• No results found

University of Groningen Genomic medicine in inflammatory bowel disease Voskuil, Michiel

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Genomic medicine in inflammatory bowel disease Voskuil, Michiel"

Copied!
11
0
0

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

Hele tekst

(1)

University of Groningen

Genomic medicine in inflammatory bowel disease

Voskuil, Michiel

DOI:

10.33612/diss.136307453

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: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Voskuil, M. (2020). Genomic medicine in inflammatory bowel disease. University of Groningen. https://doi.org/10.33612/diss.136307453

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)
(3)

TNFα-ANTAGONIST USE AND MUCOSAL

INFLAMMATION ARE ASSOCIATED WITH

INCREASED INTESTINAL EXPRESSION OF

SARS-CoV-2 HOST PROTEASE TMPRSS2 IN

PATIENTS WITH INFLAMMATORY BOWEL

DISEASE

Amber Bangma1,2*, Michiel D. Voskuil1,2*, Shixian Hu1,2, Werna T.C. Uniken Venema1,2, Arno R. Bourgonje1,

Eleonora A.M. Festen1,2, Harry van Goor3, Gerard Dijkstra1, Rinse K. Weersma1

Gastroenterology 2020

* Contributed equally to this work

1Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the

Netherlands; 2Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; 3Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands

(4)
(5)

Letter to the Editor

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral transfection is dependent on ACE2 and TMPRSS2, and increased intestinal receptor expression might support viral replication[1,2,3].

Therefore, we read with great interest the work by Krzysztof et al, reporting that anatomical location, intestinal inflammation and age are key determinants of intestinal expression of

angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) in patients

with inflammatory bowel disease (IBD)[1]. However, several additional questions remain as the

authors themselves also describe. It is important to understand the effects of immunomodulating drugs on ACE2 and TMPRSS2 expression, and the cell-type specific context in which these genes are expressed[1]. In addition, it is important to understand genetic determinants of intestinal ACE2 and TMPRSS2 expression. To address these remaining questions, we queried host genetic

data and RNA sequencing data of intestinal biopsies from patients with IBD. We independently confirm that anatomical location, intestinal inflammation and age are determinants of intestinal

ACE2 and TMPRSS2 expression. Furthermore, we demonstrate that immunomodulating drugs

are associated with increased intestinal TMPRSS2 expression, and highlight the cell-type specific context of ACE2 and TMPRSS2 expression.

We analyzed bulk RNA sequencing data of 92 ileal and 199 colonic snap frozen mucosal biopsies from 168 patients with IBD, as part of the 1000IBD project[4]. Genotypes were obtained using both

imputed Global Screening Array data and whole exome sequencing. We analyzed clinical data regarding the use of immunomodulating drugs, age, sex, diagnosis (CD, UC or IBD-U) and BMI. Multivariate linear mixed regression analyses were performed to assess the effects of the above-mentioned clinical factors on intestinal gene expression levels of ACE2 and TMPRSS2 (R v.3.6.0). In addition, we assessed the effects of host genetic variation on gene expression (cis-expression quantitative trait loci). All of the above-mentioned clinical factors, as well as sequencing batch, were included as covariates. We performed deconvolution analyses on bulk RNA sequencing data to assess cell-type-specific expression of ACE2 and TMPRSS2. We validated this approach using single-cell RNA sequencing data from an independent set of 18 colonic biopsies from 11 patients with ulcerative colitis (unpublished data).

First, we replicated the effects of anatomical location, intestinal inflammation and age on intestinal expression of ACE2 and TMPRSS2. Expression of ACE2 was higher, and TMPRSS2 was lower in ileum compared to colon (P < 2.2 x 10-16). Moreover, expression of ACE2 was lower, and expression

of TMPRSS2 was higher in inflamed ileum compared to non-inflamed ileum, independent of medication use, age, sex, diagnosis and BMI (P = 4.4 x 10-6 and P = 8.6 x 10-8, respectively) (Figure

1A). Ileal TMPRSS2 expression was associated with increasing age (P = .04) (Figure 2). Furthermore, we found increased TMPRSS2 expression in intestinal (ileal and colonic) biopsies of male patients (P = .02).

Second, we studied the effects of immunomodulating drugs on ACE2 and TMPRSS2 expression in intestinal biopsies. We found increased TMPRSS2 expression in ileal biopsies of patients using Tumor Necrosis Factor alpha (TNFα)-antagonists (P = 8.8 x 10-6) independent of intestinal

inflammation, age, sex, diagnosis and BMI (Figure 1B).

Intestinal expression of SARS-CoV-2 host protease TMPRSS2 in patients with IBD

(6)

Figure 1. Intestinal TMPRSS2 gene expression in patients with inflammatory bowel disease. Panel A: Violin plots representing TMPRSS2 gene expression levels in ileal and colonic biopsies with and without active mucosal inflammation. 60 non-inflamed ileal, 32 inflamed ileal, 118 non-inflamed colonic, and 81 inflamed colonic biopsies were included. Differences in gene expression (log2-transformed counts per million reads mapped [cpm]) between inflamed and non-inflamed biopsies were assessed for ileal and colonic biopsies separately, using multivariate linear mixed regression analyses correcting for use of immunomodulating drugs (including TNFα-antagonists, steroids, thiopurines, and aminosalicylates), age, sex, BMI, diagnosis, and RNA-sequencing batch. P < .05 was considered significant. Panel B: Violin plots representing

TMPRSS2 gene expression levels in ileal and colonic biopsies from patients with and without TNFα-antagonist use. 71

ileal biopsies from TNFα-antagonist users, 20 ileal biopsies from non-users, 161 colonic biopsies from TNFα-antagonist users, and 38 colonic biopsies from non-users were included. Differences in gene expression (log2-transformed cpm) between patients using and not using TNFα-antagonist therapy were assessed for ileal and colonic biopsies separately, with multivariate linear mixed regression analyses correcting for mucosal inflammation, use of immunomodulating drugs (including steroids, thiopurines, and aminosalicylates), age, sex, BMI, diagnosis, and RNA-sequencing batch. Data points are each represented by a dot, and refer to log2-transformed cpm. Data is presented as boxplots indicating the median and quartiles with whiskers reaching up to 1.5 times the interquartile range (horizontal dashed lines). The violin plot outlines illustrate the kernel probability density, i.e. the width of the shaded area represents the proportion of the data located there.

Ileum Colon

Non−inflamed Inflamed Non−inflamed Inflamed

6 8 10 Mucosal inflammation TMPRSS2 (log2[cpm])

A

Mucosal inflammation No Yes P = 8.6 x 10-8 n.s. Colon No Yes No Yes 6 8 10 TNFα−antagonist use TMPRSS2 (log2[cpm])

B

TNFα−antagonist use No Yes n.s. P = 8.8 x 10-6 Ileum Chapter 9

(7)

Because aminosalicylates were only used in the context of ulcerative colitis, we assessed the influence of aminosalicylates in colonic tissue only, and observed an increased TMPRSS2 expression (P = .02). The use of thiopurines or steroids was not associated with differential expression of ACE2 or TMPRSS2. Furthermore, host genetic variation was not associated with differential expression of ACE2 and TMPRSS2.

Third, we interrogated the cell-type-specific context of intestinal ACE2 and TMPRSS2 expression. Using bulk RNA sequencing data, we quantified cell-type proportions and observed that an enrichment of epithelial cells is associated with increased expression of ACE2 and TMPRSS2 in both ileum and colon (all P < .004). Using single-cell RNA sequencing data from colonic biopsies, we observed that TMPRSS2 is mainly expressed by absorptive enterocytes, and that intestinal inflammation is associated with increased TMPRSS2 expression within absorptive enterocytes (P = 3.2 x 10-22). ACE2 was also primarily expressed by absorptive enterocytes, but expression was not

affected by intestinal inflammation.

Figure 2. Intestinal TMPRSS2 gene expression by age. The x-axis of the scatterplot represents the age of the patient at time of biopsy collection, while the y-axis represents TMPRSS2 gene expression levels (log2-transformed counts per million reads mapped [cpm]). Here, 91 ileal biopsies and 191 colonic biopsies were included. Data points are each represented by a dot, and the line illustrates the best-fitting straight line through the data points using linear regression. Correlation between gene expression levels and age was assessed for ileal and colonic biopsies separately using multivariate linear mixed regression analyses correcting for mucosal inflammation, use of immunomodulating drugs (including TNFα-antagonists, steroids, thiopurines, and aminosalicylates), sex, BMI, diagnosis, and RNA-sequencing batch. P < .05 was considered significant.

Intestinal expression of SARS-CoV-2 host protease TMPRSS2 in patients with IBD

Ileum Colon 20 40 60 80 20 40 60 80 6 7 8 9 10 11 Age TMPRSS2 (log2[cpm]) P = .04 n.s.

9

(8)

In conclusion, ACE2 and TMPRSS2 are key proteins for cellular entry of SARS-CoV-2, and are highly expressed in the intestinal mucosa. Next to intestinal inflammation, age, sex and anatomical location, also the use of TNFα-antagonists and aminosalicylates influence intestinal expression of TMPRSS2. We demonstrate that intestinal inflammation is associated with increased expression of TMPRSS2 in absorptive enterocytes, suggesting that the increased expression is not merely an effect of change in cellular composition during inflammation. Altered intestinal expression could render patients with IBD particularly susceptible to COVID-19 and absorptive enterocytes could provide targets for interventional studies. Indeed, clinical studies are needed to monitor the impact of COVID-19 on patients with IBD.

Acknowledgements

We thank all the participants of the 1000IBD cohort.

Author contributions

AB and MDV contributed equally to this work. Concept and design: AB, MDV, RKW. Acquisition, analysis, or interpretation of data: AB, MDV, SH, WTCUV, ARB, RKW. Drafting of the manuscript: AB, MDV, RKW. Critical revision of the manuscript: SH, WTCUV, ARB, HvG, EAMF, GD. All authors approved the final version of this manuscript.

Conflicts of interest

EAMF received an unrestricted grant from Takeda. GD received an unrestricted research grant from Takeda, and received speaker fees from Pfizer and Janssen Pharmaceuticals. RKW acted as consultant for Takeda, received unrestricted research grants from Takeda, Johnson and Johnson, Tramedico and Ferring and received speaker fees from MSD, Abbvie and Janssen Pharmaceuticals. The remaining authors disclose no (potential) conflicts of interest.

Funding

RKW is supported by a Diagnostics Grant from the Dutch Digestive Foundation (D 16-14). EAMF is supported by a MLDS Career Development grant (CDG 14-04).

(9)

References 1 2 3 4 5

Krzysztof NJ, Christoffer LJ, Rahul K, et al. Age, inflammation and disease location are critical determinants of intestinal expression of SARS-CoV-2 receptor ACE2 and TMPRSS2 in inflammatory bowel disease. Gastroenterology. Published Online First: 12 May 2020 12. doi: 10.1053/j.gastro.2020.05.030 Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181(2):271-280.

Lamers MM, Beumer J, van der Vaart J, et al. SARS-CoV-2 productively infects human gut enterocytes. Science. Published Online First: 01 May 2020. doi: 10.1126/science.abc1669

Zhang H, Kang Z, Gong H, et al. Digestive system is a potential route of COVID-19: an analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut. 2020;69:1010-1018.

Imhann, F, van der Velde KJ, Barbieri R, et al. The 1000IBD project: multi-omics data of 1000 inflammatory bowel disease patients; data release 1. BMC Gastroenterol. 2019;19(1):5.

Intestinal expression of SARS-CoV-2 host protease TMPRSS2 in patients with IBD

(10)
(11)

Referenties

GERELATEERDE DOCUMENTEN

Lieve papa &amp; mama, ik wil jullie bedanken voor de onvoorwaardelijke steun en liefde die jullie mij mijn hele leven al hebben gegeven. Ik heb van jullie mogen leren dat ik trots

Good evidence on types of dietary fibers is lacking, hence, specific advice cannot be given to IBD patients (this thesis). If the Groningen Anti-Inflammatory Diet (GrAID)

Printing of this thesis was financially supported by the University of Groningen, the Graduate School of Medical Sciences of the University Medical Center Groningen, Noord

The final part (Therapeutic outcomes) aims to identify genetic variants predictive of adverse drug responses and show the potential of pre-treatment genetic testing in patients

Early GWAS and subsequent meta-analyses in CD revealed CD risk loci with candidate genes, encoding proteins involved in immunological pathways such as autophagy (ATG16L1 and IRGM),

We performed RNA sequencing analyses of single cells (scRNAseq) from the intestinal mucosa and peripheral blood of patients with CD to identify cells that express CD risk genes

NUDT15 codon 139 is the best pharmacogenetic marker for predicting thiopurine-induced severe adverse events in Japanese patients with inflammatory bowel disease: A

Association of genetic variants in NUDT15 with thiopurine-induced myelosuppression in patients with inflammatory bowel disease using data from the Exome Wide Association Study