Abstract

BackgroundThe participation of microglia in CNS development and homeostasis indicate that these cells are pivotal for the regeneration that occurs after demyelination. The clearance of myelin debris and the inflammatory-dependent activation of local oligodendrocyte progenitor cells in a demyelinated lesion is dependent on the activation of M2c microglia, which display both phagocytic and healing functions. Emerging interest has been raised about the role of Wnt/β-catenin signaling in oligodendrogenesis and myelination. Besides, cytokines and growth factors released by microglia can control the survival, proliferation, migration, and differentiation of neural stem cells (NSCs), contributing to remyelination through the oligodendrocyte specification of this adult neurogenic niche.MethodsTMEV-IDD model was used to study the contribution of dorsal SVZ stem cells to newly born oligodendrocytes in the corpus callosum following demyelination by (i) en-face dorsal SVZ preparations; (ii) immunohistochemistry; and (iii) cellular tracking. By RT-PCR, we analyzed the expression of Wnt proteins in demyelinated and remyelinating corpus callosum. Using in vitro approaches with microglia cultures and embryonic NSCs, we studied the role of purified myelin, Wnt proteins, and polarized microglia-conditioned medium to NSC proliferation and differentiation. One-way ANOVA followed by Bonferroni’s post-hoc test, or a Student’s t test were used to establish statistical significance.ResultsThe demyelination caused by TMEV infection is paralleled by an increase in B1 cells and pinwheels in the dorsal SVZ, resulting in the mobilization of SVZ proliferative progenitors and their differentiation into mature oligodendrocytes. Demyelination decreased the gene expression of Wnt5a and Wnt7a, which was restored during remyelination. In vitro approaches show that Wnt3a enhances NSC proliferation, while Wnt7a and myelin debris promotes oligodendrogenesis from NSCs. As phagocytic M2c microglia secrete Wnt 7a, their conditioned media was found to induce Wnt/β-Catenin signaling in NSCs promoting an oligodendroglial fate.ConclusionsWe define here the contribution of microglia to Wnt production depending on their activation state, with M1 microglia secreting the Wnt5a protein and M2c microglia secreting Wnt7a. Collectively, our data reveal the role of reparative microglia in NSC oligodendrogenesis with the involvement of Wnt7a.

Highlights

  • The participation of microglia in central nervous system (CNS) development and homeostasis indicate that these cells are pivotal for the regeneration that occurs after demyelination

  • The secretion of pro-inflammatory cytokines by microglia, including IL-1β, IL-6, TNF-α, and IFN-γ, promotes neurogenesis and oligodendrogenesis in the subventricular zone (SVZ), and in Conclusions In conclusion, our data suggest that the myelin debris generated during corpus callosum demyelination can directly affect the specification and differentiation of multipotential cells in the dorsal SVZ

  • Stem cells generate Oligodendrocyte intermediate progenitor cell (oIPC) that are committed to the oligodendroglial lineage and that migrate to the lesion site, where they differentiate into mature remyelinating oligodendrocytes

Read more

Summary

Introduction

The participation of microglia in CNS development and homeostasis indicate that these cells are pivotal for the regeneration that occurs after demyelination. Cytokines and growth factors released by microglia can control the survival, proliferation, migration, and differentiation of neural stem cells (NSCs), contributing to remyelination through the oligodendrocyte specification of this adult neurogenic niche. The SVZ is a specialized niche in the walls of the lateral ventricles of the forebrain that contents multipotent cells known as neural stem cells (NSCs), cells that can self-renew and differentiate into neurons, astrocytes, or oligodendrocytes [3, 4]. Adult NSCs are slowly dividing progenitors referred to as B1 cells, and they generate transit amplifying progenitors (type-C cells) that in turns differentiate into neuroblasts (type-A cells). These neuroblasts generally migrate to the olfactory bulb via the rostral migratory stream (RMS) to replace local interneurons [5]

Methods
Results
Discussion
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