Abstract

The schizophrenia risk gene NRG1 controls the formation of excitatory and inhibitory synapses in cortical circuits. While the expression of different NRG1 isoforms occurs during development, adult neurons primarily express the CRD-NRG1 isoform characterized by a highly conserved intracellular domain (NRG1-ICD). We and others have demonstrated that Nrg1 intracellular signaling promotes dendrite elongation and excitatory connections during neuronal development. However, the role of Nrg1 intracellular signaling in adult neurons and pathological conditions remains largely unaddressed. Here, we investigated the role of Nrg1 intracellular signaling in neuroprotection and stroke. Our bioinformatic analysis revealed the evolutionary conservation of the NRG1-ICD and a decrease in NRG1 expression with age in the human frontal cortex. Hence, we first evaluated whether Nrg1 signaling may affect pathological hallmarks in an in vitro model of neuronal senescence; however, our data failed to reveal a role for Nrg1 in the activation of the stress-related pathway p38 MAPK and DNA damage. Previous studies demonstrated that the soluble EGF domain of Nrg1 alleviated brain ischemia, a pathological process involving the generation of free radicals, reactive oxygen species (ROS), and excitotoxicity. Hence, we tested the hypothesis that Nrg1 intracellular signaling could be neuroprotective in stroke. We discovered that Nrg1 expression significantly increased neuronal survival upon oxygen-glucose deprivation (OGD), an established in vitro model for stroke. Notably, the specific activation of Nrg1 intracellular signaling by expression of the Nrg1-ICD protected neurons from OGD. Additionally, time-lapse experiments confirmed that Nrg1 intracellular signaling increased the survival of neurons exposed to OGD. Finally, we investigated the relevance of Nrg1 intracellular signaling in stroke in vivo. Using viral vectors, we expressed the Nrg1-ICD in cortical neurons and subsequently challenged them by a focal hemorrhagic stroke; our data indicated that Nrg1 intracellular signaling improved neuronal survival in the infarcted area. Altogether, these data highlight Nrg1 intracellular signaling as neuroprotective upon ischemic lesion both in vitro and in vivo. Given the complexity of the neurotoxic effects of stroke and the involvement of various mechanisms, such as the generation of ROS, excitotoxicity, and inflammation, further studies are required to determine the molecular bases of the neuroprotective effect of Nrg1 intracellular signaling. In conclusion, our research highlights the stimulation of Nrg1 intracellular signaling as a promising target for cortical stroke treatment.

Highlights

  • The major synaptogenic protein Neuregulin 1 (Nrg1) controls the formation of excitatory and inhibitory synapses in the cortex [1,2,3]

  • Given the GFP tagging of Nrg1-ICD and Nrg1 full-length (Nrg1-FL), we directly quantified the fractions of surviving neurons expressing Nrg1 as compared to GFP-expressing controls. We discovered that both the expression of Nrg1-FL and the specific activation of Nrg1 intracellular signaling improved the survival of neurons exposed to oxygen-glucose deprivation (OGD) when compared to controls (Figure 5)

  • Nrg1 is a major regulator of cortical circuit development; we know relatively little regarding its role in mature neurons

Read more

Summary

Introduction

The major synaptogenic protein Neuregulin 1 (Nrg1) controls the formation of excitatory and inhibitory synapses in the cortex [1,2,3]. The Nrg gene encodes more than 20 isoforms grouped into six types of proteins. While the Nrg extracellular domain displays high variability, all isoforms contain the epidermal growth factor- (EGF-) like domain located in the extracellular domain that is necessary and sufficient for activation of the ErbB4 receptor. Many Nrg isoforms possess common transmembrane and intracellular domains that elicit Nrg intracellular (or noncanonical) signaling [1]. To Notch signaling, the transmembrane domain of Nrg is first cleaved by alpha- or beta-secretases and subsequently by gamma-secretase [4, 5]. Neuronal activity or binding to the ErbB4 receptor triggers this regulated processing [2, 6,7,8], releasing the resulting intracellular domain of Nrg (Nrg1-ICD) for translocation to the nucleus

Methods
Results
Conclusion
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