Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism

Abstract Individuals with Autism Spectrum Disorder (ASD; autism) commonly present with gastrointestinal (GI) illness in addition to core diagnostic behavioural traits. The appendix, or cecum in mice, is important for GI homeostasis via its function as a key site for fermentation and a microbial rese...

Full description

Bibliographic Details
Main Authors: Chalystha Yie Qin Lee, Gayathri K. Balasuriya, Madushani Herath, Ashley E. Franks, Elisa L. Hill-Yardin
Format: Article
Language:English
Published: Nature Portfolio 2023-08-01
Series:Scientific Reports
Online Access:https://doi.org/10.1038/s41598-023-39555-y
_version_ 1797559825770479616
author Chalystha Yie Qin Lee
Gayathri K. Balasuriya
Madushani Herath
Ashley E. Franks
Elisa L. Hill-Yardin
author_facet Chalystha Yie Qin Lee
Gayathri K. Balasuriya
Madushani Herath
Ashley E. Franks
Elisa L. Hill-Yardin
author_sort Chalystha Yie Qin Lee
collection DOAJ
description Abstract Individuals with Autism Spectrum Disorder (ASD; autism) commonly present with gastrointestinal (GI) illness in addition to core diagnostic behavioural traits. The appendix, or cecum in mice, is important for GI homeostasis via its function as a key site for fermentation and a microbial reservoir. Even so, the role of the appendix and cecum in autism-associated GI symptoms remains uninvestigated. Here, we studied mice with an autism-associated missense mutation in the post-synaptic protein neuroligin-3 (Nlgn3 R451C ), which impacts brain and enteric neuronal activity. We assessed for changes in cecal motility using a tri-cannulation video-imaging approach in ex vivo preparations from wild-type and Nlgn3 R451C mice. We investigated cecal permeability and neurally-evoked secretion in wild-type and Nlgn3 R451C tissues using an Ussing chamber set-up. The number of cecal patches in fresh tissue samples were assessed and key immune populations including gut macrophages and dendritic cells were visualised using immunofluorescence. Nlgn3 R451C mice displayed accelerated cecal motor complexes and reduced cecal weight in comparison to wildtype littermates. Nlgn3 R451C mice also demonstrated reduced neurally-evoked cecal secretion in response to the nicotinic acetylcholine receptor agonist 1,1-dimethyl-4-phenylpiperazinium (DMPP), but permeability was unchanged. We observed an increase in the number of cecal patches in Nlgn3 R451C mice, however the cellular morphologies of key immune populations studied were not significantly altered. We show that the R451C nervous system mutation leads to cecal dysmotility, impaired secretion, and neuro-immune alterations. Together, these results suggest that the R451C mutation disrupts the gut-brain axis with GI dysfunction in autism.
first_indexed 2024-03-10T17:50:47Z
format Article
id doaj.art-520ff9eac8c641d186e95bfb648f0bd0
institution Directory Open Access Journal
issn 2045-2322
language English
last_indexed 2024-03-10T17:50:47Z
publishDate 2023-08-01
publisher Nature Portfolio
record_format Article
series Scientific Reports
spelling doaj.art-520ff9eac8c641d186e95bfb648f0bd02023-11-20T09:21:17ZengNature PortfolioScientific Reports2045-23222023-08-0113111810.1038/s41598-023-39555-yImpaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autismChalystha Yie Qin Lee0Gayathri K. Balasuriya1Madushani Herath2Ashley E. Franks3Elisa L. Hill-Yardin4School of Health and Biomedical Sciences, RMIT UniversityGraduate School of Medicine, Kobe UniversityDepartment of Pathology and Immunology, Baylor College of MedicineSchool of Life Sciences, La Trobe UniversitySchool of Health and Biomedical Sciences, RMIT UniversityAbstract Individuals with Autism Spectrum Disorder (ASD; autism) commonly present with gastrointestinal (GI) illness in addition to core diagnostic behavioural traits. The appendix, or cecum in mice, is important for GI homeostasis via its function as a key site for fermentation and a microbial reservoir. Even so, the role of the appendix and cecum in autism-associated GI symptoms remains uninvestigated. Here, we studied mice with an autism-associated missense mutation in the post-synaptic protein neuroligin-3 (Nlgn3 R451C ), which impacts brain and enteric neuronal activity. We assessed for changes in cecal motility using a tri-cannulation video-imaging approach in ex vivo preparations from wild-type and Nlgn3 R451C mice. We investigated cecal permeability and neurally-evoked secretion in wild-type and Nlgn3 R451C tissues using an Ussing chamber set-up. The number of cecal patches in fresh tissue samples were assessed and key immune populations including gut macrophages and dendritic cells were visualised using immunofluorescence. Nlgn3 R451C mice displayed accelerated cecal motor complexes and reduced cecal weight in comparison to wildtype littermates. Nlgn3 R451C mice also demonstrated reduced neurally-evoked cecal secretion in response to the nicotinic acetylcholine receptor agonist 1,1-dimethyl-4-phenylpiperazinium (DMPP), but permeability was unchanged. We observed an increase in the number of cecal patches in Nlgn3 R451C mice, however the cellular morphologies of key immune populations studied were not significantly altered. We show that the R451C nervous system mutation leads to cecal dysmotility, impaired secretion, and neuro-immune alterations. Together, these results suggest that the R451C mutation disrupts the gut-brain axis with GI dysfunction in autism.https://doi.org/10.1038/s41598-023-39555-y
spellingShingle Chalystha Yie Qin Lee
Gayathri K. Balasuriya
Madushani Herath
Ashley E. Franks
Elisa L. Hill-Yardin
Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
Scientific Reports
title Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
title_full Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
title_fullStr Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
title_full_unstemmed Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
title_short Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism
title_sort impaired cecal motility and secretion alongside expansion of gut associated lymphoid tissue in the nlgn3 r451c mouse model of autism
url https://doi.org/10.1038/s41598-023-39555-y
work_keys_str_mv AT chalysthayieqinlee impairedcecalmotilityandsecretionalongsideexpansionofgutassociatedlymphoidtissueinthenlgn3r451cmousemodelofautism
AT gayathrikbalasuriya impairedcecalmotilityandsecretionalongsideexpansionofgutassociatedlymphoidtissueinthenlgn3r451cmousemodelofautism
AT madushaniherath impairedcecalmotilityandsecretionalongsideexpansionofgutassociatedlymphoidtissueinthenlgn3r451cmousemodelofautism
AT ashleyefranks impairedcecalmotilityandsecretionalongsideexpansionofgutassociatedlymphoidtissueinthenlgn3r451cmousemodelofautism
AT elisalhillyardin impairedcecalmotilityandsecretionalongsideexpansionofgutassociatedlymphoidtissueinthenlgn3r451cmousemodelofautism