Picking a bone with heterotopic ossification: translational progress current and future
Letter to the Editor

Picking a bone with heterotopic ossification: translational progress current and future

Jonathan R. Peterson1, Shailesh Agarwal2, Shawn J. Loder1, Oluwatobi Eboda1, Paul S. Cederna2, Steven R. Buchman2, Chuanwu Xi3, Stewart C. Wang1, Benjamin Levi2

1Department of Surgery, 2Department of Plastic Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA; 3Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA

Correspondence to: Benjamin Levi, MD. Department of Plastic Surgery, University of Michigan Medical School, 1150 W Medical Center Dr, Room A552 MSRB2, Ann Arbor, MI 48105, USA. Email: blevi@med.umich.edu.

Submitted Jul 24, 2015. Accepted for publication Jul 24, 2015.

doi: 10.3978/j.issn.2305-5839.2015.07.27


Heterotopic ossification (HO) represents a serious clinical and basic science conundrum. It is a frequent complication of some of the most common orthopaedic procedures including joint replacement, fracture repair, and amputations. Furthermore it causes costly debilitation following trauma, burns, brain and spinal injuries, and is one of the defining injuries of modern military conflicts (1). However, despite the commonality and burden of HO, relatively little is known about its mechanism. This leads to a dearth of effective clinical prophylactic and therapeutic strategies.

A recent editorial discussing new additions to the bank of knowledge on HO highlighted some of the questions that remain and were indeed posed by our recent article on targeting ATP hydrolysis and BMP signaling blockade as potential therapeutic pathways to prevent ectopic bone formation (2,3). The purpose of this article is to expound upon some of those questions and lay a framework for some of the future translational research that is critically needed in HO.

Broadly speaking, we know that HO is a reactive process in response to inflammatory insult. Furthermore, because ectopic bone is a highly differentiated tissue that develops in an endochondral fashion and is complete with its own marrow cavity, it likely develops from multipotent stem cells capable of forming bone. We have shown that histologically and spectroscopically HO resembles mature orthotopic bone but, because it is a dysregulated process, there are differences in the mineral content, function, and strength (4). To date there is no clear consensus on the specific identity of the multipotent cells responsible for forming HO, despite many elegant studies suggesting a range of local or circulating progenitors (5).

We have used adipose stromal cells (ASC) as a proxy for the still unidentified HO progenitor cell because it is well-described and easily manipulated mesenchymal stem cell. Hence, we believe that until the HO progenitor is definitively identified, ASCs represent an adequate vehicle for in vitro study of biologic mechanism and osteogenic modifiers. We do not suggest that ASCs are ultimately the primary cells responsible for HO.

One of the interesting results in our study was the effect of a remote burn injury on ASCs distant from the burn site. We saw increased osteogenecity of the ASCs from mice that sustained a burn injury. This highlights the importance of a systemic inflammatory insult to the permissive niche necessary for ectopic bone formation.

These results are consistent with the more clinically relevant data we presented on gene expression in adipose tissue from burn patients in which osteogenic genes, including SMADs and RUNX2, were upregulated within the first 96 hours following burn injury. While we agree with Kan et al. (2) that these samples were not pure MSC populations, the significantly elevated expression of osteogenic genes in a heterogenous population of cells highlights how influential the post-injury inflammatory environment is to the signaling pathways that lead to bone formation. Similarly, in our mouse model of HO formation, which consists of a focused musculoskeletal injury (achilles tenotomy) and a distant burn injury, we noted significant, but not complete, reductions in HO development by treating the burn injury site with a targeted anti-inflammatory drug—apyrase. One important implication of these results is that the initial mitigation of inflammation may be of paramount importance to the prevention of HO.

While the general anti-inflammatory effects of apyrase are certainly contributing to the overall reduction in reactive osteogenicity, we also believe that its specific action—inactivation of ATP through cleavage of phosphate groups—is central to its efficacy in preventing HO while not inhibiting wound healing. We were somewhat surprised at the intracellular signaling effects of apyrase which itself primarily functions extracellularly. Cyclic adenosine monophosphate (cAMP) is known to inhibit phosphorylation of SMAD1/5/8 and thereby prevent osteogenesis (6). Increasing cAMP activity intracellularly has also been seen with phosphodiesterase inhibitors which suppress osteogenic differentiation of MSCs (7,8).

While delineating the precise mechanistic details between extracellular ATP hydrolysis and intracellular cAMP signaling was not a goal of our study, one link was provided by the increase in signaling through adenosine receptors A2A and A2B. These receptors are important for purine metabolism and also for the inflammatory response in general (9). In regards to osteogenic differentiation of MSCs, the A2B receptor has been shown to increase osteogenic tendency. Knockout mice lacking A2B develop osteopenia with poor osteoblast activity (10). Conversely the A2A receptor promotes adipogenesis and blockade of A2A causes increases osteogenicity (11). We saw a reduction in both A2A and A2B in MSCs from mice that had suffered a burn injury, with restitution of these levels following apyrase treatment at the burn site. Clearly there is impetus for further exploration of the effects of these and the other two types of adenosine receptors (A1 and A3) and their differential roles in osteogenicity and ectopic bone formation. In this respect our findings raise more questions than answers as to the specific role of adenosine receptors, however a clinically relevant connection may be found.

While the exact mechanism whereby apyrase increases cAMP levels intracellularly are not fully delineated, the terminal effect resulting in decreased osteogenesis through reduction in SMAD phosphorylation is clear. One of the exciting themes of recent HO research is the developing body of work showing that trauma induced HO has many histologic and mechanistic similarities to genetic forms of HO including fibrodysplasia ossificans progressive (FOP). It has been established that trauma induced HO develops in an endochondral fashion, as is the case in FOP. Much research has been completed showing that ectopic bone in FOP is caused by a point mutation (R206H) in the type 1 BMP receptor ALK2 causing it to be constitutively active (12,13). Similarly, BMP signaling is critical in trauma induced HO. We saw a striking reduction in ectopic bone formation with the application of a small molecule inhibitor of BMP type 1 receptors ALK2 and ALK3-LDN-193189. This compound, which is a derivative of dorsomorphin, was originally developed as a potential treatment for FOP (14). In a similar vein, much of the well-delineated mechanistic details of ectopic bone formation in FOP may be similar to the pathways in trauma-induced HO. Future translational research in reactive bone formation may benefit greatly by taking advantage of the robust body of work already completed on FOP.

There are several major questions of critical importance that still remain to be answered including the identification of the progenitor cell line(s) responsible for HO, understanding the signaling mechanisms responsible for progenitor cell differentiation which link inflammation and bone formation, and understanding how diverse injuries create the permissive niche that induces ectopic bone formation. Some leading hypotheses of progenitor cell etiology include endothelial cells; bone marrow derived mesenchymal cells locally or circulating, macrophages, and circulating hematopoietic cells (15-19).

Elegant studies using tracers for Tie-2 have suggested that at least 50% of HO is derived from vasculogenic precursors, which may be explained by other findings that an endothelial to mesenchymal transition can produce HO (20). Local vasculogenic factors including vascular endothelial growth factor (VEGF), and platelet derived growth factor (PDGF) are crucial components of the permissive niche for HO (16,21). There are also important connections between these vasculogenic cytokines and BMP signaling which seems to be the final common pathway through which vasculogenic, osteogenic, and purine signaling functions. Future directions may also benefit from a focus on chondrogenesis which has consistently been shown to be the first step in ectopic bone formation. Given the endochondral nature of HO development, preventing cartilage formation may logically be the step at which prophylactic intervention would be the most efficacious.

Clinically, there are many promising developments on the horizon. Because calcified bone is a highly specialized tissue for which the body does not have native capability to quickly dismantle and resorb, current treatment for HO relies on surgical excision. These procedures are difficult and carry high rates of morbidity and recurrence (22,23). For these reasons, much of the current translational research on HO focuses on prophylaxis to prevent ectopic bone formation and early diagnostic modalities.

Current prophylactic regimens rely on NSAIDs (e.g., indomethacin), radiation, or bisphosphonates; all of which have their own divers efficacies and drawbacks (5). While these systemic treatments have shown general efficacy and are used clinically, there may be great benefit obtained from more localized treatment with substances such as apyrase, or from more targeted inhibitors of osteogenesis or chondrogenesis. Indeed, one of the most surprising and promising results of our findings on apyrase was the future effects of an initial application, which we have also seen improves wound healing (24). This type of targeted therapy could be very useful in burn wounds, trauma repair, and arthroplasty where the general location of potential ectopic bone formation is known. However, apyrase application may impractical for HO resulting from TBI, poly-trauma, and other inflammatory states stemming from more global insults.

We certainly agree that it would be prudent and of potential great benefit to further study the efficacy and mechanistic details of apyrase use at both a basic science and clinical level (13). Furthermore we believe it would be wise to continue to include advancements in both the fields of HO and FOP in future projects and hypotheses given the general concordance in results thus far. Ultimately it will require a multifaceted approach for these advances in translational medicine to make a difference in the clinical treatment of HO.


Acknowledgements

None.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Alfieri KA, Forsberg JA, Potter BK. Blast injuries and heterotopic ossification. Bone Joint Res 2012;1:192-7. [PubMed]
  2. Kan C, Kan L. The burning questions of heterotopic ossification. Ann Transl Med 2015;3:14. [PubMed]
  3. Peterson JR, De La Rosa S, Eboda O, et al. Treatment of heterotopic ossification through remote ATP hydrolysis. Sci Transl Med 2014;6:255ra132.
  4. Peterson JR, Okagbare PI, De La Rosa S, et al. Early detection of burn induced heterotopic ossification using transcutaneous Raman spectroscopy. Bone 2013;54:28-34. [PubMed]
  5. Ranganathan K, Loder S, Agarwal S, et al. Heterotopic Ossification: Basic-Science Principles and Clinical Correlates. J Bone Joint Surg Am 2015;97:1101-11. [PubMed]
  6. Zhang S, Kaplan FS, Shore EM. Different roles of GNAS and cAMP signaling during early and late stages of osteogenic differentiation. Horm Metab Res 2012;44:724-31. [PubMed]
  7. Gimble JM, Youkhana K, Hua X, et al. Adipogenesis in a myeloid supporting bone marrow stromal cell line. J Cell Biochem 1992;50:73-82. [PubMed]
  8. Ninomiya Y, Sugahara-Yamashita Y, Nakachi Y, et al. Development of a rapid culture method to induce adipocyte differentiation of human bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun 2010;394:303-8. [PubMed]
  9. Regateiro FS, Cobbold SP, Waldmann H. CD73 and adenosine generation in the creation of regulatory microenvironments. Clin Exp Immunol 2013;171:1-7. [PubMed]
  10. Carroll SH, Wigner NA, Kulkarni N, et al. A2B adenosine receptor promotes mesenchymal stem cell differentiation to osteoblasts and bone formation in vivo. J Biol Chem 2012;287:15718-27. [PubMed]
  11. Costa MA, Barbosa A, Neto E, et al. On the role of subtype selective adenosine receptor agonists during proliferation and osteogenic differentiation of human primary bone marrow stromal cells. J Cell Physiol 2011;226:1353-66. [PubMed]
  12. Shore EM, Xu M, Feldman GJ, et al. A recurrent mutation in the BMP type I receptor ACVR1 causes inherited and sporadic fibrodysplasia ossificans progressiva. Nat Genet 2006;38:525-7. [PubMed]
  13. Liang OD, Reginato AM, Medici D. Apyrase as a novel therapeutic inhibitor of heterotopic ossification. Ann Transl Med 2015;3:S32. [PubMed]
  14. Yu PB, Deng DY, Lai CS, et al. BMP type I receptor inhibition reduces heterotopic [corrected] ossification. Nat Med 2008;14:1363-9. [PubMed]
  15. Wu X, Walters TJ, Rathbone CR. Skeletal muscle satellite cell activation following cutaneous burn in rats. Burns 2013;39:736-44. [PubMed]
  16. Medici D, Shore EM, Lounev VY, et al. Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med 2010;16:1400-6. [PubMed]
  17. Suda RK, Billings PC, Egan KP, et al. Circulating osteogenic precursor cells in heterotopic bone formation. Stem Cells 2009;27:2209-19. [PubMed]
  18. Kaplan FS, Glaser DL, Shore EM, et al. Hematopoietic stem-cell contribution to ectopic skeletogenesis. J Bone Joint Surg Am 2007;89:347-57. [PubMed]
  19. Kan L, Liu Y, McGuire TL, et al. Dysregulation of local stem/progenitor cells as a common cellular mechanism for heterotopic ossification. Stem Cells 2009;27:150-6. [PubMed]
  20. Medici D, Olsen BR. The role of endothelial-mesenchymal transition in heterotopic ossification. J Bone Miner Res 2012;27:1619-22. [PubMed]
  21. Dilling CF, Wada AM, Lazard ZW, et al. Vessel formation is induced prior to the appearance of cartilage in BMP-2-mediated heterotopic ossification. J Bone Miner Res 2010;25:1147-56. [PubMed]
  22. Tsionos I, Leclercq C, Rochet JM. Heterotopic ossification of the elbow in patients with burns. Results after early excision. J Bone Joint Surg Br 2004;86:396-403. [PubMed]
  23. Lee EK, Namdari S, Hosalkar HS, et al. Clinical results of the excision of heterotopic bone around the elbow: a systematic review. J Shoulder Elbow Surg 2013;22:716-22. [PubMed]
  24. Bayliss J, Delarosa S, Wu J, et al. Adenosine triphosphate hydrolysis reduces neutrophil infiltration and necrosis in partial-thickness scald burns in mice. J Burn Care Res 2014;35:54-61. [PubMed]
Cite this article as: Peterson JR, Agarwal S, Loder SJ, Eboda O, Cederna PS, Buchman SR, Xi C, Wang SC, Levi B. Picking a bone with heterotopic ossification: translational progress current and future. Ann Transl Med 2015;3(13):188. doi: 10.3978/j.issn.2305-5839.2015.07.27

Download Citation