Skip to main content
Erschienen in: BMC Gastroenterology 1/2017

Open Access 01.12.2017 | Research article

Genetic variation and expression levels of tight junction genes identifies association between MAGI3 and inflammatory bowel disease

verfasst von: Elisabeth Norén, Sven Almer, Jan Söderman

Erschienen in: BMC Gastroenterology | Ausgabe 1/2017

Abstract

Background

Inflammatory bowel disease (IBD) is associated with increased intestinal permeability, which involves paracellular passage regulated through tight junctions (TJ). The aim of the study was to investigate single nucleotide polymorphisms (SNP) located in genes encoding interacting TJ proteins and corresponding expressions, in relation to IBD.

Methods

Allelic associations between TJ-related genes (F11R, MAGI1, MAGI2, MAGI3, PARD3, PTEN, and TJP1) and IBD, Crohn’s disease (CD), or ulcerative colitis (UC) were investigated. PTPN22 was included since it’s located in the same genetic region as MAGI3. Gene expression levels were investigated in relation to genotype, inflammatory status, phenotype, and medical treatment.

Results

The two strongest allelic associations were observed between IBD and SNPs in MAGI2 (rs6962966) and MAGI3 (rs1343126). Another MAGI3 SNP marker (rs6689879) contributed to increased ileal MAGI3 expression level in non-IBD controls. Furthermore, association between inflammation and decreased expression levels of MAGI3, PTEN, and TJP1 in colonic IBD as well as UC mucosa, and between inflammation and increased expression of PTPN22 in colonic IBD mucosa, was observed.

Conclusions

Our findings lend support to a genetic basis for modulation of intestinal epithelial barrier in IBD, and we have identified MAGI3 as a new candidate gene for IBD.

Background

Inflammatory bowel disease (IBD), including Crohn’s disease (CD) and ulcerative colitis (UC), are complex diseases thought to result from loss of homeostasis between the intestinal microbial milieu, the immune system, and a genetic predisposition [1]. Genetic association in IBD has been the focus of much research [2, 3] leading to the recent identification of a large number of susceptibility loci [35]. Still, the hitherto identified genetic associations only account for a small fraction of the heritability of CD and UC [3].
IBD has been linked to increased paracellular permeability [6, 7]. The intestinal permeability is further increased in both CD patients and their relatives, indicating underlying hereditary factors [8]. However, it remains controversial whether this affected permeability is primary, caused by genetic factors, and/or secondary to inflammation or environmental factors. The tight junction (TJ) structure is critical for the permeability properties of the intestine [9] and several studies provide support for a genetic basis in increased permeability [10].
Wapenaar et al. [11] have described an association between celiac disease and single nucleotide polymorphism (SNP) markers in MAGI2 and PARD3. This MAGI2 marker was also associated with UC [11]. Additionally, McGovern et al. [12] identified a link between genetic variation in MAGI2 and both CD and UC.
The aim of this study was to investigate relations between IBD and MAGI2 and PARD3, as well as other TJ genes (F11R, MAGI1, MAGI3, PTEN, and TJP1) encoding products interacting with each other (Fig. 1) [1315]. Additionally, PTPN22 was included since it is located in the same genetic region as MAGI3 (Fig. 2) and has previously been described in relation to IBD [5, 16]. To gain a better understanding of the pathogenic mechanism of IBD we further analyzed ileal and colonic gene expression in relation to genotype, inflammatory status, phenotype, and ongoing medical treatment.

Methods

Study subjects

Genetic association was investigated in adult Swedish patients with IBD (138 and 157 patients with CD or UC, respectively) and controls from an anonymized regional DNA bank consisting of randomly selected individuals (n = 423) living in the southeastern part of Sweden was used after verbal permission from Prof. Peter Söderkvist (Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden) (Table 1, subgroup 1).
Table 1
Summary of study participants in subgroup 1
Cohort and diseasea
Number of individuals
Number of women
Control subjects
423
223 (52.7%)
CD cases
138
72 (52.2%)
UC cases
157
70 (44.6%)
CD Crohn’s disease, UC ulcerative colitis
aControl subjects are from an anonymized regional DNA bank consisting of randomly selected individuals living in the southeastern part of Sweden
A second Swedish cohort (Table 2, subgroup 2), from which both DNA and RNA were available, was recruited to follow up the case–control study of subgroup 1. Blood samples and intestinal biopsy specimens were obtained from adult IBD patients and non-IBD controls. Each intestinal biopsy was categorized as inflamed or non-inflamed based on a compound evaluation of endoscopic findings assessed by one experienced endoscopist (S.A.) and routine histopathologic assessment for inflammation. Only biopsies with concordant results were analyzed further. In total the study included biopsies from 52 Swedish IBD patients (42 inflamed biopsies and 55 non-inflamed biopsies), including 21 CD patients (16 inflamed biopsies and 24 non-inflamed biopsies), 29 UC patients (24 inflamed biopsies and 29 non-inflamed biopsies), 2 IBD-type unclassified (IBDU; 2 inflamed biopsies and 2 non-inflamed biopsies), and 33 non-inflamed non-IBD controls (86 biopsies).
Table 2
Summary of study participants in subgroup 2
Disease and subgroupsa
Number of individuals (women)
Number of non-inflamed biopsies
Number of inflamed biopsies
IBD
52 (29)
  
 Ileum
 
24
7
 Colon
 
31
35
CD
21 (13)
  
 Ileum
 
11
7
 Colon
 
13
9
UC
29 (15)
  
 Ileum
 
12
0
 Colon
 
17
24
Non-IBD
33 (20)
  
 Ileum
 
24
0
 Colon
 
62
0
IBD inflammatory bowel disease, CD Crohn’s disease, UC ulcerative colitis, IBDU IBD-type unclassified, non-IBD non-inflamed non-IBD controls
aThe total IBD group included CD (n=21), UC (n=29), and IBDU (n=2) patients

SNP selection for genetic association studies

TJ-related genes (F11R, MAGI1, MAGI2, MAGI3, PARD3, PTEN, and TJP1) encoding products interacting with each other [13, 14] were investigated (Fig. 1). Additionally, PTPN22 was included since it is located in the same genetic region as MAGI3 (Fig. 2) and has previously been described in relation to IBD [5, 16]. All SNP markers are given in Additional file 1: Table S1.
SNP markers (minor allele frequency ≥10%, pair-wise r 2 ≥ 0.8) of F11R, MAGI1, MAGI3, and PTEN were selected using SNPbrowser Software version 4.0 (Applied Biosystems, Foster City, CA). SNP markers for MAGI1 and MAGI3 were limited to exons and exon-intron boundaries, due to the large size of these genes. All SNP markers in the genetic region of MAGI2 [11, 12], MAGI3-PTPN22 [5], PARD3 [11], TJP1 [11], and an additional marker for MAGI1 (rs9880851) [11] were selected from the literature.

Definition of genetic blocks of MAGI3 and PTPN22

Blocks of genetic linkage disequilibrium in the region of MAGI3 and PTPN22 were identified using data from HapMap (available at: www.​hapmap.​org; HapMap Data Rel. 28 Phase II + III, August10) (Fig. 2). The blocks were visualized using Haploview and the algorithm by Gabriel et al. [17] based on 95% confidence bounds on D’ (Haploview version 4.2, available at: www.​broad.​mit.​edu/​haploview).

Genotyping

DNA was isolated from buffy coat or whole blood (EDTA blood) using the MagNA Pure LC DNA Isolation Kit and MagNA Pure extraction robot (Roche, Basel, Switzerland). In patients where no DNA from blood was available, the genotyping was performed using DNA isolated from intestinal biopsies (isolation previously described [18]). Allelic discrimination was carried out using either the TaqMan OpenArray system or real-time PCR, as well as TaqMan SNP genotyping assays (Applied Biosystems, Foster City, CA) (Additional file 1: Table S1).
Allelic discrimination using TaqMan OpenArray Genotyping Plates, TaqMan OpenArray Genotyping Master Mix, GeneAmp PCR System 9700, and OpenArray NT Imager (Applied Biosystems) was in accordance with the manufacturer’s recommendation. Genotype data were analyzed using OpenArray SNP Genotyping Analysis Software version 1.0.3 and TaqMan Genotyper Software v.1.3 (Applied Biosystems).
The allelic discrimination using real-time PCR was performed in a total reaction volume of 10 μL, consisting of TaqMan SNP genotyping assays and TaqMan Genotyping Master Mix using either the 7500 Fast Real-Time PCR System (Applied Biosystems) or the CFX96 Real-time System, C1000 Touch Thermal Cycler (Bio-Rad Laboratories Inc., Hercules, CA). The genotype data was analyzed using 7500 Software version 2.0.6 (Applied Biosystems) or Bio-Rad CFX Manager 3.1 (Bio-Rad Laboratories Inc.).

Gene expression analysis

For genes with a significant genetic association to IBD, CD, or UC (in this study), gene expression was analyzed in relation to genotype, inflammatory status, phenotype, and ongoing medical treatment.
RNA purification was performed as described previously [18]. cDNA was synthesized from 2 μg RNA in a total volume of 40 μL using High Capacity cDNA Reverse Transcription kit with RNase Inhibitor according to the manufacturer’s instructions (Invitrogen, Carlsbad, CA). The cDNA was diluted to 25 ng/μL using 0.1 × TE buffer and stored at −80 °C until analysis.
Gene expression of F11R (Hs00170991_m1), MAGI2 (Hs00202321_m1), MAGI3 (Hs00326365_m1), PTEN (Hs02621230_s1), PTPN22 (Hs01587518_m1), and TJP1 (Hs01551861_m1) was analyzed using TaqMan Gene Expression Assay (Applied Biosystems), TaqMan Universal Mastermix (Applied Biosystems), and a 7500 Fast Real-Time PCR System (Applied Biosystems). Each individual reaction contained 10 ng cDNA in a total reaction volume of 20 μL.
Threshold cycle (CT) values were established (ExpressionSuite Software Version 1.0.3; Applied Biosystems) and normalized to the average of selected reference genes (CASC3 (Hs00201226_m1), UBA52 (Hs03004332_g1), and POP4 (Hs00198357_m1) [18]) generating delta-CT (ΔCT) values. Relative quantification (RQ) values were further established, according to Livak et al. [19], by relating the ΔCT value to the sample with the lowest gene expression for each gene (using Microsoft Office Excel; Microsoft Corporation, Redmond, WA). Group differences in gene expression were calculated as a fold change (fc) based on RQ values.

Statistical analysis

Allelic odds ratios (OR) and p-values, based on chi-squared (χ2) tests, were calculated using JMP Genomics 6.0 (JMP Genomics 6.0; SAS Institute Inc., Cary, NC). Deviation from Hardy-Weinberg equilibrium was analyzed using the exact test implemented in Haploview version 4.2 (available at: http://​www.​broad.​mit.​edu/​haploview). For these statistical tests, p < 0.05 was considered significant.
Genes with significant association to IBD, CD, or UC were further analyzed. Group differences in gene expression were investigated using Kruskal-Wallis ANOVA or Mann–Whitney U test (Statistica 12; StatSoft Inc., Tulsa, OK). Gene expression levels were investigated in relation to genotype using Spearman’s rank correlation test (Statistica version 12.7; StatSoft Inc., Tulsa, OK) and also in relation inflammatory status, phenotype, and ongoing medical treatment using logistic regression (Statistica version 12.7; StatSoft Inc.). The analyses of the group differences of gene expressions as well as the logistic regressions were performed using ΔCT values. Box plots were created using Statistica version 12.7 (StatSoft Inc.). A Bonferroni adjusted p < 0.008 (based on the number of analyzed genes; n = 6) was considered significant for statistical analysis based on gene expression levels.

Ethical considerations

This study was approved by the ethics committee of Linköping University (dnr. M35-07, dnr. 2011/201-31). Written and informed consent were obtained from all study participants.

Results

Genetic association – case–control approach

Of the 64 SNP markers, twelve were excluded due to failed genotyping or absence of Hardy-Weinberg equilibrium (Additional file 1: Table S1). The two strongest associations were observed between IBD overall and SNP markers in MAGI2 (rs6962966; susceptibility allele A; p = 0.004) and MAGI3 (rs1343126; susceptibility allele T; p = 0.004) (Additional file 2: Table S2). These two markers were also associated with CD (p = 0.008 and p = 0.051, respectively) and UC (p = 0.045 and p = 0.011, respectively) individually, even if the association between CD and the MAGI3 SNP marker was borderline significant. Associations were also observed between IBD overall and MAGI3 (rs12119076, p = 0.022), as well as between UC and F11R (rs7546890, p = 0.043), MAGI2 (rs7803276, p = 0.031), MAGI3 (rs6689879, p = 0.043), PTEN (rs1234226, p = 0.046), and TJP1 (rs260526, p = 0.010).

Gene expression

Biopsies from different colonic segments (cecum, ascending colon, transverse colon, descending colon, sigmoid colon, and rectum) were treated as biological replicates since all genes except TJP1 were expressed at equal levels at these locations in non-IBD controls (data not shown). TJP1 showed slightly higher expression in the sigmoid colon compared to ascending colon (p = 0.006; fc = 1.16). The biopsy specimens from ileum and colon were analyzed separately.

Gene expression versus genotype

Carriage of at least one UC susceptibility allele (C) of MAGI3 rs6689879 contributed to increased MAGI3 expression level in ileal non-IBD mucosa (p = 0.002; fc = 1.7) (Table 3). No other SNP markers contributed significantly to the gene expression level of corresponding genes.
Table 3
For genes with a significant genetic association to IBD, CD, or UC, gene expression (ΔCt values) was analyzed in relation to genotype, in intestinal biopsies from the different subgroups, using Spearman’s rank correlation test
  
Non-IBD
Non-inflamed IBD
Inflamed IBD
Gene
Gene SNPa
Ileum
Colon
Ileum
Colon
Ileum
Colon
  
p-valueb
p-valueb
p-valueb
p-valueb
p-valueb
p-valueb
F11R
rs7546890 (T)
0.144 (5, 14, 5)
0.112 (6, 19, 8)
0.167 (5, 13, 6)
0.541 (8, 15, 6)
0.140 (1, 4, 1)
0.347 (7, 16, 5)
MAGI2
rs7803276 (C)
0.009 (10, 8, 6)
0.603 (11, 15, 7)
0.604 (5, 11, 8)
0.029 (8, 14, 7)
0.637 (3, 2, 1)
0.285 (6, 13, 9)
rs6962966 (A)
0.797 (11, 8, 5)
0.657 (14, 11, 8)
0.266 (5, 11, 8)
0.868 (6, 16, 7)
0.954 (3, 1, 2)
0.817 (6, 13, 9)
MAGI3
rs6689879 (C)
0.002 (4, 7, 13)
0.050 (5, 10, 18)
0.497 (1, 8, 15)
0.785 (2, 13, 14)
0.021 (0, 3, 3)
0.355 (1, 13, 14)
rs1343126 (T)
1.000 (0, 14, 10)
0.916 (0, 16, 17)
0.044 (1, 15, 8)
0.737 (2, 14, 13)
0.594 (1, 3, 2)
0.780 (0, 18, 10)
rs12119076 (C)
0.978 (0, 13, 11)
0.777 (0, 15, 18)
0.044 (1, 15, 8)
0.737 (2, 14, 13)
0.594 (1, 3, 2)
0.780 (0, 18, 10)
rs6679677d
0.266 (18, 6, 0)
0.018 (25, 8, 0)
0.477 (22, 2, 0)
0.834 (26, 3, 0)
NAc (6, 0, 0)
0.047 (25, 3, 0)
PTEN
rs1234224 (G)
0.554 (2, 12, 10)
0.458 (3, 16, 14)
0.851 (1, 9, 14)
0.772 (2, 15, 12)
NAc (0, 0, 6)
0.695 (3, 14, 11)
PTPN22
rs6689879 (C)
0.697 (4, 7, 13)
0.509 (5, 10, 18)
0.634 (1, 8, 15)
0.129 (2, 13, 14)
0.854 (0, 3, 3)
0.764 (1, 13, 14)
rs1343126 (T)
0.227 (0, 14, 10)
0.025 (0, 16, 17)
0.870 (1, 15, 8)
0.822 (2, 14, 13)
0.908 (1, 3, 2)
0.963 (0, 18, 10)
rs12119076 (C)
0.555 (0, 13, 11)
0.089 (0, 15, 18)
0.870 (1, 15, 8)
0.822 (2, 14, 13)
0.908 (1, 3, 2)
0.963 (0, 18, 10)
rs6679677d
0.128 (18, 6, 0)
0.183 (25, 8, 0)
0.147 (22, 2, 0)
0.291 (26, 3, 0)
NAc (6, 0, 0)
0.745 (25, 3, 0)
TJP1
rs260526 (G)
0.285 (21, 3, 0)
0.052 (29, 4, 0)
0.582 (21, 3, 0)
0.396 (24, 5, 0)
NAc (6, 0, 0)
0.517 (22, 5, 1)
IBD inflammatory bowel disease, CD Crohn’s disease, UC ulcerative colitis
aThe susceptibility allele is shown in parentheses. bNumber of individuals in each genotype group is shown in parentheses (individuals homozygous for the susceptibility allele, heterozygous individuals, and individuals homozygous for the non-susceptibility allele). cNA; not applicable. dThe susceptibility allele (C) is defined from Jostins et. al [5]

Gene expression versus inflammation

Biopsies from inflamed colonic IBD and UC mucosa expressed significant lower levels of MAGI3, PTEN, and TJP1, compared to non-inflamed colonic mucosa from the same groups of patients (Table 4). There was no overlap between the expression levels of PTEN in inflamed colonic mucosa of CD patients compared to non-inflamed CD mucosa (Fig. 3). Furthermore, biopsies from inflamed colonic IBD mucosa expressed a significantly higher level of PTPN22, compared to non-inflamed colonic IBD mucosa.
Table 4
For genes with a significant genetic association to IBD, CD, or UC, gene expression (ΔCt values) was analyzed in relation to presence of inflammation, using logistic regression. ΔCt values are inversely related to gene expression values. The estimates represent the natural logarithm of the odds ratio with a negative value corresponding to increased odds for inflammation, while a positive value corresponds to decreased odds
Gene expression
Single logistic regression
p value
Estimate
Estimate (95% CI)
Nagelkerke R2
IBD; colonic biopsies a
F11R
0.139
1.04
-0.34–2.43
0.05
MAGI2
0.115
0.93
-0.22–2.08
0.06
MAGI3
1.53×10-4
3.38
1.63–5.13
0.52
PTEN
6.72×10-5
10.25
5.21–15.29
0.69
PTPN22
0.005
-1.76
-2.98– -0.54
0.22
TJP1
1.56×10-4
4.97
2.39–7.55
0.48
CD; ileal biopsies b
F11R
0.768
0.34
-1.92–2.61
0.01
MAGI2
0.065
-2.48
-5.11–0.15
0.35
MAGI3
0.107
1.97
-0.43–4.36
0.28
PTEN
0.343
3.35
-3.58–10.27
0.08
PTPN22
0.388
0.56
-0.71–1.82
0.06
TJP1
0.230
2.07
-1.84–5.97
0.09
CD; colonic biopsies c
F11R
0.357
1.30
-1.47–4.06
0.06
MAGI2
0.118
1.94
-0.49–4.38
0.21
MAGI3
0.047
3.01
0.03–5.98
0.39
PTEN
e
e
e
e
PTPN22
0.029
-2.82
-5.35– -0.29
0.45
TJP1
0.028
5.13
0.56-9.69
0.50
UC; colonic biopsies d
F11R
0.408
0.70
-0.96–2.35
0.03
MAGI2
0.684
0.30
-1.16–1.77
0.01
MAGI3
0.003
3.39
1.19–5.59
0.55
PTEN
0.002
7.95
2.82–13.08
0.57
PTPN22
0.048
-1.54
-3.06– -0.01
0.18
TJP1
0.003
4.83
1.61-8.05
0.47
IBD inflammatory bowel disease, CD Crohn’s disease, UC ulcerative colitis
annon-inflamed=29 and ninflamed=28. bnnon-inflamed=11 and ninflamed=6. cnnon-inflamed=12 and ninflamed=7. dnnon-inflamed=16 and ninflamed=19. eComplete separation and thereby uncertain data (Mann-Whitney U-test showed decreased gene expression level in inflamed mucosa, compared to non-inflamed mucosa; p=0.0004)

Gene expression versus phenotype

The gene expression levels of analyzed genes were not different between any of the phenotypes (CD, UC, or non-IBD) (Additional file 3: Table S3).

Gene expression versus medication

Expression of investigated genes in non-inflamed and inflamed IBD biopsy specimens was not affected by ongoing medical treatment (thiopurines, aminosalicylates, or glucocorticosteroids) (Additional file 4: Table S4). Too few individuals underwent anti-TNF-\( \alpha \)-antibody treatment for meaningful statistical analysis.

Discussion

IBD has been associated with an increased paracellular permeability of the intestinal epithelium and a dysfunctional barrier is considered important in the pathogenesis of IBD [6, 7]. The TJ structure is critical for the paracellular barrier properties [9]. Although a few IBD susceptibility markers have been linked to barrier related genes, e.g. CLDN2 and MAGI2 [11, 12, 20], the extent to which a dysfunctional barrier is a consequence of genetic factors remains unclear.
The large numbers of susceptibility loci identified in relation to IBD [35] only explain 10.9% and 7.7% of the heritability of CD and UC, respectively [3]. Regional heterogeneity of allele frequencies among different sub-populations has been observed [4, 5, 21], and indicates the importance of identifying associations in homogenous populations to increase study power [21] and to explain the genetic variance in IBD. To further clarify the role of the paracellular barrier in the etiology of IBD, a Swedish population of IBD patients and non-IBD controls was genotyped for several TJ related genes and analyzed for expression in the intestinal mucosa.
One strong association was observed between the T-allele of the MAGI3 SNP marker rs1343126 and IBD, CD, and UC. Further, the C-allele of another MAGI3 SNP marker (rs6689879) was found to be significantly associated with UC. Presence of this marker also resulted in an increased MAGI3 expression in ileal non-IBD mucosa, which might contribute to the restriction of inflammation in UC to the colon. To the best of our knowledge we have for the first time demonstrated an association between MAGI3 and IBD. However, Jostins et al. have previously demonstrated association between this genetic region and CD, but primarily highlighted PTPN22 [5].
In our study, carriage of at least one IBD associated allele at rs1343126 was nominally associated to a decreased colonic expression of PTPN22 (located in the same genetic region as MAGI3) in the non-IBD subgroup. The corresponding relationship was not identified in inflamed or non-inflamed mucosa of IBD patients. In fact, all relationships between gene expression levels and the susceptibility alleles were observed in mucosal biopsies from non-inflamed, non-IBD controls. Even though the expression of several genes were affected in inflamed mucosa, it is unlikely that genotype – gene expression relationships were concealed in the IBD patients by clinical or subclinical inflammation in the non-inflamed biopsies, since these biopsies were obtained from IBD mucosa that were assessed as histopathologically normal. It is, however, possible that the investigated genes were affected by other factors (e.g. microRNA) that persist in non-inflamed IBD-mucosa [2224].
Jostins et al. previously identified the C-allele of rs6679677 (MAGI3-PTPN22) as a susceptibility allele of CD, but a protective allele with respect to UC [5], which was not confirmed by our study. Nevertheless, it is possible that our study missed a possible association due to a comparatively small sample size.
Significant associations were observed between an SNP marker in MAGI2 (rs6962966) and IBD, CD, as well as UC. Wapenaar et al. [11] previously described an association between this marker and UC. Their study revealed, however, an association to the opposite allele (G) compared to our study (A). Presently we have no explanation for this discrepancy.
In our association study we applied a significance level of p < 0.05, and the findings were further followed up by analyzing for a genotype-gene expression correlation in a second independent cohort. However, even if we apply a more stringent p-value, by making a Bonferroni adjustment based on the number of analyzed genes (n = 8; p < 0.006), both MAGI3 (rs1343126) and MAGI2 (rs6962966) would still remain significantly associated to IBD.
One limitation of our study was that MAGI1 and MAGI3 were only investigated for SNP markers of exons and exon-intron-boundaries, due to the large size of these genes, and that the selection of SNP markers of MAGI2, MAGI3-PTPN22, PARD3, and TJP1 were based on reports in the literature.
MAGI3 was expressed to a lower level in inflamed, compared to non-inflamed, colonic mucosa from IBD and UC patients. MAGI3 is involved in suppression of the PI3K/Akt pathway due to interaction with PTEN [25], and also in the Wnt/β-catenin pathway [26, 27]. T cells with a high β-catenin level induce T cell activation and intestinal inflammation [28]. Thus, regulation of these pathways might constitute a mechanism by which decreased MAGI3 expression promotes intestinal inflammation. Furthermore, the PTPN22 gene expression was shown to be increased in inflamed colonic IBD mucosa, compared to non-inflamed mucosa, which corroborates previous results reported by Chen et al. [29]. The PTPN22 gene is expressed at a higher level in immune cells compared to non-immune cells [30], and further PTPN22 negatively regulates T cell receptor signaling [31]. In our study gene expression was determined using biopsies, and therefore we are unable to determine the exact cellular origin of the increased expression in inflamed mucosa. It may, however, be due to increased quantities of PTPN22-expressing immune cells during inflammation.
Decreased PTEN expression was observed in inflamed colonic mucosa of IBD, CD, and UC patients, compared to non-inflamed mucosa. Furthermore, there was no overlap between the PTEN expression levels in inflamed colonic mucosa of CD patients, compared to non-inflamed CD mucosa, suggesting PTEN as a marker for inflammatory response in colonic CD mucosa. PTEN is described to play a role in the regulation of intestinal permeability [32, 33], and is also defined as a tumor suppressor molecule inhibiting inflammatory response, cell migration, and proliferation via PI3K/Akt pathway [34, 35]. An increased level of microRNA-21 has been described in colonic biopsies from UC patients, compared to normal colonic mucosa, and expression of microRNA-21 in the Caco-2 cell line, a colonic epithelial cell model, resulted in an increased permeability [36]. The mechanism of microRNA-21 has been investigated in Caco-2 cells, and may involve an effect on paracellular permeability via the PTEN level and Akt phosphorylation [33]. Moreover, activation of PI3K/Akt pathway has been demonstrated to inhibit the differentiation of T cells towards regulatory T cells [37] and previously a decreased PTEN expression level was observed in intestinal mucosal lymphocytes in CD patients, compared to controls [38].
The decreased TJP1 expression level observed in inflamed IBD and UC mucosa, compared to non-inflamed mucosa, is consistent with a loss of ZO-1 protein (encoded by TJP1) in dextran sulfate sodium induced colitis in mice [39]. Poritz et al. further observed a correlation between loss of ZO-1 and increased paracellular permeability [39].
Gene expression was not affected by ongoing medical treatment (thiopurines, aminosalicylates, or glucocorticosteroids), irrespective of the presence of inflammation. Nevertheless, other studies have shown that infliximab, an anti-TNF-antibody, can impact the expression of genes associated with intestinal permeability [40] and mucosal cytokine expression [41]. Among our patients, too few were treated with infliximab to allow meaningful analysis with this drug.

Conclusions

In conclusion, our findings support a genetic basis for modulation of the intestinal epithelial barrier in IBD, and point to a complex regulation of TJ gene expression through a secondary role of inflammation. By focusing on a Swedish population and a network of interacting TJ components we have identified MAGI3 as a new candidate gene for IBD.

Acknowledgments

We would like to thank laboratory technician Lena Svensson (Department of Clinical and Experimental Medicine, Linköping University, Sweden) for excellent technical assistance and support.

Funding

This work was supported by FORSS, the Medical Research Council of South-Eastern Sweden [grant No 236541-2012 and 235131-2012], Futurum - the Academy for Healthcare, Region Jönköping County [grant No FUTURUM-338631], Bengt Ihre-Fonden [grant No 2012-SLS 254491], and Karolinska Institutets Forskningsfonder [grant No 2014fobi42063]. The funding bodies had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

The datasets during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors’ contributions

All authors conceived of the study; all authors designed the study; EN contributed to all laboratory work; EN contributed to data analysis and drafted the manuscript; SA provided blood samples and patient data; EN performed the statistical analysis; all authors contributed to the context of the manuscript; all authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
This study was approved by the ethics committee of Linköping University (dnr. M35-07, dnr. 2011/201-31). Written and informed consent were obtained from all study participants.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Peloquin JM, Nguyen DD. The microbiota and inflammatory bowel disease: insights from animal models. Anaerobe. 2013;24:102–6.CrossRefPubMed Peloquin JM, Nguyen DD. The microbiota and inflammatory bowel disease: insights from animal models. Anaerobe. 2013;24:102–6.CrossRefPubMed
3.
Zurück zum Zitat Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet. 2016;48(5):150–8.CrossRef Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet. 2016;48(5):150–8.CrossRef
4.
Zurück zum Zitat Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979–86.CrossRefPubMedPubMedCentral Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979–86.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–24.CrossRefPubMedPubMedCentral Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–24.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat John LJ, Fromm M, Schulzke JD. Epithelial barriers in intestinal inflammation. Antioxid Redox Signal. 2011;15(5):1255–70.CrossRefPubMed John LJ, Fromm M, Schulzke JD. Epithelial barriers in intestinal inflammation. Antioxid Redox Signal. 2011;15(5):1255–70.CrossRefPubMed
7.
Zurück zum Zitat Lee SH. Intestinal permeability regulation by tight junction: implication on inflammatory bowel diseases. Intestinal res. 2015;13(1):11–8.CrossRef Lee SH. Intestinal permeability regulation by tight junction: implication on inflammatory bowel diseases. Intestinal res. 2015;13(1):11–8.CrossRef
8.
Zurück zum Zitat Hollander D, Vadheim CM, Brettholz E, Petersen GM, Delahunty T, Rotter JI. Increased intestinal permeability in patients with Crohn’s disease and their relatives. A possible etiologic factor. Ann Intern Med. 1986;105(6):883–5.CrossRefPubMed Hollander D, Vadheim CM, Brettholz E, Petersen GM, Delahunty T, Rotter JI. Increased intestinal permeability in patients with Crohn’s disease and their relatives. A possible etiologic factor. Ann Intern Med. 1986;105(6):883–5.CrossRefPubMed
9.
Zurück zum Zitat Vetrano S, Danese S. The role of JAM-A in inflammatory bowel disease: unrevealing the ties that bind. Ann N Y Acad Sci. 2009;1165:308–13.CrossRefPubMed Vetrano S, Danese S. The role of JAM-A in inflammatory bowel disease: unrevealing the ties that bind. Ann N Y Acad Sci. 2009;1165:308–13.CrossRefPubMed
10.
Zurück zum Zitat Buhner S, Buning C, Genschel J, Kling K, Herrmann D, Dignass A, et al. Genetic basis for increased intestinal permeability in families with Crohn's disease: role of CARD15 3020insC mutation? Gut. 2006;55(3):342–7.CrossRefPubMedPubMedCentral Buhner S, Buning C, Genschel J, Kling K, Herrmann D, Dignass A, et al. Genetic basis for increased intestinal permeability in families with Crohn's disease: role of CARD15 3020insC mutation? Gut. 2006;55(3):342–7.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Wapenaar MC, Monsuur AJ, van Bodegraven AA, Weersma RK, Bevova MR, Linskens RK, et al. Associations with tight junction genes PARD3 and MAGI2 in Dutch patients point to a common barrier defect for coeliac disease and ulcerative colitis. Gut. 2008;57(4):463–7.CrossRefPubMed Wapenaar MC, Monsuur AJ, van Bodegraven AA, Weersma RK, Bevova MR, Linskens RK, et al. Associations with tight junction genes PARD3 and MAGI2 in Dutch patients point to a common barrier defect for coeliac disease and ulcerative colitis. Gut. 2008;57(4):463–7.CrossRefPubMed
12.
Zurück zum Zitat McGovern DP, Taylor KD, Landers C, Derkowski C, Dutridge D, Dubinsky M, et al. MAGI2 genetic variation and inflammatory bowel disease. Inflamm Bowel Dis. 2009;15(1):75–83.CrossRefPubMedPubMedCentral McGovern DP, Taylor KD, Landers C, Derkowski C, Dutridge D, Dubinsky M, et al. MAGI2 genetic variation and inflammatory bowel disease. Inflamm Bowel Dis. 2009;15(1):75–83.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43(Database issue):D447–452.CrossRefPubMed Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43(Database issue):D447–452.CrossRefPubMed
16.
Zurück zum Zitat Rivas MA, Beaudoin M, Gardet A, Stevens C, Sharma Y, Zhang CK, et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet. 2011;43(11):1066–73.CrossRefPubMedPubMedCentral Rivas MA, Beaudoin M, Gardet A, Stevens C, Sharma Y, Zhang CK, et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet. 2011;43(11):1066–73.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, et al. The structure of haplotype blocks in the human genome. Science. 2002;296(5576):2225–9.CrossRefPubMed Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, et al. The structure of haplotype blocks in the human genome. Science. 2002;296(5576):2225–9.CrossRefPubMed
18.
Zurück zum Zitat Söderman J, Berglind L, Almer S. Gene Expression-Genotype Analysis Implicates GSDMA, GSDMB, and LRRC3C as Contributors to Inflammatory Bowel Disease Susceptibility. Biomed Res Int. 2015;2015:834805.CrossRefPubMedPubMedCentral Söderman J, Berglind L, Almer S. Gene Expression-Genotype Analysis Implicates GSDMA, GSDMB, and LRRC3C as Contributors to Inflammatory Bowel Disease Susceptibility. Biomed Res Int. 2015;2015:834805.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 (−Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 (−Delta Delta C(T)) Method. Methods. 2001;25(4):402–8.CrossRefPubMed
20.
Zurück zum Zitat Soderman J, Noren E, Christiansson M, Bragde H, Thiebaut R, Hugot JP, et al. Analysis of single nucleotide polymorphisms in the region of CLDN2-MORC4 in relation to inflammatory bowel disease. World J gastroenterol : WJG. 2013;19(30):4935–43.CrossRefPubMedPubMedCentral Soderman J, Noren E, Christiansson M, Bragde H, Thiebaut R, Hugot JP, et al. Analysis of single nucleotide polymorphisms in the region of CLDN2-MORC4 in relation to inflammatory bowel disease. World J gastroenterol : WJG. 2013;19(30):4935–43.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Tian C, Gregersen PK, Seldin MF. Accounting for ancestry: population substructure and genome-wide association studies. Hum Mol Genet. 2008;17(R2):R143–150.CrossRefPubMedPubMedCentral Tian C, Gregersen PK, Seldin MF. Accounting for ancestry: population substructure and genome-wide association studies. Hum Mol Genet. 2008;17(R2):R143–150.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Fasseu M, Treton X, Guichard C, Pedruzzi E, Cazals-Hatem D, Richard C, et al. Identification of restricted subsets of mature microRNA abnormally expressed in inactive colonic mucosa of patients with inflammatory bowel disease. PLoS one. 2010;5(10):e13160. doi:10.1371/journal.pone.0013160. Fasseu M, Treton X, Guichard C, Pedruzzi E, Cazals-Hatem D, Richard C, et al. Identification of restricted subsets of mature microRNA abnormally expressed in inactive colonic mucosa of patients with inflammatory bowel disease. PLoS one. 2010;5(10):e13160. doi:10.​1371/​journal.​pone.​0013160.
23.
Zurück zum Zitat Planell N, Lozano JJ, Mora-Buch R, Masamunt MC, Jimeno M, Ordas I, et al. Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013;62(7):967–76.CrossRefPubMed Planell N, Lozano JJ, Mora-Buch R, Masamunt MC, Jimeno M, Ordas I, et al. Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013;62(7):967–76.CrossRefPubMed
24.
Zurück zum Zitat Peloquin J, Goel G, Huang H, Haritunians T, Sartor R, Daly M, et al. O-002 Genes in IBD-Associated Risk Loci Demonstrate Genotype-, Tissue-, and Inflammation-Specific Patterns of Expression in Terminal Ileum and Colon Mucosal Tissue. Inflamm Bowel Dis. 2016;22 Suppl 1:S1.CrossRef Peloquin J, Goel G, Huang H, Haritunians T, Sartor R, Daly M, et al. O-002 Genes in IBD-Associated Risk Loci Demonstrate Genotype-, Tissue-, and Inflammation-Specific Patterns of Expression in Terminal Ileum and Colon Mucosal Tissue. Inflamm Bowel Dis. 2016;22 Suppl 1:S1.CrossRef
25.
Zurück zum Zitat Wu Y, Dowbenko D, Spencer S, Laura R, Lee J, Gu Q, et al. Interaction of the tumor suppressor PTEN/MMAC with a PDZ domain of MAGI3, a novel membrane-associated guanylate kinase. J Biol Chem. 2000;275(28):21477–85.CrossRefPubMed Wu Y, Dowbenko D, Spencer S, Laura R, Lee J, Gu Q, et al. Interaction of the tumor suppressor PTEN/MMAC with a PDZ domain of MAGI3, a novel membrane-associated guanylate kinase. J Biol Chem. 2000;275(28):21477–85.CrossRefPubMed
26.
Zurück zum Zitat Ma Q, Zhang Y, Meng R, Xie KM, Xiong Y, Lin S, et al. MAGI3 Suppresses Glioma Cell Proliferation via Upregulation of PTEN Expression. Biomed environ sci : BES. 2015;28(7):502–9.PubMed Ma Q, Zhang Y, Meng R, Xie KM, Xiong Y, Lin S, et al. MAGI3 Suppresses Glioma Cell Proliferation via Upregulation of PTEN Expression. Biomed environ sci : BES. 2015;28(7):502–9.PubMed
27.
Zurück zum Zitat Ma Q, Yang Y, Feng D, Zheng S, Meng R, Fa P, et al. MAGI3 negatively regulates Wnt/beta-catenin signaling and suppresses malignant phenotypes of glioma cells. Oncotarget. 2015;6(34):35851–65.PubMedPubMedCentral Ma Q, Yang Y, Feng D, Zheng S, Meng R, Fa P, et al. MAGI3 negatively regulates Wnt/beta-catenin signaling and suppresses malignant phenotypes of glioma cells. Oncotarget. 2015;6(34):35851–65.PubMedPubMedCentral
28.
Zurück zum Zitat Keerthivasan S, Aghajani K, Dose M, Molinero L, Khan MW, Venkateswaran V, et al. beta-Catenin promotes colitis and colon cancer through imprinting of proinflammatory properties in T cells. Sci Transl Med. 2014;6(225):225–8.CrossRef Keerthivasan S, Aghajani K, Dose M, Molinero L, Khan MW, Venkateswaran V, et al. beta-Catenin promotes colitis and colon cancer through imprinting of proinflammatory properties in T cells. Sci Transl Med. 2014;6(225):225–8.CrossRef
29.
Zurück zum Zitat Chen Z, Zhang H, Xia B, Wang P, Jiang T, Song M, et al. Association of PTPN22 gene (rs2488457) polymorphism with ulcerative colitis and high levels of PTPN22 mRNA in ulcerative colitis. Int J Color Dis. 2013;28(10):1351–8.CrossRef Chen Z, Zhang H, Xia B, Wang P, Jiang T, Song M, et al. Association of PTPN22 gene (rs2488457) polymorphism with ulcerative colitis and high levels of PTPN22 mRNA in ulcerative colitis. Int J Color Dis. 2013;28(10):1351–8.CrossRef
30.
Zurück zum Zitat Arimura Y, Yagi J. Comprehensive expression profiles of genes for protein tyrosine phosphatases in immune cells. Sci Signal. 2010;3(137):rs1.CrossRefPubMed Arimura Y, Yagi J. Comprehensive expression profiles of genes for protein tyrosine phosphatases in immune cells. Sci Signal. 2010;3(137):rs1.CrossRefPubMed
31.
Zurück zum Zitat Sharp RC, Abdulrahim M, Naser ES, Naser SA. Genetic Variations of PTPN2 and PTPN22: Role in the Pathogenesis of Type 1 Diabetes and Crohn’s Disease. Front Cell Infect Microbiol. 2015;5:95.CrossRefPubMedPubMedCentral Sharp RC, Abdulrahim M, Naser ES, Naser SA. Genetic Variations of PTPN2 and PTPN22: Role in the Pathogenesis of Type 1 Diabetes and Crohn’s Disease. Front Cell Infect Microbiol. 2015;5:95.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Langlois MJ, Bergeron S, Bernatchez G, Boudreau F, Saucier C, Perreault N, et al. The PTEN phosphatase controls intestinal epithelial cell polarity and barrier function: role in colorectal cancer progression. PLoS One. 2010;5(12):e15742.CrossRefPubMedPubMedCentral Langlois MJ, Bergeron S, Bernatchez G, Boudreau F, Saucier C, Perreault N, et al. The PTEN phosphatase controls intestinal epithelial cell polarity and barrier function: role in colorectal cancer progression. PLoS One. 2010;5(12):e15742.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zhang L, Shen J, Cheng J, Fan X. MicroRNA-21 regulates intestinal epithelial tight junction permeability. Cell Biochem Funct. 2015;33(4):235–40.CrossRefPubMed Zhang L, Shen J, Cheng J, Fan X. MicroRNA-21 regulates intestinal epithelial tight junction permeability. Cell Biochem Funct. 2015;33(4):235–40.CrossRefPubMed
34.
Zurück zum Zitat Hu Y, Li Z, Guo L, Wang L, Zhang L, Cai X, et al. MAGI-2 Inhibits cell migration and proliferation via PTEN in human hepatocarcinoma cells. Arch Biochem Biophys. 2007;467(1):1–9.CrossRefPubMed Hu Y, Li Z, Guo L, Wang L, Zhang L, Cai X, et al. MAGI-2 Inhibits cell migration and proliferation via PTEN in human hepatocarcinoma cells. Arch Biochem Biophys. 2007;467(1):1–9.CrossRefPubMed
35.
Zurück zum Zitat Tokuhira N, Kitagishi Y, Suzuki M, Minami A, Nakanishi A, Ono Y, et al. PI3K/AKT/PTEN pathway as a target for Crohn’s disease therapy (Review). Int J Mol Med. 2015;35(1):10–6.PubMed Tokuhira N, Kitagishi Y, Suzuki M, Minami A, Nakanishi A, Ono Y, et al. PI3K/AKT/PTEN pathway as a target for Crohn’s disease therapy (Review). Int J Mol Med. 2015;35(1):10–6.PubMed
36.
Zurück zum Zitat Yang Y, Ma Y, Shi C, Chen H, Zhang H, Chen N, et al. Overexpression of miR-21 in patients with ulcerative colitis impairs intestinal epithelial barrier function through targeting the Rho GTPase RhoB. Biochem Biophys Res Commun. 2013;434(4):746–52.CrossRefPubMed Yang Y, Ma Y, Shi C, Chen H, Zhang H, Chen N, et al. Overexpression of miR-21 in patients with ulcerative colitis impairs intestinal epithelial barrier function through targeting the Rho GTPase RhoB. Biochem Biophys Res Commun. 2013;434(4):746–52.CrossRefPubMed
37.
Zurück zum Zitat Sauer S, Bruno L, Hertweck A, Finlay D, Leleu M, Spivakov M, et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci U S A. 2008;105(22):7797–802.CrossRefPubMedPubMedCentral Sauer S, Bruno L, Hertweck A, Finlay D, Leleu M, Spivakov M, et al. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci U S A. 2008;105(22):7797–802.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Long SH, He Y, Chen MH, Cao K, Chen YJ, Chen BL, et al. Activation of PI3K/Akt/mTOR signaling pathway triggered by PTEN downregulation in the pathogenesis of Crohn’s disease. J Dig Dis. 2013;14(12):662–9.CrossRefPubMed Long SH, He Y, Chen MH, Cao K, Chen YJ, Chen BL, et al. Activation of PI3K/Akt/mTOR signaling pathway triggered by PTEN downregulation in the pathogenesis of Crohn’s disease. J Dig Dis. 2013;14(12):662–9.CrossRefPubMed
39.
Zurück zum Zitat Poritz LS, Garver KI, Green C, Fitzpatrick L, Ruggiero F, Koltun WA. Loss of the tight junction protein ZO-1 in dextran sulfate sodium induced colitis. J Surg Res. 2007;140(1):12–9.CrossRefPubMed Poritz LS, Garver KI, Green C, Fitzpatrick L, Ruggiero F, Koltun WA. Loss of the tight junction protein ZO-1 in dextran sulfate sodium induced colitis. J Surg Res. 2007;140(1):12–9.CrossRefPubMed
40.
Zurück zum Zitat Toedter G, Li K, Sague S, Ma K, Marano C, Macoritto M, et al. Genes associated with intestinal permeability in ulcerative colitis: changes in expression following infliximab therapy. Inflamm Bowel Dis. 2012;18(8):1399–410.CrossRefPubMed Toedter G, Li K, Sague S, Ma K, Marano C, Macoritto M, et al. Genes associated with intestinal permeability in ulcerative colitis: changes in expression following infliximab therapy. Inflamm Bowel Dis. 2012;18(8):1399–410.CrossRefPubMed
41.
Zurück zum Zitat Arijs I, De Hertogh G, Machiels K, Van Steen K, Lemaire K, Schraenen A, et al. Mucosal gene expression of cell adhesion molecules, chemokines, and chemokine receptors in patients with inflammatory bowel disease before and after infliximab treatment. Am J Gastroenterol. 2011;106(4):748–61.CrossRefPubMed Arijs I, De Hertogh G, Machiels K, Van Steen K, Lemaire K, Schraenen A, et al. Mucosal gene expression of cell adhesion molecules, chemokines, and chemokine receptors in patients with inflammatory bowel disease before and after infliximab treatment. Am J Gastroenterol. 2011;106(4):748–61.CrossRefPubMed
Metadaten
Titel
Genetic variation and expression levels of tight junction genes identifies association between MAGI3 and inflammatory bowel disease
verfasst von
Elisabeth Norén
Sven Almer
Jan Söderman
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Gastroenterology / Ausgabe 1/2017
Elektronische ISSN: 1471-230X
DOI
https://doi.org/10.1186/s12876-017-0620-y

Weitere Artikel der Ausgabe 1/2017

BMC Gastroenterology 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.