AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (10.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Tppp3+ synovial/tendon sheath progenitor cells contribute to heterotopic bone after trauma

Ji-Hye Yea1,Mario Gomez-Salazar1,Sharon Onggo1Zhao Li1Neelima Thottappillil1Masnsen Cherief1Stefano Negri1,2Xin Xing1Qizhi Qin1Robert Joel Tower3Chen-Ming Fan4Benjamin Levi3Aaron W. James1 ( )
Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
Orthopaedic and Trauma Surgery Unit, Department of Surgery, Dentistry, Paediatrics and Gynaecology of the University of Verona, Verona, Italy
Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, USA
Carnegie Institution for Science, Baltimore, MD, USA

These authors contributed equally: Ji-Hye Yea, Mario Gomez-Salazar

Show Author Information

Abstract

Heterotopic ossification (HO) is a pathological process resulting in aberrant bone formation and often involves synovial lined tissues. During this process, mesenchymal progenitor cells undergo endochondral ossification. Nonetheless, the specific cell phenotypes and mechanisms driving this process are not well understood, in part due to the high degree of heterogeneity of the progenitor cells involved. Here, using a combination of lineage tracing and single-cell RNA sequencing (scRNA-seq), we investigated the extent to which synovial/tendon sheath progenitor cells contribute to heterotopic bone formation. For this purpose, Tppp3 (tubulin polymerization-promoting protein family member 3)-inducible reporter mice were used in combination with either Scx (Scleraxis) or Pdgfra (platelet derived growth factor receptor alpha) reporter mice. Both tendon injury- and arthroplasty-induced mouse experimental HO models were utilized. ScRNA-seq of tendon-associated traumatic HO suggested that Tppp3 is an early progenitor cell marker for either tendon or osteochondral cells. Upon HO induction, Tppp3 reporter+ cells expanded in number and partially contributed to cartilage and bone formation in either tendon- or joint-associated HO. In double reporter animals, both Pdgfra+Tppp3+ and Pdgfra+Tppp3- progenitor cells gave rise to HO-associated cartilage. Finally, analysis of human samples showed a substantial population of TPPP3-expressing cells overlapping with osteogenic markers in areas of heterotopic bone. Overall, these data demonstrate that synovial/tendon sheath progenitor cells undergo aberrant osteochondral differentiation and contribute to HO after trauma.

References

1

Negri, S. et al. Acetabular reaming is a reliable model to produce and characterize periarticular heterotopic ossification of the hip. Stem Cell Transl. Med. 11, 876–888 (2022).

2

McCarthy, E. & Sundaram, M. Heterotopic ossification: a review. Skeletal Radiol. 34, 609–619 (2005).

3

Lin, L., Shen, Q., Xue, T. & Yu, C. Heterotopic ossification induced by Achilles tenotomy via endochondral bone formation: expression of bone and cartilage related genes. Bone 46, 425–431 (2010).

4

Meyers, C. et al. Heterotopic Ossification: A Comprehensive Review. JBMR Plus. 3, e10172 (2019).

5

Wittenberg, R. H., Peschke, U. & Botel, U. Heterotopic ossification after spinal-cord injury-epidemiology and risk-factors. J. Bone Joint Surgery-British Volume 74, 215–218 (1992).

6

Garland, D. E. Clinical observations on fractures and heterotopic ossification in the spinal-cord and traumatic brain injured populations. Clin. Orthop. Relat. R 233, 86–101 (1988).

7

Kan, C. et al. Microenvironmental factors that regulate mesenchymal stem cells: lessons learned from the study of heterotopic ossification. Histol. Histopathol. 32, 977–985 (2017).

8

Nelson, E. R., Wong, V. W., Krebsbach, P. H., Wang, S. C. & Levi, B. Heterotopic ossification following burn injury: the role of stem cells. J. Burn Care Res. 33, 463–470 (2012).

9

Zhang, Q., Zhou, D., Wang, H. T. & Tan, J. Heterotopic ossification of tendon and ligament. J. Cellular Mol. Med. 24, 5428–5437 (2020).

10

Agarwal, S. et al. Scleraxis-lineage cells contribute to ectopic bone formation in muscle and tendon. Stem Cells 35, 705–710 (2017).

11

Pagani, C. A. et al. Novel lineage-tracing system to identify site-specific ectopic bone precursor cells. Stem Cell Rep.16, 626–640 (2021).

12

Regard, J. B. et al. Activation of hedgehog signaling by loss of GNAS causes heterotopic ossification. Nat. Med. 19, 1505–1512 (2013).

13

Dey, D. et al. Two tissue-resident progenitor lineages drive distinct phenotypes of heterotopic ossification. Sci. Transl. Medicine 8, 366ra163 (2016).

14

Kan, C. et al. Gli1-labeled adult mesenchymal stem/progenitor cells and hedgehog signaling contribute to endochondral heterotopic ossification. Bone 109, 71–79 (2018).

15

Farahani, R. M. & Xaymardan, M. Platelet-derived growth factor receptor alpha as a marker of mesenchymal stem cells in development and stem cell biology. Stem Cells Int. 2015, 362753 (2015).

16

Bi, Y. et al. Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat. Med. 13, 1219–1227 (2007).

17

Lee, C. H. et al. Harnessing endogenous stem/progenitor cells for tendon regeneration. J. Clinical Invest. 125, 2690–2701 (2015).

18

Gumucio, J. P., Schonk, M. M., Kharaz, Y. A., Comerford, E. & Mendias, C. L. Scleraxis is required for the growth of adult tendons in response to mechanical loading. JCI Insight 5, e138295 (2020).

19

Harvey, T., Flamenco, S. & Fan, C. M. A Tppp3(+)Pdgfra(+) tendon stem cell population contributes to regeneration and reveals a shared role for PDGF signalling in regeneration and fibrosis. Nat. Cell Biol. 21, 1490–1503 (2019).

20

Lee, S. Y. et al. NGF-TrkA signaling dictates neural in growth and aberrant osteochondral differentiation after soft tissue trauma. Nat. Commun. 12, 4939 (2021).

21

Staverosky, J. A., Pryce, B. A., Watson, S. S. & Schweitzer, R. Tubulin polymerization‐promoting protein family member 3, Tppp3, is a specific marker of the differentiating tendon sheath and synovial joints. Dev. Dyn. 238, 685–692 (2009).

22

Zhang, Y.-J. et al. Concise review: stem cell fate guided by bioactive molecules for tendon regeneration. Stem Cell Transl. Med. 7, 404–414 (2018).

23

Convente, M. R., Wang, H. T., Pignolo, R. J., Kaplan, F. S. & Shore, E. M. The immunological contribution to heterotopic ossification disorders. Curr. Osteoporos. Rep. 13, 116–124 (2015).

24

Hwang, C. et al. Mesenchymal VEGFA induces aberrant differentiation in heterotopic ossification. Bone Res. 7, 36 (2019).

25

Yang, L., Tsang, K. Y., Tang, H. C., Chan, D. & Cheah, K. S. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc. Natl. Acad. Sci. 111, 12097–12102 (2014).

26

Tarafder, S. et al. Tendon stem/progenitor cells regulate inflammation in tendon healing via JNK and STAT3 signaling. Faseb J. 31, 3991–3998 (2017).

27

Hsu, G. C. Y. et al. Endogenous CCN family member WISP1 inhibits traumainduced heterotopic ossification. JCI Insight 5, e135432 (2020).

28

Yea, J.-H., Bae, T. S., Kim, B. J., Cho, Y. W. & Jo, C. H. Regeneration of the Rotator Cuff Tendon-to-Bone Interface using Umbilical Cord-Derived Mesenchymal Stem Cells and Gradient Extracellular Matrix Scaffolds from Adipose Tissue in a Rat Model. Acta Biomater. 114, 104–116 (2020).

29

Yea, J.-H. et al. Regeneration of a full-thickness defect of rotator cuff tendon with freshly thawed umbilical cord-derived mesenchymal stem cells in a rat model. Stem Cell Res. Ther. 11, 1–13 (2020).

30

Yea, J. H. et al. Regeneration of a full-thickness defect in rotator cuff tendon with umbilical cord-derived mesenchymal stem cells in a rat model. PLoS One 15, e0235239 (2020).

31

Qin, Q. Z. et al. Neuron-to-vessel signaling is a required feature of aberrant stem cell commitment after soft tissue trauma. Bone Res. 10, 43 (2022).

32

Qiu, X. J. et al. Reversed graph embedding resolves complex single-cell trajectories. Nat. Methods 14, 979–982 (2017).

33

Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

Bone Research
Article number: 39
Cite this article:
Yea J-H, Gomez-Salazar M, Onggo S, et al. Tppp3+ synovial/tendon sheath progenitor cells contribute to heterotopic bone after trauma. Bone Research, 2023, 11: 39. https://doi.org/10.1038/s41413-023-00272-x

128

Views

1

Downloads

6

Crossref

6

Web of Science

6

Scopus

Altmetrics

Received: 30 December 2022
Revised: 18 May 2023
Accepted: 28 May 2023
Published: 21 July 2023
© The Author(s) 2023

This 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 license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license 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 license, visit http://creativecommons.org/licenses/by/4.0/.

Return