Cargando…

Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells

BACKGROUND: Rapidly expanding clones (RECs) are one of the single-cell-derived mesenchymal stem cell clones sorted from human bone marrow mononuclear cells (BMMCs), which possess advantageous features. The RECs exhibit long-lasting proliferation potency that allows more than 10 repeated serial passa...

Descripción completa

Detalles Bibliográficos
Autores principales: Suyama, Takashi, Takemoto, Yuto, Miyauchi, Hiromi, Kato, Yuko, Matsuzaki, Yumi, Kato, Ryuji
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9526913/
https://www.ncbi.nlm.nih.gov/pubmed/36182958
http://dx.doi.org/10.1186/s41232-022-00214-w
_version_ 1784800976024109056
author Suyama, Takashi
Takemoto, Yuto
Miyauchi, Hiromi
Kato, Yuko
Matsuzaki, Yumi
Kato, Ryuji
author_facet Suyama, Takashi
Takemoto, Yuto
Miyauchi, Hiromi
Kato, Yuko
Matsuzaki, Yumi
Kato, Ryuji
author_sort Suyama, Takashi
collection PubMed
description BACKGROUND: Rapidly expanding clones (RECs) are one of the single-cell-derived mesenchymal stem cell clones sorted from human bone marrow mononuclear cells (BMMCs), which possess advantageous features. The RECs exhibit long-lasting proliferation potency that allows more than 10 repeated serial passages in vitro, considerably benefiting the manufacturing process of allogenic MSC-based therapeutic products. Although RECs aid the preparation of large-variation clone libraries for a greedy selection of better-quality clones, such a selection is only possible by establishing multiple-candidate cell banks for quality comparisons. Thus, there is a high demand for a novel method that can predict “low-risk and high-potency clones” early and in a feasible manner given the excessive cost and effort required to maintain such an establishment. METHODS: LNGFR and Thy-1 co-positive cells from BMMCs were single-cell-sorted into 96-well plates, and only fast-growing clones that reached confluency in 2 weeks were picked up and passaged as RECs. Fifteen RECs were prepared as passage 3 (P3) cryostock as the primary cell bank. From this cryostock, RECs were passaged until their proliferation limitation; their serial-passage limitation numbers were labeled as serial-passage potencies. At the P1 stage, phase-contrast microscopic images were obtained over 6–90 h to identify time-course changes of 24 morphological descriptors describing cell population information. Machine learning models were constructed using the morphological descriptors for predicting serial-passage potencies. The time window and field-of-view-number effects were evaluated to identify the most efficient image data usage condition for realizing high-performance serial-passage potency models. RESULTS: Serial-passage test results indicated variations of 7–13-repeated serial-passage potencies within RECs. Such potency values were predicted quantitatively with high performance (RMSE < 1.0) from P1 morphological profiles using a LASSO model. The earliest and minimum effort predictions require 6–30 h with 40 FOVs and 6–90 h with 15 FOVs, respectively. CONCLUSION: We successfully developed a noninvasive morphology-based machine learning model to enhance the efficiency of establishing cell banks with single-cell-derived RECs for quantitatively predicting the future serial-passage potencies of clones. Conventional methods that can make noninvasive and quantitative predictions without wasting precious cells in the early stage are lacking; the proposed method will provide a more efficient and robust cell bank establishment process for allogenic therapeutic product manufacturing. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s41232-022-00214-w.
format Online
Article
Text
id pubmed-9526913
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-95269132022-10-03 Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells Suyama, Takashi Takemoto, Yuto Miyauchi, Hiromi Kato, Yuko Matsuzaki, Yumi Kato, Ryuji Inflamm Regen Research Article BACKGROUND: Rapidly expanding clones (RECs) are one of the single-cell-derived mesenchymal stem cell clones sorted from human bone marrow mononuclear cells (BMMCs), which possess advantageous features. The RECs exhibit long-lasting proliferation potency that allows more than 10 repeated serial passages in vitro, considerably benefiting the manufacturing process of allogenic MSC-based therapeutic products. Although RECs aid the preparation of large-variation clone libraries for a greedy selection of better-quality clones, such a selection is only possible by establishing multiple-candidate cell banks for quality comparisons. Thus, there is a high demand for a novel method that can predict “low-risk and high-potency clones” early and in a feasible manner given the excessive cost and effort required to maintain such an establishment. METHODS: LNGFR and Thy-1 co-positive cells from BMMCs were single-cell-sorted into 96-well plates, and only fast-growing clones that reached confluency in 2 weeks were picked up and passaged as RECs. Fifteen RECs were prepared as passage 3 (P3) cryostock as the primary cell bank. From this cryostock, RECs were passaged until their proliferation limitation; their serial-passage limitation numbers were labeled as serial-passage potencies. At the P1 stage, phase-contrast microscopic images were obtained over 6–90 h to identify time-course changes of 24 morphological descriptors describing cell population information. Machine learning models were constructed using the morphological descriptors for predicting serial-passage potencies. The time window and field-of-view-number effects were evaluated to identify the most efficient image data usage condition for realizing high-performance serial-passage potency models. RESULTS: Serial-passage test results indicated variations of 7–13-repeated serial-passage potencies within RECs. Such potency values were predicted quantitatively with high performance (RMSE < 1.0) from P1 morphological profiles using a LASSO model. The earliest and minimum effort predictions require 6–30 h with 40 FOVs and 6–90 h with 15 FOVs, respectively. CONCLUSION: We successfully developed a noninvasive morphology-based machine learning model to enhance the efficiency of establishing cell banks with single-cell-derived RECs for quantitatively predicting the future serial-passage potencies of clones. Conventional methods that can make noninvasive and quantitative predictions without wasting precious cells in the early stage are lacking; the proposed method will provide a more efficient and robust cell bank establishment process for allogenic therapeutic product manufacturing. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s41232-022-00214-w. BioMed Central 2022-10-02 /pmc/articles/PMC9526913/ /pubmed/36182958 http://dx.doi.org/10.1186/s41232-022-00214-w Text en © The Author(s) 2022 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/) .
spellingShingle Research Article
Suyama, Takashi
Takemoto, Yuto
Miyauchi, Hiromi
Kato, Yuko
Matsuzaki, Yumi
Kato, Ryuji
Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title_full Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title_fullStr Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title_full_unstemmed Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title_short Morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
title_sort morphology-based noninvasive early prediction of serial-passage potency enhances the selection of clone-derived high-potency cell bank from mesenchymal stem cells
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9526913/
https://www.ncbi.nlm.nih.gov/pubmed/36182958
http://dx.doi.org/10.1186/s41232-022-00214-w
work_keys_str_mv AT suyamatakashi morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells
AT takemotoyuto morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells
AT miyauchihiromi morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells
AT katoyuko morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells
AT matsuzakiyumi morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells
AT katoryuji morphologybasednoninvasiveearlypredictionofserialpassagepotencyenhancestheselectionofclonederivedhighpotencycellbankfrommesenchymalstemcells