and experimental colitis. Clin Exp Immunol. 2005, 140: 547-555. 10.1111/j.1365-2249.2005.02775.x.CrossRefPubMedPubMedCentral Paul G, Bataille F, Obermeier F, Bock J, Klebl F, Strauch U, et al: Analysis of intestinal haem-oxygenase-1 (HO-1) in clinical and experimental colitis. Clin Exp Immunol. 2005, 140: 547-555. 10.1111/j.1365-2249.2005.02775.x.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Wang WP, Guo X, Koo MW, Wong BC, Lam SK, Ye YN, et al: Protective role of heme oxygenase-1 on trinitrobenzene sulfonic acid-induced colitis in rats. Am J Physiol Gastrointest Liver Physiol. 2001, 281: G586-G594.PubMed Wang WP, Guo X, Koo MW, Wong BC, Lam SK, Ye YN, et al: Protective role of heme oxygenase-1 on trinitrobenzene sulfonic acid-induced colitis in rats. Am J Physiol Gastrointest Liver Physiol. 2001, 281: G586-G594.PubMed
46.
Zurück zum Zitat Hegazi RA, Rao KN, Mayle A, Sepulveda AR, Otterbein LE, Plevy SE: Carbon monoxide ameliorates chronic murine colitis through a heme oxygenase 1-dependent pathway. J Exp Med. 2005, 19 (202): 1703-1713. 10.1084/jem.20051047.CrossRef Hegazi RA, Rao KN, Mayle A, Sepulveda AR, Otterbein LE, Plevy SE: Carbon monoxide ameliorates chronic murine colitis through a heme oxygenase 1-dependent pathway. J Exp Med. 2005, 19 (202): 1703-1713. 10.1084/jem.20051047.CrossRef
47.
Zurück zum Zitat Takagi T, Naito Y, Mizushima K, Nukigi Y, Okada H, Suzuki T, et al: Increased intestinal expression of heme oxygenase-1 and its localization in patients with ulcerative colitis. J Gastroenterol Hepatol. 2008, 23 (Suppl 2): S229-S233. 10.1111/j.1440-1746.2008.05443.x.CrossRefPubMed Takagi T, Naito Y, Mizushima K, Nukigi Y, Okada H, Suzuki T, et al: Increased intestinal expression of heme oxygenase-1 and its localization in patients with ulcerative colitis. J Gastroenterol Hepatol. 2008, 23 (Suppl 2): S229-S233. 10.1111/j.1440-1746.2008.05443.x.CrossRefPubMed
48.
Zurück zum Zitat Ono K, Goto Y, Takagi S, Baba S, Tago N, Nonogi H, et al: A promoter variant of the heme oxygenase-1 gene may reduce the incidence of ischemic heart disease in Japanese. Atherosclerosis. 2004, 173: 315-319. 10.1016/j.atherosclerosis.2003.11.021.CrossRefPubMed Ono K, Goto Y, Takagi S, Baba S, Tago N, Nonogi H, et al: A promoter variant of the heme oxygenase-1 gene may reduce the incidence of ischemic heart disease in Japanese. Atherosclerosis. 2004, 173: 315-319. 10.1016/j.atherosclerosis.2003.11.021.CrossRefPubMed
49.
Zurück zum Zitat Hausmann M, Paul G, Kellermeier S, Frey I, Scholmerich J, Falk W, et al: (GT)N dinucleotide repeat polymorphism of haem oxygenase-1 promotor region is not associated with inflammatory bowel disease risk or disease course. Clin Exp Immunol. 2008, 153: 81-85. 10.1111/j.1365-2249.2008.03674.x.CrossRefPubMedPubMedCentral Hausmann M, Paul G, Kellermeier S, Frey I, Scholmerich J, Falk W, et al: (GT)N dinucleotide repeat polymorphism of haem oxygenase-1 promotor region is not associated with inflammatory bowel disease risk or disease course. Clin Exp Immunol. 2008, 153: 81-85. 10.1111/j.1365-2249.2008.03674.x.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Mahid SS, Minor KS, Soto RE, Hornung CA, Galandiuk S: Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc. 2006, 81: 1462-1471. 10.4065/81.11.1462.CrossRefPubMed Mahid SS, Minor KS, Soto RE, Hornung CA, Galandiuk S: Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc. 2006, 81: 1462-1471. 10.4065/81.11.1462.CrossRefPubMed
51.
Zurück zum Zitat Kikuchi H, Itoh J, Fukuda S: Chronic nicotine stimulation modulates the immune response of mucosal T cells to Th1-dominant pattern via nAChR by upregulation of Th1-specific transcriptional factor. Neurosci Lett. 2008, 432: 217-221. 10.1016/j.neulet.2007.12.027.CrossRefPubMed Kikuchi H, Itoh J, Fukuda S: Chronic nicotine stimulation modulates the immune response of mucosal T cells to Th1-dominant pattern via nAChR by upregulation of Th1-specific transcriptional factor. Neurosci Lett. 2008, 432: 217-221. 10.1016/j.neulet.2007.12.027.CrossRefPubMed
52.
Zurück zum Zitat Podolsky DK: Inflammatory bowel disease. N Engl J Med. 2002, 347: 417-429. 10.1056/NEJMra020831.CrossRefPubMed Podolsky DK: Inflammatory bowel disease. N Engl J Med. 2002, 347: 417-429. 10.1056/NEJMra020831.CrossRefPubMed
53.
Zurück zum Zitat Satsangi J, Silverberg MS, Vermeire S, Colombel JF: The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006, 55: 749-753. 10.1136/gut.2005.082909.CrossRefPubMedPubMedCentral Satsangi J, Silverberg MS, Vermeire S, Colombel JF: The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006, 55: 749-753. 10.1136/gut.2005.082909.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Macarthur M, Sharp L, Hold GL, Little J, El-Omar EM: The role of cytokine gene polymorphisms in colorectal cancer and their interaction with aspirin use in the northeast of Scotland. Cancer Epidemiol Biomarkers Prev. 2005, 14: 1613-1618. 10.1158/1055-9965.EPI-04-0878.CrossRefPubMed Macarthur M, Sharp L, Hold GL, Little J, El-Omar EM: The role of cytokine gene polymorphisms in colorectal cancer and their interaction with aspirin use in the northeast of Scotland. Cancer Epidemiol Biomarkers Prev. 2005, 14: 1613-1618. 10.1158/1055-9965.EPI-04-0878.CrossRefPubMed
55.
Zurück zum Zitat Vogel U, Christensen J, Wallin H, Friis S, Nexo BA, Tjonneland A: Polymorphisms in COX-2, NSAID use and risk of basal cell carcinoma in a prospective study of Danes. Mutat Res. 2007, 617: 138-146.CrossRefPubMed Vogel U, Christensen J, Wallin H, Friis S, Nexo BA, Tjonneland A: Polymorphisms in COX-2, NSAID use and risk of basal cell carcinoma in a prospective study of Danes. Mutat Res. 2007, 617: 138-146.CrossRefPubMed
56.
Zurück zum Zitat Greenland S: Dose-response and trend analysis in epidemiology: alternatives to categorical analysis. Epidemiology. 1995, 6: 356-365. 10.1097/00001648-199507000-00005.CrossRefPubMed Greenland S: Dose-response and trend analysis in epidemiology: alternatives to categorical analysis. Epidemiology. 1995, 6: 356-365. 10.1097/00001648-199507000-00005.CrossRefPubMed
58.
Zurück zum Zitat Tedde A, Laura PA, Bagnoli S, Congregati C, Milla M, Sorbi S, et al: Interleukin-10 promoter polymorphisms influence susceptibility to ulcerative colitis in a gender-specific manner. Scand J Gastroenterol. 2008, 43: 712-718. 10.1080/00365520701885507.CrossRefPubMed Tedde A, Laura PA, Bagnoli S, Congregati C, Milla M, Sorbi S, et al: Interleukin-10 promoter polymorphisms influence susceptibility to ulcerative colitis in a gender-specific manner. Scand J Gastroenterol. 2008, 43: 712-718. 10.1080/00365520701885507.CrossRefPubMed
59.
Zurück zum Zitat El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, et al: Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000, 404: 398-402. 10.1038/35006081.CrossRefPubMed El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, et al: Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000, 404: 398-402. 10.1038/35006081.CrossRefPubMed
60.
Zurück zum Zitat Vogel U, Christensen J, Wallin H, Friis S, Nexo BA, Raaschou-Nielsen O, et al: Polymorphisms in genes involved in the inflammatory response and interaction with NSAID use or smoking in relation to lung cancer risk in a prospective study. Mutat Res. 2008, 639: 89-100.CrossRefPubMed Vogel U, Christensen J, Wallin H, Friis S, Nexo BA, Raaschou-Nielsen O, et al: Polymorphisms in genes involved in the inflammatory response and interaction with NSAID use or smoking in relation to lung cancer risk in a prospective study. Mutat Res. 2008, 639: 89-100.CrossRefPubMed
61.
Zurück zum Zitat Birrenbach T, Bocker U: Inflammatory bowel disease and smoking: a review of epidemiology, pathophysiology, and therapeutic implications. Inflamm Bowel Dis. 2004, 10: 848-859. 10.1097/00054725-200411000-00019.CrossRefPubMed Birrenbach T, Bocker U: Inflammatory bowel disease and smoking: a review of epidemiology, pathophysiology, and therapeutic implications. Inflamm Bowel Dis. 2004, 10: 848-859. 10.1097/00054725-200411000-00019.CrossRefPubMed
62.
Zurück zum Zitat Aldhous MC, Prescott RJ, Roberts S, Samuel K, Waterfall M, Satsangi J: Does nicotine influence cytokine profile and subsequent cell cycling/apoptotic responses in inflammatory bowel disease?. Inflamm Bowel Dis. 2008, 14: 1469-1482. 10.1002/ibd.20523.CrossRefPubMed Aldhous MC, Prescott RJ, Roberts S, Samuel K, Waterfall M, Satsangi J: Does nicotine influence cytokine profile and subsequent cell cycling/apoptotic responses in inflammatory bowel disease?. Inflamm Bowel Dis. 2008, 14: 1469-1482. 10.1002/ibd.20523.CrossRefPubMed
63.
Zurück zum Zitat Karban A, Eliakim R: Effect of smoking on inflammatory bowel disease: Is it disease or organ specific?. World J Gastroenterol. 2007, 13: 2150-2152.CrossRefPubMedPubMedCentral Karban A, Eliakim R: Effect of smoking on inflammatory bowel disease: Is it disease or organ specific?. World J Gastroenterol. 2007, 13: 2150-2152.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Fowler EV, Doecke J, Simms LA, Zhao ZZ, Webb PM, Hayward NK, et al: ATG16L1 T300A shows strong associations with disease subgroups in a large Australian IBD population: further support for significant disease heterogeneity. Am J Gastroenterol. 2008, 103: 2519-2526. 10.1111/j.1572-0241.2008.02023.x.CrossRefPubMed Fowler EV, Doecke J, Simms LA, Zhao ZZ, Webb PM, Hayward NK, et al: ATG16L1 T300A shows strong associations with disease subgroups in a large Australian IBD population: further support for significant disease heterogeneity. Am J Gastroenterol. 2008, 103: 2519-2526. 10.1111/j.1572-0241.2008.02023.x.CrossRefPubMed
65.
Zurück zum Zitat Achkar JP, Dassopoulos T, Silverberg MS, Tuvlin JA, Duerr RH, Brant SR, et al: Phenotype-stratified genetic linkage study demonstrates that IBD2 is an extensive ulcerative colitis locus. Am J Gastroenterol. 2006, 101: 572-580. 10.1111/j.1572-0241.2006.00451.x.CrossRefPubMed Achkar JP, Dassopoulos T, Silverberg MS, Tuvlin JA, Duerr RH, Brant SR, et al: Phenotype-stratified genetic linkage study demonstrates that IBD2 is an extensive ulcerative colitis locus. Am J Gastroenterol. 2006, 101: 572-580. 10.1111/j.1572-0241.2006.00451.x.CrossRefPubMed
Metadaten
Titel
The polymorphism rs3024505 proximal to IL-10 is associated with risk of ulcerative colitis and Crohns disease in a Danish case-control study
verfasst von
Vibeke Andersen
Anja Ernst
Jane Christensen
Mette Østergaard
Bent A Jacobsen
Anne Tjønneland
Henrik B Krarup
Ulla Vogel
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
BMC Medical Genetics / Ausgabe 1/2010
Elektronische ISSN: 1471-2350
DOI
https://doi.org/10.1186/1471-2350-11-82

Weitere Artikel der Ausgabe 1/2010

BMC Medical Genetics 1/2010 Zur Ausgabe