Skip to main content

GENERAL COMMENTARY article

Front. Cell Dev. Biol., 28 May 2021
Sec. Stem Cell Research

Commentary: In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates

\nXiaoqin ZhangXiaoqin ZhangFenghua ChenFenghua ChenYoucui Wang
Youcui Wang*
  • Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China

A Commentary on
In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates

by Ge, L-J., Yang, F-H., Li, W., Wang, T., Lin, Y., Feng, J., et al. (2020). Front. Cell Dev. Biol. 8:590008. doi: 10.3389/fcell.2020.590008

Introduction

The loss of neurons is the most common cause of nervous system disorders, such as stroke, spinal cord injury (SCI), and neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Given that neurons cannot divide to regenerate themselves, together with the fact that only a small number of new neurons derived from external cell transplantation can survive in vivo (Goldman, 2016), few neuroregenerative strategies have succeeded in adult mammalian brains. In order to avoid the limitations of cell transplantation therapy, such as ethical issues and transplantation rejection (Chen et al., 2019), there are research groups that have developed a technology that allows reprogramming of astrocytes into functional new neurons in situ. For example, the single neural transcription factor NeuroD1-based gene therapy can successfully reprogram astrocytes into functional neurons in AD mouse brains (Guo et al., 2014), damaged spinal cord (Puls et al., 2020), and stroke mouse brains, as well as promote functional recovery in a mouse stroke model (Chen et al., 2020). Besides, astrocytes in the striatum of HD mice can be directly converted into GABAergic medium spinal neurons by co-overexpressing NeuroD1 and Dlx2, leading to motor function recovery and longer life spans (Wu et al., 2020). Three groups have shown that astrocytes also can be converted into dopaminergic neurons, which led to behavioral improvement in a mouse PD model by highly expressing three transcription factors, NeuroD1, Ascl1, and Lmx1a, and the microRNA, miR218, collectively referred to as NeAL218 (Fyfe, 2017; Rivetti di Val Cervo et al., 2017), or by depleting an RNA-binding protein called PTB (Arenas, 2020; Qian et al., 2020; Zhou et al., 2020). Additionally, in an adult rat model of SCI, astrocytes can be reprogrammed into neurons by overexpressing SOX2, leading to motor function recovery (Su et al., 2014), or into neural stem cells by highly expressing Zfp521 (Zarei-Kheirabadi et al., 2019). Notably, it has been confirmed that human astrocytes can be successfully converted into neurons or neuroblasts in vitro (Corti et al., 2012; Ghasemi-Kasman et al., 2015; Zhang et al., 2015; Li et al., 2016; Rivetti di Val Cervo et al., 2017; Yin et al., 2019; Qian et al., 2020). These studies provide a potential alternative approach to regenerate functional new neurons in the central nervous system of adult mammals by directly reprogramming glial cells in situ into neurons.

Advantages of Neurod1-Mediated Astrocyte-To-Neuron Conversion For Evaluating Therapeutic Treatment After Stroke In Adult Non-Human Primates

Recently, Chen's research group firstly reported that an in situ neuronal regeneration approach using AAV NeuroD1-based gene therapy could repair damaged brains in adult non-human primates (NHPs) with ischemic stroke (Ge et al., 2020). These findings were based on earlier successful investigations in vitro and in vivo (Guo et al., 2014; Brulet et al., 2017; Chen et al., 2020; Wu et al., 2020; Zhang et al., 2020).

The results have shown that high expression of a neural transcription factor NeuroD1 in astrocytes can successfully reprogram nearly 90% of infected astrocytes into neurons in the monkey cortex following ischemic stroke, and those neurons could survive over 1 year. The neurons from NeuroD1-mediated astrocyte-to-neuron (AtN) conversion displayed Tbr1+ cortical neuron identity and were mostly located in the monkey gray matter (Ge et al., 2020), which is entirely consistent with earlier findings in rodent ischemic stroke models (Chen et al., 2020; Liu et al., 2020). Interestingly, the number and intrinsic proliferative property of astrocytes in the converted areas were not changed compared with those in the control side.

Moreover, it was extremely attractive that the NeuroD1-mediated AtN conversion not only significantly increased the cortical neuronal density, dendritic marker, and synaptic marker levels in the ischemic injured areas of the monkey cortex but also improved the microenvironment, including reducing the number of reactive microglia and macrophages (Ge et al., 2020). The improvement of the inflammatory microenvironment was beneficial for the survival of parvalbumin-positive GABAergic interneurons that were badly damaged by ischemic injury (Povysheva et al., 2019). Another interesting finding different from that in rodent animal models was that the expression of NeuroD1 was significantly decreased in newly regenerated neurons after 6 months of AAV injection, which may be beneficial in future clinical trials (Ge et al., 2020). Furthermore, this study reported that this treatment had a broad time window from 10 to 30 days following ischemic stroke, and the cell conversion effect is long-lasting, ranging from 2 months to 1 year after viral infection (Ge et al., 2020), suggesting that the NeuroD1-mediated AtN conversion therapy might be a potential approach for neural repair in NHP brain.

Discussion

Many clinical trials of treatment for cerebral ischemia based on preclinical parameters obtained from rodent animal models have failed in the past (Turner et al., 2013), because the successful discoveries in rodent animal models may not be replicated in primates. The main reason for such problems is that the human brain is largely different from rodent brain, while the brains of large animal models, such as monkeys, are more similar to those of humans (Modo et al., 2018). Therefore, in order to confirm the validity of in vivo astrocyte conversion in human clinical trials, it is very important and necessary to first test such findings in adult NHP models, such as in monkeys. Previously, acute NHP stroke models often were used to test drug effects, and the drug administration time was typically within a few hours after stroke (Takamatsu et al., 2001; Cook and Tymianski, 2012, Cook et al., 2017). In this study, a focal stroke model with a very low mortality rate was employed through intracranial injection of endothelin-1 to induce blood vessel constriction in the motor cortex of rhesus macaque monkeys. This study demonstrated that in situ NeuroD1-mediated AtN conversion therapy had a broad time window from 10 to 30 days following ischemic stroke (Ge et al., 2020).

Nevertheless, in this study, some neuronal functions of converted neurons were not investigated, such as neuronal excitability, production, and release neurotransmitter. Furthermore, there is a question as to why and how NeuroD1-mediated AtN conversion could reduce the number of microglia and macrophages; unfortunately, the mechanisms were not investigated, and further studies will be required. However, there is mounting evidence that the expression of NeuroD1 is often high during early brain development and is lower in the adult brain (Pataskar et al., 2016); further studies are needed to clarify the mechanism of the significant decrease in NeuroD1 expression following AtN conversion in the monkey cortex over time. There was no evidence about a correlation with behavioral improvement in adult NHPs after stroke, and to our knowledge, neuronal recovery at the cellular level needs to be accompanied by behavioral improvement.

Taken together, this is the first report about successful application of NeuroD1-mediated AtN conversion technology in NHP models, which might fill some gaps between in vivo rodent models and in vitro human astrocyte culture models, as well as take an important step toward future clinical trials using such technology for nervous system diseases with neuronal loss.

Author Contributions

XZ and FC wrote the first draft. YW reviewed and critiqued the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by grants from the National Natural Science Foundation of China (31900745), Shandong Provincial Natural Science Foundation (ZR2017BC031), and Special Fund for Youth of Applied Foundational Research Program of Qingdao (No. 18-2-2-44-jch).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Arenas, E. (2020). Method to combat Parkinson's disease by astrocyte-to-neuron conversion. Nature 582, 489–490. doi: 10.1038/d41586-020-01817-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Brulet, R., Matsuda, T., Zhang, L., Miranda, C., Giacca, M., Kaspar, B. K., et al. (2017). NEUROD1 instructs neuronal conversion in non-reactive astrocytes. Stem Cell Rep. 8, 1506–1515. doi: 10.1016/j.stemcr.2017.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, W., Huang, Q., Ma, S., and Li, M. (2019). Progress in dopaminergic cell replacement and regenerative strategies for Parkinson's disease. ACS Chem. Neurosci. 10, 839–851. doi: 10.1021/acschemneuro.8b00389

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y. C., Ma, N. X., Pei, Z. F., Wu, Z., Do-Monte, F. H., Keefe, S., et al. (2020). A NeuroD1 AAV-based gene therapy for functional brain repair after ischemic injury through in vivo astrocyte-to-neuron conversion. Mol. Ther. 28, 217–234. doi: 10.1016/j.ymthe.2019.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Cook, D. J., Nguyen, C., Chun, H. N. L., Llorente, I., Chiu, A. S., Machnicki, M., et al. (2017). Hydrogel-delivered brain-derived neurotrophic factor promotes tissue repair and recovery after stroke. J. Cereb. Blood Flow Metab. 37, 1030–1045. doi: 10.1177/0271678X16649964

PubMed Abstract | CrossRef Full Text | Google Scholar

Cook, D. J., and Tymianski, M. (2012). Nonhuman primate models of stroke for translational neuroprotection research. Neurotherapeutics 9, 371–379. doi: 10.1007/s13311-012-0115-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Corti, S., Nizzardo, M., Simone, C., Falcone, M., Donadoni, C., Salani, S., et al. (2012). Direct reprogramming of human astrocytes into neural stem cells and neurons. Exp. Cell Res. 318, 1528–1541. doi: 10.1016/j.yexcr.2012.02.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Fyfe, I. (2017). Parkinson disease: in situ astrocyte reprogramming - simpler cell replacement therapy in PD? Nat. Rev. Neurol. 13, 318–319. doi: 10.1038/nrneurol.2017.60

PubMed Abstract | CrossRef Full Text | Google Scholar

Ge, L. J., Yang, F. H., Li, W., Wang, T., Lin, Y., Feng, J., et al. (2020). In vivo neuroregeneration to treat ischemic stroke through NeuroD1 AAV-based gene therapy in adult non-human primates. Front. Cell Dev. Biol. 8:590008. doi: 10.3389/fcell.2020.590008

PubMed Abstract | CrossRef Full Text | Google Scholar

Ghasemi-Kasman, M., Hajikaram, M., Baharvand, H., and Javan, M. (2015). MicroRNA-mediated in vitro and in vivo direct conversion of astrocytes to neuroblasts. PLoS ONE 10:e0127878. doi: 10.1371/journal.pone.0127878

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldman, S. A. (2016). Stem and progenitor cell-based therapy of the central nervous system: hopes, hype, and wishful thinking. Cell Stem Cell 18, 174–188. doi: 10.1016/j.stem.2016.01.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Z., Zhang, L., Wu, Z., Chen, Y., Wang, F., and Chen, G. (2014). In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer's disease model. Cell Stem Cell 14, 188–202. doi: 10.1016/j.stem.2013.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Kozielski, K., Cheng, Y. H., Liu, H., Zamboni, C. G., Green, J., et al. (2016). Nanoparticle-mediated conversion of primary human astrocytes into neurons and oligodendrocytes. Biomater. Sci. 4, 1100–1112. doi: 10.1039/c6bm00140h

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, M. H., Li, W., Zheng, J. J., Xu, Y. G., He, Q., and Chen, G. (2020). Differential neuronal reprogramming induced by NeuroD1 from astrocytes in grey matter versus white matter. Neural Regen. Res. 15, 342–351. doi: 10.4103/1673-5374.265185

PubMed Abstract | CrossRef Full Text | Google Scholar

Modo, M. M., Jolkkonen, J., Zille, M., and Boltze, J. (2018). Future of animal modeling for poststroke tissue repair. Stroke 49, 1099–1106. doi: 10.1161/strokeaha.117.018293

PubMed Abstract | CrossRef Full Text | Google Scholar

Pataskar, A., Jung, J., Smialowski, P., Noack, F., Calegari, F., Straub, T., et al. (2016). NeuroD1 reprograms chromatin and transcription factor landscapes to induce the neuronal program. EMBO J. 35, 24–45. doi: 10.15252/embj.201591206

PubMed Abstract | CrossRef Full Text | Google Scholar

Povysheva, N., Nigam, A., Brisbin, A. K., Johnson, J. W., and Barrionuevo, G. (2019). Oxygen-glucose deprivation differentially affects neocortical pyramidal neurons and parvalbumin-positive interneurons. Neuroscience 412, 72–82. doi: 10.1016/j.neuroscience. 2019.05.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Puls, B., Ding, Y., Zhang, F., Pan, M., Lei, Z., Pei, Z., et al. (2020). Regeneration of functional neurons after spinal cord injury via in situ NeuroD1-mediated astrocyte-to-neuron conversion. Front. Cell Dev. Biol. 8:591883. doi: 10.3389/fcell.2020.591883

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, H., Kang, X., Hu, J., Zhang, D., Liang, Z., Meng, F., et al. (2020). Reversing a model of Parkinson's disease with in situ converted nigral neurons. Nature 582, 550–556. doi: 10.1038/s41586-020-2388-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Rivetti di Val Cervo, P., Romanov, R. A., Spigolon, G., Masini, D., Martín-Montañez, E., Toledo, E. M., et al. (2017). Induction of functional dopamine neurons from human astrocytes in vitro and mouse astrocytes in a Parkinson's disease model. Nat. Biotechnol. 35, 444–452. doi: 10.1038/nbt.3835

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, Z., Niu, W., Liu, M. L., Zou, Y., and Zhang, C. L. (2014). In vivo conversion of astrocytes to neurons in the injured adult spinal cord. Nat. Commun. 5:3338. doi: 10.1038/ncomms4338

PubMed Abstract | CrossRef Full Text | Google Scholar

Takamatsu, H., Tsukada, H., Noda, A., Kakiuchi, T., Nishiyama, S., Nishimura, S., et al. (2001). FK506 attenuates early ischemic neuronal death in a monkey model of stroke. J. Nucl. Med. 42, 1833–1840.

PubMed Abstract | Google Scholar

Turner, R. C., Dodson, S. C., Rosen, C. L., and Huber, J. D. (2013). The science of cerebral ischemia and the quest for neuroprotection: navigating past failure to future success. J. Neurosurg. 118, 1072–1085. doi: 10.3171/2012.11.jns12408

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Z., Parry, M., Hou, X. Y., Liu, M. H., Wang, H., Cain, R., et al. (2020). Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington's disease. Nat. Commun. 11:1105. doi: 10.1038/s41467-020-14855-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, J. C., Zhang, L., Ma, N. X., Wang, Y., Lee, G., Hou, X. Y., et al. (2019). Chemical conversion of human fetal astrocytes into neurons through modulation of multiple signaling pathways. Stem Cell Rep. 12, 488–501. doi: 10.1016/j.stemcr.2019.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Zarei-Kheirabadi, M., Hesaraki, M., Kiani, S., and Baharvand, H. (2019). In vivo conversion of rat astrocytes into neuronal cells through neural stem cells in injured spinal cord with a single zinc-finger transcription factor. Stem Cell Res. Ther. 10:380. doi: 10.1186/s13287-019-1448-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Lei, Z., Guo, Z., Pei, Z., Chen, Y., Zhang, F., et al. (2020). Development of neuroregenerative gene therapy to reverse glial scar tissue back to neuron-enriched tissue. Front. Cell Neurosci. 14:594170. doi: 10.3389/fncel.2020.594170

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Yin, J. C., Yeh, H., Ma, N. X., Lee, G., Chen, X. A., et al. (2015). Small molecules efficiently reprogram human astroglial cells into functional neurons. Cell Stem Cell 17, 735–747. doi: 10.1016/j.stem.2015.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, H., Su, J., Hu, X., Zhou, C., Li, H., Chen, Z., et al. (2020). Glia-to-neuron conversion by CRISPR-CasRx alleviates symptoms of neurological disease in mice. Cell 181, 590.e16–603.e16. doi: 10.1016/j.cell.2020.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: NeuroD1, astrocyte-to-neuron, non-human primates, in vivo, stroke

Citation: Zhang X, Chen F and Wang Y (2021) Commentary: In vivo Neuroregeneration to Treat Ischemic Stroke Through NeuroD1 AAV-Based Gene Therapy in Adult Non-human Primates. Front. Cell Dev. Biol. 9:648020. doi: 10.3389/fcell.2021.648020

Received: 31 December 2020; Accepted: 12 April 2021;
Published: 28 May 2021.

Edited by:

Erdal Karaoz, Istinye University, Turkey

Reviewed by:

Juergen Hescheler, University of Cologne, Germany
Ozgur Tataroglu, University of Istinye, Turkey

Copyright © 2021 Zhang, Chen and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Youcui Wang, wangyoucui@qdu.edu.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.