Inflammatory phase of bone healing initiates the regenerative healing cascade

Bone healing commences with an inflammatory reaction which initiates the regenerative healing process leading in the end to reconstitution of bone. An unbalanced immune reaction during this early bone healing phase is hypothesized to disturb the healing cascade in a way that delays bone healing and jeopardizes the successful healing outcome. The immune cell composition and expression pattern of angiogenic factors were investigated in a sheep bone osteotomy model and compared to a mechanically-induced impaired/delayed bone healing group. In the impaired/delayed healing group, significantly higher T cell percentages were present in the bone hematoma and the bone marrow adjacent to the osteotomy gap when compared to the normal healing group. This was mirrored in the higher cytotoxic T cell percentage detected under delayed bone healing conditions indicating longer pro-inflammatory processes. The highly activated periosteum adjourning the osteotomy gap showed lower expression of hematopoietic stem cell markers and angiogenic factors such as heme oxygenase and vascular endothelial growth factor. This indicates a deferred revascularization of the injured area due to ongoing pro-inflammatory processes in the delayed healing group. Results from this study suggest that there are unfavorable immune cells and factors participating in the initial healing phase. In conclusion, identifying beneficial aspects may lead to promising therapeutical approaches that might benefit further by eliminating the unfavorable factors.

[1]  D. Marsh,et al.  Inflammatory cells in normal human fracture healing. , 1994, Acta orthopaedica Scandinavica.

[2]  P. Collin‐Osdoby,et al.  Bone cell function, regulation, and communication: A role for nitric oxide , 1995, Journal of cellular biochemistry.

[3]  T A Einhorn,et al.  The expression of cytokine activity by fracture callus , 1995, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[4]  T. Einhorn,et al.  Significant advances have been made in the way surgeons treat fractures. , 1998, Clinical orthopaedics and related research.

[5]  A. Singer,et al.  Cutaneous wound healing. , 1999, The New England journal of medicine.

[6]  M. Klagsbrun,et al.  Cartilage to bone—Angiogenesis leads the way , 1999, Nature Medicine.

[7]  S. Opal Phylogenetic and functional relationships between coagulation and the innate immune response. , 2000, Critical care medicine.

[8]  M. Tsunoda,et al.  Role of fracture hematoma and periosteum during fracture healing in rats: interaction of fracture hematoma and the periosteum in the initial step of the healing process , 2000, Journal of orthopaedic science : official journal of the Japanese Orthopaedic Association.

[9]  T. Suda,et al.  Role of Hematopoietic Stem Cells in Angiogenesis , 2001, International journal of hematology.

[10]  T A Einhorn,et al.  Expression of Osteoprotegerin, Receptor Activator of NF‐κB Ligand (Osteoprotegerin Ligand) and Related Proinflammatory Cytokines During Fracture Healing , 2001, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[11]  P. Allavena,et al.  Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. , 2002, Trends in immunology.

[12]  D. Graves,et al.  Impaired Fracture Healing in the Absence of TNF‐α Signaling: The Role of TNF‐α in Endochondral Cartilage Resorption , 2003 .

[13]  T A Einhorn,et al.  Impaired fracture healing in the absence of TNF-alpha signaling: the role of TNF-alpha in endochondral cartilage resorption. , 2003, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[14]  B. Rinker,et al.  Intractable wounds and infections: the role of impaired vascularity and advanced surgical methods for treatment. , 2004, American journal of surgery.

[15]  John Savill,et al.  Resolution of inflammation: the beginning programs the end , 2005, Nature Immunology.

[16]  Gang Li,et al.  Nonsteroidal anti-inflammatory drug-induced fracture nonunion: an inhibition of angiogenesis? , 2006, The Journal of bone and joint surgery. American volume.

[17]  Georg N Duda,et al.  Instability prolongs the chondral phase during bone healing in sheep. , 2006, Bone.

[18]  C. Colnot,et al.  Analyzing the cellular contribution of bone marrow to fracture healing using bone marrow transplantation in mice. , 2006, Biochemical and biophysical research communications.

[19]  M. Filbin How inflammation promotes regeneration , 2006, Nature Neuroscience.

[20]  Georg N Duda,et al.  Timely fracture-healing requires optimization of axial fixation stability. , 2007, The Journal of bone and joint surgery. American volume.

[21]  Frank A. Liporace,et al.  Complications of Ankle Fracture in Patients With Diabetes , 2008, The Journal of the American Academy of Orthopaedic Surgeons.

[22]  A. Agarwal,et al.  Heme oxygenase-1 and carbon monoxide in vascular pathobiology: focus on angiogenesis. , 2008, Circulation.

[23]  Georg N Duda,et al.  Mechanical induction of critically delayed bone healing in sheep: radiological and biomechanical results. , 2008, Journal of biomechanics.

[24]  Qingbo Xu,et al.  Vascular repair by endothelial progenitor cells. , 2008, Cardiovascular research.

[25]  Antonios G Mikos,et al.  Modulation of the inflammatory response for enhanced bone tissue regeneration. , 2008, Tissue engineering. Part B, Reviews.

[26]  A. Friedman Cell cycle and developmental control of hematopoiesis by Runx1 , 2009, Journal of cellular physiology.

[27]  Mark Sweeney,et al.  Molecular analysis of endothelial progenitor cell (EPC) subtypes reveals two distinct cell populations with different identities , 2010, BMC Medical Genomics.

[28]  Georg N Duda,et al.  Differential regulation of blood vessel formation between standard and delayed bone healing , 2009, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[29]  D. Hutmacher,et al.  Influences of age and mechanical stability on volume, microstructure, and mineralization of the fracture callus during bone healing: is osteoclast activity the key to age-related impaired healing? , 2010, Bone.

[30]  D. Hu,et al.  Rejuvenation of the inflammatory system stimulates fracture repair in aged mice , 2010, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[31]  Georg N Duda,et al.  Insight into the molecular pathophysiology of delayed bone healing in a sheep model. , 2010, Tissue engineering. Part A.

[32]  P. Lacativa,et al.  Osteoporosis and inflammation. , 2010, Arquivos brasileiros de endocrinologia e metabologia.

[33]  A. Józkowicz,et al.  Heme oxygenase-1 in neovascularisation: A diabetic perspective , 2010, Thrombosis and Haemostasis.

[34]  H. Kimura,et al.  Malfunction of bone marrow-derived osteoclasts and the delay of bone fracture healing in diabetic mice. , 2010, Bone.

[35]  T. Rőszer,et al.  Inflammation as death or life signal in diabetic fracture healing , 2010, Inflammation Research.

[36]  David M. Mosser,et al.  Exploring the full spectrum of macrophage activation , 2010, Nature Reviews Immunology.

[37]  R. Beelen,et al.  Macrophages in skin injury and repair. , 2011, Immunobiology.