DDX3 modulates neurite development via translationally activating an RNA regulon involved in Rac1 activation

HH Chen, HI Yu, WY Tarn - Journal of Neuroscience, 2016 - Soc Neuroscience
HH Chen, HI Yu, WY Tarn
Journal of Neuroscience, 2016Soc Neuroscience
The RNA helicase DDX3 is a component of neuronal granules, and its gene mutations are
linked to intellectual disability (ID). Here we demonstrate that DDX3 depletion in neurons
impairs neurite development by downregulating Rac1 level and activation. Moreover, DDX3
activates the translation of functionally coherent mRNAs involved in Rac1 activation
including Rac1. Among the DDX3 regulon, Prkaca encodes the catalytic subunit of PKA, a
potential activator of Rac1 in neurons. DDX3-modulated PKAcα and Rac1 expression tunes …
The RNA helicase DDX3 is a component of neuronal granules, and its gene mutations are linked to intellectual disability (ID). Here we demonstrate that DDX3 depletion in neurons impairs neurite development by downregulating Rac1 level and activation. Moreover, DDX3 activates the translation of functionally coherent mRNAs involved in Rac1 activation including Rac1. Among the DDX3 regulon, Prkaca encodes the catalytic subunit of PKA, a potential activator of Rac1 in neurons. DDX3-modulated PKAcα and Rac1 expression tunes the strength of PKA–Rac1 signaling and thereby contributes to neurite outgrowth and dendritic spine formation. Inhibition of DDX3 activity or expression in neonatal mice impaired dendritic outgrowth and spine formation of hippocampal neurons, echoing neuronal deficits underling DDX3 mutation-associated ID. Finally, we provide evidence that DDX3 activates local protein synthesis through a 5′ UTR-dependent mechanism in neurons. The novel DDX3 regulon may conduct a spatial and temporal control of Rac1 signaling to regulate neurite development.
SIGNIFICANCE STATEMENT DDX3X mutations are linked to intellectual disability (ID). We provide first evidence that DDX3 is required for neurite outgrowth and dendritic spine formation in vitro and in vivo. We identified a DDX3 regulon constituting functionally cohesive mRNAs involved in Rac1 signaling, which contributes to DDX3-modulated neurite development. Inhibition or ablation of DDX3 in vivo shortened neurite lengths and impaired dendritic spine formation in hippocampal neurons, reflecting the prevalence of ID-associated DDX3X mutations in the helicase domain. Mechanistically, DDX3 activates local protein synthesis of mRNAs sharing similar 5′ UTR structures and therefore controls Rac1 signaling strength in neurites.
Soc Neuroscience