6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Recapitulation of physiological spatiotemporal signals promotes in vitro formation of phenotypically stable human articular cartilage

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Despite significant efforts, stable and organized human cartilage has not been grown from human mesenchymal stem cells in vitro. We report the formation of organized cartilage discs that resemble the articular cartilage from self-assembling human mesenchymal stem cells by implementing spatiotemporal regulation in vitro that mimics native development. Selective application of chondrogenic and hypertrophic induction regimens enabled the maintenance of functional hyaline cartilage and progressive deep-zone mineralization. We demonstrate that this simple biomimetic approach helped mature the cartilage discs and enabled them to remain stable and organized following implantation. These findings highlight the limitations of current isotropic culture, and could greatly accelerate the development of new therapeutic modalities for cartilage repair from a patient’s own cells.

          Abstract

          Standard isotropic culture fails to recapitulate the spatiotemporal gradients present during native development. Cartilage grown from human mesenchymal stem cells (hMSCs) is poorly organized and unstable in vivo. We report that human cartilage with physiologic organization and in vivo stability can be grown in vitro from self-assembling hMSCs by implementing spatiotemporal regulation during induction. Self-assembling hMSCs formed cartilage discs in Transwell inserts following isotropic chondrogenic induction with transforming growth factor β to set up a dual-compartment culture. Following a switch in the basal compartment to a hypertrophic regimen with thyroxine, the cartilage discs underwent progressive deep-zone hypertrophy and mineralization. Concurrent chondrogenic induction in the apical compartment enabled the maintenance of functional and hyaline cartilage. Cartilage homeostasis, chondrocyte maturation, and terminal differentiation markers were all up-regulated versus isotropic control groups. We assessed the in vivo stability of the cartilage formed under different induction regimens. Cartilage formed under spatiotemporal regulation in vitro resisted endochondral ossification, retained the expression of cartilage markers, and remained organized following s.c. implantation in immunocompromised mice. In contrast, the isotropic control groups underwent endochondral ossification. Cartilage formed from hMSCs remained stable and organized in vivo. Spatiotemporal regulation during induction in vitro recapitulated some aspects of native cartilage development, and potentiated the maturation of self-assembling hMSCs into stable and organized cartilage resembling the native articular cartilage.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: found
          • Article: not found

          The control of chondrogenesis.

          Chondrogenesis is the earliest phase of skeletal development, involving mesenchymal cell recruitment and migration, condensation of progenitors, and chondrocyte differentiation, and maturation and resulting in the formation of cartilage and bone during endochondral ossification. This process is controlled exquisitely by cellular interactions with the surrounding matrix, growth and differentiation factors, and other environmental factors that initiate or suppress cellular signaling pathways and transcription of specific genes in a temporal-spatial manner. Vertebrate limb development is controlled by interacting patterning systems involving prominently the fibroblast growth factor (FGF), bone morphogenetic protein (BMP), and hedgehog pathways. Both positive and negative signaling kinases and transcription factors, such as Sox9 and Runx2, and interactions among them determine whether the differentiated chondrocytes remain within cartilage elements in articular joints or undergo hypertrophic maturation prior to ossification. The latter process requires extracellular matrix remodeling and vascularization controlled by mechanisms that are not understood completely. Recent work has revealed novel roles for mediators such as GADD45beta, transcription factors of the Dlx, bHLH, leucine zipper, and AP-1 families, and the Wnt/beta-catenin pathway that interact at different stages during chondrogenesis. (c) 2005 Wiley-Liss, Inc.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Premature induction of hypertrophy during in vitro chondrogenesis of human mesenchymal stem cells correlates with calcification and vascular invasion after ectopic transplantation in SCID mice.

            Functional suitability and phenotypic stability of ectopic transplants are crucial factors in the clinical application of mesenchymal stem cells (MSCs) for articular cartilage repair, and might require a stringent control of chondrogenic differentiation. This study evaluated whether human bone marrow-derived MSCs adopt natural differentiation stages during induction of chondrogenesis in vitro, and whether they can form ectopic stable cartilage that is resistant to vascular invasion and calcification in vivo. During in vitro chondrogenesis of MSCs, the expression of 44 cartilage-, stem cell-, and bone-related genes and the deposition of aggrecan and types II and X collagen were determined. Similarly treated, expanded articular chondrocytes served as controls. MSC pellets were allowed to differentiate in chondrogenic medium for 3-7 weeks, after which the chondrocytes were implanted subcutaneously into SCID mice; after 4 weeks in vivo, samples were evaluated by histology. The 3-stage chondrogenic differentiation cascade initiated in MSCs was primarily characterized by sequential up-regulation of common cartilage genes. Premature induction of hypertrophy-related molecules (type X collagen and matrix metalloproteinase 13) occurred before production of type II collagen and was followed by up-regulation of alkaline phosphatase activity. In contrast, hypertrophy-associated genes were not induced in chondrocyte controls. Whereas control chondrocyte pellets resisted calcification and vascular invasion in vivo, most MSC pellets mineralized, in spite of persisting proteoglycan and type II collagen content. An unnatural pathway of differentiation to chondrocyte-like cells was induced in MSCs by common in vitro protocols. MSC pellets transplanted to ectopic sites in SCID mice underwent alterations related to endochondral ossification rather than adopting a stable chondrogenic phenotype. Further studies are needed to evaluate whether a more stringent control of MSC differentiation to chondrocytes can be achieved during cartilage repair in a natural joint environment.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Functional characterization of hypertrophy in chondrogenesis of human mesenchymal stem cells.

              Mesenchymal stem cells (MSCs) are promising candidate cells for cartilage tissue engineering. Expression of cartilage hypertrophy markers (e.g., type X collagen) by MSCs undergoing chondrogenesis raises concern for a tissue engineering application for MSCs, because hypertrophy would result in apoptosis and ossification. To analyze the biologic basis of MSC hypertrophy, we examined the response of chondrifying MSCs to culture conditions known to influence chondrocyte hypertrophy, using an array of hypertrophy-associated markers. Human MSC pellet cultures were predifferentiated for 2 weeks in a chondrogenic medium, and hypertrophy was induced by withdrawing transforming growth factor beta (TGFbeta), reducing the concentration of dexamethasone, and adding thyroid hormone (T3). Cultures were characterized by histologic, immunohistochemical, and biochemical methods, and gene expression was assessed using quantitative reverse transcription-polymerase chain reaction. The combination of TGFbeta withdrawal, a reduction in the level of dexamethasone, and the addition of T3 was essential for hypertrophy induction. Cytomorphologic changes were accompanied by increased alkaline phosphatase activity, matrix mineralization, and changes in various markers of hypertrophy, including type X collagen, fibroblast growth factor receptors 1-3, parathyroid hormone-related protein receptor, retinoic acid receptor gamma, matrix metalloproteinase 13, Indian hedgehog, osteocalcin, and the proapoptotic gene p53. However, hypertrophy was not induced uniformly throughout the pellet culture, and distinct regions of dedifferentiation were observed. Chondrogenically differentiating MSCs behave in a manner functionally similar to that of growth plate chondrocytes, expressing a very similar hypertrophic phenotype. Under the in vitro culture conditions used here, MSC-derived chondrocytes underwent a differentiation program analogous to that observed during endochondral embryonic skeletal development, with the potential for terminal differentiation. This culture system is applicable for the screening of hypertrophy-inhibitory conditions and agents that may be useful to enhance MSC performance in cartilage tissue engineering.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                7 March 2017
                22 February 2017
                : 114
                : 10
                : 2556-2561
                Affiliations
                [1] aDepartment of Biomedical Engineering, Columbia University , New York, NY 10032;
                [2] bDepartment of Medicine, Columbia University , New York, NY 10032
                Author notes
                2To whom correspondence should be addressed. Email: gv2131@ 123456columbia.edu .

                Edited by Kristi S. Anseth, Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO, and approved January 25, 2017 (received for review July 17, 2016)

                Author contributions: J.J.N. and G.V.-N. designed research; J.J.N., Y.W., B.Z., S.R., and A.B. performed research; J.J.N., Y.W., B.Z., J.B., S.R., X.E.G., and G.V.-N. analyzed data; and J.J.N. and G.V.-N. wrote the paper.

                1J.J.N. and Y.W. contributed equally to this work.

                Author information
                http://orcid.org/0000-0002-5170-154X
                http://orcid.org/0000-0002-9382-1574
                Article
                PMC5347558 PMC5347558 5347558 201611771
                10.1073/pnas.1611771114
                5347558
                28228529
                7f34ae94-b349-46b7-89fb-ab2f962f7f27
                History
                Page count
                Pages: 6
                Funding
                Funded by: HHS | NIH | National Institute of Biomedical Imaging and Bioengineering (NIBIB) 100000070
                Award ID: EB002520
                Award ID: EB015888
                Funded by: NIDCR
                Award ID: DE016525
                Funded by: NIAMS
                Award ID: AR061988
                Funded by: ASTAR
                Award ID: NSS scholarship
                Categories
                Biological Sciences
                Applied Biological Sciences

                tissue engineering,cartilage development,regenerative medicine,biomimetic,cartilage repair

                Comments

                Comment on this article