Cargando…

Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation

Transplantation of mesenchymal stromal cells (MSCs) improves functional recovery in experimental models of spinal cord injury (SCI), but the mechanism is not fully understood. Activation of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2), a collagen-modifying enzyme, reportedly follows MS...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Fang, Liu, Hua, Zhang, Kun, Xiao, Dong-Jie, Wang, Chang, Wang, Yun-Shan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7906172/
https://www.ncbi.nlm.nih.gov/pubmed/33495412
http://dx.doi.org/10.18632/aging.202399
_version_ 1783655239646183424
author Li, Fang
Liu, Hua
Zhang, Kun
Xiao, Dong-Jie
Wang, Chang
Wang, Yun-Shan
author_facet Li, Fang
Liu, Hua
Zhang, Kun
Xiao, Dong-Jie
Wang, Chang
Wang, Yun-Shan
author_sort Li, Fang
collection PubMed
description Transplantation of mesenchymal stromal cells (MSCs) improves functional recovery in experimental models of spinal cord injury (SCI), but the mechanism is not fully understood. Activation of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2), a collagen-modifying enzyme, reportedly follows MSC transplantation in an SCI animal model. We investigated the regulation of PLOD2 expression and its potential contribution to the neuroprotective effects of adipose-derived stromal cells (ADSCs) following mechanical injury to neurons in vitro and SCI in vivo. ADSCs enhanced wound healing in vitro and promoted functional recovery after their implantation near injury sites in a rat SCI model. These effects correlated with upregulation of PLOD2, MAP2, NSE and GAP43, and downregulation of GFAP, which is indicative of improved neuronal survival and axonal regeneration as well as reduced glial scar formation. The neurorestorative effect of ADSCs was weakened after inhibition of PLOD2 expression. ADSCs appeared to induce PLOD2 upregulation via TGF-β1 secretion, as ADSC-mediated PLOD2 expression, neuronal survival, and functional recovery after SCI were largely prevented by SB431542, a TGF-(1 receptor inhibitor. These findings indicate that ADSCs reduce lesion size and promote functional recovery after SCI mainly through activation of a TGF-β1/P-Samd3/PLOD2 pathway in spinal cord neurons.
format Online
Article
Text
id pubmed-7906172
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-79061722021-03-04 Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation Li, Fang Liu, Hua Zhang, Kun Xiao, Dong-Jie Wang, Chang Wang, Yun-Shan Aging (Albany NY) Research Paper Transplantation of mesenchymal stromal cells (MSCs) improves functional recovery in experimental models of spinal cord injury (SCI), but the mechanism is not fully understood. Activation of procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2), a collagen-modifying enzyme, reportedly follows MSC transplantation in an SCI animal model. We investigated the regulation of PLOD2 expression and its potential contribution to the neuroprotective effects of adipose-derived stromal cells (ADSCs) following mechanical injury to neurons in vitro and SCI in vivo. ADSCs enhanced wound healing in vitro and promoted functional recovery after their implantation near injury sites in a rat SCI model. These effects correlated with upregulation of PLOD2, MAP2, NSE and GAP43, and downregulation of GFAP, which is indicative of improved neuronal survival and axonal regeneration as well as reduced glial scar formation. The neurorestorative effect of ADSCs was weakened after inhibition of PLOD2 expression. ADSCs appeared to induce PLOD2 upregulation via TGF-β1 secretion, as ADSC-mediated PLOD2 expression, neuronal survival, and functional recovery after SCI were largely prevented by SB431542, a TGF-(1 receptor inhibitor. These findings indicate that ADSCs reduce lesion size and promote functional recovery after SCI mainly through activation of a TGF-β1/P-Samd3/PLOD2 pathway in spinal cord neurons. Impact Journals 2021-01-20 /pmc/articles/PMC7906172/ /pubmed/33495412 http://dx.doi.org/10.18632/aging.202399 Text en Copyright: © 2021 Li et al. https://creativecommons.org/licenses/by/3.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Li, Fang
Liu, Hua
Zhang, Kun
Xiao, Dong-Jie
Wang, Chang
Wang, Yun-Shan
Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title_full Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title_fullStr Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title_full_unstemmed Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title_short Adipose-derived stromal cells improve functional recovery after spinal cord injury through TGF-β1/Smad3/PLOD2 pathway activation
title_sort adipose-derived stromal cells improve functional recovery after spinal cord injury through tgf-β1/smad3/plod2 pathway activation
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7906172/
https://www.ncbi.nlm.nih.gov/pubmed/33495412
http://dx.doi.org/10.18632/aging.202399
work_keys_str_mv AT lifang adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation
AT liuhua adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation
AT zhangkun adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation
AT xiaodongjie adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation
AT wangchang adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation
AT wangyunshan adiposederivedstromalcellsimprovefunctionalrecoveryafterspinalcordinjurythroughtgfb1smad3plod2pathwayactivation