PUBLICATION

Generation of novel genetic models to dissect resistance to thyroid hormone receptor alpha in zebrafish

Authors
Han, C.R., Holmsen, E., Carrington, B., Bishop, K., Zhu, Y.J., Starost, M., Meltzer, P., Sood, R., Liu, P., Cheng, S.Y.
ID
ZDB-PUB-200119-3
Date
2020
Source
Thyroid : official journal of the American Thyroid Association   30(2): 314-328 (Journal)
Registered Authors
Cheng, Sheue-yann, Han, Cho Rong, Sood, Raman
Keywords
none
MeSH Terms
  • Animals
  • Animals, Genetically Modified
  • Disease Models, Animal
  • Genes, erbA/genetics*
  • Growth Disorders/genetics
  • Growth Disorders/metabolism*
  • Growth Hormone/genetics
  • Growth Hormone/metabolism
  • Insulin-Like Growth Factor I/genetics
  • Insulin-Like Growth Factor I/metabolism
  • Thyroid Hormone Receptors alpha/genetics
  • Thyroid Hormone Receptors alpha/metabolism*
  • Thyroid Hormone Resistance Syndrome/genetics
  • Thyroid Hormone Resistance Syndrome/metabolism*
  • Zebrafish/genetics
  • Zebrafish/metabolism*
PubMed
31952464 Full text @ Thyroid
Abstract
Background: Patients with mutations of the thyroid hormone receptor alpha (THRA) gene show resistance to thyroid hormone alpha (RTHα). No amendable mouse models are currently available to elucidate deleterious effects of TRα1 mutants during early development. Zebrafish with transient suppressed expression by morpholino knockdown and ectopic expression of TRα1 mutants in the embryos have been reported. However, zebrafish with germline transmittable mutations have not been reported. The stable expression of thra mutants from embryos to adulthood facilitated the study of molecular actions of TRα1 mutants during development. Methods: In contrast to human and mice, the thra gene is duplicated in zebrafish, thraa, and thrab. Using CRISPR/Cas9-mediated targeted mutagenesis, we created dominant negative mutations in the two duplicated thra genes. We comprehensively analyzed the molecular and phenotypic characteristics of mutant fish during development. Results: Adult and juvenile homozygous thrab 1-bp ins (m/m) mutants exhibited severe growth retardation, but adult homozygous thraa 8-bp ins (m/m) mutants had very mild growth impairment. Expression of the growth hormone (gh1) and insulin-like growth factor 1 was markedly suppressed in homozygous thrab 1-bp ins (m/m) mutants. Decreased messenger RNA and protein levels of triiodothyronine-regulated keratin genes and inhibited keratinocyte proliferation resulted in hypoplasia of the epidermis in adult and juvenile homozygous thrab 1-bp ins (m/m) mutants, but not homozygous thraa 8-bp ins (m/m) mutants. RNA-seq analysis showed that homozygous thrab 1-bp ins (m/m) mutation had global impact on the functions of the adult pituitary. However, no morphological defects nor any changes in the expression of gh1 and keratin genes were observed in the embryos and early larvae. Thus, mutations of either the thraa or thrab gene did not affect initiation of embryogenesis. But the mutation of the thrab gene, but not the thraa gene, is detrimental in postlarval growth and skin development. Conclusions: The thra duplicated genes are essential to control temporal coordination in postlarval growth and development in a tissue-specific manner. We uncovered novel functions of the duplicated thra genes in zebrafish in development. These mutant zebrafish could be used as a model for further analysis of TRα1 mutant actions and for rapid screening of therapeutics for RTHα.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping