Cargando…

Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential

Engineering functional tissues of clinically relevant size (in mm-scale) in vitro is still a challenge in tissue engineering due to low oxygen diffusion and lack of vascularization. To address these limitations, a perfusion bioreactor was used to generate contractile engineered muscles of a 3 mm-thi...

Descripción completa

Detalles Bibliográficos
Autores principales: Borisov, Vladislav, Gili Sole, Laia, Reid, Gregory, Milan, Giulia, Hutter, Gregor, Grapow, Martin, Eckstein, Friedrich Stefan, Isu, Giuseppe, Marsano, Anna
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10376693/
https://www.ncbi.nlm.nih.gov/pubmed/37508827
http://dx.doi.org/10.3390/bioengineering10070800
_version_ 1785079335167721472
author Borisov, Vladislav
Gili Sole, Laia
Reid, Gregory
Milan, Giulia
Hutter, Gregor
Grapow, Martin
Eckstein, Friedrich Stefan
Isu, Giuseppe
Marsano, Anna
author_facet Borisov, Vladislav
Gili Sole, Laia
Reid, Gregory
Milan, Giulia
Hutter, Gregor
Grapow, Martin
Eckstein, Friedrich Stefan
Isu, Giuseppe
Marsano, Anna
author_sort Borisov, Vladislav
collection PubMed
description Engineering functional tissues of clinically relevant size (in mm-scale) in vitro is still a challenge in tissue engineering due to low oxygen diffusion and lack of vascularization. To address these limitations, a perfusion bioreactor was used to generate contractile engineered muscles of a 3 mm-thickness and a 8 mm-diameter. This study aimed to upscale the process to 50 mm in diameter by combining murine skeletal myoblasts (SkMbs) with human adipose-derived stromal vascular fraction (SVF) cells, providing high neuro-vascular potential in vivo. SkMbs were cultured on a type-I-collagen scaffold with (co-culture) or without (monoculture) SVF. Large-scale muscle-like tissue showed an increase in the maturation index over time (49.18 ± 1.63% and 76.63 ± 1.22%, at 9 and 11 days, respectively) and a similar force of contraction in mono- (43.4 ± 2.28 µN) or co-cultured (47.6 ± 4.7 µN) tissues. Four weeks after implantation in subcutaneous pockets of nude rats, the vessel length density within the constructs was significantly higher in SVF co-cultured tissues (5.03 ± 0.29 mm/mm(2)) compared to monocultured tissues (3.68 ± 0.32 mm/mm(2)) (p < 0.005). Although no mature neuromuscular junctions were present, nerve-like structures were predominantly observed in the engineered tissues co-cultured with SVF cells. This study demonstrates that SVF cells can support both in vivo vascularization and innervation of contractile muscle-like tissues, making significant progress towards clinical translation.
format Online
Article
Text
id pubmed-10376693
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-103766932023-07-29 Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential Borisov, Vladislav Gili Sole, Laia Reid, Gregory Milan, Giulia Hutter, Gregor Grapow, Martin Eckstein, Friedrich Stefan Isu, Giuseppe Marsano, Anna Bioengineering (Basel) Article Engineering functional tissues of clinically relevant size (in mm-scale) in vitro is still a challenge in tissue engineering due to low oxygen diffusion and lack of vascularization. To address these limitations, a perfusion bioreactor was used to generate contractile engineered muscles of a 3 mm-thickness and a 8 mm-diameter. This study aimed to upscale the process to 50 mm in diameter by combining murine skeletal myoblasts (SkMbs) with human adipose-derived stromal vascular fraction (SVF) cells, providing high neuro-vascular potential in vivo. SkMbs were cultured on a type-I-collagen scaffold with (co-culture) or without (monoculture) SVF. Large-scale muscle-like tissue showed an increase in the maturation index over time (49.18 ± 1.63% and 76.63 ± 1.22%, at 9 and 11 days, respectively) and a similar force of contraction in mono- (43.4 ± 2.28 µN) or co-cultured (47.6 ± 4.7 µN) tissues. Four weeks after implantation in subcutaneous pockets of nude rats, the vessel length density within the constructs was significantly higher in SVF co-cultured tissues (5.03 ± 0.29 mm/mm(2)) compared to monocultured tissues (3.68 ± 0.32 mm/mm(2)) (p < 0.005). Although no mature neuromuscular junctions were present, nerve-like structures were predominantly observed in the engineered tissues co-cultured with SVF cells. This study demonstrates that SVF cells can support both in vivo vascularization and innervation of contractile muscle-like tissues, making significant progress towards clinical translation. MDPI 2023-07-04 /pmc/articles/PMC10376693/ /pubmed/37508827 http://dx.doi.org/10.3390/bioengineering10070800 Text en © 2023 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Borisov, Vladislav
Gili Sole, Laia
Reid, Gregory
Milan, Giulia
Hutter, Gregor
Grapow, Martin
Eckstein, Friedrich Stefan
Isu, Giuseppe
Marsano, Anna
Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title_full Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title_fullStr Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title_full_unstemmed Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title_short Upscaled Skeletal Muscle Engineered Tissue with In Vivo Vascularization and Innervation Potential
title_sort upscaled skeletal muscle engineered tissue with in vivo vascularization and innervation potential
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10376693/
https://www.ncbi.nlm.nih.gov/pubmed/37508827
http://dx.doi.org/10.3390/bioengineering10070800
work_keys_str_mv AT borisovvladislav upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT gilisolelaia upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT reidgregory upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT milangiulia upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT huttergregor upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT grapowmartin upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT ecksteinfriedrichstefan upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT isugiuseppe upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential
AT marsanoanna upscaledskeletalmuscleengineeredtissuewithinvivovascularizationandinnervationpotential