Affiliations 

  • 1 Department of Applied Biology, College of Sciences, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
  • 2 Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543 Singapore
  • 3 Department of Biomedical and Clinical Sciences (BKV), Linköping University, P.O. Box 581 83, Linköping, Sweden
  • 4 Research Institute of Science and Engineering, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
  • 5 Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
  • 6 Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
Cell Biosci, 2020;10:126.
PMID: 33133516 DOI: 10.1186/s13578-020-00487-z

Abstract

Introduction: Mesenchymal stem cells (MSCs) isolated from bone marrow have different developmental origins, including neural crest. MSCs can differentiate into neural progenitor-like cells (NPCs) under the influence of bFGF and EGF. NPCs can terminally differentiate into neurons that express beta-III-tubulin and elicit action potential. The main aim of the study was to identify key genetic markers involved in differentiation of MSCs into NPCs through transcriptomic analysis.

Method: Total RNA was isolated from MSCs and MSCs-derived NPCs followed by cDNA library construction for transcriptomic analysis. Sample libraries that passed the quality and quantity assessments were subjected to high throughput mRNA sequencing using NextSeq®500. Differential gene expression analysis was performed using the DESeq2 R package with MSC samples being a reference group. The expression of eight differentially regulated genes was counter validated using real-time PCR.

Results: In total, of the 3,252 differentially regulated genes between MSCs and NPCs with two or more folds, 1,771 were upregulated genes, whereas 1,481 were downregulated in NPCs. Amongst these differential genes, 104 transcription factors were upregulated, and 45 were downregulated in NPCs. Neurogenesis related genes were upregulated in NPCs and the main non-redundant gene ontology (GO) terms enriched in NPCs were the autonomic nervous system, cell surface receptor signalling pathways), extracellular structure organisation, and programmed cell death. The main non-redundant GO terms enriched in MSCs included cytoskeleton organisation cytoskeleton structural constituent, mitotic cell cycle), and the mitotic cell cycle process Gene set enrichment analysis also confirmed cell cycle regulated pathways as well as Biocarta integrin pathway were upregulated in MSCs. Transcription factors enrichment analysis by ChEA3 revealed Foxs1 and HEYL, amongst the top five transcription factors, inhibits and enhances, respectively, the NPCs differentiation of MSCs.

Conclusions: The vast differences in the transcriptomic profiles between NPCs and MSCs revealed a set of markers that can identify the differentiation stage of NPCs as well as provide new targets to enhance MSCs differentiation into NPCs.

* Title and MeSH Headings from MEDLINE®/PubMed®, a database of the U.S. National Library of Medicine.