Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome

Dax-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital region on X-chromosome gene 1) blocks 17β-estradiol biosynthesis and its knockdown would be expected to increase 17β-estradiol production. We hypothesized that knockdown of Dax-1 in a conditionally immortalized neural stem cell (NSC)...

Full description

Bibliographic Details
Main Authors: Shalmali Patkar, Dafe Uwanogho, Michel Modo, Rothwelle J. Tate, Robin Plevin, Hilary V. O. Carswell
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-07-01
Series:Frontiers in Cellular Neuroscience
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fncel.2022.917181/full
_version_ 1811288184087117824
author Shalmali Patkar
Dafe Uwanogho
Michel Modo
Michel Modo
Rothwelle J. Tate
Robin Plevin
Hilary V. O. Carswell
author_facet Shalmali Patkar
Dafe Uwanogho
Michel Modo
Michel Modo
Rothwelle J. Tate
Robin Plevin
Hilary V. O. Carswell
author_sort Shalmali Patkar
collection DOAJ
description Dax-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital region on X-chromosome gene 1) blocks 17β-estradiol biosynthesis and its knockdown would be expected to increase 17β-estradiol production. We hypothesized that knockdown of Dax-1 in a conditionally immortalized neural stem cell (NSC) line, MHP36, is a useful approach to increase 17β-estradiol production. Short hairpin (sh) RNA targeted to Dax-1 in NSCs, namely MHP36-Dax1KD cells, resulted in the degradation of Dax-1 RNA and attenuation of Dax-1 protein expression. In vitro, MHP36-Dax1KD cells exhibited overexpression of aromatase and increased 17β-estradiol secretion compared to MHP36 cells. As 17β-estradiol has been shown to promote the efficacy of cell therapy, we interrogated the application of 17β-estradiol-enriched NSCs in a relevant in vivo disease model. We hypothesized that MHP36-Dax1KD cells will enhance functional recovery after transplantation in a stroke model. C57BL/6 male adult mice underwent ischemia/reperfusion by left middle cerebral artery occlusion for 45 min using an intraluminal thread. Two days later male mice randomly received vehicle, MHP36 cells, MHP36-Dax1KD cells, and MHP36 cells suspended in 17β-estradiol (100 nm) or 17β-estradiol alone (100 nm) with serial behavioral testing over 28 days followed by post-mortem histology and blinded analysis. Recovery of sensorimotor function was accelerated and enhanced, and lesion volume was reduced by MHP36-Dax1KD transplants. Regarding mechanisms, immunofluorescence indicated increased synaptic plasticity and neuronal differentiation after MHP36-Dax1KD transplants. In conclusion, knockdown of Dax-1 is a useful target to increase 17β-estradiol biosynthesis in NSCs and improves functional recovery after stroke in vivo, possibly mediated through neuroprotection and improved synaptic plasticity. Therefore, targeting 17β-estradiol biosynthesis in stem cells may be a promising therapeutic strategy for enhancing the efficacy of stem cell-based therapies for stroke.
first_indexed 2024-04-13T03:32:36Z
format Article
id doaj.art-61187a76e35041b3a5cd3c7189a35120
institution Directory Open Access Journal
issn 1662-5102
language English
last_indexed 2024-04-13T03:32:36Z
publishDate 2022-07-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Cellular Neuroscience
spelling doaj.art-61187a76e35041b3a5cd3c7189a351202022-12-22T03:04:25ZengFrontiers Media S.A.Frontiers in Cellular Neuroscience1662-51022022-07-011610.3389/fncel.2022.917181917181Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcomeShalmali Patkar0Dafe Uwanogho1Michel Modo2Michel Modo3Rothwelle J. Tate4Robin Plevin5Hilary V. O. Carswell6Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United KingdomDepartment of Neuroscience, James Black Centre, King’s College London, London, United KingdomDepartment of Neuroscience, James Black Centre, King’s College London, London, United KingdomDepartment of Radiology, University of Pittsburgh, Pittsburgh, PA, United StatesStrathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United KingdomStrathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United KingdomStrathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow, United KingdomDax-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital region on X-chromosome gene 1) blocks 17β-estradiol biosynthesis and its knockdown would be expected to increase 17β-estradiol production. We hypothesized that knockdown of Dax-1 in a conditionally immortalized neural stem cell (NSC) line, MHP36, is a useful approach to increase 17β-estradiol production. Short hairpin (sh) RNA targeted to Dax-1 in NSCs, namely MHP36-Dax1KD cells, resulted in the degradation of Dax-1 RNA and attenuation of Dax-1 protein expression. In vitro, MHP36-Dax1KD cells exhibited overexpression of aromatase and increased 17β-estradiol secretion compared to MHP36 cells. As 17β-estradiol has been shown to promote the efficacy of cell therapy, we interrogated the application of 17β-estradiol-enriched NSCs in a relevant in vivo disease model. We hypothesized that MHP36-Dax1KD cells will enhance functional recovery after transplantation in a stroke model. C57BL/6 male adult mice underwent ischemia/reperfusion by left middle cerebral artery occlusion for 45 min using an intraluminal thread. Two days later male mice randomly received vehicle, MHP36 cells, MHP36-Dax1KD cells, and MHP36 cells suspended in 17β-estradiol (100 nm) or 17β-estradiol alone (100 nm) with serial behavioral testing over 28 days followed by post-mortem histology and blinded analysis. Recovery of sensorimotor function was accelerated and enhanced, and lesion volume was reduced by MHP36-Dax1KD transplants. Regarding mechanisms, immunofluorescence indicated increased synaptic plasticity and neuronal differentiation after MHP36-Dax1KD transplants. In conclusion, knockdown of Dax-1 is a useful target to increase 17β-estradiol biosynthesis in NSCs and improves functional recovery after stroke in vivo, possibly mediated through neuroprotection and improved synaptic plasticity. Therefore, targeting 17β-estradiol biosynthesis in stem cells may be a promising therapeutic strategy for enhancing the efficacy of stem cell-based therapies for stroke.https://www.frontiersin.org/articles/10.3389/fncel.2022.917181/fullcerebral ischemiaestrogensteroidogenesiscell therapytransplantationneuroprotection
spellingShingle Shalmali Patkar
Dafe Uwanogho
Michel Modo
Michel Modo
Rothwelle J. Tate
Robin Plevin
Hilary V. O. Carswell
Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
Frontiers in Cellular Neuroscience
cerebral ischemia
estrogen
steroidogenesis
cell therapy
transplantation
neuroprotection
title Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
title_full Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
title_fullStr Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
title_full_unstemmed Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
title_short Targeting 17β-estradiol biosynthesis in neural stem cells improves stroke outcome
title_sort targeting 17β estradiol biosynthesis in neural stem cells improves stroke outcome
topic cerebral ischemia
estrogen
steroidogenesis
cell therapy
transplantation
neuroprotection
url https://www.frontiersin.org/articles/10.3389/fncel.2022.917181/full
work_keys_str_mv AT shalmalipatkar targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT dafeuwanogho targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT michelmodo targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT michelmodo targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT rothwellejtate targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT robinplevin targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome
AT hilaryvocarswell targeting17bestradiolbiosynthesisinneuralstemcellsimprovesstrokeoutcome