Cargando…

Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts

BACKGROUND: Myogenic transdifferentiation can be accomplished through ectopic MYOD1 expression, which is facilitated by various signaling pathways associated with myogenesis. In this study, we attempted to transdifferentiate pig embryonic fibroblasts (PEFs) myogenically into skeletal muscle through...

Descripción completa

Detalles Bibliográficos
Autores principales: Jeong, Jinsol, Choi, Kwang-Hwan, Kim, Seung-Hun, Lee, Dong-Kyung, Oh, Jong-Nam, Lee, Mingyun, Choe, Gyung Cheol, Lee, Chang-Kyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8117598/
https://www.ncbi.nlm.nih.gov/pubmed/33980301
http://dx.doi.org/10.1186/s40104-021-00583-1
_version_ 1783691613803905024
author Jeong, Jinsol
Choi, Kwang-Hwan
Kim, Seung-Hun
Lee, Dong-Kyung
Oh, Jong-Nam
Lee, Mingyun
Choe, Gyung Cheol
Lee, Chang-Kyu
author_facet Jeong, Jinsol
Choi, Kwang-Hwan
Kim, Seung-Hun
Lee, Dong-Kyung
Oh, Jong-Nam
Lee, Mingyun
Choe, Gyung Cheol
Lee, Chang-Kyu
author_sort Jeong, Jinsol
collection PubMed
description BACKGROUND: Myogenic transdifferentiation can be accomplished through ectopic MYOD1 expression, which is facilitated by various signaling pathways associated with myogenesis. In this study, we attempted to transdifferentiate pig embryonic fibroblasts (PEFs) myogenically into skeletal muscle through overexpression of the pig MYOD1 gene and modulation of the FGF, TGF-β, WNT, and cAMP signaling pathways. RESULTS: The MYOD1 overexpression vector was constructed based on comparative sequence analysis, demonstrating that pig MYOD1 has evolutionarily conserved domains across various species. Although forced MYOD1 expression through these vectors triggered the expression of endogenous muscle markers, transdifferentiated muscle cells from fibroblasts were not observed. Therefore, various signaling molecules, including FGF2, SB431542, CHIR99021, and forskolin, along with MYOD1 overexpression were applied to enhance the myogenic reprogramming. The modified conditions led to the derivation of myotubes and activation of muscle markers in PEFs, as determined by qPCR and immunostaining. Notably, a sarcomere-like structure was observed, indicating that terminally differentiated skeletal muscle could be obtained from transdifferentiated cells. CONCLUSIONS: In summary, we established a protocol for reprogramming MYOD1-overexpressing PEFs into the mature skeletal muscle using signaling molecules. Our myogenic reprogramming can be used as a cell source for muscle disease models in regenerative medicine and the production of cultured meat in cellular agriculture.
format Online
Article
Text
id pubmed-8117598
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-81175982021-05-13 Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts Jeong, Jinsol Choi, Kwang-Hwan Kim, Seung-Hun Lee, Dong-Kyung Oh, Jong-Nam Lee, Mingyun Choe, Gyung Cheol Lee, Chang-Kyu J Anim Sci Biotechnol Research BACKGROUND: Myogenic transdifferentiation can be accomplished through ectopic MYOD1 expression, which is facilitated by various signaling pathways associated with myogenesis. In this study, we attempted to transdifferentiate pig embryonic fibroblasts (PEFs) myogenically into skeletal muscle through overexpression of the pig MYOD1 gene and modulation of the FGF, TGF-β, WNT, and cAMP signaling pathways. RESULTS: The MYOD1 overexpression vector was constructed based on comparative sequence analysis, demonstrating that pig MYOD1 has evolutionarily conserved domains across various species. Although forced MYOD1 expression through these vectors triggered the expression of endogenous muscle markers, transdifferentiated muscle cells from fibroblasts were not observed. Therefore, various signaling molecules, including FGF2, SB431542, CHIR99021, and forskolin, along with MYOD1 overexpression were applied to enhance the myogenic reprogramming. The modified conditions led to the derivation of myotubes and activation of muscle markers in PEFs, as determined by qPCR and immunostaining. Notably, a sarcomere-like structure was observed, indicating that terminally differentiated skeletal muscle could be obtained from transdifferentiated cells. CONCLUSIONS: In summary, we established a protocol for reprogramming MYOD1-overexpressing PEFs into the mature skeletal muscle using signaling molecules. Our myogenic reprogramming can be used as a cell source for muscle disease models in regenerative medicine and the production of cultured meat in cellular agriculture. BioMed Central 2021-05-13 /pmc/articles/PMC8117598/ /pubmed/33980301 http://dx.doi.org/10.1186/s40104-021-00583-1 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Jeong, Jinsol
Choi, Kwang-Hwan
Kim, Seung-Hun
Lee, Dong-Kyung
Oh, Jong-Nam
Lee, Mingyun
Choe, Gyung Cheol
Lee, Chang-Kyu
Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title_full Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title_fullStr Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title_full_unstemmed Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title_short Combination of cell signaling molecules can facilitate MYOD1-mediated myogenic transdifferentiation of pig fibroblasts
title_sort combination of cell signaling molecules can facilitate myod1-mediated myogenic transdifferentiation of pig fibroblasts
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8117598/
https://www.ncbi.nlm.nih.gov/pubmed/33980301
http://dx.doi.org/10.1186/s40104-021-00583-1
work_keys_str_mv AT jeongjinsol combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT choikwanghwan combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT kimseunghun combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT leedongkyung combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT ohjongnam combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT leemingyun combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT choegyungcheol combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts
AT leechangkyu combinationofcellsignalingmoleculescanfacilitatemyod1mediatedmyogenictransdifferentiationofpigfibroblasts