The University of Southampton
University of Southampton Institutional Repository

Deleterious genetic variation across the NOD-signaling pathway is associated with reduced NFKB-signaling transcription and upregulation of alternative inflammatory transcripts in paediatric inflammatory bowel disease

Deleterious genetic variation across the NOD-signaling pathway is associated with reduced NFKB-signaling transcription and upregulation of alternative inflammatory transcripts in paediatric inflammatory bowel disease
Deleterious genetic variation across the NOD-signaling pathway is associated with reduced NFKB-signaling transcription and upregulation of alternative inflammatory transcripts in paediatric inflammatory bowel disease

Background: Inflammatory bowel disease may arise with inadequate immune response to intestinal bacteria. NOD2 is an established gene in Crohn's disease pathogenesis, with deleterious variation associated with reduced NFKB signaling. We hypothesized that deleterious variation across the NOD2 signaling pathway impacts on transcription. Methods: Treatment-naïve pediatric inflammatory bowel disease patients had ileal biopsies for targeted autoimmune RNA-sequencing and blood for whole exome sequencing collected at diagnostic endoscopy. Utilizing GenePy, a per-individual, per-gene score, genes within the NOD signaling pathway were assigned a quantitative score representing total variant burden. Where multiple genes formed complexes, GenePy scores were summed to create a "complex"score. Normalized transcript expression of 95 genes within this pathway was retrieved. Regression analysis was performed to determine the impact of genomic variation on gene transcription. Results: Thirty-nine patients were included. Limited clustering of patients based on NOD signaling transcripts was related to underlying genomic variation. Patients harboring deleterious variation in NOD2 had reduced NOD2 (β = -0.702, P = 4.3 × 10-5) and increased NFKBIA (β = 0.486, P =. 001), reflecting reduced NFKB signal activation. Deleterious variation in the NOD2-RIPK2 complex was associated with increased NLRP3 (β = 0.8, P = 3.1475 × 10-8) and TXN (β = -0.417, P = 8.4 × 10-5) transcription, components of the NLRP3 inflammasome. Deleterious variation in the TAK1-TAB complex resulted in reduced MAPK14 transcription (β = -0.677, P = 1.7 × 10-5), a key signal transduction protein in the NOD2 signaling cascade and increased IFNA1 (β = 0.479, P =. 001), indicating reduced transcription of NFKB activators and alternative interferon transcription in these patients. Conclusions: Data integration identified perturbation of NOD2 signaling transcription correlated with genomic variation. A hypoimmune NFKB signaling transcription response was observed. Alternative inflammatory pathways were activated and may represent therapeutic targets in specific patients.

Crohn’s disease, IBD, NOD2, WES, transcriptome
1536-4844
912-922
Ashton, James
03369017-99b5-40ae-9a43-14c98516f37d
Boukas, Konstantinos
d6d89940-c494-4ce4-a3e8-ad1372dcee00
Stafford, Imogen, Sian
50987dc1-3772-408f-9093-9124f3d6b2cd
Cheng, Guo
fdfb3e03-f185-49b1-9c53-05b93bb6c8d0
Haggarty, Rachel
d79fd59d-2cdc-465a-93e4-b0aeb78583c6
Coelho, Tracy
a78b627c-ea78-41e1-9553-0390921e3c93
Batra, Akshay
822f891e-87ca-41d9-b68d-27c395e88809
Afzal, Nadeem A.
62505946-2503-42ba-9b02-85513bb3ec87
Williams, Anthony
973ff46f-46f1-4d7c-b27d-0f53221e4c44
Polak, Marta
e0ac5e1a-7074-4776-ba23-490bd4da612d
Beattie, R. Mark
9a66af0b-f81c-485c-b01d-519403f0038a
Ennis, Sarah
7b57f188-9d91-4beb-b217-09856146f1e9
Ashton, James
03369017-99b5-40ae-9a43-14c98516f37d
Boukas, Konstantinos
d6d89940-c494-4ce4-a3e8-ad1372dcee00
Stafford, Imogen, Sian
50987dc1-3772-408f-9093-9124f3d6b2cd
Cheng, Guo
fdfb3e03-f185-49b1-9c53-05b93bb6c8d0
Haggarty, Rachel
d79fd59d-2cdc-465a-93e4-b0aeb78583c6
Coelho, Tracy
a78b627c-ea78-41e1-9553-0390921e3c93
Batra, Akshay
822f891e-87ca-41d9-b68d-27c395e88809
Afzal, Nadeem A.
62505946-2503-42ba-9b02-85513bb3ec87
Williams, Anthony
973ff46f-46f1-4d7c-b27d-0f53221e4c44
Polak, Marta
e0ac5e1a-7074-4776-ba23-490bd4da612d
Beattie, R. Mark
9a66af0b-f81c-485c-b01d-519403f0038a
Ennis, Sarah
7b57f188-9d91-4beb-b217-09856146f1e9

Ashton, James, Boukas, Konstantinos, Stafford, Imogen, Sian, Cheng, Guo, Haggarty, Rachel, Coelho, Tracy, Batra, Akshay, Afzal, Nadeem A., Williams, Anthony, Polak, Marta, Beattie, R. Mark and Ennis, Sarah (2022) Deleterious genetic variation across the NOD-signaling pathway is associated with reduced NFKB-signaling transcription and upregulation of alternative inflammatory transcripts in paediatric inflammatory bowel disease. Inflammatory Bowel Diseases, 28 (6), 912-922. (doi:10.1093/ibd/izab318).

Record type: Article

Abstract

Background: Inflammatory bowel disease may arise with inadequate immune response to intestinal bacteria. NOD2 is an established gene in Crohn's disease pathogenesis, with deleterious variation associated with reduced NFKB signaling. We hypothesized that deleterious variation across the NOD2 signaling pathway impacts on transcription. Methods: Treatment-naïve pediatric inflammatory bowel disease patients had ileal biopsies for targeted autoimmune RNA-sequencing and blood for whole exome sequencing collected at diagnostic endoscopy. Utilizing GenePy, a per-individual, per-gene score, genes within the NOD signaling pathway were assigned a quantitative score representing total variant burden. Where multiple genes formed complexes, GenePy scores were summed to create a "complex"score. Normalized transcript expression of 95 genes within this pathway was retrieved. Regression analysis was performed to determine the impact of genomic variation on gene transcription. Results: Thirty-nine patients were included. Limited clustering of patients based on NOD signaling transcripts was related to underlying genomic variation. Patients harboring deleterious variation in NOD2 had reduced NOD2 (β = -0.702, P = 4.3 × 10-5) and increased NFKBIA (β = 0.486, P =. 001), reflecting reduced NFKB signal activation. Deleterious variation in the NOD2-RIPK2 complex was associated with increased NLRP3 (β = 0.8, P = 3.1475 × 10-8) and TXN (β = -0.417, P = 8.4 × 10-5) transcription, components of the NLRP3 inflammasome. Deleterious variation in the TAK1-TAB complex resulted in reduced MAPK14 transcription (β = -0.677, P = 1.7 × 10-5), a key signal transduction protein in the NOD2 signaling cascade and increased IFNA1 (β = 0.479, P =. 001), indicating reduced transcription of NFKB activators and alternative interferon transcription in these patients. Conclusions: Data integration identified perturbation of NOD2 signaling transcription correlated with genomic variation. A hypoimmune NFKB signaling transcription response was observed. Alternative inflammatory pathways were activated and may represent therapeutic targets in specific patients.

Text
Deleterious genetic variation within the NOD_Untracked - Accepted Manuscript
Download (278kB)
Text
izab318 - Version of Record
Download (8MB)

More information

Accepted/In Press date: 13 November 2021
e-pub ahead of print date: 3 January 2022
Published date: 3 January 2022
Additional Information: © 2022 Crohn’s & Colitis Foundation. Published by Oxford University Press on behalf of Crohn’s & Colitis Foundation.
Keywords: Crohn’s disease, IBD, NOD2, WES, transcriptome

Identifiers

Local EPrints ID: 453106
URI: http://eprints.soton.ac.uk/id/eprint/453106
ISSN: 1536-4844
PURE UUID: 12a6f929-e496-45ff-810b-ad7f410c0ebc
ORCID for James Ashton: ORCID iD orcid.org/0000-0003-0348-8198
ORCID for Imogen, Sian Stafford: ORCID iD orcid.org/0000-0003-1666-1906
ORCID for Sarah Ennis: ORCID iD orcid.org/0000-0003-2648-0869

Catalogue record

Date deposited: 08 Jan 2022 21:22
Last modified: 17 Mar 2024 06:58

Export record

Altmetrics

Contributors

Author: James Ashton ORCID iD
Author: Konstantinos Boukas
Author: Imogen, Sian Stafford ORCID iD
Author: Guo Cheng
Author: Rachel Haggarty
Author: Tracy Coelho
Author: Akshay Batra
Author: Nadeem A. Afzal
Author: Marta Polak
Author: R. Mark Beattie
Author: Sarah Ennis ORCID iD

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×