PUBLICATION

Intestinal dysmotility in a zebrafish (Danio rerio) shank3a;shank3b mutant model of autism

Authors
James, D.M., Kozol, R.A., Kajiwara, Y., Wahl, A.L., Storrs, E.C., Buxbaum, J.D., Klein, M., Moshiree, B., Dallman, J.E.
ID
ZDB-PUB-190209-15
Date
2019
Source
Molecular autism   10: 3 (Journal)
Registered Authors
Dallman, Julia, James, David
Keywords
Digestive transit, Enteroendocrine, Peristaltic rate, Phelan-McDermid syndrome
MeSH Terms
  • Animals
  • Autistic Disorder/genetics*
  • Autistic Disorder/physiopathology
  • Enteric Nervous System/cytology
  • Enteric Nervous System/metabolism
  • Enteroendocrine Cells/metabolism
  • Gastrointestinal Motility*
  • Intestinal Mucosa/metabolism
  • Intestines/cytology
  • Intestines/growth & development
  • Intestines/physiology
  • Mutation
  • Nerve Tissue Proteins/genetics*
  • Neurons/metabolism
  • Protein Isoforms/genetics
  • Protein Isoforms/metabolism
  • Serotonin/metabolism
  • Zebrafish
  • Zebrafish Proteins/genetics*
PubMed
30733854 Full text @ Mol Autism
Abstract
Autism spectrum disorder (ASD) is currently estimated to affect more than 1% of the world population. For people with ASD, gastrointestinal (GI) distress is a commonly reported but a poorly understood co-occurring symptom. Here, we investigate the physiological basis for GI distress in ASD by studying gut function in a zebrafish model of Phelan-McDermid syndrome (PMS), a condition caused by mutations in the SHANK3 gene.
To generate a zebrafish model of PMS, we used CRISPR/Cas9 to introduce clinically related C-terminal frameshift mutations in shank3a and shank3b zebrafish paralogues (shank3abΔC). Because PMS is caused by SHANK3 haploinsufficiency, we assessed the digestive tract (DT) structure and function in zebrafish shank3abΔC+/- heterozygotes. Human SHANK3 mRNA was then used to rescue DT phenotypes in larval zebrafish.
Significantly slower rates of DT peristaltic contractions (p < 0.001) with correspondingly prolonged passage time (p < 0.004) occurred in shank3abΔC+/- mutants. Rescue injections of mRNA encoding the longest human SHANK3 isoform into shank3abΔC+/- mutants produced larvae with intestinal bulb emptying similar to wild type (WT), but still deficits in posterior intestinal motility. Serotonin-positive enteroendocrine cells (EECs) were significantly reduced in both shank3abΔC+/- and shank3abΔC-/- mutants (p < 0.05) while enteric neuron counts and overall structure of the DT epithelium, including goblet cell number, were unaffected in shank3abΔC+/- larvae.
Our data and rescue experiments support mutations in SHANK3 as causal for GI transit and motility abnormalities. Reductions in serotonin-positive EECs and serotonin-filled ENS boutons suggest an endocrine/neural component to this dysmotility. This is the first study to date demonstrating DT dysmotility in a zebrafish single gene mutant model of ASD.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping