Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Generation of articular chondrocytes from human pluripotent stem cells

Abstract

The replacement of articular cartilage through transplantation of chondrogenic cells or preformed cartilage tissue represents a potential new avenue for the treatment of degenerative joint diseases. Although many studies have described differentiation of human pluripotent stem cells (hPSCs) to the chondrogenic lineage, the generation of chondrocytes able to produce stable articular cartilage in vivo has not been demonstrated. Here we show that activation of the TGFβ pathway in hPSC-derived chondrogenic progenitors promotes the efficient development of articular chondrocytes that can form stable cartilage tissue in vitro and in vivo. In contrast, chondrocytes specified by BMP4 signaling display characteristics of hypertrophy and give rise to cartilage tissues that initiate the endochondral ossification process in vivo. These findings provide a simple serum-free and efficient approach for the routine generation of hPSC-derived articular chondrocytes for modeling diseases of the joint and developing cell therapy approaches to treat them.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Generation of paraxial mesoderm and chondrocyte progenitors from hPSCs.
Figure 2: TGFβ3 and BMP4 specify distinct populations of chondrocytes and cartilage tissues.
Figure 3: Comparison of hESC-derived cartilage tissues and the developing fetal femur.
Figure 4: hPSC-derived chondrocytes generate cartilage tissue in vivo.
Figure 5: hPSC-derived chondrocytes maintain articular and hypertrophic phenotypes in vivo.
Figure 6: hPSC-derived articular-like cartilage responds to IL1β.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

References

  1. Archer, C.W., Dowthwaite, G.P. & Francis-West, P. Development of synovial joints. Birth Defects Res. C Embryo Today 69, 144–155 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Colnot, C. Cellular and molecular interactions regulating skeletogenesis. J. Cell. Biochem. 95, 688–697 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Pacifici, M. et al. Cellular and molecular mechanisms of synovial joint and articular cartilage formation. Ann. NY Acad. Sci. 1068, 74–86 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Pelttari, K. et al. 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. Arthritis Rheum. 54, 3254–3266 (2006).

    Article  CAS  PubMed  Google Scholar 

  5. Pelttari, K., Steck, E. & Richter, W. The use of mesenchymal stem cells for chondrogenesis. Injury 39 (suppl. 1), S58–S65 (2008).

    Article  PubMed  Google Scholar 

  6. Steinert, A.F. et al. Major biological obstacles for persistent cell-based regeneration of articular cartilage. Arthritis Res. Ther. 9, 213 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Murry, C.E. & Keller, G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132, 661–680 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Oldershaw, R.A. et al. Directed differentiation of human embryonic stem cells toward chondrocytes. Nat. Biotechnol. 28, 1187–1194 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Umeda, K. et al. Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci. Rep. 2, 455 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Hwang, N.S. et al. In vivo commitment and functional tissue regeneration using human embryonic stem cell-derived mesenchymal cells. Proc. Natl. Acad. Sci. USA 105, 20641–20646 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Toh, W.S. et al. Cartilage repair using hyaluronan hydrogel-encapsulated human embryonic stem cell-derived chondrogenic cells. Biomaterials 31, 6968–6980 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Koyama, E. et al. A distinct cohort of progenitor cells participates in synovial joint and articular cartilage formation during mouse limb skeletogenesis. Dev. Biol. 316, 62–73 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Spagnoli, A. et al. TGF-beta signaling is essential for joint morphogenesis. J. Cell Biol. 177, 1105–1117 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, T. et al. Joint TGF-beta type II receptor-expressing cells: ontogeny and characterization as joint progenitors. Stem Cells Dev. 22, 1342–1359 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Serra, R. et al. Expression of a truncated, kinase-defective TGF-beta type II receptor in mouse skeletal tissue promotes terminal chondrocyte differentiation and osteoarthritis. J. Cell Biol. 139, 541–552 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Evseenko, D. et al. Mapping the first stages of mesoderm commitment during differentiation of human embryonic stem cells. Proc. Natl. Acad. Sci. USA 107, 13742–13747 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Craft, A.M. et al. Specification of chondrocytes and cartilage tissues from embryonic stem cells. Development 140, 2597–2610 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Kattman, S.J. et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8, 228–240 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Tanaka, M. et al. BMP inhibition stimulates WNT-dependent generation of chondrogenic mesoderm from embryonic stem cells. Stem Cell Res. (Amst.) 3, 126–141 (2009).

    Article  CAS  Google Scholar 

  20. Roark, E.F. & Greer, K. Transforming growth factor-beta and bone morphogenetic protein-2 act by distinct mechanisms to promote chick limb cartilage differentiation in vitro. Dev. Dyn. 200, 103–116 (1994).

    Article  CAS  PubMed  Google Scholar 

  21. Mackay, A.M. et al. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng. 4, 415–428 (1998).

    Article  CAS  PubMed  Google Scholar 

  22. Enomoto-Iwamoto, M. et al. Bone morphogenetic protein signaling is required for maintenance of differentiated phenotype, control of proliferation, and hypertrophy in chondrocytes. J. Cell Biol. 140, 409–418 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Volk, S.W., Luvalle, P., Leask, T. & Leboy, P.S.A. BMP responsive transcriptional region in the chicken type X collagen gene. J. Bone Miner. Res. 13, 1521–1529 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. Kronenberg, H.M. & Chung, U. The parathyroid hormone-related protein and Indian hedgehog feedback loop in the growth plate. Novartis Foundation symposium 232, 144–152 discussion 152–147 (2001).

    CAS  PubMed  Google Scholar 

  25. Bernardo, B.C. et al. Cartilage intermediate layer protein 2 (CILP-2) is expressed in articular and meniscal cartilage and down-regulated in experimental osteoarthritis. J. Biol. Chem. 286, 37758–37767 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Koch, M. et al. A novel marker of tissue junctions, collagen XXII. J. Biol. Chem. 279, 22514–22521 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Iwamoto, M. et al. Transcription factor ERG and joint and articular cartilage formation during mouse limb and spine skeletogenesis. Dev. Biol. 305, 40–51 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Guo, S. et al. Missense mutations in IHH impair Indian Hedgehog signaling in C3H10T1/2 cells: Implications for brachydactyly type A1, and new targets for Hedgehog signaling. Cell. Mol. Biol. Lett. 15, 153–176 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Zhang, Q. et al. Expression of doublecortin reveals articular chondrocyte lineage in mouse embryonic limbs. Genesis 49, 75–82 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Fukunaga, T. et al. Connective tissue growth factor mRNA expression pattern in cartilages is associated with their type I collagen expression. Bone 33, 911–918 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Wang, X., Li, F., Fan, C., Wang, C. & Ruan, H. Effects and relationship of ERK1 and ERK2 in interleukin-1beta-induced alterations in MMP3, MMP13, type II collagen and aggrecan expression in human chondrocytes. Int. J. Mol. Med. 27, 583–589 (2011).

    CAS  PubMed  Google Scholar 

  32. Goldring, M.B. et al. Roles of inflammatory and anabolic cytokines in cartilage metabolism: signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur. Cell. Mater. 21, 202–220 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Janicki, P., Kasten, P., Kleinschmidt, K., Luginbuehl, R. & Richter, W. Chondrogenic pre-induction of human mesenchymal stem cells on beta-TCP: enhanced bone quality by endochondral heterotopic bone formation. Acta Biomater. 6, 3292–3301 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Farrell, E. et al. In-vivo generation of bone via endochondral ossification by in-vitro chondrogenic priming of adult human and rat mesenchymal stem cells. BMC Musculoskelet. Disord. 12, 31 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Scotti, C. et al. Engineering of a functional bone organ through endochondral ossification. Proc. Natl. Acad. Sci. USA 110, 3997–4002 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sasano, Y. et al. Chondrocytes synthesize type I collagen and accumulate the protein in the matrix during development of rat tibial articular cartilage. Anat. Embryol. (Berl.) 194, 247–252 (1996).

    Article  CAS  Google Scholar 

  37. Kennedy, M., D'Souza, S.L., Lynch-Kattman, M., Schwantz, S. & Keller, G. Development of the hemangioblast defines the onset of hematopoiesis in human ES cell differentiation cultures. Blood 109, 2679–2687 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Witty, A.D. et al. Generation of the epicardial lineage from human pluripotent stem cells. Nat. Biotechnol. 32, 1026–1035 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Park, I.H. et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141–146 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Yang, L. et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 453, 524–528 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Saeed, A.I. et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34, 374–378 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Taylor for providing primary human chondrocyte samples, H. Whetstone for histological expertise and helpful discussions, and A. Grigoriadis, F. Beier and members of the Keller laboratory for critical reading of the manuscript. We thank T. Araki and B. Neel (Ontario Cancer Institute, Toronto) for the iBJ hiPSC line and M. Warman (Boston Children's Hospital) and G. Jay (Lifespan Health System, RI) for the lubricin antibody. This work was supported by a grant from Canadian Institutes of Health Research (MOP 219710) to B.A.A. and G.M.K., and a generous contribution from the Krembil Foundation to A.M.C. and G.M.K.

Author information

Authors and Affiliations

Authors

Contributions

A.M.C. jointly conceived the study, interpreted results and wrote the paper with G.M.K. A.M.C. also designed and performed experiments and analyzed data; J.S.R. analyzed data and discussed results; Y.N. performed experiments and analyzed data; B.A.A. and R.A.K. provided primary human samples, gave conceptual advice, discussed results and edited the manuscript.

Corresponding authors

Correspondence to April M Craft or Gordon M Keller.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of paraxial mesoderm from the H7 hESC line.

H7 hESCs were differentiated as shown in Figure 1a. (a) Flow cytometric analyses of the proportion of CD56+, PDGFRα+, and KDR+ cells in day 4 EBs. (b) Flow cytometric analyses showing the proportion of CD73+, CD105+, and PDGFRβ+ cells in day 15 populations specified as indicated (MOCK indicates no DM and no bFGF). qRT-PCR-based expression analyses of indicated genes on day 8 (c-d) and 15 (e-h) of culture. Each point represents the copy number mRNA relative to TBP (n = 3 biological replicates), the line indicates the mean. (i-j) Percentage of cardiac troponin T (cTnT) positive cells on day 15 of culture was measured by intracellular flow cytometry. (i) Representative contour plots of day 15 cultures treated with indicated factors. (j) Quantification of cTnT-positive cells from three independent experiments (points), the line indicates the mean. Significance was calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 2 Generation of paraxial mesoderm from human induced pluripotent stem cells (iBJ hiPSCs).

iBJ hiPSCs were differentiated with the following modifications of the protocol shown in Figure 1a; the Wnt pathway agonist CHIR99061 (1 μM) was added to the stage 1 cultures and stage 1 was shortened from 3 to 2 days. The paraxial mesoderm fate was specified in the monolayer cultures by treatment with Dorsomorphin (DM) from day 3 to day 5, and FGF from day 3 to day 14 (Stage 2). Experimental cell treatment(s) during stage 2 include: no additional factors (MOCK), and Dorsomorphin and bFGF (DM+FGF). Inhibition of the Wnt signaling pathway from day 3 to 5 improved the efficiency of paraxial mesoderm specification in some cases. (a) Flow cytometric analyses of the proportion of CD56+, PDGFRα+, and KDR+ cells in day 3 EBs. qRT-PCR-based expression analyses of indicated genes on day 6 (b, n = 4) and 15 (c,d; MOCK n = 3, DM+FGF n = 5) of culture. Boxplots depict the copy number mRNA relative to TBP. The box extends from the 25th to 75th percentiles, the line indicates the median, and the Tukey method was used to plot the whiskers and the outliers. (e) Flow cytometric analyses showing the proportion of CD73+, CD105+, PDGFRβ+, and CD31+ cells in day 14 populations specified as indicated. (f) Intracellular flow cytometric analysis of the percentage of cardiac troponin T+ cardiomyocytes in the day 14 monolayer populations. Points on graph indicate the average percentage of troponin T+ cells in the day 14 populations for 3 independent experiments, the line indicates the mean. Significance between Mock and DM+FGF groups was calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 3 Generation of paraxial mesoderm and chondrocytes from human induced pluripotent stem cells (Y2-1 line).

Y2-1 hiPSCs were differentiated with the following modifications of the protocol shown in Figure 1a; stage 1 was shortened from 3 to 2 days. The paraxial mesoderm fate was specified in the monolayer cultures by treatment with Dorsomorphin (DM) from day 3 to day 5, and FGF from day 3 to day 14 (Stage 2). Experimental cell treatment(s) during stage 2 include: no additional factors (MOCK), and Dorsomorphin and bFGF (DM+FGF). Inhibition of the Wnt signaling pathway from day 3 to 5 improved the efficiency of paraxial mesoderm specification in some cases. (a) Flow cytometric analyses of the proportion of CD56+, PDGFRα+, and KDR+ cells in day 3 EBs. (b) qRT-PCR-based expression analyses of indicated genes on day 8 of culture. Boxplots depict the copy number mRNA relative to TBP (MOCK, n = 8; DM+FGF, n = 10). (c) Flow cytometric analyses showing the proportion of CD73+, CD105+, and PDGFRβ+ cells in day 14 populations specified as indicated. (d) qRT-PCR-based expression analyses of indicated genes on day 14 of culture. Boxplots depict relative gene expression (MOCK, n = 4; DM+FGF, n = 5). The box extends from the 25th to 75th percentiles, the line indicates the median, and the Tukey method was used to plot the whiskers and the outliers. (e) Intracellular flow cytometric analysis of the percentage of cardiac troponin T+ cardiomyocytes in the day 14 monolayer populations. Histological images of micromass tissues derived from Y2-1 hiPSCs in the presence of TGFβ3 (f) or BMP4 (g) for 12 weeks. Sections stained metachromatically with Toluidine blue dye. Scale bar, 200 μm. (h-r) qRT-PCR-based expression analyses of indicated genes in micromass tissue cultured in the presence of TGFβ3 or BMP4 for 6 weeks (h-o) or 12 weeks (p-r). Points represent copy number mRNA relative to TBP (n = 3), lines indicate the mean. Significance was calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 4 TGFβ3 and BMP4 specify distinct populations of chondrocytes and cartilage tissues from hiPSC-derived progenitors (iBJ hiPSC line).

(a) Photomicrographs of chondrocytes generated following 6 weeks of culture in TGFβ3 or BMP4. Scale bar, 100 μm. (b) Flow cytometric analysis of forward (cell size) and side scatter (cell granularity) parameters of chondrocytes isolated from 12-week-old tissue. (c-n) qRT-PCR-based expression analyses of indicated genes in micromass tissue cultured in the presence of TGFβ3 or BMP4 for 6 weeks (c-k, n = 4) or 10 weeks (l-n, n = 5). Boxplots depict the copy number mRNA relative to TBP. The box extends from the 25th to 75th percentiles, the line indicates the median, and the Tukey method was used to plot the whiskers and the outliers. Significance calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 5 Primary human fetal chondrocytes generate cartilage tissues in vitro.

Primary fetal (F) chondrocytes (passage 0) isolated from the knee joints at 16.5 and 18 weeks of gestation were cultured as micromass in the presence of TGFβ3 or BMP4 (a) Flow cytometric analysis of forward (FSC-A) and side (SSC-A) scatter parameters of primary fetal chondrocytes isolated from micromass tissue cultured in TGFβ3 or BMP4 for 12 weeks. (b-t) qRT-PCR-based expression analyses of indicated genes in micromass tissue at 1, 4, 6, 8, and 12 weeks of culture. Values represent copy number mRNA relative to TBP (n = 6 from two biologically independent experiments). Bars represent standard error of the mean. Significance calculated by student’s t-test and compared TGFβ3 treated micromass cultures to BMP4-treated micromass cultures at indicated time points. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 6 hESC-derived paraxial/chondrogenic mesoderm expresses CD73, CD105 and PDGFRβ.

(a) Flow cytometric analyses of DM+FGF-treated monolayer cultures during stage 2 of differentiation (T6 to T15). T (time) indicates day of differentiation. (b) Cells that expressed the combination of CD73 and either CD105 or PDGFRβ generated tissues in vitro. Double positive (CD73+CD105+ and CD73+PDGFRβ+) populations from day 15 (T15) cultures were isolated from double negative populations by cell sorting and cultured as micromass. Micrographs of cells derived from sorted populations after 14 days (d, Scale bar, 200 μm) of micromass culture. (c) Tissues derived from sorted populations after 5 weeks of micromass culture in standard 24-well tissue culture plates.

Supplementary Figure 7 Gene expression analyses of hESC-derived chondrocytes (HES2 hESC line).

qRT-PCR-based expression analyses of indicated genes in day 15 populations and derivative micromass tissue at different times. Values represent copy number mRNA relative to TBP (n = 4-9 biological replicates), and are compared to primary fetal chondrocytes (aged 16-19 weeks, n = 4), primary healthy adult articular chondrocytes (n = 2), and hypertrophic chondrocytes isolated from the iliac crest of a pediatric patient (n = 1). T15 Mesoderm indicates day 15 hESC-derived paraxial mesoderm (DM+FGF-treated). Bars represent standard error of the mean. n.d. indicates no data due to unavailability of primary tissue sample. Significance between TGFβ3 and BMP4 treatments at indicated time points was calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 8 Proliferation and apoptosis during micromass culture.

(a) Growth curve of hPSC-derived cells during micromass culture (Stage 4). The initial number of cells plated in micromass for each experiment was 200,000 (week 0). Micromass tissues were digested with 0.2% collagenase for 1-4 hours at 37 degrees Celcius at indicated timepoints (n 3 biologically independent samples per timepoint). Bars indicate standard error of the mean. Significance calculated by student’s t-test, **p<0.01, ***p<0.001. Analysis of BMP4-treated cultures began at 2 weeks of culture (approximately 4 days after BMP4 treatment began). (b) Apoptotic cells were quantified by annexin V staining using flow cytometry after 10 weeks of micromass culture. Representative contour plots are shown.

Supplementary Figure 9 Generation of chondrocytes from the H7 hESC line.

H7 hESCs were differentiated as shown in Figure 1a. (a-m) qRT-PCR-based expression analyses of indicated genes in micromass tissue cultured in the presence of TGFβ3 or BMP4 for 6 weeks. Boxplots depict the copy number mRNA relative to TBP (n = 4 biological replicates). The box extends from the 25th to 75th percentiles, the line indicates the median, and the Tukey method was used to plot the whiskers and the outliers. Significance was calculated by student’s t-test. *p<0.05, **p<0.01, ***p<0.001.

Supplementary Figure 10 TGFβ1, TGFβ2, and TGFβ3 have similar effects on generating articular chondrocytes from hPSC-derived paraxial mesoderm.

COL2A1, PRG4, and CILP2 gene expression after 12 weeks of micromass culture in the presence of TGFβ agonists as indicated (10 ng/ml). Points on graphs indicate the copy number mRNA relative to TBP (n = 3). Lines indicate the mean. Significance was calculated by one-way ANOVA followed by Tukey’s post-hoc test (no significant differences were observed).

Supplementary Figure 11 hESC-derived chondrocytes derived in the presence of GDF5 do not acquire an articular chondrocyte phenotype.

(a-d) qRT-PCR-based expression analyses of indicated genes after 6 weeks of micromass culture in the presence of indicated factors. Day 15 DM+FGF treated chondrogenic mesoderm was plated in micromass culture in the presence of TGFβ3 for 10 days. Micromasses were then either maintained in TGFβ3-containing media or switched to media containing BMP4 (50 ng/ml), GDF5 (50 ng/ml) or a combination (G1aC) of GDF5, soluble BMPR1a (500 ng/ml) and cyclopamine (2.5 μM; a Hedgehog pathway inhibitor) for 5 additional weeks. Points on graphs indicate the copy number mRNA relative to TBP (n = 3 biological replicates), lines indicate the mean and error bars indicate the standard deviation. Significance calculated by one-way ANOVA followed by Tukey’s post hoc test. *p<0.05, **p<0.01, ***p<0.001. Toluidine blue staining of cartilage tissues derived in the presence of GDF5 (e) or G1aC (f) for 6 weeks. Scale bar, 200 μm.

Supplementary Figure 12 BMP4-treated chondrocytes have the capacity to generate grafts containing developing bone ossicles and hematopoietic cells after 12 weeks in vivo.

Hematoxylin and eosin (H & E; a-c) and TRAP (d, e) staining of graft 2 shown in Figure 5. (d, e) TRAP staining indicates the presence of osteoclasts (purple stain). Double arrowheads indicate hypertrophic chondrocytes, grey filled arrowheads indicate developing bone ossicles, black filled arrows indicate hematopoietic cells, black stylized arrowheads indicate erythrocytes (red blood cells). Scale bar (a) 200 μm, (b, d) 100 μm, (c, e) 50 μm.

Supplementary Figure 13 Presence of osteoclasts and vascularization within grafts generated by BMP4-treated chondrocytes after 12 weeks.

Grafts generated by TGFβ3-treated chondrocytes (a, b, f, g) and BMP4-treated chondrocytes (c-e, h-j) were analyzed histologically after 12 weeks. Two representative high magnification fields are shown for BMP4 grafts. (a-e) Identification of osteoclasts by TRAP staining (purple, arrows). (f-j) The presence of CD31 (PECAM)-positive endothelial cells by immunohistochemistry (brown staining) indicates vascularization of the graft. No TRAP-positive or CD31-positive cells were observed in grafts generated by TGFβ3-treated chondrocytes. Scale bar (a, c, f, g), 100 μm; Scale bar (b, d, e, g, i, j), 50 μm.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 (PDF 9198 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Craft, A., Rockel, J., Nartiss, Y. et al. Generation of articular chondrocytes from human pluripotent stem cells. Nat Biotechnol 33, 638–645 (2015). https://doi.org/10.1038/nbt.3210

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3210

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing