Abstract

Schizophrenia (SZ) is a neurodevelopmental genetic disorder in which maternal immune activation (MIA) and increased tumor necrosis factor-α (TNF-α) may contribute. Previous studies using iPSC-derived cerebral organoids and neuronal cells demonstrated developmental malformation and transcriptional dysregulations, including TNF receptors and their signaling genes, common to SZ patients with diverse genetic backgrounds. In the present study, we examined the significance of the common TNF receptor dysregulations by transiently exposing cerebral organoids from embryonic stem cells (ESC) and from representative control and SZ patient iPSCs to TNF. In control iPSC organoids, TNF produced malformations qualitatively similar in, but generally less pronounced than, the malformations of the SZ iPSC-derived organoids. TNF and SZ alone disrupted subcortical rosettes and dispersed proliferating Ki67+ neural progenitor cells (NPC) from the organoid ventricular zone (VZ) into the cortical zone (CZ). In the CZ, the absence of large ramified pan-Neu+ neurons coincided with loss of myelinated neurites despite increased cortical accumulation of O4+ oligodendrocytes. The number of calretinin+ interneurons increased; however, they lacked the preferential parallel orientation to the organoid surface. SZ and SZ+TNF affected fine cortical and subcortical organoid structure by replacing cells with extracellular matrix (ECM)-like fibers The SZ condition increased developmental vulnerability to TNF, leading to more pronounced changes in NPC, pan-Neu+ neurons, and interneurons. Both SZ- and TNF-induced malformations were associated with the loss of nuclear (n)FGFR1 form in the CZ and its upregulation in deep IZ regions, while in earlier studies blocking nFGFR1 reproduced cortical malformations observed in SZ. Computational analysis of ChiPseq and RNAseq datasets shows that nFGFR1 directly targets neurogenic, oligodendrogenic, cell migration, and ECM genes, and that the FGFR1-targeted TNF receptor and signaling genes are overexpressed in SZ NPC. Through these changes, the developing brain with the inherited SZ genome dysregulation may suffer increased vulnerability to TNF and thus, MIA.

Highlights

  • Schizophrenia (SZ) is a neurodevelopmental psychotic disorder associated with fine changes in the brain cortical structure (Harrison, 1999)

  • This study aimed to explore a potential interaction between genetic disease modeled in SZ induced pluripotent stem cell (iPSC) cerebral organoids and in maternal immune activation (MIA) modeled by organoid exposure to the cytokine TNF

  • We recently reported that dysregulation of nuclear FGFR1 (nFGFR1) in the developing neural progenitor cells (NPC) progeny directly affected genes representing additional ontogenic processes, including cell migration and extracellular matrix-like (ECM), all of which were disrupted in SZ and in organoids exposed to TNF (Stachowiak et al, 2017)

Read more

Summary

Introduction

Schizophrenia (SZ) is a neurodevelopmental psychotic disorder associated with fine changes in the brain cortical structure (Harrison, 1999) Both genetic factors, as well as physical and psychosocial environmental factors play a role in the development of SZ. In cellular development, controlling signals propagate through diverse pathways to their end-point transcription factors. In parallel, these signals are fed forward by nFGFR1 directly to CREB binding protein (CBP), allowing genes to respond in a coordinated manner (Stachowiak and Stachowiak, 2016). Evidence indicates that in SZ, mutations of genes in diverse signaling pathways integrated by nFGFR1 disrupt the INFS function and neural development (Narla et al, 2018). Whether pathological environmental factors that contribute to SZ affect nFGFR1, developmental function is unknown

Objectives
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