Elsevier

Acta Biomaterialia

Volume 20, 1 July 2015, Pages 39-50
Acta Biomaterialia

Delineation of in vitro chondrogenesis of human synovial stem cells following preconditioning using decellularized matrix

https://doi.org/10.1016/j.actbio.2015.04.001Get rights and content

Abstract

As a tissue-specific stem cell for chondrogenesis, synovium-derived stem cells (SDSCs) are a promising cell source for cartilage repair. However, a small biopsy can only provide a limited number of cells. Cell senescence from both in vitro expansion and donor age presents a big challenge for stem cell based cartilage regeneration. Here we found that expansion on decellularized extracellular matrix (dECM) full of three-dimensional nanostructured fibers provided SDSCs with unique surface profiles, low elasticity but large volume as well as a fibroblast-like shape. dECM expanded SDSCs yielded larger pellets with intensive staining of type II collagen and sulfated glycosaminoglycans compared to those grown on plastic flasks while SDSCs grown in ECM yielded 28-day pellets with minimal matrix as evidenced by pellet size and chondrogenic marker staining, which was confirmed by both biochemical data and real-time PCR data. Our results also found lower levels of inflammatory genes in dECM expanded SDSCs that might be responsible for enhanced chondrogenic differentiation. Despite an increase in type X collagen in chondrogenically induced cells, dECM expanded cells had significantly lower potential for endochondral bone formation. Wnt and MAPK signals were actively involved in both expansion and chondrogenic induction of dECM expanded cells. Since young and healthy people can be potential donors for this matrix expansion system and decellularization can minimize immune concerns, human SDSCs expanded on this future commercially available dECM could be a potential cell source for autologous cartilage repair.

Introduction

Articular cartilage has a limited capacity for self-repair. Once damaged, the injured cartilage will develop defects. Despite promising results from autologous chondrocyte implantation [1], donor tissue availability is a challenge. Recent advances make adult stem cells an attractive cell source for cartilage regeneration, especially tissue-specific stem cells such as synovium-derived stem cells (SDSCs) [2], [3]. Since in vitro expansion is a necessary step before in vivo application, accompanying cell senescence and dedifferentiation represent a formidable challenge for stem cell-based cartilage repair [4].

We found that decellularized extracellular matrix (dECM) deposited by mesenchymal stem cells could rejuvenate stem cells [5], [6], [7], [8], [9], [10], [11] and primary cells [12], [13], [14] in both proliferation and differentiation capacity. For instance, dECM deposited by SDSCs significantly promoted expanded porcine SDSCs (pSDSCs) in both proliferation and chondrogenic potential [5]. In vivo transplantation of dECM expanded pSDSCs demonstrated efficacy in promoting cartilage regeneration in a partial thickness cartilage defect porcine model [15].

Our recent reports suggested that this in vitro cell expansion system also benefits human SDSC (hSDSC) expansion and rejuvenation of chondrogenic potential [16], [17], which brings hope for the potential use of this approach in clinical treatment [18], [19]. However, a concomitant up-regulation of type X collagen (COL10A1) was also observed in chondrogenically differentiated hSDSCs that have undergone dECM expansion, indicating a tendency toward chondrogenic hypertrophy.

In this study, we fully characterized cell morphology, volume, elasticity, and surface phenotypes in hSDSCs following dECM expansion. We not only defined proliferation and chondrogenic potential in dECM expanded hSDSCs but also examined whether increased expression of COL10A1 could be a sign of endochondral bone formation. Since both the mitogen-activated protein kinase (MAPK) and Wnt signals are critical pathways for chondrogenesis and have crosstalk in stem cell mediated cartilage regeneration [20], these two signals were evaluated for their changes in both cell expansion and chondrogenic induction of hSDSCs after preconditioning using dECM and conventional plastic flasks, which might provide evidence for further investigation of potential mechanisms underlying the rejuvenation of hSDSCs by dECM expansion.

Section snippets

SDSC culture

Adult human synovial fibroblasts (4 donors, two male and two female, average 43 years old, all had no known joint disease), referred to as hSDSCs [16], [17], were obtained from Asterand (North America Laboratories, Detroit, MI). Human SDSCs were plated and cultured in a growth medium [alpha minimum essential medium (αMEM) containing 10% fetal bovine serum (FBS), 100 U/mL penicillin, 100 μg/mL streptomycin, and 0.25 μg/mL fungizone (Invitrogen, Carlsbad, CA)] at 37 °C in a humidified 5% CO2 and 21% O2

Expansion on dECM changed cell morphology and increased cell proliferation

Before plating cells, SEM data showed that, compared with a flat surface on Plastic substrate, dECM coated flasks exhibited a three-dimensional structure with interconnected nanofibers (Fig. 1(A)) that was corroborated by morphological data from AFM (Fig. 1(B)), in which the diameter of matrix fibers was 109.5 ± 39.6 nm (n = 82). SEM data also showed that hSDSCs displayed a well-organized fibroblast-like cell shape when expanded on dECM rather than a broad and flat cell shape when grown on Plastic

Discussion

Our previous work found that dECM could rejuvenate pSDSCs for both proliferation and chondrogenic potential. We recently found this in vitro cell expansion system also applied to hSDSCs. In order to make this approach applicable to future clinical treatment, in this study, we characterized dECM and expanded cells using both SEM and AFM and compared them to the Plastic control; we also characterized proliferation and chondrogenic capacity of adult hSDSCs after expansion on dECM and determined

Conclusion and perspectives

Our study demonstrated that dECM deposited by hSDSCs formed a three-dimensional nanostructured matrix; expansion on dECM yielded a large quantity of hSDSCs with enhanced chondrogenic potential; despite an increase of type X collagen in chondrogenically induced cells, dECM expanded cells had significantly lower potential for endochondral bone formation. Even though the composition of dECM has been characterized in a recent report [16], research on the crosstalk between expanded SDSCs and dECMs

Competing Interest Statement

The authors do not have any conflicts of interest.

Acknowledgments

The authors thank Suzanne Danley for help in editing the manuscript. We also thank Ting Zheng and Dr. Cerasela Zoica Dinu for their help with the AFM instrument. This project was partially supported by research grants from the AO Foundation (S-12-19P) and the National Institutes of Health (NIH) (1 R03 AR062763-01A1 and P20 GM103434).

References (57)

  • M. Almeida et al.

    Wnt proteins prevent apoptosis of both uncommitted osteoblast progenitors and differentiated osteoblasts by beta-catenin-dependent and -independent signaling cascades involving Src/ERK and phosphatidylinositol 3-kinase/AKT

    J Biol Chem

    (2005)
  • J.S. Lunn et al.

    A spatial and temporal map of FGF/Erk1/2 activity and response repertoires in the early chick embryo

    Dev Biol

    (2007)
  • C.D. Oh et al.

    Opposing role of mitogen-activated protein kinase subtypes, erk-1/2 and p38, in the regulation of chondrogenesis of mesenchymes

    J Biol Chem

    (2000)
  • K. Nakamura et al.

    P38 mitogen-activated protein kinase functionally contributes to chondrogenesis induced by growth/differentiation factor-5 in ATDC5 cells

    Exp Cell Res

    (1999)
  • L.A. Stanton et al.

    MAP kinases in chondrocyte differentiation

    Dev Biol

    (2003)
  • J. Karnes et al.

    Cell therapy for the creation of cartilage and related clinical trials

  • B. Jones et al.

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

    Tissue Eng Part B Rev

    (2012)
  • T. Pizzute et al.

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

    Stem Cell Rev Rep

    (2014)
  • J.T. Li et al.

    Cell senescence: A challenge in cartilage engineering and regeneration

    Tissue Eng Part B

    (2012)
  • F. He et al.

    Reconstruction of an in vitro tissue-specific microenvironment to rejuvenate synovium-derived stem cells for cartilage tissue engineering

    Tissue Eng Part A

    (2009)
  • F. He et al.

    Extracellular matrix enhances differentiation of adipose stem cells from infrapatellar fat pad toward chondrogenesis

    J Tissue Eng Regener Med

    (2013)
  • J.T. Li et al.

    Creation of an in vitro microenvironment to enhance human fetal synovium-derived stem cell chondrogenesis

    Cell Tissue Res

    (2011)
  • J.T. Li et al.

    Optimization of an in vitro three-dimensional microenvironment to reprogram synovium-derived stem cells for cartilage tissue engineering

    Tissue Eng Part A

    (2011)
  • M. Pei et al.

    Expansion on extracellular matrix deposited by human bone marrow stromal cells facilitates stem cell proliferation and tissue-specific lineage potential

    Tissue Eng Part A

    (2011)
  • M. Pei et al.

    Three dimensional cell expansion substrate for cartilage tissue engineering and regeneration: a comparison in decellularized matrix deposited by synovium-derived stem cells and chondrocytes

    J Tissue Sci Eng

    (2011)
  • M. Pei et al.

    Expansion on matrix deposited by nonchondrogenic urine stem cells strengthens repeated passage bone marrow stromal cells’ chondrogenic capacity

    Cell Tissue Res

    (2014)
  • F. He et al.

    Rejuvenation of nucleus pulposus cells using extracellular matrix deposited by synovium-derived stem cells

    Spine

    (2012)
  • M. Pei et al.

    Extracellular matrix deposited by synovium-derived stem cells delays chondrocyte dedifferentiation and enhances redifferentiation

    J Cell Physiol

    (2012)
  • Cited by (29)

    • Repair of osteochondral defects mediated by double-layer scaffolds with natural osteochondral-biomimetic microenvironment and interface

      2022, Materials Today Bio
      Citation Excerpt :

      As shown in Fig. 6, for chondrogenic induction, the results showed that 3378 DEGs were detected after culture in the ACM-added medium, with 2101 genes up-regulated and 1277 genes down-regulated (Fig. 6B1). KEGG pathway analysis and GO functional enrichment of up-regulated DEGs results (Fig. 6C1, D1, Table S2A) found that several cartilage-related pathways and bioprocesses, as well as cartilage-specific genes (COL II, ACAN, and SOX9), were significantly activated, such as the MAPK signaling pathway [51–53], cartilage development, glycosaminoglycan biosynthetic process, and negative regulation of smooth muscle cell proliferation. These signaling pathways, bioprocesses, and specific genes were related to the positive regulation of the chondrogenic differentiation, cartilage development, ECM synthesis, and negative regulation of angiogenesis, implying that ACM powder promoted chondrogenic differentiation and cartilage regeneration of BMSCs and inhibited the development of blood vessel, which benefited enhancing the stability of BMSC-regenerated cartilage.

    • Synovium stem cell-derived matrix enhances anti-inflammatory properties of rabbit articular chondrocytes via the SIRT1 pathway

      2020, Materials Science and Engineering C
      Citation Excerpt :

      Compared with tissue-derived DECM, cell-derived DECM has recently gained a lot of interest as an instructive culture platform for in vitro MSC expansion, which does not have the issues of limited organ availability, disease transmission, donor-site morbidity, or immune reactions. More interestingly, DECM-expanded MSCs showed a superior lineage-specific differentiation potential than those cultured on traditional culture substrates (i.e., TCPS or coatings of individual matrix proteins on plastic surfaces), because native ECM provides a complex of structural, physical, biochemical signals that cannot be substituted for simple substrates [25]. When culturing on the DECM, articular chondrocytes maintained their original round morphology and the different stiffness between the DECM and TCPS might be responsible for the alternation of chondrocyte phenotype.

    • Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources

      2018, Acta Biomaterialia
      Citation Excerpt :

      Interestingly, some studies showed higher levels of chondrogenesis with nanofiber scaffolds compared with microfiber scaffolds [125,131], despite contrasting studies showing that chondrogenic gene expression was significantly enhanced on microfiber scaffolds compared with nanofiber scaffolds [134,135]. Recent studies have demonstrated that chondrocytes/stem cells cultured on cartilage T-dECMs and C-dECMs, which were made of nanoscale ECM fibers, exerted obvious cell proliferation and chondrogenic differentiation [31,140,141], also indicating that fiber diameter may be attributed to the differences in chondrogenesis. Increasing evidence shows that fiber diameter could influence scaffold pore size which, in turn, affects chondrogenesis.

    View all citing articles on Scopus
    View full text