• No results found

University of Groningen Genetic susceptibility for inflammatory bowel disease across ethnicities and diseases van Sommeren, Suzanne

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Genetic susceptibility for inflammatory bowel disease across ethnicities and diseases van Sommeren, Suzanne"

Copied!
13
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Genetic susceptibility for inflammatory bowel disease across ethnicities and diseases

van Sommeren, Suzanne

DOI:

10.33612/diss.100597247

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

van Sommeren, S. (2019). Genetic susceptibility for inflammatory bowel disease across ethnicities and

diseases. Rijksuniversiteit Groningen. https://doi.org/10.33612/diss.100597247

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

CHAPTER

8

Conclusion, discussion and future

perspectives

(3)
(4)

113

CONCLUSION

Our first goal of this thesis was to increase the knowledge of the genetic basis of the inflam-matory bowel diseases (IBD) by identifying new genetic risk loci. We did this in multiple ways: at the time the first genome-wide association stud-ies (GWAS) were published, there was a need for replication of their findings in an independent cohort. In chapter 2 we could confirm two out of three loci that were previously reported in an ulcerative colitis (UC) GWAS in our independent Dutch cohort, one locus harbouring HNF4A and one harbouring CDH1. We did not replicate the third locus, containing candidate gene LAMB1. We suggest this may be due to phenotype dif-ferences compared to the identification cohort, our cohort contained more severely affected patients. After generating more extensive data of this cohort, none of the single nucleotide polymorphisms (SNPs) in this locus turned out to be associated with UC, which underscores that this SNP covers the locus well and no tech-nical error was made. The LAMB1 locus has been established in other cohorts in later years. All three genes play roles in the integrity of the intestinal barrier.

Next, in chapter 3 we performed a large trans-ethnic meta-analysis including almost 100,000 individuals from European, Indian, Iranian and East-Asian descent. We identified 38  new genetic risk loci for Crohn’s disease (CD), UC or IBD, increasing the number of in-dependently associated SNPs to 231 in 200 ge-netic loci. Furthermore, we demonstrated that there is extensive sharing of genetic risk across populations, showing that combining multiple populations is helpful for increasing much needed statistical power. There are some striking exceptions for a few loci which can be attributed to differences in occurrence or effect size of these loci in different populations.

In the period before the large GWAS meta- analyses, when sample sizes were relatively small (meaning low statistical power) and costs for genotyping were relatively high, it was important to select SNPs for testing for association. In chapter 4 we developed a new

method to prioritise SNPs that did not reach the genome-wide significant threshold in a published GWAS for replication. Our method is based on the known effect of these SNPs on gene expression, hypothesising that SNPs with an effect on gene expression were more likely to be involved in disease pathogenesis than SNPs with no such effect. In this way, we identified two new CD loci, harbouring candidate genes UBE2L3 and BCL3.

Because it is expected that genetic risk factors affect gene expression and we see an enrich-ment of eQTLs among disease associated SNPs we wanted to link genetic risk to gene expres-sion by focusing on a defined pathway both ge-netically and functionally strongly implicated in IBD: the Th17/IL23 pathway. In chapter 5 we combined the genetic risk of 10 Th17/IL23 pathway associated SNPs and correlated that to gene expression of 9 representative Th17/IL23 genes in peripheral blood mononuclear cells (PBMCs) of 80 IBD patients and 40 controls. We observed a lower baseline expression of IL6 in IBD patients compared to controls. Upon stim-ulation of T cells, we saw a smaller increase in IL23A gene expression in cases compared to controls, and a larger decrease in RORC gene expression in IBD patients compared to controls. We did not find a correlation between genetic risk load and gene expression. One explanation for this lack of association would be that alter-ations in gene expression are cell specific and becomes overshadowed in a mixed collection of cells like PBMCs.

In order to expand the knowledge of the ge-netic background shared with other diseases, we first addressed the overlap between IBD and its extraintestinal manifestations (EIM), be-cause we believe they share pathogenetic links. We demonstrated a large overlap in genetic risk loci between IBD and the EIM in chapter 6. Furthermore, based on co-expression of genes and protein-protein interactions, we showed that candidate genes residing in associated loci for the different diseases work together in pathways.

Finally, we aimed to identify genetic risk loci in a disease occurring after allogeneic

(5)

114

Chapter 8

hematopoietic cell transplantation and clinically resembling Crohn’s disease: gastro- intestinal graft-vs.-host disease. We found no significant associations in our small cohort, but found sug-gestive signals in interesting loci like JAK2, IL2 and IL2RA. This work is presented in chapter 7.

DISCUSSION AND FUTURE

PERSPECTIVES

The wide variety of the IBD disease spectrum and the complexity of disease pathogenesis make the disease both a heaven and a hell for research purposes. On the one hand, we can study the disease from many perspectives and use multiple techniques that evolve along the way. On the other hand, we are hampered by the many different factors influencing disease pathogenesis, making it hard to distinguish the importance of each separate component and unravel how different components influence each other. Although genetic research in IBD has delivered many results, there are many more questions remaining in our research field regarding identification of genetic risk loci and causal variants, correlating genetic risk fac-tors to biological processes and pathways that eventually lead to disease, and the influence of environmental factors and the microbiome on this process. My thesis has focused on differ-ent subsets of this field, therefore I will focus my discussion on several of these components, highlighting things I have learned or that have fascinated me over the years.

Identification of genetic risk loci

Genetic studies in IBD have been extremely successful and the insights they have provided are undeniable. At the start of my PhD the first GWAS in IBD were published, and needed rep-lication in independent cohorts, to which we contributed with the work presented in chap-ter 2. Furthermore we provided a strategy to prioritise candidate SNPs based on eQTL effects to test for association (chapter 4). Our biggest contribution to the identification of genetic risk loci was by identifying 38 new IBD risk loci in

the transethnic meta-analysis, raising the total number to 200 genetic loci (chapter 3). The dis-ease variance explained based on these genetic risk factors went to 13.1% for Crohn’s disease and 8.2% for ulcerative colitis. Although we are still not able to explain the full genetic background of IBD, we provided biological meaning by reinforcing the importance of the autophagy pathway by identifying candidate gene ATG4B, which plays a crucial role in this process. Furthermore, we can now implicate all three components of T cell activation (TCR ligation, co-stimulation and interleukin-2 sig-nalling) in disease pathogenesis by identifying LY75, CD28, CCL20, NFKBIZ, AHR and NFATC1. One could think that the maximum number of associated genetic loci will once be reached. However, it is now clear that the number of IBD risk loci still has the potential to grow, as long as we are able to increase statistical power with extra samples to discover new loci with smaller effect sizes. The current number of IBD risk loci is 2411 and a large new GWAS meta-analysis is

underway within the Internation IBD Genetics Consortium. Although the genetic variance ex-plained does not increase much with new loci, finding new (candidate) genes keeps enhancing our understanding of the pathogenesis of IBD by reinforcing existing or discovering new biologi-cal pathways that underlie disease pathogenesis. Performing genetic studies has become more generally available over the last decade. Ge-notyping arrays that enable us to study multi-ple genetic loci at the same time have become cheaper, making it possible to include a large number of samples and making genetic studies more available for study groups that focus on non-Caucasian populations. Furthermore, the experience of performing genetic studies has grown by cooperating and sharing knowledge. Our trans-ethnic meta-analysis showcases the result of such cooperation. By combining multiple ethnic populations we were able to demonstrate for the first time that most of the genetic risk is shared across populations. Be-cause we have more statistical power with this large dataset, we were able to identify new IBD risk loci. I believe this is great progress, but I

(6)

115

also think that the real added value of

incor-porating multiple ethnic populations will be beyond locus identification, and can be helpful in identifying causal variants and unravelling the effect of environmental factors on disease pathogenesis. It is clear that the overall focus of genetic research is no longer on locus iden-tification but on discovering the implications of genetic risk factors.

The need for large samples sizes results from the fact that IBD has a complex pathogenesis with multiple genetic and non-genetic factors. To elucidate one of these factors with limited effect, studies require large numbers. This is not only the case for genetic studies focusing on locus identification, but it also hampers other fields of IBD research like identification of causal variants and gene expression studies. It might be rewarding to use patients with ex-treme phenotypes (severely affected individuals, individuals with early onset disease) where one would expect that individual risk factors play larger roles compared to mildly affected patients.

Furthermore, IBD has a wide variety of dis-ease localisation and severity. Most of the ge-netic studies have applied a simple case-control strategy comparing CD or UC affected patients with healthy controls. Cleynen et al. went be-yond this and undertook an interesting study to investigate the correlation between genetic risk factors and disease location and disease be-haviour. They identified associations between SNPs in the MST1, NOD2 and MHC loci and age at diagnosis and disease location of CD and UC. They also showed that genetic risk scores dif-fer between ileal CD, ileocolonic CD, colonic CD and UC. Moreover, they found that genetic risk loci associated with UC correlate better with CD localisation (colonic vs non-colonic) than risk loci associated with CD, suggesting that genetic variation contributes significantly to disease location.2 Also, genetic variation

con-tributing to disease progression in CD has been described.3 Because of the wide variety in the

phenotype, I believe it would be very interesting to investigate further how genetic factors lead to particular phenotypes or disease behaviour. With the sample sizes we are accustomed to, we

should be able to have enough statistical power to detect correlations with a subset of individ-uals. The real challenge is to deeply phenotype individuals, which is a challenge many groups are willing to take on.

Identification of causal variants

In order to fully comprehend how associated genetic risk loci contribute to disease patho-genesis it is crucial to identify causal variants. Multiple approaches have been applied. One of them is fine-mapping loci by using SNP data; however, this is complicated by the strong correlation structure of the genome (linkage disequilibrium – LD). The Immunochip accom-modates fine-mapping by the dense coverage of relevant loci which allows researches to look for subtle differences of association within the locus. Huang et al. performed fine-mapping for 94 IBD signals in high density loci and found 139 independent signals. For each of these sig-nals a set of SNPs (credible sets) was selected that was > 95% likely to contain the causal variant. For 18 signals, only one variant was selected, 23 had 2–5 variants that were likely to contain the causal variant, for only 23 signals the set contained more than 50 SNPs. There is an en-richment of functional elements. Furthermore, by identifying single or functional variants that are likely to underlie disease association, the number of candidate genes can be reduced.4

An approach based on conditioning analy-sis on association signals of (predominantly) classical HLA-alleles was used to fine-map the HLA region, a region with a large effect in predominantly ulcerative colitis but with very strong LD. In this study, Goyette et al. showed amongst others things that the disease variance explained can be increased by using causal vari-ants (classical HLA alleles) instead of tag SNPs associations, which underscores the idea that part of the ‘hidden heritability’ phenomena is explained by incomplete covering of the associa-tion signal by the tag SNP.5 Another approach is

sequencing interesting regions, for which mul-tiple approaches can be used. Pooled targeted sequencing identified rare variants in for ex-ample MUC2,6 CARD9, IL18RAP and PTPN22.7

(7)

116

Chapter 8

Over the years, cost of sequencing has decreased dramatically, permitting researchers to increase sample size and genome coverage. Although some newly associated rare variants are being identified, pointing out the candidate gene in the locus relevant for follow up, the number of identified causal variants remains low and does not (yet) clarify the genetic base of the majority of associated loci.

I believe we can benefit greatly from the use of multiple ethnic populations in fine-mapping analyses. LD structures can differ between pop-ulations and especially poppop-ulations with smaller LD blocks (e.g. African ancestry) can aid to downgrade large stretches of LD. This method has proven to be successful in traits like adipos-ity and fasting glucose and insulin levels8,9 and

would be very interesting to perform on a large scale in IBD.

Population specific loci?

Interestingly, in the transethnic meta-analysis we have observed genetic differences between populations, some strongly associated ‘Eu-ropean’ loci don’t seem to play an important role in the Asian population and some loci contribute largely to the variance explained in the Asian population but have limited effect in the European population. Some of these can be explained by differences in occurrence of SNPs across populations and some by dif-ferences in effect size. A striking example is NOD2, the strongest associated locus in Eu-ropean Crohn’s disease. All three previously described causal variants are monomorphic, in other words non-existing, in the Asian popu-lation. Associations with other NOD2 markers have been described in Asian populations, for example JW1 and P268S; however, they are performed in small cohorts and/or could not be replicated.10,11 We could also not replicate

the findings of these variants in our data, nor found any strong signal in the locus (lowest p-value 7.18 × 10–4). The other autophagy loci

(IRGM and ATG16L1) both show lower asso-ciation signals in the Asian population com-pared to the European; for the IRGM SNP this can be attributed to a difference in effect size.

Other SNPs in this locus show some convincing signals in our largest cohort, the East Asian, 138 kb downstream of the European top SNP

(lowest p-value at rs2287720 7.66 × 10–8). For

the ATG16L1 SNP, the difference can be at-tributed to differences in both effect size and allele frequency. Furthermore, in the complete ATG16L1 locus only nominal signals are found (lowest p-value at rs6758317 0.0016). Of course it is too early to speculate that autophagy would not play a role in Asian IBD. We lack functional studies on autophagy in Asian populations, so we don’t know the role of autophagy or ge-netic variation in autophagy associated loci in Asian IBD, which would be very interesting to follow up.

In contrast, the locus containing TNFSF8/ TNFSF15 has a much larger effect size in the East Asian population compared to the European population (for example rs4246905 European OR = 1.15 and East Asian OR = 1.75). It might be more rewarding investigating a causal variant in this locus in the East Asian population rather than in the European population. Of course we need to take into account that possible (rare) causal variants might be population specific, but finding causal variants in a specific popu-lation could provide insight into overall disease pathogenesis.

From genetic risk to biological implication In order to really understand disease pathogene-sis we need to correlate genetic risk factors with biological processes to try and understand how these processes get disrupted. As mentioned before, this can be done in many different ways. One important factor is how genetic risk factors influence gene expression. Many GWAS asso-ciated loci harbour eQTLs, but for most GWAS hits an eQTL effect is not described. We have in-vestigated the influence of multiple known IBD risk factors on gene expression of the Th17/IL23 pathway that where shown to be very relevant for disease pathogenesis from both a genetic perspective (enriched in genetic risk factors) and a biological perspective. Furthermore, one of the treatment options is directly targeting this pathway (Ustekinumab; targeted against IL23).

(8)

117

The results of this experiment are described in

chapter 5. Although we found some alterations in gene expression between cases and controls in stimulated and unstimulated PBMCs, we found no correlation between genetic risk and altered gene expression. This was not the result we had hoped for. There are several possible reasons that would explain why we did not find more correlations, one of course being statisti-cal power. One other important reason would be that alterations in gene expression are very cell specific and because we used PBMCs, which is a mix of cells, we were not able to pick up the rele-vant signal. This is underscored by the fact that we did not find any relation between genetic risk and gene expression in a larger database of PBMCs (not IBD patients). This was also found by Huang et al. when they searched for enrich-ment of eQTLs among their credible sets after finemapping: they did not find enrichment of eQTLs in PBMCs; however, when they analysed eQTLs in specific celltypes (CD4+ cells or ileal cells) they did find enrichment of eQTLs (Huang). Therefore, I believe it is extremely important to use specific cells in relevant tissue (for exam-ple T-cells in inflamed colon) and in this way investigate how genetic risk factors influence gene expression. Luckily, with advances in tech-niques like cell sorting and single cell mRNA sequencing, such an effort can be made. Other ways to uncover functional consequences of ge-netic variation are identifying protein coding variants, disruption of enhancers, promoters and transcription-factor binding sites. In the last decade much has been discovered in this area.12

The next step is how genes work together in pathways. We know several relevant pathways for IBD pathogenesis such as the Th17/IL23R pathway and autophagy and we are currently at the point that we are able to identify pathways relevant for disease pathogenesis, but many questions remain unanswered: how does ge-netic variation influence the function of such a pathway? What is the contribution of a certain pathway to disease pathogenesis? Does this differ between subgroups of patients (certain phenotypes) or between populations? How do biological pathways work together? How do

other factors (for example environmental fac-tors) influence pathways? Which pathways are shared between CD and UC, and other diseases and which are specific for a certain trait? In or-der to answer all these questions we will even-tually need functional studies incorporating multiple layers of data (multi-omics).

Shared genetics and pathogenesis

It has been known that the disease spectrum of IBD comprises extra-intestinal manifestations and IBD co-occurs with other immune medi-ated diseases. A logical explanation would be that these diseases share genetic risk factors and biological pathways in which they contrib-ute to disease pathogenesis. It is estimated that around 70% of IBD risk loci are shared with other immune mediated diseases like celiac dis-ease, ankylosing spondylitis, primary sclerosing cholangitis and type 1 diabetes mellitus.13 In

chapter 6, we investigated the overlap between IBD and its extra-intestinal manifestations and found substantial overlap in genetic fac-tors and biological pathways. This overlap was also described well in a recent review.14 On the

other hand there must be factors that drive the occurrence of a specific EIM or co-occurrence of another immune mediated disease, and not lead to another EIM of immune mediated dis-ease. In line with this is the co-occurrence of PSC and IBD: 75% of PSC patients also have a form of IBD; however, up until now the genetic overlap is limited.15 Therefore, there must be

specific factors that drive the development of this disease phenotype in contrast to developing for example IBD only.

To further investigate the overlapping patho-genesis it is possible to take several approaches. On the one hand, we can combine IBD data with data from related disorders to search for over-lapping signals. On the other hand, if we are able to deeply phenotype our IBD cases, we can focus on patients who develop (certain) EIM and investigate what genetic background or other factor drives a particular disease course.

In chapter 7, we focus our attention on the overlap with graft-versus-host disease after allogenic stemcell transplantation, a disease

(9)

118

Chapter 8

in which the gastrointestinal form resembles Crohn’s disease. Again we found substantial overlap. Interestingly, we found a suggestive signal at JAK2 in GVHD, a protein that is tar-geted by Tofacitinib, a recently registered treat-ment option in IBD. Because of the overlap be-tween IBD and many other diseases, it is logical that we can learn and benefit from each other. For example, functional work done for another disease can also aid in our understanding of IBD. Furthermore, registered drugs for other diseases can also be interesting treatment op-tions for IBD (drug repositioning).

Exposome and microbiome

As mentioned in the introduction a lot of re-search has been done on environmental factors influencing IBD (exposome) and more recently the role of the microbiome has been highlighted. The interaction between the genetic background, the exposome and microbiome is not yet under-stood. I believe that including multiple different populations can significantly aid in answering this question. Where environmental factors dif-fer between populations, we have shown that genetic background greatly overlaps, providing great opportunities for further research.

We do need to take into account that the influence of environmental factors might be different in other populations compared to Eu-ropean studies. For example, cigarette smoking increases the risk off Crohn’s disease in Western countries, but not in Eastern Asian countries and among immigrants from the Middle East.16

In developing countries, measures of hygiene have not demonstrated the inverse association reported in the West and have in fact been asso-ciated with an increased risk of UC.17

Intriguing is the overall first occurrence of UC and later occurrence of CD, both in the past in European and now in industrialising coun-tries.18,19 This leads to the question why: do

environmental factors drive this difference? In European populations the heritability of UC is lower compared to CD, if this is the same for industrialising populations, a greater contribu-tion of environmental factors to disease patho-genesis of UC compared to CD would explain

this. We don’t know the exact mechanisms how environmental factors contribute to dis-ease pathogenesis, and we know even less about differences in exposome for UC and CD. One exception is smoking, which, in European pop-ulations increases the risk of CD and quitting smoking increases UC. Part of industrialising is an increasing number of smokers; in our Asian cohort we also saw a higher occurrence of smokers. One would expect to see a higher incidence of CD. Another hypothesis is that the genetic background of UC in for example Asian populations would resemble European UC more than in CD. Other possible explanations would be a better recognition of UC than CD, because clinical aspects are more pronounced. None-theless, a thorough investigation of the genetic basis and the interaction with the exposome between populations will be needed.

There is still limited access to non-European cohorts, so efforts will have to be made to in-clude more non-European cases and controls. There are some factors that we have to account for when using different populations: (I) geno-typing platforms were primarily designed to cover the ‘European genome’; therefore genetic variation in other population might not be cov-ered well. A striking example is the Immunochip, where one fifth of the technically successful gen-otyped SNPs are monomorphic in our East Asian cohort. (II) imputing genotypes is a very success-ful method of improving genotyping coverage, but because we are dealing with population specific variation, the reference genomes used for imputation need to originate from the same population. (III) the statistical method used needs to take into account genetic differences between populations. (IV) differences in cohort size. (V) differences in clinical aspects Systems biology

It is clear that in order to provide answers re-lated to disease pathogenesis, behaviour and response to therapy we will need many different types of data (omics): different types of genetic data (genotype data, sequence data), gene ex-pression data in relevant tissues, microbiome data and detailed phenotype data including

(10)

119

information about the disease, other diseases,

family history and detailed environmental expo-sure. Ideally, all data should originate from the same samples. In our own centre (UMCG), such data collection is already being undertaken: for 1,000 IBD patients, different forms of genetic data (GWAS, Immunochip, whole exome se-quence data), gene expression data (RNAseq on intestinal biopsies) and microbiome data (16S and full sequence data) are being collected in conjunction with clinical, exposome and dietary data.20 Furthermore, we will need robust

statis-tical approaches that can deal with all these lay-ers of data and provide answlay-ers to the question how the different omics work together and lead to the disease. Moreover, over the last few years different molecular profiles have been associ-ated with a particular IBD subphenotype, like a specific localisation or disease behaviour.21

Such a dataset provides opportunities to fully investigate such questions.

Genetic information currently used in clinical practice

In addition to understanding the pathogene-sis of IBD, we can use genetic information in daily practice. Genetic riskmodels are currently not useful in order to distinguish IBD affected individuals from non-IBD affected individu-als. This is probably due to the fact that many more non-genetic factors contribute to disease pathogenesis. If we get a better grasp of these other factors, as stated above, we might be able to better predict IBD occurrence, and perhaps disease behaviour and progression and response to therapy.

Another purpose genetic information can serve is found in the field of pharmacogenetics, where genetic factors predict the effect or side effects of drug therapies in IBD, as has been described for the development of thiopurine induced myelotoxicity with certain TPMT and NUDT15 genotypes,22,23 the development of

thiopurine induced pancreatitis with certain HLA variants24 and anti-TNF antibody

devel-opment associated with both genetic variants25

and gene expression profiles.26

Final remarks

After successfully identifying numerous IBD genetic risk loci, the research focus will shift to-wards finding underlying causal variants and in-vestigating the functional consequences for dis-ease pathogenesis. Furthermore, the influence of environmental factors and the microbiome will have to be clarified. I believe characterising multiple ethnic populations and incorporating information from other related (immune me-diated) diseases can aid in achieving this. The integration of multiple omics will be necessary to start to comprehend the complex interaction of the different components. I also believe that the attention should focus on subphenotypic aspects like disease localisation and behaviour. In order to do this, detailed phenotype information will be needed. The scale of this challenge is incredi-ble, making close collaboration necessary. How-ever, the goal of understanding and predicting disease behaviour and offering a personalised therapeutic strategy for each IBD affected patient makes this tremendous effort worthwhile.

REFERENCES

1. de Lange KM, Moutsianas L, Lee JC, et al. Genome- wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet. 2017;49:256–61.

2. Cleynen I, Boucher G, Jostins L, et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet. 2016;387:156–67.

3. Lee JC, Biasci D, Robeters R, et al. Genome-wide association study identifies distinct genetic contributions to prognosis and susceptibility in Crohn’s disease.

4. Huang H, Fang M, Jostins L, et al. Fine-mapping inflammatory bowel disease loci to single variant resolution. Nature 2017;547:173–8.

5. Goyette P, Boucher G, Dermot M, et al. High- density mapping of the MHC identifies a shared role for HLA-DRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcer-ative colitis. Nat genet 2015;47:172–9. 6. Visschedijk MC, Alberts R, Mucha S, et al. Pooled

(11)

120

Chapter 8

large Dutch cohort suggests population-specific associations of rare variants in MUC2. PLoS ONE 2016;11: e0159609

7. Rivas MA, Beaudoin M, Gardet A, et al. Deep resequencing of GWAS loci identifies indepen-dent rare variants associated with inflammatory bowel disease. Nat Genet 2011;43:1066–73. 8. Ng MCY, Graff M, Lu Y, et al. Discovery and

fine-mapping of adiposity loci using high density imputation of genome-wide association studies in individuals of African ancestry: African an-cestry anthropometry genetics consortium. PLoS Genet 2017;13: e1006719.

9. Liu C, Raghavan S, Maruthur N, et al. Trans- ethnic meta-analysis and functional annotation illuminates the genetic architecture of fasting glucose and insulin. Am J Hum Genet 2016;99: 56–75.

10. Chua KH, Hilmi I, Ng CC, Eng TL, Palaniappan S, Lee WS, Goh KL. Identification of NOD2/ CARD15 mutations in Malaysian patients with Crohn’s disease. J Dig Dis 2009;10–124–30. 11. Fuyono Y, Yamazaki K, Takahashi A, et al.

Ge-netic characteristics of inflammatory bowel dis-ease in a Japanese population. J Gastroenterol. 2016;51:672–681.

12. The ENCODE project consortium. An integrated encyclopedia of DNA elements in the human ge-nome. Nature 2012;489:57–74

13. Zhernakova A, van Diemen CC, Wijmenga C. Detecting shared pathogenesis from the shared genetics of immune-related diseases. Nat Rev Genet 2009;10:43–5.

14. Greuter T, Vavricka SR. Extraintestinal manifes-tations in inflammatory bowel disease – epide-miology, genetics, and pathogenesis. Exert Rev Gastro Hepatol 2019;14:307–17.

15. Ji SG, Juran BD, Mucha S, et al. Genome-wide as-sociation study of primary sclerosing cholangitis identifies new risk loci and quantifies the genetic relationship with inflammatory bowel disease. Nat Genet 2017;49:269–73.

16. Ng SC, Leong RW, Ko Y, et al. Environmental risk factors in inflammatory bowel disease in Asia-Pacific: a population-based case-control study. Gastroenterology 2014;1:S771–2. 17. Sood A, Mare D, Midha V, et al. Low hygiene and

exposure to infections may be associated with increased risk for ulcerative colitis in a North Indian population. Ann Gastro 2014;27:219–23. 18. Ekbom A, Helmick C, Zack M, et al. The epide-miology of inflammatory bowel disease: a large,

population-based study in Sweden. Gastroenter-ology 1991;100:350–8.

19. Ng SC, Tang W, Ching JY, et al. Incidence and phenotype of inflammatory bowel disease based on results from the Asia-Pacific Crohn’s and colitis epidemiology study. Gastroenterology 2013;145:158–65.

20. Imhann F, van der Velde KJ, Barbieri R, et al. The 1000IBD project: multi-omics data of 1000 in-flammatory bowel disease patients; data release 1. BMC Gastroenterology 2019;19:5–14. 21. Furey TS, Sethupathy P, Sheikh SZ. Redefining

the IBDs using genome-scale molecular pheno-typing. Nat Rev Gastroenterol Hepatol 2019 22. Lennard L, van Loon JA, Weinshilboum RM.

Pharmacogenetics of acute azathioprine toxic-ity: relationship to thiopurine methyltransfer-ase genetic polymorphism. Clin Pharmacol Ther 1989;46:149–54.

23. Yang SK, Hong M, Baek J, et al. A common mis-sense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet 2014;46:1017–20.

24. Heap GA, Weedon MN, Bewshea CM, et al. Nat Genet 2014;46:1131–1134.

25. Sazonovs A, Kennedy NA, Bewshea C, et al. HLA-DQA1 contributes to the development of antibod-ies to anti-TNF therapy in Crohn’s disease. ECCO oral presentation OP013 2018.

26. Gaujoux R, StarosvetskyE, Maimon N, et al. Cell-centred meta-analysis reveals baseline pre-dictors of anti-TNFA non-response in biopsy and blood of patients with IBD. Gut 2018;68:604–14.

(12)
(13)

Referenties

GERELATEERDE DOCUMENTEN

Further- more, although we tried to investigate a homo- geneous cohort of patients by including only patients with a severe form of GI GVHD, it might be possible that GVHD remains

De projecten in dit proefschrift zijn mogelijk gemaakt met behulp van samples van het Nederlands Initiatief voor Crohn en Colitis (ICC) en samples verzameld binnen het

Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations Jimmy Z Liu*, Suzanne van Sommeren*,

Identifying an increasing number of genetic risk loci for inflam- matory bowel disease hardly increases the explained disease va- riance, but contributes to biological insights

Chapter 8 Identification of clinical and genetic parameters associated with hidradenitis suppurativa in inflammatory bowel disease. Inflamm Bowel Dis

In addition to these clinical parameters known to be associated with severe disease course, there are many factors whose effect on disease phenotype and disease course has not

Observers with less than 10 years of experience performed best at scoring disease severity (Table 4) and were significantly better at scoring UC severity than observers with

Participants: Since 2007, every patient with IBD treated in one of the eight Dutch university medical centres is asked to participate in the Dutch IBD Biobank in which 225