Retrospective Study Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 7, 2017; 23(33): 6137-6146
Published online Sep 7, 2017. doi: 10.3748/wjg.v23.i33.6137
Patients with inflammatory bowel disease have increased risk of autoimmune and inflammatory diseases
Morten L Halling, Torben Knudsen, Department of Gastroenterology and Hepatology, Hospital of Southwest Jutland, 6700 Esbjerg, Denmark
Jens Kjeldsen, Lars Koch Hansen, Department of Medical Gastroenterology S, Odense University Hospital, 5000 Odense, Denmark
Jan Nielsen, Center for Clinical Epidemiology, Odense University Hospital, 5000 Odense, Denmark
Author contributions: Halling ML, Kjeldsen J, Knudsen T and Koch Hansen L designed the study; Koch Hansen L performed data collection; Nielsen J performed statistical analyses; Halling ML and Koch Hansen L drafted the manuscript and obtained funding; all authors revised and accepted the final manuscript.
Institutional review board statement: This study was approved by the Danish Data Protection Agency (approval # 2013-41-1596).
Conflict-of-interest statement: The authors report no conflict of interest.
Data sharing statement: No additional data are available.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Morten L Halling, MD, Department of Gastroenterology and Hepatology, Hospital of Southwest Jutland, Medicinsk Gastroenterologisk Afdeling, Sydvestjysk Sygehus, Finsensgade 35, 6700 Esbjerg, Denmark. mortenhalling@gmail.com
Telephone: +45-79-183141 Fax: +45-79-183147
Received: March 2, 2017
Peer-review started: March 3, 2017
First decision: April 21, 2017
Revised: May 30, 2017
Accepted: July 12, 2017
Article in press: July 12, 2017
Published online: September 7, 2017

Abstract
AIM

To investigate whether immune mediated diseases (IMD) are more frequent in patients with inflammatory bowel disease (IBD).

METHODS

In this population based registry study, a total of 47325 patients with IBD were alive and registered in the Danish National Patient Registry on December 16, 2013. Controls were randomly selected from the Danish Civil Registration System (CRS) and matched for sex, age, and municipality. We used ICD 10 codes to identify the diagnoses of the included patients. The IBD population was divided into three subgroups: Ulcerative colitis (UC), Crohn’s disease (CD) and Both the latter referring to those registered with both diagnoses. Subsequently, odds-ratios (OR) and 95%CI were obtained separately for each group and their respective controls. The use of Bonferoni post-test correction adjusted the significance level to P < 0.00125. P-values were estimated using Fisher’s exact test.

RESULTS

There were significantly more women than men in the registry, and a greater percentage of comorbidity in the IBD groups (P < 0.05). Twenty different IMDs were all significantly more frequent in the IBD group. Sixteen were associated with UC versus twelve with CD. In both UC and CD ORs were significantly increased (P < 0.00125) for primary sclerosing cholangitis (PSC), celiac disease, type 1 diabetes (T1D), sarcoidosis, asthma, iridocyclitis, psoriasis, pyoderma gangrenosum, rheumatoid arthritis, and ankylosing spondylitis. Restricted to UC (P < 0.00125) were autoimmune hepatitis, primary biliary cholangitis, Grave’s disease, polymyalgia rheumatica, temporal arteritis , and atrophic gastritis. Restricted to CD (P < 0.00125) were psoriatic arthritis and episcleritis. Restricted to women with UC (P < 0.00125) were atrophic gastritis, rheumatoid arthritis, temporal arteritis, and polymyalgia rheumatica. Restricted to women with CD were episcleritis, rheumatoid arthritis, and psoriatic arthritis. The only disease restricted to men (P < 0.00125) was sarcoidosis.

CONCLUSION

Immune mediated diseases were significantly more frequent in patients with IBD. Our results strengthen the hypothesis that some IMDs and IBD may have overlapping pathogenic pathways.

Key Words: Immune mediated diseases, Ulcerative colitis, Risk, Prevalence, Registry, Chronic inflammatory diseases, Autoimmune diseases, Inflammatory bowel disease, Crohn’s disease, Extraintestinal manifestations

Core tip: Essential to inflammatory bowel disease (IBD) pathogenesis are environmental factors, altered gut microbiota and genetic susceptibility. The latter causing impairment of barrier function, autophagy, and Th1, 2 and 17 cell responses. Interestingly, these mechanisms are also thought important in other immune mediated diseases, as is the overlap of susceptibility genes. Besides the classic extraintestinal manifestations, we found a variety of immune mediated diseases to be more frequent in individuals with IBD. Physicians should be aware of this when treating these patients. Furthermore, these findings support the hypothesis that immune mediated diseases may have overlapping pathogeneses. Thus, understanding IBD might help us understand other immune mediated diseases and vice versa.



INTRODUCTION

Crohn’s disease (CD) and ulcerative colitis (UC) are two distinct types of chronic inflammatory bowel diseases. The insight into etiology factors and the complex pathogenetic process is not yet fully understood. The diseases are often diagnosed in young individuals and recent studies report increasing incidences of both UC and CD, not only in Denmark but globally[1]. It has been suggested that inflammatory bowel disease (IBD) may be due to an inappropriate inflammatory response to the intestinal flora in genetically susceptible individuals. So far, several susceptibility genes have been identified[2]. Many of these are also found in other immune mediated diseases (IMDs), indicating overlaps between pathogenic pathways. The identified risk genes in IBD are involved in maintaining normal microbial gut homeostasis and adequate immune response[3,4]. Mutations in these may impair mechanisms essential to innate and adaptive immune response, i.e. weakened mucosal barrier, a decrease of antibacterial agents, impaired autophagy and antigen recognition. Mutations may also cause an imbalance of pro- and anti-inflammatory cytokines related to the regulation of Th1, 2 and 17 in particular[5]. CD is considered Th1 mediated thus characterized by interferon gamma, tumor necrosis factor alpha, and IL 12. UC is associated with a Th2 response where IL 4, 5, 10 and 13 are dominant. The Th17 response is present in both CD and UC but most pronounced in CD. It is characterized by IL 17 and 23 production. Th17 can also produce interferon gamma like Th1[5-8]. It is suggested that disturbances in these mechanisms may cause a loss of self-tolerance leading towards chronic inflammation or autoimmunity[9-12].

The gut microbiota of patients with IBD has been shown to contain less diversity, a reduced number of bacteria, and an altered microbial metabolite profile compared to healthy individuals[13]. Environmental factors, i.e. medication (antibiotics, non-steroid anti-inflammatory drugs and hormones), diet, geography, and previous infections might influence this[4]. A similar etiology is believed to exist in other IMDs, i.e. rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, type 1 diabetes (T1D), and celiac disease[14,15].

It has become clear, that patients with an existing IMD are more likely to develop other IMDs, this is more evident in females than in males[16]. Apart from the extraintestinal manifestations of IBD, little is known about the association between IBD and other IMDs.

Only a few large population based studies on the subject exist. The results of these suggest that IBD is associated with asthma, rheumatoid arthritis, psoriasis, multiple sclerosis, autoimmune thyroiditis, T1D, and vasculitis[17-22]. Different study designs, varying validity of diagnoses, population sizes and confounders, i.e. ethnicity, economic and social status, all make the findings of these studies difficult to interpret.

In Denmark healthcare is free and all contacts to hospitals are registered on an individual basis based on a civil registration number together with diagnosis and procedural codes. This allows a unique access to information not confounded by economic and social status.

The aim of this study was to examine if IMDs are more frequent among patients with CD and UC compared to the background population.

MATERIALS AND METHODS

This was a cross-sectional study including all living patients with IBD who were matched with a control group to compare the point-prevalence of specific IMDs.

Identification of patients and controls

The Danish National Patient Registry include all contacts within the healthcare system both in-hospital, since 1977, and in outpatient settings since 1994. Data were retrieved on December 16, 2013 and included all patients alive registered with a diagnosis compatible with CD and UC. Patients were identified using the ICD 10 codes: CD K50.0-K50.9; UC, K51.0-K51.9). ICD 10 codes including “other” or “unspecified” were excluded to avoid inclusion of non-specific diseases and incorrect diagnosis codes.

The Danish Civil Registration System (CRS) includes all Danish inhabitants and each person has a unique 10-digit identification number. The CRS includes demographic data e.g. name, sex, date of birth, and death[23]. All IBD patients were paired (2:1) with random controls identified in the CRS and matched by sex, age (± 1 year) and municipality. Demographic data presented are based on data from the CRS.

The selected forty IMDs are all considered to be of either autoimmune or inflammatory origin. The same criteria were used for the IMDs. ICD 10 codes for the IMDs are listed in the Supplementary Table 1.

Table 1 Participants’ demographic data.
VariablesIBDControlUCControlCDControlBoth1Control
n47325928393106660951133432617229165716
Female54%55%53%53%58%58%56%56%
Male46%45%47%47%42%42%44%44%
Mean age at entry, yr5353555549494747
Mean age at onset of IBD, yr42-44-37-34-
Mean duration of IBD at entry, yr10-9-10-11-
Comorbidity 0277%83%76.50%82.00%77%85%82%87%
Comorbidity 1-218%13.50%18.00%14%18%12%15%10%
Comorbidity ≥ 35%3.50%5.50%4%5%3%3%3%

To assess comorbidity we used the Charlson comorbidity index which has been developed to estimate 1-year mortality in cancer patients. It is also useful in research to identify possible confounding diseases. It includes a number of systemic diseases associated with increased mortality, i.e. organ failure, AIDS, and cancer[24].

Ethics

This study was approved by the Danish Data Protection Agency (approval # 2013-41-1596). Approval from the Ethics Committee was not needed as this is a registry study.

Statistical analysis

The occurrence of IMDs was obtained separately for each group. Then OR and 95%CI were calculated. Fisher’s exact test was used to calculate P-values.

We used the Bonferroni post-test correction to reduce the likelihood of false positives. We did 40 comparisons (the 40 IMDs investigated) and adjusted the significance level accordingly to P < 0.00125. Calculations was made using STATA version 13.0 (StataCorp LP, TX, United States).

RESULTS

A total of 47325 patients were alive and registered with IBD on December 16, 2013. A total of 92839 controls were identified.

CD was registered in 13343 patients, UC in 31066, and 2916 were registered with both diagnoses. A total of 92839 controls were found for the IBD group, 26172 for CD, 60951 for UC and 5716 for those with both diagnoses. Due to the matching criteria, five IBD patients had only one or no controls.

There was an excess of women in all IBD groups, most pronounced in CD (P < 0.05). The mean age at onset of disease was significantly higher in UC. Comorbidity was most frequent in those with either UC or CD (P < 0.05). See Table 1.

Twenty out of forty IMDs had significantly increased ORs in the IBD groups compared to their controls (P < 0.00125). Sixteen IMDs were associated with UC and twelve with CD. See Tables 2 and 3.

Table 2 Number of immune mediated diseases.
DiseaseIBDControlCDControlUCControlBoth1Control
Primary sclerosing cholangitis25743511922301
Pyoderma gangrenosum1938601977360
Autoimmune hepatitis1243515119622132
Celiac disease280921333013258154
Ankylosing spondylitis431151189322011024117
Churg Strauss syndrome145418420
Primary biliary cholangitis7132116532571
Episcleritis5633259232183
Iridocyclitis419295148822301884125
Atrophic gastritis60471611423422
Psoriasis378345148992002293017
Polyarteritis nodosa4238159242732
Rheumatoid arthritis4464011191102503113225
Type 1 diabetes168214643594311002118010371
Sarcoidosis14112229387994149
Asthma11409813373635686957682
Giant cell arteritis193156374611614136
Psoriatic arthritis31624981931472062117
Grave's disease8175811412073945614649
Polymyalgia rheumatica46832072122242324622
Table 3 Odds-ratios for immune mediated diseases, in patients with inflammatory bowel disease.
DiseaseIBD95%CIUC95%CICD95%CIBoth195%CI
Primary sclerosing cholangitis126.7a47.2-340.3189.5a47.0-763.468.8a9.4-502.659.4a8.1-436.2
Pyoderma gangrenosum47.5a23.4-96.427.3a12.7-58.7118.2a16.4-853.336/0a2
Autoimmune hepatitis7.0a4.8-10.18.6a5.4-13.62.7b1.2-5.812.8a2.9-56.8
Celiac disease6.0a4.7-7.64.5a3.3-6.18.8a5.9-13.07.4a2.4-22.3
Ankylosing spondylitis5.6a4.7-6.83.9a3.1-4.911.7a8.1-17.14.8a2.7-8.4
Churg Strauss syndrome5.5a2.0-15.33.9b1.2-13.0-c-c
Primary biliary cholangitis4.4a2.9-6.64.2a2.6-6.73.6b1.3-9.713.8b1.7-111.9
Episcleritis3.3a2.2-5.12.1b1.2-3.95.5a2.5-11.75.2b1.4-19.8
Iridocyclitis2.8a2.4-3.32.4a2.0-2.93.6a2.7-4.73.2a2.0-5.4
Atrophic gastritis2.5a1.7-3.72.4a1.5-3.82.9b1.3-6.2-c
Psoriasis2.2a1.9-2.51.7a1.4-2.13.0a2.3-3.83.5a1.9-6.5
Polyarteritis nodosa2.2a1.4-3.41.7b1.0-3.03.3b1.4-7.5-c
Rheumatoid arthritis1.8a1.5-2.01.6a1.3-1.92.1a1.6-2.82.5a1.5-4.2
Type 1 diabetes1.7a1.6-1.91.7a1.6-1.81.7a1.4-1.92.9a2.2-3.9
Sarcoidosis1.7a1.3-2.21.7a1.2-2.2-c3.1b1.9-4.8
Asthma1.7a1.6-1.91.6a1.4-1.81.8a1.6-2.11.8a1.3-2.5
Giant cell arteritis1.6a1.3-2.01.6a1.3-2.11.6b1.0-2.4-c
Psoriatic arthritis1.5a1.3-1.81.4b1.1-1.71.7a1.3-2.32.4b1.3-4.6
Grave's disease1.4a1.3-1.61.4a1.2-1.61.3b1.1-1.71.9b1.2-2.8
Polymyalgia rheumatica1.3a1.2-1.51.5a1.2-1.7-c-c

Seven of the IMDs were considered rheumatologic diseases, included ankylosing spondylitis, rheumatoid arthritis, psoriatic arthritis, polymyalgia rheumatic, temporal arteritis, polyarteritis nodosa, and Churg Strauss Syndrome.

Five IMDs were gastrointestinal including celiac disease, atrophic gastritis, primary sclerosing cholangitis, primary biliary cholangitis, and autoimmune hepatitis.

The remaining IMDs were T1D, Grave’s disease, pyoderma gangrenosum, psoriasis, iridocyclitis, episcleritis, sarcoidosis, and asthma.

There was a trend towards significance (P = 0.00125-0.05) for Wegener’s granulomatosis, chorioretinitis, vitiligo, lichen ruber planus, scleroderma, and multiple sclerosis.

Seven IMDs were only significant in women. While only one was restricted to men. See Table 4.

Table 4 Odds-ratios for immune mediated diseases restricted to either gender.
DiseaseFemales95%CIMales95%CI
IBD
Episcleritis3.6a2.1-6.12.9b1.4-6.1
Atrophic gastritis3.5a2.1-5.9-c
Polyarteritis nodosa2.6a1.5-4.5-c
Rheumatoid arthritis1.9a1.6-2.21.4b1.1-1.9
Giant cell arteritis1.7a1.3-2.2-c
Psoriatic arthritis1.6a1.3-2.01.4b1.1-1.9
Polymyalgia rheumatica1.5a1.3-1.8-c
Sarcoidosis1.5b1.1-2.21.9a1.3-2.6
UC
Atrophic gastritis3.1a1.7-5.8-c
Rheumatoid arthritis1.7a1.4-2.1
Giant cell arteritis1.7a1.3-2.3-c
Polymyalgia rheumatica1.6a1.3-2.0-c
CD
Episclerit5.9a2.4-15.04.5b1.2-17.5
Rheumatoid arthritis2.3a1.7-3.0-c
Psoriatic arthritis2.0a1.3-2.8-c
Sarcoidosis-c3.2a1.6-6.6
Both1
Iridocyklitis3.6a1.9-6.82.71.2-6.2
Celiac disease6.0a1.9-18.63/0b2
Autoimmune hepatitis17.9a2.3-141.57.8b0.9-69.8

In general, the same pattern is seen in those registered with both CD and UC.

We did not observe any OR below one, neither did we record any cases of Sjögren’s syndrome, inclusion body myositis, eosinophilic esophagitis, or autoimmune adrenalitis.

DISCUSSION

In this study, we documented an increased frequency of twenty IMDs in patients with IBD compared to matched cohorts.

Although most of the IMDs are considered to be Th1 mediated, UC was associated with more IMDs than CD. The presence of Th17 cells in UC and their ability to induce a Th1 response might explain this. Another explanation might be that certain susceptibility genes can act differently depending on the setting[25]. A gene might increase the risk of one disease while reducing the risk of others[25-27].

Extraintestinal manifestations

Ankylosing spondylitis, pyoderma gangrenosum, psoriasis, iridocyclitis, episcleritis, and primary sclerosing cholangitis (PSC) are all well described in IBD[28]. Thus the significant associations were expected. Except from PSC, these will not be discussed further.

Primary sclerosing cholangitis and gastrointestinal immune mediated diseases

PSC is predominant in men and most frequent in UC[29]. We found PSC to be associated with both types of IBD and both genders. Most striking is the association with CD which is less often described. Studies suggest that PSC is more frequent when colon is affected and a distinct subtype, PSC-IBD has been suggested[30-33]. This study does not include data on localization, severity or extension. Several PSC risk genes are shared with IBD and other IMDs[33,34]. Gene mutations influencing IL 10 signaling are identified in CD, UC and PSC. The absence of IL 10 can cause severe CD due to lack of Th1 and macrophage inhibition[33-35]. Interestingly, hepatobiliary inflammation is thought to be induced by microbial metabolites and changes in the microbiota and this inflammation is linked to the FUT2 gene, which is also found in CD[33,34,36].

In contrast to most other studies[18,36,37], we found celiac disease to be more frequent in those with IBD regardless of type, as did another Danish study[16]. Other studies found IBD to be more common in patients with celiac disease but not vice versa[38-40]. Similarities and differences in pathogeneses might explain these conflicting results. Celiac disease is like IBD an inflammatory disorder of the intestine, often diagnosed in young individuals, more common in women, and Th1 mediated. Changes in microbiota and dysfunctional IL 18 receptor are also noted in both conditions[27,41]. Risk genes of celiac disease shared with CD relates to adaptive immunity while those shared with UC primarily relates to barrier function. Different from IBD is the absence of Th17 response, impaired autophagy and while important in celiac disease, IL 15 is not that important in IBD[41].

We found autoimmune hepatitis, primary biliary cholangitis, and atrophic gastritis to be more common in UC only. Again, results from previous studies conflict[16,18,21,42-45]. Little is known about the association with atrophic gastritis, which to our knowledge is unique to this study. Th1, 2 and 17 responses are important in IBD, PSC and primary biliary cholangitis pathogenesis. Primary biliary cholangitis and IBD have overlapping susceptibility genes, which is not the case with autoimmune hepatitis[46,47]. The pathogenesis of primary biliary cholangitis resembles those of autoimmune hepatitis and CD, dysfunctionalities in IL 12 signaling promotes a Th1 and possibly also a Th17 response, causing a granulomatous inflammation[47,48].

Endocrine diseases

UC is reported to occur more frequently in family members of patients with T1D[18,19,49]. However, three studies did not find any association[16,20,21]. This study found T1D associated with both UC and CD. Confounding due to treatment with corticosteroids is unlikely, as the mechanisms in steroid induced diabetes resemble those in type 2 diabetes[50,51]. Levels of IL 18 are elevated in CD and T1D, but not in UC. IL 18 causes a Th1 response and is likely to affect mucosal barrier function too[27,52]. PTPN2 is one of many shared risk genes[2,53]. It promotes beta cell apoptosis in T1D while causing intestinal barrier dysfunction, impaired autophagocytosis, and inhibition of Th17 in IBD[25,54]. Changes in the gut microbiota are also suggested to trigger T1D[27].

Data on autoimmune thyroiditis and IBD is sparse, similarities to IBD limited and only few risk genes overlap[55-57]. Restricted to UC only, we found OR significantly increased for Grave’s disease. None was detected for Hashimoto’s thyroiditis. Similar results are reported in two other studies[18,58]. One study reports hypothyroidism more common in CD[19]. In addition, three other studies did not find any association at all[16,17,21].

Rheumatic diseases

Rheumatoid arthritis was associated with both UC and CD while psoriatic arthritis was restricted to CD. Previously published data support this[18,20,21,59]. The microbiome of the gut and skin are possible triggers in rheumatoid arthritis and psoriatic arthritis[60]. Both types of arthritis share characteristics with CD in particular. Th1 and 17 are essential in all three pathogeneses[2,61-64].

ORs for polymyalgia rheumatica and temporal arteritis were significantly increased in the IBD and UC group, not in CD. This is supported by one study while refuted by another[16,18]. Overlapping susceptibility genes suggest that Th1, Th17 and regulatory T cells are of importance to the pathogeneses[65].

ORs for Churg Strauss Syndrome and polyarteritis nodosa were significantly increased in the overall IBD group but not in the subgroups. The low number of cases calls for careful interpretation and future studies.

Other disorders

In this study, asthma was more common in both UC and CD. Both UC and allergic asthma are considered Th2 mediated. Also, a Th17 response is described in severe asthma[66]. Risk genes are associated with IL 13 and 17 production, dysfunctional regulatory T cells and regulation of Th1, 2 and 17 responses[26,66]. Studies have not found that asthma reduces the risk of IBD[67,68], rather the opposite seems more likely[17,18,20,21].

The association of sarcoidosis and IBD were restricted to UC and males with CD. Another study confirms the linkage to UC[18]. There is not much documentation for this association. The inflammation in sarcoidosis is similar to CD; granulomatous; Th1 and 17 driven; and mutations in NOD2 and IL 23 receptor gene are identified[2,69-71].

There were no cases of Sjögren’s syndrome, inclusion body myositis, eosinophilic esophagitis, or autoimmune adrenalitis. This is unexpected. Some case reports have described the coexistence of Sjögren and primary adrenocortical insufficiency in IBD patients[16,72-74]. One case report describes eosinophilic esophagitis and CD[75]. While to our knowledge, no association between inclusion myositis and IBD has been reported. Although specific ICD 10 codes were used misclassification is still possible e.g. autoimmune adrenalitis might be registered as Addison’s disease.

Strengths and limitations

The strength of this study is that it includes all patients alive with CD or UC in Denmark. The Danish population is homogenous regarding ethnicity and religion. Health care is free to all residents; thus, NPR is not biased by inclusion of specific hospitals, age groups, insurance policies, social, or financial status. As the general practitioners do not provide data, diseases not requiring hospital treatment could be underrepresented i.e. asthma, Grave’s disease, Hashimoto’s thyroiditis, and atrophic gastritis[76].

A limitation of the study is possible bias caused by varying validity of the ICD 10 codes. Only few Danish studies have addressed this issue. The average positive predictive value (PPV) of an ICD 10 diagnosis for any medical condition in the NPR varies from 65.5 % to 81%[76].

However, the completeness is 94% for both UC and CD while the PPV of UC and CD is 90% and 97% respectively[77].

The validity of T1D is like that of IBD, very high[24,78]. The PPV of asthma among hospitalized children is 85% while 65% among adults. However, a sensitivity analysis did not find the PPV in adults, low enough to nullify the hypothesis[79,80]. As a collective group the PPV of connective tissue diseases is reported as high[24]. The PPV of rheumatoid arthritis is low[81].

Despite varying validity of ICD 10 codes, most of our findings are in alignment with those of the studies using algorithms to increase the validity. Important to this study, is the occurrence of the classic extraintestinal manifestations which indicates that our results are not too biased.

Detection bias is another concern. Patients seen on regular basis by a physician such as those with IBD are more likely to be diagnosed.

To eliminate confounders like sex, age and geography in the IBD group, we used these as matching criteria. Information regarding smoking status was not available to us, thus no correction was made.

Another confounder is drug induced autoimmunity. A wide variety of drugs are suggested to induce autoimmunity. Among these are antibiotics, statins, methotrexate, thiopurines, and biological agents (anti-TNF-α agents)[82-88]. Biological agents, which are often used to treat IBD, ankylosing spondylitis, psoriasis, and rheumatoid arthritis, are paradoxically suggested to induce IMDs. No correction was made since we do not have data regarding patients’ use of prescribed drugs.

While Bonferroni post-test correction reduced the risk of false positives, the risk of false negatives simultaneously increased. Knowing this, a low number of false positives were still preferred in this study.

In conclusion, our study emphasizes that immune mediated diseases are more frequent among patients with CD or UC. Our results strengthen the thesis of partially overlapping pathogeneses among some immune mediated diseases including IBD and emphasized the complexity of IBD pathogenesis. Our most important findings are the increased risk of celiac disease and T1D in both UC and CD, but also the increased risk of primary sclerosing cholangitis in CD although not being limited to CD. Finally, when treating patients with UC or CD one should be aware of the strong association with other immune mediated diseases.

COMMENTS
Background

Extraintestinal manifestations in Crohn’s disease (CD) and ulcerative colitis (UC) are well described. The authors aimed to investigate whether other immune mediated diseases were associated with inflammatory bowel disease (IBD).

Research frontiers

Most studies on the subject are small or case reports. Only few larger studies have been conducted. The authors aimed to estimate odds-ratios of developing an immune mediated diseases (IMD) in patients with IBD compared individuals without IBD.

Innovations and breakthroughs

This is one of few larger studies on the subject. It includes all patients alive with CD or UC in Denmark. Due to free health care to all residents the study is unbiased by inclusion of specific hospitals, age groups, insurance policies, social or financial status. The authors found several IMDs not considered classic extraintestinal manifestations to be significantly associated with IBD.

Applications

Physicians treating patients with IBD should obe aware of the increased risk of developing other IMDs than the classic extraintestinal manifestations. The findings support the hypothesis that shared pathogenic pathways among IMDs could exist.

Peer-review

It’s a well-written and interesting manuscript.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: Denmark

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Triantafillidis JK S- Editor: Gong ZM L- Editor: A E- Editor: Huang Y

References
1.  Nørgård BM, Nielsen J, Fonager K, Kjeldsen J, Jacobsen BA, Qvist N. The incidence of ulcerative colitis (1995-2011) and Crohn’s disease (1995-2012) - based on nationwide Danish registry data. J Crohns Colitis. 2014;8:1274-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
2.  Lees CW, Barrett JC, Parkes M, Satsangi J. New IBD genetics: common pathways with other diseases. Gut. 2011;60:1739-1753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 420]  [Article Influence: 32.3]  [Reference Citation Analysis (1)]
3.  Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev. 2014;13:3-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 484]  [Cited by in F6Publishing: 595]  [Article Influence: 54.1]  [Reference Citation Analysis (0)]
4.  Sheehan D, Moran C, Shanahan F. The microbiota in inflammatory bowel disease. J Gastroenterol. 2015;50:495-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 174]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
5.  Xu XR, Liu CQ, Feng BS, Liu ZJ. Dysregulation of mucosal immune response in pathogenesis of inflammatory bowel disease. World J Gastroenterol. 2014;20:3255-3264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 150]  [Cited by in F6Publishing: 156]  [Article Influence: 15.6]  [Reference Citation Analysis (1)]
6.  Zhu J. T helper 2 (Th2) cell differentiation, type 2 innate lymphoid cell (ILC2) development and regulation of interleukin-4 (IL-4) and IL-13 production. Cytokine. 2015;75:14-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 260]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
7.  Cătană CS, Berindan Neagoe I, Cozma V, Magdaş C, Tăbăran F, Dumitraşcu DL. Contribution of the IL-17/IL-23 axis to the pathogenesis of inflammatory bowel disease. World J Gastroenterol. 2015;21:5823-5830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 126]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
8.  Lyakh L, Trinchieri G, Provezza L, Carra G, Gerosa F. Regulation of interleukin-12/interleukin-23 production and the T-helper 17 response in humans. Immunol Rev. 2008;226:112-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 163]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
9.  Alexander KL, Targan SR, Elson CO 3rd. Microbiota activation and regulation of innate and adaptive immunity. Immunol Rev. 2014;260:206-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 101]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
10.  Larmonier CB, Shehab KW, Ghishan FK, Kiela PR. T Lymphocyte Dynamics in Inflammatory Bowel Diseases: Role of the Microbiome. Biomed Res Int. 2015;2015:504638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
11.  Wallace KL, Zheng LB, Kanazawa Y, Shih DQ. Immunopathology of inflammatory bowel disease. World J Gastroenterol. 2014;20:6-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 356]  [Cited by in F6Publishing: 350]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
12.  El-Khider F, McDonald C. Links of Autophagy Dysfunction to Inflammatory Bowel Disease Onset. Dig Dis. 2016;34:27-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
13.  Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology. 2014;146:1489-1499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1145]  [Cited by in F6Publishing: 1131]  [Article Influence: 113.1]  [Reference Citation Analysis (0)]
14.  Vieira SM, Pagovich OE, Kriegel MA. Diet, microbiota and autoimmune diseases. Lupus. 2014;23:518-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 113]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
15.  Tlaskalová-Hogenová H, Stěpánková R, Kozáková H, Hudcovic T, Vannucci L, Tučková L, Rossmann P, Hrnčíř T, Kverka M, Zákostelská Z. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free and gnotobiotic animal models of human diseases. Cell Mol Immunol. 2011;8:110-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 473]  [Article Influence: 36.4]  [Reference Citation Analysis (0)]
16.  Eaton WW, Rose NR, Kalaydjian A, Pedersen MG, Mortensen PB. Epidemiology of autoimmune diseases in Denmark. J Autoimmun. 2007;29:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 293]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
17.  Bernstein CN, Wajda A, Blanchard JF. The clustering of other chronic inflammatory diseases in inflammatory bowel disease: a population-based study. Gastroenterology. 2005;129:827-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 386]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
18.  Hemminki K, Li X, Sundquist K, Sundquist J. Familial association of inflammatory bowel diseases with other autoimmune and related diseases. Am J Gastroenterol. 2010;105:139-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
19.  Kappelman MD, Galanko JA, Porter CQ, Sandler RS. Association of paediatric inflammatory bowel disease with other immune-mediated diseases. Arch Dis Child. 2011;96:1042-1046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
20.  Cohen R, Robinson D Jr, Paramore C, Fraeman K, Renahan K, Bala M. Autoimmune disease concomitance among inflammatory bowel disease patients in the United States, 2001-2002. Inflamm Bowel Dis. 2008;14:738-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 88]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
21.  Weng X, Liu L, Barcellos LF, Allison JE, Herrinton LJ. Clustering of inflammatory bowel disease with immune mediated diseases among members of a northern california-managed care organization. Am J Gastroenterol. 2007;102:1429-1435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 102]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
22.  Wilson JC, Furlano RI, Jick SS, Meier CR. Inflammatory Bowel Disease and the Risk of Autoimmune Diseases. J Crohns Colitis. 2016;10:186-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 45]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
23.  Pedersen CB, Gøtzsche H, Møller JO, Mortensen PB. The Danish Civil Registration System. A cohort of eight million persons. Dan Med Bull. 2006;53:441-449.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Thygesen SK, Christiansen CF, Christensen S, Lash TL, Sørensen HT. The predictive value of ICD-10 diagnostic coding used to assess Charlson comorbidity index conditions in the population-based Danish National Registry of Patients. BMC Med Res Methodol. 2011;11:83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 733]  [Cited by in F6Publishing: 910]  [Article Influence: 70.0]  [Reference Citation Analysis (0)]
25.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
26.  Li X, Ampleford EJ, Howard TD, Moore WC, Torgerson DG, Li H, Busse WW, Castro M, Erzurum SC, Israel E. Genome-wide association studies of asthma indicate opposite immunopathogenesis direction from autoimmune diseases. J Allergy Clin Immunol. 2012;130:861-8.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 114]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
27.  Gjymishka A, Coman RM, Brusko TM, Glover SC. Influence of host immunoregulatory genes, ER stress and gut microbiota on the shared pathogenesis of inflammatory bowel disease and Type 1 diabetes. Immunotherapy. 2013;5:1357-1366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
28.  Levine JS, Burakoff R. Extraintestinal manifestations of inflammatory bowel disease. Gastroenterol Hepatol (NY). 2011;7:235-241.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Boonstra K, Beuers U, Ponsioen CY. Epidemiology of primary sclerosing cholangitis and primary biliary cirrhosis: a systematic review. J Hepatol. 2012;56:1181-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 392]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
30.  Rönnblom A, Holmström T, Tanghöj H, Rorsman F, Sjöberg D. Appearance of hepatobiliary diseases in a population-based cohort with inflammatory bowel diseases (Inflammatory Bowel Disease Cohort of the Uppsala Region). J Gastroenterol Hepatol. 2015;30:1288-1292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
31.  de Vries AB, Janse M, Blokzijl H, Weersma RK. Distinctive inflammatory bowel disease phenotype in primary sclerosing cholangitis. World J Gastroenterol. 2015;21:1956-1971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 120]  [Cited by in F6Publishing: 117]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
32.  Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology. 2011;140:1704-1712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 293]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
33.  Karlsen TH, Boberg KM. Update on primary sclerosing cholangitis. J Hepatol. 2013;59:571-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
34.  Eksteen B. Advances and controversies in the pathogenesis and management of primary sclerosing cholangitis. Br Med Bull. 2014;110:89-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
35.  Marlow GJ, van Gent D, Ferguson LR. Why interleukin-10 supplementation does not work in Crohn’s disease patients. World J Gastroenterol. 2013;19:3931-3941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 106]  [Cited by in F6Publishing: 98]  [Article Influence: 8.9]  [Reference Citation Analysis (2)]
36.  Jandaghi E, Hojatnia M, Vahedi H, Shahbaz-Khani B, Kolahdoozan S, Ansari R. Is the Prevalence of Celiac Disease Higher than the General Population in Inflammatory Bowel Diseaese? Middle East J Dig Dis. 2015;7:82-87.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Casella G, D’Incà R, Oliva L, Daperno M, Saladino V, Zoli G, Annese V, Fries W, Cortellezzi C; Italian Group - IBD. Prevalence of celiac disease in inflammatory bowel diseases: An IG-IBD multicentre study. Dig Liver Dis. 2010;42:175-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 49]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
38.  Leeds JS, Höroldt BS, Sidhu R, Hopper AD, Robinson K, Toulson B, Dixon L, Lobo AJ, McAlindon ME, Hurlstone DP. Is there an association between coeliac disease and inflammatory bowel diseases? A study of relative prevalence in comparison with population controls. Scand J Gastroenterol. 2007;42:1214-1220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 64]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
39.  Yang A, Chen Y, Scherl E, Neugut AI, Bhagat G, Green PH. Inflammatory bowel disease in patients with celiac disease. Inflamm Bowel Dis. 2005;11:528-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 62]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
40.  Kocsis D, Tóth Z, Csontos ÁA, Miheller P, Pák P, Herszényi L, Tóth M, Tulassay Z, Juhász M. Prevalence of inflammatory bowel disease among coeliac disease patients in a Hungarian coeliac centre. BMC Gastroenterol. 2015;15:141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
41.  Pascual V, Dieli-Crimi R, López-Palacios N, Bodas A, Medrano LM, Núñez C. Inflammatory bowel disease and celiac disease: overlaps and differences. World J Gastroenterol. 2014;20:4846-4856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 63]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
42.  Wong GW, Heneghan MA. Association of Extrahepatic Manifestations with Autoimmune Hepatitis. Dig Dis. 2015;33 Suppl 2:25-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
43.  Xiao WB, Liu YL. Primary biliary cirrhosis and ulcerative colitis: a case report and review of literature. World J Gastroenterol. 2003;9:878-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 34]  [Cited by in F6Publishing: 27]  [Article Influence: 1.3]  [Reference Citation Analysis (1)]
44.  Gizard E, Ford AC, Bronowicki JP, Peyrin-Biroulet L. Systematic review: The epidemiology of the hepatobiliary manifestations in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2014;40:3-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
45.  Veloso FT, Carvalho J, Magro F. Immune-related systemic manifestations of inflammatory bowel disease. A prospective study of 792 patients. J Clin Gastroenterol. 1996;23:29-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 229]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
46.  Manns MP, Lohse AW, Vergani D. Autoimmune hepatitis--Update 2015. J Hepatol. 2015;62:S100-S111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 226]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
47.  Gulamhusein AF, Juran BD, Lazaridis KN. Genome-Wide Association Studies in Primary Biliary Cirrhosis. Semin Liver Dis. 2015;35:392-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
48.  Shi T, Zhang T, Zhang L, Yang Y, Zhang H, Zhang F. The Distribution and the Fibrotic Role of Elevated Inflammatory Th17 Cells in Patients With Primary Biliary Cirrhosis. Medicine (Baltimore). 2015;94:e1888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
49.  Hemminki K, Li X, Sundquist J, Sundquist K. Familial association between type 1 diabetes and other autoimmune and related diseases. Diabetologia. 2009;52:1820-1828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
50.  Fathallah N, Slim R, Larif S, Hmouda H, Ben Salem C. Drug-Induced Hyperglycaemia and Diabetes. Drug Saf. 2015;38:1153-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 88]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
51.  van Raalte DH, Diamant M. Steroid diabetes: from mechanism to treatment? Neth J Med. 2014;72:62-72.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Nowarski R, Jackson R, Gagliani N, de Zoete MR, Palm NW, Bailis W, Low JS, Harman CC, Graham M, Elinav E. Epithelial IL-18 Equilibrium Controls Barrier Function in Colitis. Cell. 2015;163:1444-1456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 313]  [Cited by in F6Publishing: 374]  [Article Influence: 46.8]  [Reference Citation Analysis (0)]
53.  Morran MP, Vonberg A, Khadra A, Pietropaolo M. Immunogenetics of type 1 diabetes mellitus. Mol Aspects Med. 2015;42:42-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 81]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
54.  Spalinger MR, McCole DF, Rogler G, Scharl M. Protein tyrosine phosphatase non-receptor type 2 and inflammatory bowel disease. World J Gastroenterol. 2016;22:1034-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
55.  Kristensen B. Regulatory B and T cell responses in patients with autoimmune thyroid disease and healthy controls. Dan Med J. 2016;63.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Tomer Y, Dolan LM, Kahaly G, Divers J, D’Agostino RB Jr, Imperatore G, Dabelea D, Marcovina S, Black MH, Pihoker C, Hasham A, Hammerstad SS, Greenberg DA, Lotay V, Zhang W, Monti MC, Matheis N; SEARCH for Diabetes in Youth Study. Genome wide identification of new genes and pathways in patients with both autoimmune thyroiditis and type 1 diabetes. J Autoimmun. 2015;60:32-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
57.  Shizuma T. Concomitant Thyroid Disorders and Inflammatory Bowel Disease: A Literature Review. Biomed Res Int. 2016;2016:5187061.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Hemminki K, Li X, Sundquist J, Sundquist K. The epidemiology of Graves’ disease: evidence of a genetic and an environmental contribution. J Autoimmun. 2010;34:J307-J313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
59.  Li WQ, Han JL, Chan AT, Qureshi AA. Psoriasis, psoriatic arthritis and increased risk of incident Crohn’s disease in US women. Ann Rheum Dis. 2013;72:1200-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 140]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
60.  Castelino M, Eyre S, Upton M, Ho P, Barton A. The bacterial skin microbiome in psoriatic arthritis, an unexplored link in pathogenesis: challenges and opportunities offered by recent technological advances. Rheumatology (Oxford). 2014;53:777-784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
61.  Mellado M, Martínez-Muñoz L, Cascio G, Lucas P, Pablos JL, Rodríguez-Frade JM. T Cell Migration in Rheumatoid Arthritis. Front Immunol. 2015;6:384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 178]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
62.  Yamamoto K, Okada Y, Suzuki A, Kochi Y. Genetic studies of rheumatoid arthritis. Proc Jpn Acad Ser B Phys Biol Sci. 2015;91:410-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
63.  Stuart PE, Nair RP, Tsoi LC, Tejasvi T, Das S, Kang HM, Ellinghaus E, Chandran V, Callis-Duffin K, Ike R. Genome-wide Association Analysis of Psoriatic Arthritis and Cutaneous Psoriasis Reveals Differences in Their Genetic Architecture. Am J Hum Genet. 2015;97:816-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 203]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
64.  de Vlam K, Gottlieb AB, Mease PJ. Current concepts in psoriatic arthritis: pathogenesis and management. Acta Derm Venereol. 2014;94:627-634.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
65.  Carmona FD, Mackie SL, Martín JE, Taylor JC, Vaglio A, Eyre S, Bossini-Castillo L, Castañeda S, Cid MC, Hernández-Rodríguez J. A large-scale genetic analysis reveals a strong contribution of the HLA class II region to giant cell arteritis susceptibility. Am J Hum Genet. 2015;96:565-580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 114]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
66.  Cosmi L, Liotta F, Maggi E, Romagnani S, Annunziato F. Th17 cells: new players in asthma pathogenesis. Allergy. 2011;66:989-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 240]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
67.  Fenta YA, Tello N, Jung JA, Urm SH, Loftus EV Jr, Yawn BP, Li X, Juhn YJ. Inflammatory bowel disease and asthma: a population-based, case-control study. Inflamm Bowel Dis. 2010;16:1957-1962.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
68.  Yun HD, Knoebel E, Fenta Y, Gabriel SE, Leibson CL, Loftus EV Jr, Roger V, Yawn BP, Li B, Juhn YJ. Asthma and proinflammatory conditions: a population-based retrospective matched cohort study. Mayo Clin Proc. 2012;87:953-960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 64]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
69.  Caso F, Galozzi P, Costa L, Sfriso P, Cantarini L, Punzi L. Autoinflammatory granulomatous diseases: from Blau syndrome and early-onset sarcoidosis to NOD2-mediated disease and Crohn’s disease. RMD Open. 2015;1:e000097.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
70.  Fischer A, Nothnagel M, Franke A, Jacobs G, Saadati HR, Gaede KI, Rosenstiel P, Schürmann M, Müller-Quernheim J, Schreiber S. Association of inflammatory bowel disease risk loci with sarcoidosis, and its acute and chronic subphenotypes. Eur Respir J. 2011;37:610-616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
71.  Kiszałkiewicz J, Piotrowski WJ, Brzeziańska-Lasota E. Selected molecular events in the pathogenesis of sarcoidosis - recent advances. Pneumonol Alergol Pol. 2015;83:462-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
72.  Qiu Y, Mao R, Chen MH. A De Novo Arisen Case of Primary Adrenal Insufficiency in an Adolescent Patient With Crohn Disease: A Case report. Medicine (Baltimore). 2015;94:e818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
73.  Govindarajan R, Galpin OP. Coexistence of Addison’s disease, ulcerative colitis, hypothyroidism and pernicious anemia. J Clin Gastroenterol. 1992;15:82-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
74.  Triantafillidis JK, Roussou P, Manousos ON, Dadioti P, Nicolakis D. Ulcerative colitis and Sjogren’s syndrome in the same patient: report of two cases and a review of the literature. Ital J Gastroenterol. 1994;26:299-302.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Mulder DJ, Hookey LC, Hurlbut DJ, Justinich CJ. Impact of Crohn disease on eosinophilic esophagitis: evidence for an altered T(H)1-T(H)2 immune response. J Pediatr Gastroenterol Nutr. 2011;53:213-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
76.  Schmidt M, Schmidt SA, Sandegaard JL, Ehrenstein V, Pedersen L, Sørensen HT. The Danish National Patient Registry: a review of content, data quality, and research potential. Clin Epidemiol. 2015;7:449-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2012]  [Cited by in F6Publishing: 2850]  [Article Influence: 316.7]  [Reference Citation Analysis (0)]
77.  Fonager K, Sørensen HT, Rasmussen SN, Møller-Petersen J, Vyberg M. Assessment of the diagnoses of Crohn’s disease and ulcerative colitis in a Danish hospital information system. Scand J Gastroenterol. 1996;31:154-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 191]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
78.  Kristensen JK, Drivsholm TB, Carstensen B, Steding-Jensen M, Green A. [Validation of methods to identify known diabetes on the basis of health registers]. Ugeskr Laeger. 2007;169:1687-1692.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Jensen AØ, Nielsen GL, Ehrenstein V. Validity of asthma diagnoses in the Danish National Registry of Patients, including an assessment of impact of misclassification on risk estimates in an actual dataset. Clin Epidemiol. 2010;2:67-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
80.  Moth G, Vedsted P, Schiøtz PO. National registry diagnoses agree with medical records on hospitalized asthmatic children. Acta Paediatr. 2007;96:1470-1473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
81.  Pedersen M, Klarlund M, Jacobsen S, Svendsen AJ, Frisch M. Validity of rheumatoid arthritis diagnoses in the Danish National Patient Registry. Eur J Epidemiol. 2004;19:1097-1103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 67]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
82.  Perez-Alvarez R, Pérez-de-Lis M, Ramos-Casals M; BIOGEAS study group. Biologics-induced autoimmune diseases. Curr Opin Rheumatol. 2013;25:56-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
83.  Castiella A, Zapata E, Lucena MI, Andrade RJ. Drug-induced autoimmune liver disease: A diagnostic dilemma of an increasingly reported disease. World J Hepatol. 2014;6:160-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 78]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
84.  Bukhari M. Drug-induced rheumatic diseases: a review of published case reports from the last two years. Curr Opin Rheumatol. 2012;24:182-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
85.  Moran GW, Lim AW, Bailey JL, Dubeau MF, Leung Y, Devlin SM, Novak K, Kaplan GG, Iacucci M, Seow C. Review article: dermatological complications of immunosuppressive and anti-TNF therapy in inflammatory bowel disease. Aliment Pharmacol Ther. 2013;38:1002-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
86.  Radić M, Martinović Kaliterna D, Radić J. Drug-induced vasculitis: a clinical and pathological review. Neth J Med. 2012;70:12-17.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Ramos-Casals M, Brito-Zerón P, Soto MJ, Cuadrado MJ, Khamashta MA. Autoimmune diseases induced by TNF-targeted therapies. Best Pract Res Clin Rheumatol. 2008;22:847-861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 198]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
88.  Ramos-Casals M, Roberto-Perez-Alvarez, Diaz-Lagares C, Cuadrado MJ, Khamashta MA; BIOGEAS Study Group. Autoimmune diseases induced by biological agents: a double-edged sword? Autoimmun Rev. 2010;9:188-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 186]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]