Semin Plast Surg 2024; 38(01): 031-038
DOI: 10.1055/s-0043-1778039
Review Article

H Vessel Formation as a Marker for Enhanced Bone Healing in Irradiated Distraction Osteogenesis

Melissa Daniel
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
,
Nathan Sheppard
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
,
Garrison Carlos
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
,
Noah Nelson
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
,
Alex Donneys
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
,
Steven R. Buchman
1   Section of Plastic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
› Author Affiliations

Abstract

In the setting of bone defects, the injured vasculature and loss of hemodynamic inflow leads to hematoma formation and low oxygen tension which stimulates vascular expansion through the HIf-1α pathway. Most importantly, this pathway upregulates sprouting of type H vessels (CD31hiEmcnhi vessels). H vessels engage in direct interaction with perivascular osteoprogenitor cells (OPCs), osteoblasts, and preosteoclasts of bone formation and remodeling. This angiogenic-osteogenic coupling leads to synchronous propagation of vascular and bony tissue for regenerative healing. A growing body of literature demonstrates that H vessels constitute a large portion of bone's innate capacity for osteogenic healing. We believe that CD31hiEmcnhi vessels play a role in bone healing during distraction osteogenesis (DO). DO is a procedure that utilizes traction forces to facilitate induction of endogenous bone formation and regeneration of surrounding soft tissues such as skin, muscle, tendon, and neurovascular structures. While the H vessel response to mechanical injury is adequate to facilitate healing in normal healthy tissue, it remains inadequate to overcome the devastation of radiation. We posit that the destruction of CD31hiEmcnhi vessels plays a role in precluding DO's effectiveness in irradiated bone defect healing. We aim, therefore, to recapitulate the normal pathway of bony healing by utilizing the regenerative capacity of H vessels. We hypothesize that using localized application of deferoxamine (DFO) will enhance the H vessel-mediated vasculogenic response to radiation damage and ultimately enable osteogenic healing during DO. This discovery could potentially be exploited by developing translational therapeutics to hopefully accelerate bone formation and shorten the DO consolidation period, thereby potentially expanding DO's utilization in irradiated bone healing.

Sprague–Dawley rats were divided into three groups: DO, radiation with DO (xDO), and radiation with DO and DFO implantation (xDODFO). Experimental groups received 35 Gy of radiation. All groups underwent DO. The treatment group received injections into the osteotomy site, every other day, beginning on postoperative day (POD) 4 of DFO. Animals were sacrificed on POD 40. For immunohistochemical analysis, mandibles were dissected and fixed in 4% paraformaldehyde for 48 hours, decalcified in Cal-Ex II for 2 days, dehydrated through graded ethanol of increasing concentration, and then embedded in paraffin. Samples were cut into 7-μm thick longitudinally oriented sections including the metaphysis and diaphysis. CD31 and Emcn double immunofluorescent staining were performed to evaluate the extent of CD31hiEmcnhi vessel formation. Bone sections were then stained with conjugated antibodies overnight at 4°C. Nuclei were stained with Hoechst. Slides were also double stained with Osterix and CD31 to study the quantity of H vessel-mediated recruitment of OPCs to accelerate bone healing. Images were acquired with a Nikon Ti2 widefield microscope and analyzed in NIS- Elements Advanced Research 5.41.02 software. The abundance of type H vessels is represented by the area fraction of CD31 + Emcn+ vessel area inside the regenerate sample. OPC concomitant proliferation into the distraction gap is represented by the area fraction of Osterix+ cell area inside of the regenerate sample.

There were 6× more type H vessels in DO groups than in xDO groups. Localized DFO significantly increased the abundance of type H vessels of irradiated DO animals compared to xDO by 15× (p = 0.00133531). Moreover, the DO and xDODFO groups with higher abundance of type H vessels also demonstrated better angiogenesis and osteogenesis outcomes. Interestingly, xDODFO groups doubled the quantity of H vessel formation compared to DO, indicating a supraphysiologic response (p = 0.044655055). Furthermore, H vessel-mediated recruitment of OPCs mimicked the described H vessel formation trend in our study groups. Irradiated DO groups contained 3× less OPCs compared to DO controls. DFO treatment to xDO animals remediated irradiation damage by containing 12× Osterix+ cells. Finally, DFO treatment of irradiated animals quadrupled osteoprogenitor recruitment into the distraction gap compared to DO controls.

In this study, we developed a novel approach to visualize CD31hiEmcnhi in paraffin sections to study DO regeneration. Normal DO demonstrated a significant upregulation of H vessel formation and associated angiogenic-osteogenic coupling. Radiation severely decreased H vessel formation along with an associated significant diminution of new bone formation and nonunion. DFO administration, however, resulted in vascular replenishment and the restoration of high quantities of CD31hiEmcnhi and OPCs, recapitulating the normal process of bony regeneration and repair. DFO treatment remediated new bone formation and bony union in irradiated fields associated with increased H vessel angiogenic-osteogenic coupling. While further studies are required to optimize this approach, the results of this study are incredibly promising for the long-awaited translation of localized DFO into the clinical arena.



Publication History

Article published online:
19 January 2024

© 2024. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Birch JG. A brief history of limb lengthening. J Pediatr Orthop 2017; 37 (Suppl. 02) S1-S8
  • 2 McCarthy JG, Stelnicki EJ, Mehrara BJ, Longaker MT. Distraction osteogenesis of the craniofacial skeleton. Plast Reconstr Surg 2001; 107 (07) 1812-1827
  • 3 Dua G, Navin Kumar A, Roy ID, Roy SK. Maxillary distraction osteogenesis in cleft lip and palate cases with midface hypoplasia using rigid external distractor: an alternative technique. J Craniofac Surg 2014; 25 (03) 746-751
  • 4 Hopper RA, Ettinger RE, Purnell CA, Dover MS, Pereira AR, Tunçbilek G. Thirty years later: what has craniofacial distraction osteogenesis surgery replaced?. Plast Reconstr Surg 2020; 145 (06) 1073e-1088e
  • 5 Choi IH, Chung CY, Cho TJ, Yoo WJ. Angiogenesis and mineralization during distraction osteogenesis. J Korean Med Sci 2002; 17 (04) 435-447
  • 6 Kusumbe AP, Ramasamy SK, Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014; 507 (7492): 323-328
  • 7 Kusumbe AP, Ramasamy SK, Itkin T. et al. Age-dependent modulation of vascular niches for haematopoietic stem cells. Nature 2016; 532 (7599): 380-384
  • 8 Ramasamy SK, Kusumbe AP, Schiller M. et al. Blood flow controls bone vascular function and osteogenesis. Nat Commun 2016; 7: 13601
  • 9 Sivaraj KK, Adams RH. Blood vessel formation and function in bone. Development 2016; 143 (15) 2706-2715
  • 10 Greijer AE, van der Groep P, Kemming D. et al. Up-regulation of gene expression by hypoxia is mediated predominantly by hypoxia-inducible factor 1 (HIF-1). J Pathol 2005; 206 (03) 291-304
  • 11 Jones DT, Harris AL. Identification of novel small-molecule inhibitors of hypoxia-inducible factor-1 transactivation and DNA binding. Mol Cancer Ther 2006; 5 (09) 2193-2202
  • 12 Farberg AS, Jing XL, Monson LA. et al. Deferoxamine reverses radiation induced hypovascularity during bone regeneration and repair in the murine mandible. Bone 2012; 50 (05) 1184-1187
  • 13 Felice PA, Ahsan S, Donneys A, Deshpande SS, Nelson NS, Buchman SR. Deferoxamine administration delivers translational optimization of distraction osteogenesis in the irradiated mandible. Plast Reconstr Surg 2013; 132 (04) 542e-548e
  • 14 Donneys A, Weiss DM, Deshpande SS. et al. Localized deferoxamine injection augments vascularity and improves bony union in pathologic fracture healing after radiotherapy. Bone 2013; 52 (01) 318-325
  • 15 Gosain AK. Plastic Surgery Educational Foundation DATA Committee. Distraction osteogenesis of the craniofacial skeleton. Plast Reconstr Surg 2001; 107 (01) 278-280
  • 16 Nakashima K, Zhou X, Kunkel G. et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 2002; 108 (01) 17-29
  • 17 Li J, Chen X, Ren L. et al. Type H vessel/platelet-derived growth factor receptor β+ perivascular cell disintegration is involved in vascular injury and bone loss in radiation-induced bone damage. Cell Prolif 2023; 56 (07) e13406
  • 18 Wan C, Gilbert SR, Wang Y. et al. Activation of the hypoxia-inducible factor-1α pathway accelerates bone regeneration. Proc Natl Acad Sci U S A 2008; 105 (02) 686-691
  • 19 He X, Han Z, Jiang W. et al. Hypoxia improved vasculogenesis in distraction osteogenesis through Mesenchymal-Epithelial transition (MET), Wnt/β-catenin signaling pathway, and autophagy. Acta Histochem 2020; 122 (06) 151593
  • 20 Yang M, Li CJ, Sun X. et al. MiR-497∼195 cluster regulates angiogenesis during coupling with osteogenesis by maintaining endothelial Notch and HIF-1α activity. Nat Commun 2017; 8: 16003
  • 21 Ramasamy SK. Structure and functions of blood vessels and vascular niches in bone. Stem Cells Int 2017; 5046953
  • 22 Peng Y, Wu S, Li Y, Crane JL. Type H blood vessels in bone modeling and remodeling. Theranostics 2020; 10 (01) 426-436