Cargando…

Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse

Ischemic stroke is characterized by extensive neuronal loss, glial scar formation, neural tissue degeneration that leading to profound changes in the extracellular matrix, neuronal circuitry, and long‐lasting functional disabilities. Although transplanted neural stem cells (NSCs) can recover some of...

Descripción completa

Detalles Bibliográficos
Autores principales: Shabani, Zahra, Rahbarghazi, Reza, Karimipour, Mohammad, Ghadiri, Tahereh, Salehi, Roya, Sadigh‐Eteghad, Saeed, Farhoudi, Mehdi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley & Sons, Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8780906/
https://www.ncbi.nlm.nih.gov/pubmed/35111956
http://dx.doi.org/10.1002/btm2.10264
_version_ 1784637958211502080
author Shabani, Zahra
Rahbarghazi, Reza
Karimipour, Mohammad
Ghadiri, Tahereh
Salehi, Roya
Sadigh‐Eteghad, Saeed
Farhoudi, Mehdi
author_facet Shabani, Zahra
Rahbarghazi, Reza
Karimipour, Mohammad
Ghadiri, Tahereh
Salehi, Roya
Sadigh‐Eteghad, Saeed
Farhoudi, Mehdi
author_sort Shabani, Zahra
collection PubMed
description Ischemic stroke is characterized by extensive neuronal loss, glial scar formation, neural tissue degeneration that leading to profound changes in the extracellular matrix, neuronal circuitry, and long‐lasting functional disabilities. Although transplanted neural stem cells (NSCs) can recover some of the functional deficit after stroke, retrieval is not complete and repair of lost tissue is negligible. Therefore, the current challenge is to use the combination of NSCs with suitably enriched biomaterials to retain these cells within the infarct cavity and accelerate the formation of a de novo tissue. This study aimed to test the regenerative potential of polylactic‐co‐glycolic acid‐polyethylene glycol (PLGA‐PEG) micelle biomaterial enriched with Reelin and embryonic NSCs on photothrombotic stroke model of mice to gain appropriate methods in tissue engineering. For this purpose, two sets of experiments, either in vitro or in vivo models, were performed. In vitro analyses exhibited PLGA‐PEG plus Reelin‐induced proliferation rate (Ki‐67(+) NSCs) and neurite outgrowth (axonization and dendritization) compared to PLGA‐PEG + NSCs and Reelin + NSCs groups (p < 0.05). Besides, neural differentiation (Map‐2(+) cells) was high in NSCs cultured in the presence of Reelin‐loaded PLGA‐PEG micelles (p < 0.05). Double immunofluorescence staining showed that Reelin‐loaded PLGA‐PEG micelles increased the number of migrating neural progenitor cells (DCX(+) cells) and mature neurons (NeuN(+) cells) around the lesion site compared to the groups received PLGA‐PEG and Reelin alone after 1 month (p < 0.05). Immunohistochemistry results showed that the PLGA/PEG plus Reelin significantly decreased the astrocytic gliosis and increased local angiogenesis (vWF‐positive cells) relative to the other groups. These changes led to the reduction of cavity size in the Reelin‐loaded PLGA‐PEG+NSCs group. Neurobehavioral tests indicated Reelin‐loaded PLGA‐PEG+NSCs promoted neurological outcome and functional recovery (p < 0.05). These results indicated that Reelin‐loaded PLGA‐PEG is capable of promoting NSCs dynamic growth, neuronal differentiation, and local angiogenesis following ischemic injury via providing a desirable microenvironment. These features can lead to neural tissue regeneration and functional recovery.
format Online
Article
Text
id pubmed-8780906
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley & Sons, Inc.
record_format MEDLINE/PubMed
spelling pubmed-87809062022-02-01 Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse Shabani, Zahra Rahbarghazi, Reza Karimipour, Mohammad Ghadiri, Tahereh Salehi, Roya Sadigh‐Eteghad, Saeed Farhoudi, Mehdi Bioeng Transl Med Research Articles Ischemic stroke is characterized by extensive neuronal loss, glial scar formation, neural tissue degeneration that leading to profound changes in the extracellular matrix, neuronal circuitry, and long‐lasting functional disabilities. Although transplanted neural stem cells (NSCs) can recover some of the functional deficit after stroke, retrieval is not complete and repair of lost tissue is negligible. Therefore, the current challenge is to use the combination of NSCs with suitably enriched biomaterials to retain these cells within the infarct cavity and accelerate the formation of a de novo tissue. This study aimed to test the regenerative potential of polylactic‐co‐glycolic acid‐polyethylene glycol (PLGA‐PEG) micelle biomaterial enriched with Reelin and embryonic NSCs on photothrombotic stroke model of mice to gain appropriate methods in tissue engineering. For this purpose, two sets of experiments, either in vitro or in vivo models, were performed. In vitro analyses exhibited PLGA‐PEG plus Reelin‐induced proliferation rate (Ki‐67(+) NSCs) and neurite outgrowth (axonization and dendritization) compared to PLGA‐PEG + NSCs and Reelin + NSCs groups (p < 0.05). Besides, neural differentiation (Map‐2(+) cells) was high in NSCs cultured in the presence of Reelin‐loaded PLGA‐PEG micelles (p < 0.05). Double immunofluorescence staining showed that Reelin‐loaded PLGA‐PEG micelles increased the number of migrating neural progenitor cells (DCX(+) cells) and mature neurons (NeuN(+) cells) around the lesion site compared to the groups received PLGA‐PEG and Reelin alone after 1 month (p < 0.05). Immunohistochemistry results showed that the PLGA/PEG plus Reelin significantly decreased the astrocytic gliosis and increased local angiogenesis (vWF‐positive cells) relative to the other groups. These changes led to the reduction of cavity size in the Reelin‐loaded PLGA‐PEG+NSCs group. Neurobehavioral tests indicated Reelin‐loaded PLGA‐PEG+NSCs promoted neurological outcome and functional recovery (p < 0.05). These results indicated that Reelin‐loaded PLGA‐PEG is capable of promoting NSCs dynamic growth, neuronal differentiation, and local angiogenesis following ischemic injury via providing a desirable microenvironment. These features can lead to neural tissue regeneration and functional recovery. John Wiley & Sons, Inc. 2021-10-29 /pmc/articles/PMC8780906/ /pubmed/35111956 http://dx.doi.org/10.1002/btm2.10264 Text en © 2021 The Authors. Bioengineering & Translational Medicine published by Wiley Periodicals LLC on behalf of American Institute of Chemical Engineers. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Shabani, Zahra
Rahbarghazi, Reza
Karimipour, Mohammad
Ghadiri, Tahereh
Salehi, Roya
Sadigh‐Eteghad, Saeed
Farhoudi, Mehdi
Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title_full Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title_fullStr Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title_full_unstemmed Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title_short Transplantation of bioengineered Reelin‐loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
title_sort transplantation of bioengineered reelin‐loaded plga/peg micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8780906/
https://www.ncbi.nlm.nih.gov/pubmed/35111956
http://dx.doi.org/10.1002/btm2.10264
work_keys_str_mv AT shabanizahra transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT rahbarghazireza transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT karimipourmohammad transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT ghadiritahereh transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT salehiroya transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT sadigheteghadsaeed transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse
AT farhoudimehdi transplantationofbioengineeredreelinloadedplgapegmicellescanaccelerateneuraltissueregenerationinphotothromboticstrokemodelofmouse