Abstract

Overexpression of Dual‐specificity tyrosine‐phosphorylated regulated kinase 1A (DYRK1A), located on human chromosome 21, may alter molecular processes linked to developmental deficits in Down syndrome (DS). Trisomic DYRK1A is a rational therapeutic target, and although reductions in Dyrk1a genetic dosage have shown improvements in trisomic mouse models, attempts to reduce Dyrk1a activity by pharmacological mechanisms and correct these DS‐associated phenotypes have been largely unsuccessful. Epigallocatechin‐3‐gallate (EGCG) inhibits DYRK1A activity in vitro and this action has been postulated to account for improvement of some DS‐associated phenotypes that have been reported in preclinical studies and clinical trials. However, the beneficial effects of EGCG are inconsistent and there is no direct evidence that any observed improvement actually occurs through Dyrk1a inhibition. Inconclusive outcomes likely reflect a lack of knowledge about the tissue‐specific patterns of spatial and temporal overexpression and elevated activity of Dyrk1a that may contribute to emerging DS traits during development. Emerging evidence indicates that Dyrk1a expression varies over the life span in DS mouse models, yet preclinical therapeutic treatments targeting Dyrk1a have largely not considered these developmental changes. Therapies intended to improve DS phenotypes through normalizing trisomic Dyrk1a need to optimize the timing and dose of treatment to match the spatiotemporal patterning of excessive Dyrk1a activity in relevant tissues. This will require more precise identification of developmental periods of vulnerability to enduring adverse effects of elevated Dyrk1a, representing the concurrence of increased Dyrk1a expression together with hypothesized tissue‐specific‐sensitive periods when Dyrk1a regulates cellular processes that shape the long‐term functional properties of the tissue. Future efforts targeting inhibition of trisomic Dyrk1a should identify these putative spatiotemporally specific developmental sensitive periods and determine whether normalizing Dyrk1a activity then can lead to improved outcomes in DS phenotypes.

Highlights

  • Trisomy of human chromosome 21 (Hsa21) results in myriad phenotypes including cognitive impairment, cardiac abnormalities, and craniofacial features collectively referred to as Down syndrome (DS) (OMIM: 190685)

  • The temporal and spatial expression of Dyrk1a protein and kinase activity in trisomic mouse models must be characterized in greater detail, and altered regulation of pathways of downstream targets of Dyrk1a needs to be identified in those tissues during periods of excessive Dyrk1a activity

  • This can guide efforts to identify cellular and molecular signaling processes regulated by Dyrk1a that are disrupted by excessive Dyrk1a activity

Read more

Summary

REVIEW ARTICLE

Targeting trisomic treatments: optimizing Dyrk1a inhibition to improve Down syndrome deficits.

Introduction
The Role of DS Mouse Models in Finding Therapies
Hippocampus Striatum Global Global
ROA Length Improved deficits No effect
DW DW DW DW Chow DW DW DW DW
None None None
Findings
Conclusions
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call