Abstract

Cerebral ischemia and neurodegenerative diseases lead to impairment or death of neurons in the central nervous system. Stem cell based therapies are promising strategies currently under investigation. Carbon monoxide (CO) is an endogenous product of heme degradation by heme oxygenase (HO) activity. Administration of CO at low concentrations produces several beneficial effects in distinct tissues, namely anti-apoptotic and anti-inflammatory. Herein the CO role on modulation of neuronal differentiation was assessed. Three different models with increasing complexity were used: human neuroblastoma SH-S5Y5 cell line, human teratocarcinoma NT2 cell line and organotypic hippocampal slice cultures (OHSC). Cell lines were differentiated into post-mitotic neurons by treatment with retinoic acid (RA) supplemented with CO-releasing molecule A1 (CORM-A1). CORM-A1 positively modulated neuronal differentiation, since it increased final neuronal production and enhanced the expression of specific neuronal genes: Nestin, Tuj1 and MAP2. Furthermore, during neuronal differentiation process, there was an increase in proliferative cell number (ki67 mRNA expressing cells) and a decrease in cell death (lower propidium iodide (PI) uptake, limitation of caspase-3 activation and higher Bcl-2 expressing cells). CO supplementation did not increase the expression of RA receptors. In the case of SH-S5Y5 model, small amounts of reactive oxygen species (ROS) generation emerges as important signaling molecules during CO-promoted neuronal differentiation. CO’s improvement of neuronal differentiation yield was validated using OHSC as ex vivo model. CORM-A1 treatment of OHSC promoted higher levels of cells expressing the neuronal marker Tuj1. Still, CORM-A1 increased cell proliferation assessed by ki67 expression and also prevented cell death, which was followed by increased Bcl-2 expression, decreased levels of active caspase-3 and PI uptake. Likewise, ROS signaling emerged as key factors in CO’s increasing number of differentiated neurons in OHSC. In conclusion, CO’s increasing number of differentiated neurons is a novel biological role disclosed herein. CO improves neuronal yield due to its capacity to reduce cell death, promoting an increase in proliferative population. However, one cannot disregard a direct CO’s effect on specific cellular processes of neuronal differentiation. Further studies are needed to evaluate how CO can potentially modulate cell mechanisms involved in neuronal differentiation. In summary, CO appears as a promising therapeutic molecule to stimulate endogenous neurogenesis or to improve in vitro neuronal production for cell therapy strategies.

Highlights

  • Adult neurogenesis, which consists of generation of neurons from neural stem/precursor cells, occurs in specific brain regions called neurogenic zones

  • It was observed that retinoic acid (RA) treatment supplemented with CO-releasing molecule A1 (CORM-A1) at 25μM yielded a duplication of the final number of NT2 post-mitotic neurons (Fig 2A) and a 6-fold increase on SH-SY5Y post-mitotic neuronal population when compared to RA treatment without supplementation (Fig 2B)

  • Neurons obtained from treatment with RA supplemented with CORM-A1 were comparable to neurons obtained from RA treatment only (Fig 2C and 2D)

Read more

Summary

Introduction

Adult neurogenesis, which consists of generation of neurons from neural stem/precursor cells, occurs in specific brain regions called neurogenic zones. These niches are mostly located in the subventricular zone (SVZ), on the border of the lateral ventricle and striatum, and the subgranular zone of the dentate gyrus (DG) in the hippocampus [1]. At least five steps appear to be involved in the neurogenesis process: (i) proliferation of stem/progenitor cells, (ii) migration of newborn neurons, (iii) neuronal differentiation and maturation, (iv) integration into neuronal circuits and (v) survival of cells [2]. Programmed cell death is an important developmental cell process that occurs during neural development: from early embryonic proliferation stages until adult stages [3,4,5]. In genetically modified mouse models, where executor or regulatory apoptotic genes (caspase-3, caspase-9, Bak, Bax, among others) are knock out, resulted in supernumerary neurons in the brain [4]

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