Elsevier

Biomaterials

Volume 265, January 2021, 120387
Biomaterials

Leading Opinion
Matrix reverses immortalization-mediated stem cell fate determination

https://doi.org/10.1016/j.biomaterials.2020.120387Get rights and content

Abstract

Primary cell culture in vitro suffers from cellular senescence. We hypothesized that expansion on decellularized extracellular matrix (dECM) deposited by simian virus 40 large T antigen (SV40LT) transduced autologous infrapatellar fat pad stem cells (IPFSCs) could rejuvenate high-passage IPFSCs in both proliferation and chondrogenic differentiation. In the study, we found that SV40LT transduced IPFSCs exhibited increased proliferation and adipogenic potential but decreased chondrogenic potential. Expansion on dECMs deposited by passage 5 IPFSCs yielded IPFSCs with dramatically increased proliferation and chondrogenic differentiation capacity; however, this enhanced capacity diminished if IPFSCs were grown on dECM deposited by passage 15 IPFSCs. Interestingly, expansion on dECM deposited by SV40LT transduced IPFSCs yielded IPFSCs with enhanced proliferation and chondrogenic capacity but decreased adipogenic potential, particularly for the dECM group derived from SV40LT transduced passage 15 cells. Our immunofluorescence staining and proteomics data identify matrix components such as basement membrane proteins as top candidates for matrix mediated IPFSC rejuvenation. Both cell proliferation and differentiation were endorsed by transcripts measured by RNASeq during the process. This study provides a promising model for in-depth investigation of the matrix protein influence on surrounding stem cell differentiation.

Introduction

As cartilage does not readily self-heal, articular cartilage has difficulty recovering from trauma or degenerative disease. Given that autologous chondrocyte implantation, the most promising cell therapy for articular cartilage defects, has limited cell sources for clinical application [1], mesenchymal stromal/stem cells (MSCs), especially tissue-specific stem cells deposited by synovial tissue (SDSCs), have received much attention as possible cartilage repair therapies [[2], [3], [4]]. Considering the difficulty in harvesting synovium without contamination by surrounding connective tissue, the infrapatellar fat pad (IPFP), an easily accessible adjacent tissue, might serve as an alternative, large quantity autologous tissue-specific stem cell source for cartilage regeneration and repair [5,6]. However, increasing evidence shows that IPFSCs also suffer from replicative senescence after long-term ex vivo expansion [7].

To acquire a sufficient number of cells for cartilage engineering and regeneration, modification strategies, including modification of internal genomics or the external matrix microenvironment, have been proposed for cell-based therapy [8]. Simian virus 40 (SV40), a well-known oncogene, has been commonly utilized for cell immortalization. SV40 is restricted to two proteins, the large T (LT) antigen and small t antigen (ST). The former mainly influences the SV40-extended lifespan due to its ability to bind to pRb and p53 to inactivate these two tumor suppressors, causing cells to move from G1 phase into S phase thus promoting DNA replication [9]. However, malignant transformation is one of the potential risks caused by genetic manipulation [10].

Decellularized extracellular matrix (dECM), an integral part of the external matrix microenvironment, can be prepared from cell or tissue sources but they play a different role in cell functionality [11]. Briefly, cell-derived dECM has a distinct role in rejuvenation of adult stem cells, mainly promoting adult stem cells’ proliferation and differentiation capacity [[12], [13], [14]], which is different from tissue-derived dECM that largely guides tissue-specific behaviors [15]. Meanwhile, cells isolated from patients themselves for dECM preparation provide another benefit in avoiding potential immune issues [16], despite the fact that most patients are either elderly or older adults and the isolated stem cells are prone to premature senescence.

Few reports have investigated the influence of SV40LT transduction alone or a combined dECM approach on stem cells' chondrogenic potential. SV40LT transduction in “old” IPFSCs might be a novel strategy to provide “young” autologous cells for dECM preparation, in which a patient's IPFSCs could be rejuvenated. In this study, we investigated whether SV40LT transduction could promote IPFSCs' proliferation and chondrogenic potential and whether high-passage IPFSCs could be rejuvenated after expansion on dECM deposited by SV40LT transduced IPFSCs from either early or late passage IPFSCs. Given the close relationship between chondrogenesis and adipogenesis [17], we also aimed to explore whether the influence of SV40LT transduction on chondrogenesis also applied to adipogenesis. The main purpose of this study was to identify key matrix components guiding adult stem cells' differentiation preference by using both dECM and immortalization approaches, which might facilitate engineering of smart matrix materials for cartilage engineering and regeneration in the near future.

Section snippets

IPFSC culture and SV40LT transduction

The study was approved by the Institutional Review Board. Adult human infrapatellar fat pads were collected from six young patients with acute meniscus or anterior cruciate ligament tears (four male and two female, 22 years old on average). Minced infrapatellar fat pads were digested with 0.1% trypsin (Roche, Indianapolis, IN) for 30 min followed by 0.1% collagenase P (Roche) for 2 h at 37 °C before filtration. After centrifugation, IPFSCs were cultured in alpha minimum essential medium (αMEM)

Evaluation of Cas9-SV40LT transduction and effect on IPFSC proliferation

To make an immortalized cell line of human IPFSCs, SV40LT lentiviral vector was used for transduction with GFP as a control. RT-PCR data confirmed successful transduction of Cas9-SV40LT in IPFSCs, evidenced by strong expression of SV40LT in cells from the SV40 group at both passages 5 and 15, followed by weak expression in the GFP group and negligible expression in the CTR group (Fig. 1A). To determine whether SV40LT transduction affects the proliferation ability of IPFSCs, PDN results showed,

Discussion

Donor age and long-term in vitro culture are both responsible for MSC senescence and are a challenge for stem cell-based tissue engineering and regeneration [7,27]. Previous reports indicate that SV40 transduction could immortalize primary cells but there were conflicting results in differentiation capacities [8]. In this study, we found that SV40LT transduction yielded human IPFSCs with significantly improved proliferation and adipogenic differentiation ability; however, chondrogenic capacity

Conclusion

For the first time, this study indicated that SV40LT transduction in human IPFSCs not only promoted proliferation but also increased adipogenic potential and decreased chondrogenic potential. Furthermore, we found that dECM deposited by SV40LT transduced IPFSCs reversed the differentiation preference of expanded IPFSCs by promoting chondrogenic potential but decreasing adipogenic capacity. Despite the unelucidated mystery underlying the rejuvenation effect of dECM deposited by high-passage

CRediT authorship contribution statement

Yiming Wang: Methodology, Validation, Formal analysis, Investigation, Data curation, Writing - original draft, Writing - review & editing. Gangqing Hu: Methodology, Software, Validation, Formal analysis, Writing - review & editing. Ryan C. Hill: Software, Validation, Formal analysis, Writing - review & editing. Monika Dzieciatkowska: Software, Validation, Formal analysis, Writing - review & editing. Kirk C. Hansen: Software, Validation, Formal analysis, Writing - review & editing. Xiao-Bing

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

We thank Suzanne Danley and Amanda Stewart for editing the manuscript. This project was supported by Research Grants from the National Institutes of Health (1R01AR067747) and the Musculoskeletal Transplant Foundation (MTF). We also would like to acknowledge the WVU Flow Cytometry & Single Cell Core Facility and the Bioinformatics Core as well as the grants that support the facility, WV-INBRE grant P20 GM103434, TME CoBRE grant P20GM131322 and the WV CTS grant 5U54 GM104942-04.

References (55)

  • E. Fuchs

    Finding one's niche in the skin

    Cell Stem Cell

    (2009)
  • E. Fuchs et al.

    Socializing with the neighbors: stem cells and their niche

    Cell

    (2004)
  • U. Schlötzer-Schrehardt et al.

    Characterization of extracellular matrix components in the limbal epithelial stem cell compartment

    Exp. Eye Res.

    (2007)
  • M. Marinkovic et al.

    One size does not fit all: developing a cell-specific niche for in vitro study of cell behavior

    Matrix Biol.

    (2016)
  • H. Kumra et al.

    Fibronectin-targeted drug delivery in cancer

    Adv. Drug Deliv. Rev.

    (2016)
  • S. Ohno et al.

    RGD-CAP ((beta)ig-h3) is expressed in precartilage condensation and in prehypertrophic chondrocytes during cartilage development

    Biochim. Biophys. Acta

    (2002)
  • S.L. Dallas et al.

    Proteolysis of latent transforming growth factor-beta (TGF-beta)-binding protein-1 by osteoclasts. A cellular mechanism for release of TGF-beta from bone matrix

    J. Biol. Chem.

    (2002)
  • J. Karnes et al.

    Cell therapy for the creation of cartilage and related clinical trials

  • F. Barry et al.

    Mesenchymal stem cells in joint disease and repair

    Nat. Rev. Rheumatol.

    (2013)
  • B.A. Jones et al.

    Synovium-derived stem cells: a tissue-specific stem cell for cartilage engineering and regeneration

    Tissue Eng. B Rev.

    (2012)
  • T. Pizzute et al.

    Impact of tissue-specific stem cells on lineage-specific differentiation: a focus on the musculoskeletal system

    Stem Cell Rev.

    (2015)
  • Y. Sun et al.

    Comparative advantages of infrapatellar fat pad: an emerging stem cell source for regenerative medicine

    Rheumatology

    (2018)
  • T.L. Wang et al.

    Site-dependent lineage preference of adipose stem cells

    Front. Cell Dev. Biol.

    (2020)
  • J. Li et al.

    Cell senescence: a challenge in cartilage engineering and regeneration

    Tissue Eng. B Rev.

    (2012)
  • N. Li et al.

    Genetically transforming human mesenchymal stem cells to sarcomas: changes in cellular phenotype and multilineage differentiation potential

    Cancer

    (2009)
  • M. Pei et al.

    A review of decellularized stem cell matrix: a novel cell expansion system for cartilage tissue engineering

    Eur. Cell. Mater.

    (2011)
  • S. Zhou et al.

    Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis

    Cell. Mol. Life Sci.

    (2019)
  • Cited by (14)

    • Matrix from urine stem cells boosts tissue-specific stem cell mediated functional cartilage reconstruction

      2023, Bioactive Materials
      Citation Excerpt :

      The Genomics Core of Marshall University sequenced the libraries by using HiSeq 2500. RNA-Seq data analysis was conducted as previously described [5,25]. Briefly, reads were aligned to the Oryctolagus cuniculus genome (oryCun2; rabbit) with subread-align version 2.0.1.

    • Stem cells immortalized by hTERT perform differently from those immortalized by SV40LT in proliferation, differentiation, and reconstruction of matrix microenvironment

      2021, Acta Biomaterialia
      Citation Excerpt :

      Besides DNA replication, oxidative stress also participates in telomere loss [37]; hTERT overexpression endowed MSCs with increased ability to resist oxidative damage [38], which could explain the increased aggrecan deposition after hTERT transduction. Comparatively, SV40LT transduced IPFSCs exhibited significantly decreased chondrogenic potential, consistent with our previous report [7]. Cell condensation is a pre-requisite for chondrogenesis [39].

    View all citing articles on Scopus
    1

    The first two authors contributed equally.

    View full text