Skip to main content
Top
Published in: Current Osteoporosis Reports 2/2018

Open Access 01-04-2018 | Orthopedic Management of Fractures (S Bukata and L Gerstenfeld, Section Editors)

The Role of the Immune Cells in Fracture Healing

Published in: Current Osteoporosis Reports | Issue 2/2018

Login to get access

Abstract

Purpose of review

Bone fracture healing is a complex physiological process relying on numerous cell types and signals. Inflammatory factors secreted by immune cells help to control recruitment, proliferation, differentiation, and activation of hematopoietic and mesenchymal cells. Within this review we will discuss the functional role of immune cells as it pertains to bone fracture healing. In doing so, we will outline the cytokines secreted and their effects within the healing fracture callus.

Recent findings

Macrophages have been found to play an important role in fracture healing. These immune cells signal to other cells of the fracture callus, modulating bone healing.

Summary

Cytokines and cellular signals within fracture healing continue to be studied. The findings from this work have helped to reinforce the importance of osteoimmunity in bone fracture healing. Owing to these efforts, immunomodulation is emerging as a potential therapeutic target to improve bone fracture healing.
Literature
1.
2.
go back to reference Clement ND, Beauchamp NJ, Duckworth AD, McQueen MM, Court-Brown CM. The outcome of tibial diaphyseal fractures in the elderly. Bone Joint J. 2013;95-B(9):1255–62.CrossRefPubMed Clement ND, Beauchamp NJ, Duckworth AD, McQueen MM, Court-Brown CM. The outcome of tibial diaphyseal fractures in the elderly. Bone Joint J. 2013;95-B(9):1255–62.CrossRefPubMed
3.
go back to reference Miranda MAMSM. Treatment strategy for non unions and malunions. New York: Thieme; 2007. Miranda MAMSM. Treatment strategy for non unions and malunions. New York: Thieme; 2007.
4.
go back to reference Ng AH, Baht GS, Alman BA, Grynpas MD. Bone marrow stress decreases osteogenic progenitors. Calcif Tissue Int. 2015;97(5):476–86.CrossRefPubMed Ng AH, Baht GS, Alman BA, Grynpas MD. Bone marrow stress decreases osteogenic progenitors. Calcif Tissue Int. 2015;97(5):476–86.CrossRefPubMed
5.
go back to reference Xing Z, Lu C, Hu D, Miclau T III, Marcucio RS. Rejuvenation of the inflammatory system stimulates fracture repair in aged mice. J Orthop Res. 2010;28(8):1000–6.PubMedPubMedCentral Xing Z, Lu C, Hu D, Miclau T III, Marcucio RS. Rejuvenation of the inflammatory system stimulates fracture repair in aged mice. J Orthop Res. 2010;28(8):1000–6.PubMedPubMedCentral
6.
go back to reference Schmidt-Bleek K, Schell H, Schulz N, Hoff P, Perka C, Buttgereit F, et al. Inflammatory phase of bone healing initiates the regenerative healing cascade. Cell Tissue Res. 2012;347(3):567–73.CrossRefPubMed Schmidt-Bleek K, Schell H, Schulz N, Hoff P, Perka C, Buttgereit F, et al. Inflammatory phase of bone healing initiates the regenerative healing cascade. Cell Tissue Res. 2012;347(3):567–73.CrossRefPubMed
7.
go back to reference Lienau J, Schmidt-Bleek K, Peters A, Haschke F, Duda GN, Perka C, et al. Differential regulation of blood vessel formation between standard and delayed bone healing. J Orthop Res. 2009;27(9):1133–40.CrossRefPubMed Lienau J, Schmidt-Bleek K, Peters A, Haschke F, Duda GN, Perka C, et al. Differential regulation of blood vessel formation between standard and delayed bone healing. J Orthop Res. 2009;27(9):1133–40.CrossRefPubMed
8.
go back to reference Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: The cellular picture. Semin Cell Dev Biol. 2008;19(5):459–66.CrossRefPubMed Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture repair: The cellular picture. Semin Cell Dev Biol. 2008;19(5):459–66.CrossRefPubMed
10.
go back to reference Ai-Aql ZS, Alagl AS, Graves DT, Gerstenfeld LC, Einhorn TA. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res. 2008;87(2):107–18.CrossRefPubMedPubMedCentral Ai-Aql ZS, Alagl AS, Graves DT, Gerstenfeld LC, Einhorn TA. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res. 2008;87(2):107–18.CrossRefPubMedPubMedCentral
12.
go back to reference Ozaki A, Tsunoda M, Kinoshita S, Saura R. 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. J Orthop Sci. 2000;5(1):64–70.CrossRefPubMed Ozaki A, Tsunoda M, Kinoshita S, Saura R. 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. J Orthop Sci. 2000;5(1):64–70.CrossRefPubMed
13.
go back to reference Kolar P, Schmidt-Bleek K, Schell H, Gaber T, Toben D, Schmidmaier G, et al. The early fracture hematoma and its potential role in fracture healing. Tissue Eng Part B Rev. 2010;16(4):427–34.CrossRefPubMed Kolar P, Schmidt-Bleek K, Schell H, Gaber T, Toben D, Schmidmaier G, et al. The early fracture hematoma and its potential role in fracture healing. Tissue Eng Part B Rev. 2010;16(4):427–34.CrossRefPubMed
15.
go back to reference Einhorn TA. The cell and molecular biology of fracture healing. Clin Orthop Relat Res. 1998;(355 Suppl):S7–21. Einhorn TA. The cell and molecular biology of fracture healing. Clin Orthop Relat Res. 1998;(355 Suppl):S7–21.
16.
go back to reference Colnot C, Huang S, Helms J. Analyzing the cellular contribution of bone marrow to fracture healing using bone marrow transplantation in mice. Biochem Biophys Res Commun. 2006;350(3):557–61.CrossRefPubMed Colnot C, Huang S, Helms J. Analyzing the cellular contribution of bone marrow to fracture healing using bone marrow transplantation in mice. Biochem Biophys Res Commun. 2006;350(3):557–61.CrossRefPubMed
17.
go back to reference Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105(10):4120–6.CrossRefPubMed Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 2005;105(10):4120–6.CrossRefPubMed
18.
go back to reference Nemeth K, Keane-Myers A, Brown JM, Metcalfe DD, Gorham JD, Bundoc VG, et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc Natl Acad Sci USA. 2010;107(12):5652–7.CrossRefPubMedPubMedCentral Nemeth K, Keane-Myers A, Brown JM, Metcalfe DD, Gorham JD, Bundoc VG, et al. Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc Natl Acad Sci USA. 2010;107(12):5652–7.CrossRefPubMedPubMedCentral
19.
go back to reference Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–49. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Parmelee A, Doi K, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med. 2009;15(1):42–49.
20.
go back to reference DelaRosa O, Lombardo E, Beraza A, Mancheno-Corvo P, Ramirez C, Menta R, et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng. Part A. 2009;15(10):2795–806.CrossRefPubMed DelaRosa O, Lombardo E, Beraza A, Mancheno-Corvo P, Ramirez C, Menta R, et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng. Part A. 2009;15(10):2795–806.CrossRefPubMed
21.
go back to reference Rafei M, Campeau PM, Aguilar-Mahecha A, Buchanan M, Williams P, Birman E, et al. Mesenchymal stromal cells ameliorate experimental autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC chemokine ligand 2-dependent manner. J Immunol. 2009;182(10):5994–6002.CrossRefPubMed Rafei M, Campeau PM, Aguilar-Mahecha A, Buchanan M, Williams P, Birman E, et al. Mesenchymal stromal cells ameliorate experimental autoimmune encephalomyelitis by inhibiting CD4 Th17 T cells in a CC chemokine ligand 2-dependent manner. J Immunol. 2009;182(10):5994–6002.CrossRefPubMed
22.
23.
go back to reference Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury-International J Care Injured. 2005;36(12):1392–404.CrossRef Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury-International J Care Injured. 2005;36(12):1392–404.CrossRef
24.
go back to reference Yang L, Tsang KY, Tang HC, Chan D, Cheah KS. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc Natl Acad Sci USA. 2014;111(33):12097–102.CrossRefPubMedPubMedCentral Yang L, Tsang KY, Tang HC, Chan D, Cheah KS. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc Natl Acad Sci USA. 2014;111(33):12097–102.CrossRefPubMedPubMedCentral
25.
go back to reference McKibbin B. The biology of fracture healing in long bones. J Bone Joint Surg Br. 1978;60-B(2):150–62.CrossRefPubMed McKibbin B. The biology of fracture healing in long bones. J Bone Joint Surg Br. 1978;60-B(2):150–62.CrossRefPubMed
26.
go back to reference Roldan JC, Jepsen S, Miller J, Freitag S, Rueger DC, Acil Y, et al. Bone formation in the presence of platelet-rich plasma vs. bone morphogenetic protein-7. Bone. 2004;34(1):80–90.CrossRefPubMed Roldan JC, Jepsen S, Miller J, Freitag S, Rueger DC, Acil Y, et al. Bone formation in the presence of platelet-rich plasma vs. bone morphogenetic protein-7. Bone. 2004;34(1):80–90.CrossRefPubMed
27.
go back to reference Jingushi S, Scully SP, Joyce ME, Sugioka Y, Bolander ME. Transforming growth factor-beta 1 and fibroblast growth factors in rat growth plate. J Orthop Res. 1995;13(5):761–8.CrossRefPubMed Jingushi S, Scully SP, Joyce ME, Sugioka Y, Bolander ME. Transforming growth factor-beta 1 and fibroblast growth factors in rat growth plate. J Orthop Res. 1995;13(5):761–8.CrossRefPubMed
28.
go back to reference Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury. 2005;36(12):1392–404.CrossRefPubMed Dimitriou R, Tsiridis E, Giannoudis PV. Current concepts of molecular aspects of bone healing. Injury. 2005;36(12):1392–404.CrossRefPubMed
29.
go back to reference Dulgeroglu TC, Metineren H. Evaluation of the effect of platelet-rich fibrin on long bone healing: An experimental rat model. Orthopedics. 2017;40(3):e479–e84.CrossRefPubMed Dulgeroglu TC, Metineren H. Evaluation of the effect of platelet-rich fibrin on long bone healing: An experimental rat model. Orthopedics. 2017;40(3):e479–e84.CrossRefPubMed
32.
go back to reference Bastian OW, Koenderman L, Alblas J, Leenen LP, Blokhuis TJ. Neutrophils contribute to fracture healing by synthesizing fibronectin+ extracellular matrix rapidly after injury. Clin Immunol. 2016;164:78–84.CrossRefPubMed Bastian OW, Koenderman L, Alblas J, Leenen LP, Blokhuis TJ. Neutrophils contribute to fracture healing by synthesizing fibronectin+ extracellular matrix rapidly after injury. Clin Immunol. 2016;164:78–84.CrossRefPubMed
33.
go back to reference Xian CJ, Zhou FH, McCarty RC, Foster BK. Intramembranous ossification mechanism for bone bridge formation at the growth plate cartilage injury site. J Orthop Res. 2004;22(2):417–26.CrossRefPubMed Xian CJ, Zhou FH, McCarty RC, Foster BK. Intramembranous ossification mechanism for bone bridge formation at the growth plate cartilage injury site. J Orthop Res. 2004;22(2):417–26.CrossRefPubMed
35.
go back to reference Timlin M, Toomey D, Condron C, Power C, Street J, Murray P, et al. Fracture hematoma is a potent proinflammatory mediator of neutrophil function. J Trauma. 2005;58(6):1223–9.CrossRefPubMed Timlin M, Toomey D, Condron C, Power C, Street J, Murray P, et al. Fracture hematoma is a potent proinflammatory mediator of neutrophil function. J Trauma. 2005;58(6):1223–9.CrossRefPubMed
36.
go back to reference Soehnlein O, Lindbom L, Weber C. Mechanisms underlying neutrophil-mediated monocyte recruitment. Blood. 2009;114(21):4613–23.CrossRefPubMed Soehnlein O, Lindbom L, Weber C. Mechanisms underlying neutrophil-mediated monocyte recruitment. Blood. 2009;114(21):4613–23.CrossRefPubMed
37.
go back to reference Claes L, Recknagel S, Ignatius A. Fracture healing under healthy and inflammatory conditions. Nat Rev Rheumatol. 2012;8(3):133–43.CrossRefPubMed Claes L, Recknagel S, Ignatius A. Fracture healing under healthy and inflammatory conditions. Nat Rev Rheumatol. 2012;8(3):133–43.CrossRefPubMed
38.
go back to reference Kovtun A, Bergdolt S, Wiegner R, Radermacher P, Huber-Lang M, Ignatius A. The crucial role of neutrophil granulocytes in bone fracture healing. Eur Cell Mater. 2016;32:152–62.CrossRefPubMed Kovtun A, Bergdolt S, Wiegner R, Radermacher P, Huber-Lang M, Ignatius A. The crucial role of neutrophil granulocytes in bone fracture healing. Eur Cell Mater. 2016;32:152–62.CrossRefPubMed
39.
go back to reference Scapini P, Laudanna C, Pinardi C, Allavena P, Mantovani A, Sozzani S, et al. Neutrophils produce biologically active macrophage inflammatory protein-3alpha (MIP-3alpha)/CCL20 and MIP-3beta/CCL19. Eur J Immunol. 2001;31(7):1981–8.CrossRefPubMed Scapini P, Laudanna C, Pinardi C, Allavena P, Mantovani A, Sozzani S, et al. Neutrophils produce biologically active macrophage inflammatory protein-3alpha (MIP-3alpha)/CCL20 and MIP-3beta/CCL19. Eur J Immunol. 2001;31(7):1981–8.CrossRefPubMed
40.
go back to reference • Baht GS, Silkstone D, Vi L, Nadesan P, Amani Y, Whetstone H, et al. Erratum: Exposure to a youthful circulation rejuvenates bone repair through modulation of beta-catenin. Nat Commun. 2015;6:7761. Using parabiosis, bone marrow transplantation, and conditioned media, the authors show that hematapoeitec (CD45+) play an important role in fracture healing and osteoblastic differentiation. • Baht GS, Silkstone D, Vi L, Nadesan P, Amani Y, Whetstone H, et al. Erratum: Exposure to a youthful circulation rejuvenates bone repair through modulation of beta-catenin. Nat Commun. 2015;6:7761. Using parabiosis, bone marrow transplantation, and conditioned media, the authors show that hematapoeitec (CD45+) play an important role in fracture healing and osteoblastic differentiation.
41.
go back to reference Raggatt LJ, Wullschleger ME, Alexander KA, Wu AC, Millard SM, Kaur S, et al. Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification. Am J Pathol. 2014;184(12):3192–204.CrossRefPubMed Raggatt LJ, Wullschleger ME, Alexander KA, Wu AC, Millard SM, Kaur S, et al. Fracture healing via periosteal callus formation requires macrophages for both initiation and progression of early endochondral ossification. Am J Pathol. 2014;184(12):3192–204.CrossRefPubMed
42.
go back to reference •• Chang MK, Raggatt LJ, Alexander KA, Kuliwaba JS, Fazzalari NL, Schroder K, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol. 2008;181(2):1232–44. This work indicates, for the first time, the presence of (bone) tissue-specific macrophages. •• Chang MK, Raggatt LJ, Alexander KA, Kuliwaba JS, Fazzalari NL, Schroder K, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol. 2008;181(2):1232–44. This work indicates, for the first time, the presence of (bone) tissue-specific macrophages.
44.
45.
46.
go back to reference Juban G, Chazaud B. Metabolic regulation of macrophages during tissue repair: insights from skeletal muscle regeneration. FEBS Lett. 2017. Juban G, Chazaud B. Metabolic regulation of macrophages during tissue repair: insights from skeletal muscle regeneration. FEBS Lett. 2017.
47.
go back to reference Godwin JW, Pinto AR, Rosenthal NA. Chasing the recipe for a pro-regenerative immune system. Semin Cell Dev Biol. 2017;61:71–9.CrossRefPubMed Godwin JW, Pinto AR, Rosenthal NA. Chasing the recipe for a pro-regenerative immune system. Semin Cell Dev Biol. 2017;61:71–9.CrossRefPubMed
48.
go back to reference Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci USA. 2014;111(45):16029–34.CrossRefPubMedPubMedCentral Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci USA. 2014;111(45):16029–34.CrossRefPubMedPubMedCentral
49.
go back to reference Pettit AR, Chang MK, Hume DA, Raggatt LJ. Osteal macrophages: a new twist on coupling during bone dynamics. Bone. 2008;43(6):976–82.CrossRefPubMed Pettit AR, Chang MK, Hume DA, Raggatt LJ. Osteal macrophages: a new twist on coupling during bone dynamics. Bone. 2008;43(6):976–82.CrossRefPubMed
50.
go back to reference Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, et al. Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells. Proc Natl Acad Sci USA. 1990;87(18):7260–4.CrossRefPubMedPubMedCentral Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishihara T, et al. Origin of osteoclasts: mature monocytes and macrophages are capable of differentiating into osteoclasts under a suitable microenvironment prepared by bone marrow-derived stromal cells. Proc Natl Acad Sci USA. 1990;87(18):7260–4.CrossRefPubMedPubMedCentral
51.
go back to reference Wu Y, Humphrey MB, Nakamura MC. Osteoclasts – the innate immune cells of the bone. Autoimmunity. 2008;41(3):183–94.CrossRefPubMed Wu Y, Humphrey MB, Nakamura MC. Osteoclasts – the innate immune cells of the bone. Autoimmunity. 2008;41(3):183–94.CrossRefPubMed
52.
go back to reference Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003;423(6937):337–42.CrossRefPubMed Boyle WJ, Simonet WS, Lacey DL. Osteoclast differentiation and activation. Nature. 2003;423(6937):337–42.CrossRefPubMed
53.
go back to reference Baht GS, O'Young J, Borovina A, Chen H, Tye CE, Karttunen M, et al. Phosphorylation of Ser136 is critical for potent bone sialoprotein-mediated nucleation of hydroxyapatite crystals. Biochem J. 2010;428(3):385–95.CrossRefPubMed Baht GS, O'Young J, Borovina A, Chen H, Tye CE, Karttunen M, et al. Phosphorylation of Ser136 is critical for potent bone sialoprotein-mediated nucleation of hydroxyapatite crystals. Biochem J. 2010;428(3):385–95.CrossRefPubMed
54.
55.
go back to reference Toben D, Schroeder I, El Khassawna T, Mehta M, Hoffmann JE, Frisch JT, et al. Fracture healing is accelerated in the absence of the adaptive immune system. J Bone Miner Res. 2011;26(1):113–24.CrossRefPubMed Toben D, Schroeder I, El Khassawna T, Mehta M, Hoffmann JE, Frisch JT, et al. Fracture healing is accelerated in the absence of the adaptive immune system. J Bone Miner Res. 2011;26(1):113–24.CrossRefPubMed
56.
go back to reference Nam D, Mau E, Wang Y, Wright D, Silkstone D, Whetstone H, et al. T-lymphocytes enable osteoblast maturation via IL-17F during the early phase of fracture repair. PLoS One. 2012;7(6):e40044.CrossRefPubMedPubMedCentral Nam D, Mau E, Wang Y, Wright D, Silkstone D, Whetstone H, et al. T-lymphocytes enable osteoblast maturation via IL-17F during the early phase of fracture repair. PLoS One. 2012;7(6):e40044.CrossRefPubMedPubMedCentral
57.
go back to reference El Khassawna T, Serra A, Bucher CH, Petersen A, Schlundt C, Konnecke I, et al. T lymphocytes influence the mineralization process of bone. Front Immunol. 2017;8:562.CrossRefPubMedPubMedCentral El Khassawna T, Serra A, Bucher CH, Petersen A, Schlundt C, Konnecke I, et al. T lymphocytes influence the mineralization process of bone. Front Immunol. 2017;8:562.CrossRefPubMedPubMedCentral
58.
go back to reference Konnecke I, Serra A, El Khassawna T, Schlundt C, Schell H, Hauser A, et al. T and B cells participate in bone repair by infiltrating the fracture callus in a two-wave fashion. Bone. 2014;64:155–65.CrossRefPubMed Konnecke I, Serra A, El Khassawna T, Schlundt C, Schell H, Hauser A, et al. T and B cells participate in bone repair by infiltrating the fracture callus in a two-wave fashion. Bone. 2014;64:155–65.CrossRefPubMed
59.
go back to reference Sun GJ, Wang YC, Ti YF, Wang J, Zhao JN, Qian HB. Regulatory B cell is critical in bone union process through suppressing proinflammatory cytokines and stimulating Foxp3 in Treg cells. Clin Exp Pharmacol P. 2017;44(4):455–62.CrossRef Sun GJ, Wang YC, Ti YF, Wang J, Zhao JN, Qian HB. Regulatory B cell is critical in bone union process through suppressing proinflammatory cytokines and stimulating Foxp3 in Treg cells. Clin Exp Pharmacol P. 2017;44(4):455–62.CrossRef
60.
go back to reference Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, et al. Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature. 1999;402(6759):304–9.CrossRefPubMed Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, et al. Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature. 1999;402(6759):304–9.CrossRefPubMed
61.
go back to reference Manabe N, Kawaguchi H, Chikuda H, Miyaura C, Inada M, Nagai R, et al. Connection between B lymphocyte and osteoclast differentiation pathways. J Immunol. 2001;167(5):2625–31.CrossRefPubMed Manabe N, Kawaguchi H, Chikuda H, Miyaura C, Inada M, Nagai R, et al. Connection between B lymphocyte and osteoclast differentiation pathways. J Immunol. 2001;167(5):2625–31.CrossRefPubMed
62.
go back to reference Takayanagi H. Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems. Nat Rev Immunol. 2007;7(4):292–304.CrossRefPubMed Takayanagi H. Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems. Nat Rev Immunol. 2007;7(4):292–304.CrossRefPubMed
63.
go back to reference Han X, Yang Q, Lin L, Xu C, Zheng C, Chen X, et al. Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death Differ. 2014;21(11):1758–68.CrossRefPubMedPubMedCentral Han X, Yang Q, Lin L, Xu C, Zheng C, Chen X, et al. Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death Differ. 2014;21(11):1758–68.CrossRefPubMedPubMedCentral
64.
go back to reference Huang H, Kim HJ, Chang EJ, Lee ZH, Hwang SJ, Kim HM, et al. IL-17 stimulates the proliferation and differentiation of human mesenchymal stem cells: implications for bone remodeling. Cell Death Differ. 2009;16(10):1332–43.CrossRefPubMed Huang H, Kim HJ, Chang EJ, Lee ZH, Hwang SJ, Kim HM, et al. IL-17 stimulates the proliferation and differentiation of human mesenchymal stem cells: implications for bone remodeling. Cell Death Differ. 2009;16(10):1332–43.CrossRefPubMed
66.
go back to reference Hauser CJ, Joshi P, Jones Q, Zhou X, Livingston DH, Lavery RF. Suppression of natural killer cell activity in patients with fracture/soft tissue injury. Arch Surg. 1997;132(12):1326–30.CrossRefPubMed Hauser CJ, Joshi P, Jones Q, Zhou X, Livingston DH, Lavery RF. Suppression of natural killer cell activity in patients with fracture/soft tissue injury. Arch Surg. 1997;132(12):1326–30.CrossRefPubMed
67.
go back to reference Soderstrom K, Stein E, Colmenero P, Purath U, Muller-Ladner U, de Matos CT, et al. Natural killer cells trigger osteoclastogenesis and bone destruction in arthritis. Proc Natl Acad Sci USA. 2010;107(29):13028–33.CrossRefPubMed Soderstrom K, Stein E, Colmenero P, Purath U, Muller-Ladner U, de Matos CT, et al. Natural killer cells trigger osteoclastogenesis and bone destruction in arthritis. Proc Natl Acad Sci USA. 2010;107(29):13028–33.CrossRefPubMed
68.
go back to reference Almeida CR, Caires HR, Vasconcelos DP, Barbosa MA. NAP-2 secreted by human NK cells can stimulate mesenchymal stem/stromal cell recruitment. Stem Cell Reports. 2016;6(4):466–73.CrossRefPubMedPubMedCentral Almeida CR, Caires HR, Vasconcelos DP, Barbosa MA. NAP-2 secreted by human NK cells can stimulate mesenchymal stem/stromal cell recruitment. Stem Cell Reports. 2016;6(4):466–73.CrossRefPubMedPubMedCentral
69.
go back to reference Krampera M. Mesenchymal stromal cell 'licensing': a multistep process. Leukemia. 2011;25(9):1408–14.CrossRefPubMed Krampera M. Mesenchymal stromal cell 'licensing': a multistep process. Leukemia. 2011;25(9):1408–14.CrossRefPubMed
70.
go back to reference Dorronsoro A, Ferrin I, Salcedo JM, Jakobsson E, Fernandez-Rueda J, Lang V, et al. Human mesenchymal stromal cells modulate T-cell responses through TNF-alpha-mediated activation of NF-kappaB. Eur J Immunol. 2014;44(2):480–8.CrossRefPubMed Dorronsoro A, Ferrin I, Salcedo JM, Jakobsson E, Fernandez-Rueda J, Lang V, et al. Human mesenchymal stromal cells modulate T-cell responses through TNF-alpha-mediated activation of NF-kappaB. Eur J Immunol. 2014;44(2):480–8.CrossRefPubMed
71.
go back to reference Polchert D, Sobinsky J, Douglas G, Kidd M, Moadsiri A, Reina E, et al. IFN-gamma activation of mesenchymal stem cells for treatment and prevention of graft versus host disease. Eur J Immunol. 2008;38(6):1745–55.CrossRefPubMedPubMedCentral Polchert D, Sobinsky J, Douglas G, Kidd M, Moadsiri A, Reina E, et al. IFN-gamma activation of mesenchymal stem cells for treatment and prevention of graft versus host disease. Eur J Immunol. 2008;38(6):1745–55.CrossRefPubMedPubMedCentral
72.
go back to reference Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2(2):141–50.CrossRefPubMed Ren G, Zhang L, Zhao X, Xu G, Zhang Y, Roberts AI, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2(2):141–50.CrossRefPubMed
73.
go back to reference Richardson J, Hill AM, Johnston CJ, McGregor A, Norrish AR, Eastwood D, et al. Fracture healing in HIV-positive populations. J Bone Joint Surg Br. 2008;90(8):988–94.CrossRefPubMed Richardson J, Hill AM, Johnston CJ, McGregor A, Norrish AR, Eastwood D, et al. Fracture healing in HIV-positive populations. J Bone Joint Surg Br. 2008;90(8):988–94.CrossRefPubMed
74.
go back to reference Al-Sebaei MO, Daukss DM, Belkina AC, Kakar S, Wigner NA, Cusher D, et al. Role of Fas and Treg cells in fracture healing as characterized in the fas-deficient (lpr) mouse model of lupus. J Bone Miner Res. 2014;29(6):1478–91.CrossRefPubMedPubMedCentral Al-Sebaei MO, Daukss DM, Belkina AC, Kakar S, Wigner NA, Cusher D, et al. Role of Fas and Treg cells in fracture healing as characterized in the fas-deficient (lpr) mouse model of lupus. J Bone Miner Res. 2014;29(6):1478–91.CrossRefPubMedPubMedCentral
75.
go back to reference Briot K, Geusens P, Em Bultink I, Lems WF, Roux C. Inflammatory diseases and bone fragility. Osteoporos Int. 2017. Briot K, Geusens P, Em Bultink I, Lems WF, Roux C. Inflammatory diseases and bone fragility. Osteoporos Int. 2017.
76.
go back to reference Kayal RA, Siqueira M, Alblowi J, McLean J, Krothapalli N, Faibish D, et al. TNF-alpha mediates diabetes-enhanced chondrocyte apoptosis during fracture healing and stimulates chondrocyte apoptosis through FOXO1. J Bone Miner Res. 2010;25(7):1604–15.CrossRefPubMedPubMedCentral Kayal RA, Siqueira M, Alblowi J, McLean J, Krothapalli N, Faibish D, et al. TNF-alpha mediates diabetes-enhanced chondrocyte apoptosis during fracture healing and stimulates chondrocyte apoptosis through FOXO1. J Bone Miner Res. 2010;25(7):1604–15.CrossRefPubMedPubMedCentral
77.
go back to reference Kayal RA, Tsatsas D, Bauer MA, Allen B, Al-Sebaei MO, Kakar S, et al. Diminished bone formation during diabetic fracture healing is related to the premature resorption of cartilage associated with increased osteoclast activity. J Bone Miner Res. 2007;22(4):560–8.CrossRefPubMedPubMedCentral Kayal RA, Tsatsas D, Bauer MA, Allen B, Al-Sebaei MO, Kakar S, et al. Diminished bone formation during diabetic fracture healing is related to the premature resorption of cartilage associated with increased osteoclast activity. J Bone Miner Res. 2007;22(4):560–8.CrossRefPubMedPubMedCentral
78.
go back to reference Tevlin R, Seo EY, Marecic O, McArdle A, Tong X, Zimdahl B, et al. Pharmacological rescue of diabetic skeletal stem cell niches. Sci Transl Med. 2017;9(372). Tevlin R, Seo EY, Marecic O, McArdle A, Tong X, Zimdahl B, et al. Pharmacological rescue of diabetic skeletal stem cell niches. Sci Transl Med. 2017;9(372).
79.
go back to reference Franz S, Rammelt S, Scharnweber D, Simon JC. Immune responses to implants - a review of the implications for the design of immunomodulatory biomaterials. Biomaterials. 2011;32(28):6692–709.CrossRefPubMed Franz S, Rammelt S, Scharnweber D, Simon JC. Immune responses to implants - a review of the implications for the design of immunomodulatory biomaterials. Biomaterials. 2011;32(28):6692–709.CrossRefPubMed
80.
go back to reference Fuchs AK, Syrovets T, Haas KA, Loos C, Musyanovych A, Mailander V, et al. Carboxyl- and amino-functionalized polystyrene nanoparticles differentially affect the polarization profile of M1 and M2 macrophage subsets. Biomaterials. 2016;85:78–87.CrossRefPubMed Fuchs AK, Syrovets T, Haas KA, Loos C, Musyanovych A, Mailander V, et al. Carboxyl- and amino-functionalized polystyrene nanoparticles differentially affect the polarization profile of M1 and M2 macrophage subsets. Biomaterials. 2016;85:78–87.CrossRefPubMed
81.
go back to reference Spiller KL, Nassiri S, Witherel CE, Anfang RR, Ng J, Nakazawa KR, et al. Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds. Biomaterials. 2015;37:194–207.CrossRefPubMed Spiller KL, Nassiri S, Witherel CE, Anfang RR, Ng J, Nakazawa KR, et al. Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds. Biomaterials. 2015;37:194–207.CrossRefPubMed
83.
go back to reference Griffin XL, Wallace D, Parsons N, Costa ML. Platelet rich therapies for long bone healing in adults. Cochrane Database Syst Rev. 2012;7:CD009496. Griffin XL, Wallace D, Parsons N, Costa ML. Platelet rich therapies for long bone healing in adults. Cochrane Database Syst Rev. 2012;7:CD009496.
84.
go back to reference Wei LC, Lei GH, Sheng PY, Gao SG, Xu M, Jiang W, et al. Efficacy of platelet-rich plasma combined with allograft bone in the management of displaced intra-articular calcaneal fractures: A prospective cohort study. J Orthop Res. 2012;30(10):1570–6.CrossRefPubMed Wei LC, Lei GH, Sheng PY, Gao SG, Xu M, Jiang W, et al. Efficacy of platelet-rich plasma combined with allograft bone in the management of displaced intra-articular calcaneal fractures: A prospective cohort study. J Orthop Res. 2012;30(10):1570–6.CrossRefPubMed
85.
go back to reference Golos J, Walinski T, Piekarczyk P, Kwiatkowski K. Results of the use of platelet rich plasma in the treatment of delayed union of long bones. Ortop Traumatol Rehabil. 2014;16(4):397–406.CrossRefPubMed Golos J, Walinski T, Piekarczyk P, Kwiatkowski K. Results of the use of platelet rich plasma in the treatment of delayed union of long bones. Ortop Traumatol Rehabil. 2014;16(4):397–406.CrossRefPubMed
86.
go back to reference Malhotra R, Kumar V, Garg B, Singh R, Jain V, Coshic P, et al. Role of autologous platelet-rich plasma in treatment of long-bone nonunions: a prospective study. Musculoskelet Surg. 2015;99(3):243–8. Malhotra R, Kumar V, Garg B, Singh R, Jain V, Coshic P, et al. Role of autologous platelet-rich plasma in treatment of long-bone nonunions: a prospective study. Musculoskelet Surg. 2015;99(3):243–8.
Metadata
Title
The Role of the Immune Cells in Fracture Healing
Publication date
01-04-2018
Published in
Current Osteoporosis Reports / Issue 2/2018
Print ISSN: 1544-1873
Electronic ISSN: 1544-2241
DOI
https://doi.org/10.1007/s11914-018-0423-2

Other articles of this Issue 2/2018

Current Osteoporosis Reports 2/2018 Go to the issue

Regenerative Biology and Medicine in Osteoporosis (T Webster, Section Editor)

Progress of Regenerative Therapy in Orthopedics

Regenerative Biology and Medicine in Osteoporosis (T Webster, Section Editor)

Preclinical and Translational Studies in Small Ruminants (Sheep and Goat) as Models for Osteoporosis Research

Bone Marrow and Adipose Tissue (G Duque and B Lecka-Czernik, Section Editors)

Good, Bad, or Ugly: the Biological Roles of Bone Marrow Fat

Orthopedic Management of Fractures (S Bukata and L Gerstenfeld, Section Editors)

Clinical and Research Approaches to Treat Non-union Fracture