All-trans retinoic acid induces reprogramming of canine dedifferentiated cells into neuron-like cells

PLoS One. 2020 Mar 31;15(3):e0229892. doi: 10.1371/journal.pone.0229892. eCollection 2020.

Abstract

The specification of cell identity depends on the exposure of cells to sequences of bioactive ligands. All-trans retinoic acid (ATRA) affects neuronal development in the early stage, and it is involved in neuronal lineage reprogramming. We previously established a fibroblast-like dedifferentiated fat cells (DFATs) derived from highly homogeneous mature adipocytes, which are more suitable for the study of cellular reprogramming. Canine cognitive dysfunction is similar to human cognitive dysfunction, suggesting that dogs could be a pathological and pharmacological model for human neuronal diseases. However, the effect of ATRA on neuronal reprogramming in dogs has remained unclear. Therefore, in this study, we investigated the effect of ATRA on the neuronal reprogramming of canine DFATs. ATRA induced the expression of neuronal marker mRNA/protein. The neuron-like cells showed Ca2+ influx with depolarization (50 mM KCl; 84.75 ± 4.05%) and Na+ channel activation (50 μM veratridine; 96.02 ± 2.02%). Optical imaging of presynaptic terminal activity and detection of neurotransmitter release showed that the neuron-like cells exhibited the GABAergic neuronal property. Genome-wide RNA-sequencing analysis shows that the transcriptome profile of canine DFATs is effectively reprogrammed towards that of cortical interneuron lineage. Collectively, ATRA can produce functional GABAergic cortical interneuron-like cells from canine DFATs, exhibiting neuronal function with > 80% efficiency. We further demonstrated the contribution of JNK3 to ATRA-induced neuronal reprogramming in canine DFATs. In conclusion, the neuron-like cells from canine DFATs could be a powerful tool for translational research in cell transplantation therapy, in vitro disease modeling, and drug screening for neuronal diseases.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Dedifferentiation / drug effects*
  • Cell Differentiation / drug effects
  • Cellular Reprogramming / drug effects
  • Dogs
  • Neurogenesis / drug effects*
  • Neurogenesis / genetics
  • Neurons / drug effects*
  • RNA, Messenger / genetics
  • Synapses / drug effects
  • Synapses / genetics
  • Tretinoin / pharmacology*

Substances

  • RNA, Messenger
  • Tretinoin

Grants and funding

This work was supported in part by a Grant-in-Aid for Scientific Research (#18K14594; RN) from the Ministry of Education, Science, Sports, and Culture of Japan (https://www.jsps.go.jp/j-grantsinaid/). This work was supported in part by a Nihon University Chairman of the Board of Trustees Grant (#2018-2020; TM) from Nihon University (http://www.nihon-u.ac.jp/). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.