PUBLICATION

Signaling Events Downstream of AHR Activation That Contribute to Toxic Responses: The Functional Role of an AHR-Dependent Long Noncoding RNA ( slincR) Using the Zebrafish Model

Authors
Garcia, G.R., Shankar, P., Dunham, C.L., Garcia, A., La Du, J.K., Truong, L., Tilton, S.C., Tanguay, R.L.
ID
ZDB-PUB-181107-8
Date
2018
Source
Environmental health perspectives   126: 117002 (Journal)
Registered Authors
La Du, Jane K., Shankar, Prarthana, Tanguay, Robyn L., Tilton, Susan C.
Keywords
none
Datasets
GEO:GSE106131
MeSH Terms
  • Animals
  • Cartilage/abnormalities
  • Cartilage/drug effects
  • Embryo, Nonmammalian/abnormalities
  • Embryo, Nonmammalian/drug effects
  • Humans
  • Polychlorinated Dibenzodioxins/toxicity
  • Polycyclic Aromatic Hydrocarbons/toxicity
  • RNA, Long Noncoding/genetics
  • RNA, Long Noncoding/physiology*
  • Receptors, Aryl Hydrocarbon/physiology*
  • SOX9 Transcription Factor/biosynthesis*
  • SOX9 Transcription Factor/metabolism
  • Signal Transduction
  • Zebrafish/embryology
  • Zebrafish Proteins/metabolism
PubMed
30398377 Full text @ Environ. Health Perspect.
CTD
30398377
Abstract
A structurally diverse group of chemicals, including dioxins [e.g., 2,3,7,8-tetrachlorodibenzo- p-dioxin (TCDD)] and polycyclic aromatic hydrocarbons (PAHs), can xenobiotically activate the aryl hydrocarbon receptor (AHR) and contribute to adverse health effects in humans and wildlife. In the zebrafish model, repression of sox9b has a causal role in several AHR-mediated toxic responses, including craniofacial cartilage malformations; however, the mechanism of sox9b repression remains unknown. We previously identified a long noncoding RNA, sox9b long intergenic noncoding RNA ( slincR), which is increased (in an AHR-dependent manner) by multiple AHR ligands and is required for the AHR-activated repression of sox9b.
Using the zebrafish model, we aimed to enhance our understanding of the signaling events downstream of AHR activation that contribute to toxic responses by identifying: a) whether slincR is enriched on the sox9b locus, b) slincR's functional contributions to TCDD-induced toxicity, c) PAHs that increase slincR expression, and d) mammalian orthologs of slincR.
We used capture hybridization analysis of RNA targets (CHART), qRT-PCR, RNA sequencing, morphometric analysis of cartilage structures, and hemorrhaging screens.
The slincR transcript was enriched at the 5' untranslated region (UTR) of the sox9b locus. Transcriptome profiling and human ortholog analyses identified processes related to skeletal and cartilage development unique to TCDD-exposed controls, and angiogenesis and vasculature development unique to TCDD-exposed zebrafish that were injected with a splice-blocking morpholino targeting slincR. In comparison to TCDD exposed control morphants, slincR morphants exposed to TCDD resulted in abnormal cartilage structures and a smaller percentage of animals displaying the hemorrhaging phenotype. In addition, slincR expression was significantly increased in six out of the sixteen PAHs we screened.
Our study establishes that in zebrafish, slincR is recruited to the sox9b 5' UTR to repress transcription, can regulate cartilage development, has a causal role in the TCDD-induced hemorrhaging phenotype, and is up-regulated by multiple environmentally relevant PAHs. These findings have important implications for understanding the ligand-specific mechanisms of AHR-mediated toxicity. https://doi.org/10.1289/EHP3281.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping