Skip to main content
Erschienen in: Clinical Orthopaedics and Related Research® 5/2016

25.11.2015 | Basic Research

Does Sclerostin Depletion Stimulate Fracture Healing in a Mouse Model?

verfasst von: Mohammad M. Alzahrani, MD, MSc, Frank Rauch, MD, Reggie C. Hamdy, MB, MSc(Ortho), FRCS(C)

Erschienen in: Clinical Orthopaedics and Related Research® | Ausgabe 5/2016

Einloggen, um Zugang zu erhalten

Abstract

Background

Sclerostin is a secreted glycoprotein that inhibits the intracellular Wnt signaling pathway, which, when inactivated, stimulates bone formation. This has been seen in fracture studies, which have shown larger and stronger calluses with accelerated fracture healing in sclerostin knockout and sclerostin antibody injection models. However, the effects of these two mechanisms have not been compared in the context of fracture healing.

Questions/purposes

We sought to determine the degree to which sclerostin inhibition (Scl-Ab injection) and complete sclerostin depletion inhibit fracture healing in a mouse model as evaluated by (1) morphometric trabecular bone measures at the fracture site, and (2) fracture site structural strength.

Methods

Ten-week-old male sclerostin knockout (n = 20) and wild type (n = 40) mice underwent insertion of a tibial intramedullary pin after which a midshaft tibial osteotomy was performed. The mice were divided in three groups: sclerostin knockout (n = 20), wild type with sclerostin antibody injection (intravenous dose of 100 mg/kg weekly) (n = 20), and wild type with saline injection (n = 20). The mice for each group where subdivided and euthanized at 14, 21, 28, and 35 days after surgery, at which time the fractured tibias were assessed with microCT (to assess morphometric trabecular bone measures: bone volume to total volume (BV/TV), trabecular thickness, trabecular number, and structural model index at the fracture site. Biomechanical testing in the form of three-point bending also was done to assess fracture site structural strength. A difference greater than 3.7% in our primary outcome (BV/TV) would be required to detect a difference between groups with a power of 80%, as per our power analysis.

Results

The wild type with sclerostin antibody and the sclerostin knockout groups showed increased trabecular BV/TV at the fracture site compared with the wild type group with saline at all times, however no difference was seen between the treatment groups with the numbers available, except at 28 days postoperatively when the sclerostin knockout group showed greater BV/TV than the wild type sclerostin antibody group (47.0 ± 3.5 vs 40.1 ± 2.1; p < 0.05). On biomechanical testing the wild type sclerostin antibody showed increased stiffness at Days 14 and 28 compared with the wild type with saline group (70.9 ± 6.4 vs 14.8 ± 8.1; p = 0.001), (106.8 ± 24.3 vs 74.9 ± 16.0; p = 0.004); respectively. However, with the numbers available, no differences were detected between the wild type with sclerostin antibody and the sclerostin knockout groups in terms of whole-bone structural strength.

Conclusions

Sclerostin antibody injections showed promising results, which were not different with the numbers available, from results achieved with complete depletion of sclerostin, especially at earlier stages of the healing process, and therefore completed the healing process at an earlier time.

Clinical Relevance

Sclerostin antibody injections appear to enhance fracture healing to a degree that is not different than complete sclerostin depletion, but larger animal studies are required to assess the accurate dosage and timing of administration in the fracture healing process to further evaluate its potential clinical utility to enhance fracture healing.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Axelrad TW, Kakar S, Einhorn TA, New technologies for the enhancement of skeletal repair. Injury. 2007;38(suppl 1):S49–62.CrossRefPubMed Axelrad TW, Kakar S, Einhorn TA, New technologies for the enhancement of skeletal repair. Injury. 2007;38(suppl 1):S49–62.CrossRefPubMed
3.
Zurück zum Zitat Bonnarens F, Einhorn TA, Production of a standard closed fracture in laboratory animal bone. J Orthop Res. 1984;2:97–101.CrossRefPubMed Bonnarens F, Einhorn TA, Production of a standard closed fracture in laboratory animal bone. J Orthop Res. 1984;2:97–101.CrossRefPubMed
4.
Zurück zum Zitat Chen Y, Whetstone HC, Lin AC, Nadesan P, Wei Q, Poon R, Alman BA. Beta-catenin signaling plays a disparate role in different phases of fracture repair: implications for therapy to improve bone healing. PLoS Med. 2007;4:e249.CrossRefPubMedPubMedCentral Chen Y, Whetstone HC, Lin AC, Nadesan P, Wei Q, Poon R, Alman BA. Beta-catenin signaling plays a disparate role in different phases of fracture repair: implications for therapy to improve bone healing. PLoS Med. 2007;4:e249.CrossRefPubMedPubMedCentral
5.
6.
Zurück zum Zitat Cui L, Cheng H, Song C, Li C, Simonet WS, Ke HZ, Li G. Time-dependent effects of sclerostin antibody on a mouse fracture healing model. J Musculoskelet Neuronal Interact. 2013;13:178–184.PubMed Cui L, Cheng H, Song C, Li C, Simonet WS, Ke HZ, Li G. Time-dependent effects of sclerostin antibody on a mouse fracture healing model. J Musculoskelet Neuronal Interact. 2013;13:178–184.PubMed
7.
Zurück zum Zitat French DM, Kaul RJ, D’Souza AL, Crowley CW, Bao M, Frantz GD, Filvaroff EH, Desnoyers L. WISP-1 is an osteoblastic regulator expressed during skeletal development and fracture repair. Am J Pathol. 2004;165:855–867.CrossRefPubMedPubMedCentral French DM, Kaul RJ, D’Souza AL, Crowley CW, Bao M, Frantz GD, Filvaroff EH, Desnoyers L. WISP-1 is an osteoblastic regulator expressed during skeletal development and fracture repair. Am J Pathol. 2004;165:855–867.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Gamie Z, Korres N, Leonidou A, Gray AC, Tsiridis E. Sclerostin monoclonal antibodies on bone metabolism and fracture healing. Expert Opin Investig Drugs. 2012;21:1523–1534.CrossRefPubMed Gamie Z, Korres N, Leonidou A, Gray AC, Tsiridis E. Sclerostin monoclonal antibodies on bone metabolism and fracture healing. Expert Opin Investig Drugs. 2012;21:1523–1534.CrossRefPubMed
9.
Zurück zum Zitat Garrison KR, Donell S, Ryder J, Shemilt I, Mugford M, Harvey I, Song F. Clinical effectiveness and cost-effectiveness of bone morphogenetic proteins in the non-healing of fractures and spinal fusion: a systematic review. Health Technol Assess. 2007;11:1–150, iii–iv. Garrison KR, Donell S, Ryder J, Shemilt I, Mugford M, Harvey I, Song F. Clinical effectiveness and cost-effectiveness of bone morphogenetic proteins in the non-healing of fractures and spinal fusion: a systematic review. Health Technol Assess. 2007;11:1–150, iii–iv.
10.
Zurück zum Zitat Govender S, Csimma C, Genant HK, Valentin-Opran A, Amit Y, Arbel R, Aro H, Atar D, Bishay M, Borner MG, Chiron P, Choong P, Cinats J, Courtenay B, Feibel R, Geulette B, Gravel C, Haas N, Raschke M, Hammacher E, van der Velde D, Hardy P, Holt M, Josten C, Ketterl RL, Lindeque B, Lob G, Mathevon H, McCoy G, Marsh D, Miller R, Munting E, Oevre S, Nordsletten L, Patel A, Pohl A, Rennie W, Reynders P, Rommens PM, Rondia J, Rossouw WC, Daneel PJ, Ruff S, Ruter A, Santavirta S, Schildhauer TA, Gekle C, Schnettler R, Segal D, Seiler H, Snowdowne RB, Stapert J, Taglang G, Verdonk R, Vogels L, Weckbach A, Wentzensen A, Wisniewski T; BMP-2 Evaluation in Surgery for Tibial Trauma (BESTT) Study Group. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg Am. 2002;84:2123–2134. Govender S, Csimma C, Genant HK, Valentin-Opran A, Amit Y, Arbel R, Aro H, Atar D, Bishay M, Borner MG, Chiron P, Choong P, Cinats J, Courtenay B, Feibel R, Geulette B, Gravel C, Haas N, Raschke M, Hammacher E, van der Velde D, Hardy P, Holt M, Josten C, Ketterl RL, Lindeque B, Lob G, Mathevon H, McCoy G, Marsh D, Miller R, Munting E, Oevre S, Nordsletten L, Patel A, Pohl A, Rennie W, Reynders P, Rommens PM, Rondia J, Rossouw WC, Daneel PJ, Ruff S, Ruter A, Santavirta S, Schildhauer TA, Gekle C, Schnettler R, Segal D, Seiler H, Snowdowne RB, Stapert J, Taglang G, Verdonk R, Vogels L, Weckbach A, Wentzensen A, Wisniewski T; BMP-2 Evaluation in Surgery for Tibial Trauma (BESTT) Study Group. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg Am. 2002;84:2123–2134.
11.
Zurück zum Zitat Jho EH, Zhang T, Domon C, Joo CK, Freund JN, Costantini F. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 2002;22: 1172–1183.CrossRefPubMedPubMedCentral Jho EH, Zhang T, Domon C, Joo CK, Freund JN, Costantini F. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol Cell Biol. 2002;22: 1172–1183.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Kramer I, Loots GG, A. Studer A, Keller H, Kneissel M. Parathyroid hormone (PTH)-induced bone gain is blunted in SOST overexpressing and deficient mice. J Bone Miner Res. 2010;25:178–189.CrossRefPubMedPubMedCentral Kramer I, Loots GG, A. Studer A, Keller H, Kneissel M. Parathyroid hormone (PTH)-induced bone gain is blunted in SOST overexpressing and deficient mice. J Bone Miner Res. 2010;25:178–189.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Li C, Ominsky MS, Tan HL, Barrero M, Niu QT, Asuncion FJ, Lee E, Liu M, Simonet WS, Paszty C, Ke HZ. Increased callus mass and enhanced strength during fracture healing in mice lacking the sclerostin gene. Bone. 2011;49:1178–1185.CrossRefPubMed Li C, Ominsky MS, Tan HL, Barrero M, Niu QT, Asuncion FJ, Lee E, Liu M, Simonet WS, Paszty C, Ke HZ. Increased callus mass and enhanced strength during fracture healing in mice lacking the sclerostin gene. Bone. 2011;49:1178–1185.CrossRefPubMed
15.
Zurück zum Zitat Li X, Ominsky MS, Niu QT, Sun N, Daugherty B, D’Agostin D, Kurahara C, Gao Y, Cao J, Gong J, Asuncion F, Barrero M, Warmington K, Dwyer D, Stolina M, Morony S, Sarosi I, Kostenuik PJ, Lacey DL, Simonet WS, Ke HZ, Paszty C. Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J Bone Miner Res. 2008;23:860–869.CrossRefPubMed Li X, Ominsky MS, Niu QT, Sun N, Daugherty B, D’Agostin D, Kurahara C, Gao Y, Cao J, Gong J, Asuncion F, Barrero M, Warmington K, Dwyer D, Stolina M, Morony S, Sarosi I, Kostenuik PJ, Lacey DL, Simonet WS, Ke HZ, Paszty C. Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. J Bone Miner Res. 2008;23:860–869.CrossRefPubMed
16.
Zurück zum Zitat Li X, Ominsky MS, Warmington KS, Morony S, Gong J, Cao J, Gao Y, Shalhoub V, Tipton B, Haldankar R, Chen Q, Winters A, Boone T, Geng Z, Niu QT, Ke HZ, Kostenuik PJ, Simonet WS, Lacey DL, Paszty C. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Miner Res. 2009;24:578–588.CrossRefPubMed Li X, Ominsky MS, Warmington KS, Morony S, Gong J, Cao J, Gao Y, Shalhoub V, Tipton B, Haldankar R, Chen Q, Winters A, Boone T, Geng Z, Niu QT, Ke HZ, Kostenuik PJ, Simonet WS, Lacey DL, Paszty C. Sclerostin antibody treatment increases bone formation, bone mass, and bone strength in a rat model of postmenopausal osteoporosis. J Bone Miner Res. 2009;24:578–588.CrossRefPubMed
17.
Zurück zum Zitat Makhdom AM, Hamdy RC. The role of growth factors on acceleration of bone regeneration during distraction osteogenesis. Tissue Eng Part B Rev. 2013;19:442–453.CrossRefPubMed Makhdom AM, Hamdy RC. The role of growth factors on acceleration of bone regeneration during distraction osteogenesis. Tissue Eng Part B Rev. 2013;19:442–453.CrossRefPubMed
18.
Zurück zum Zitat Makhdom AM, Rauch F, Lauzier D, Hamdy RC. The effect of systemic administration of sclerostin antibody in a mouse model of distraction osteogenesis. J Musculoskelet Neuronal Interact. 2014;14:124–130.PubMed Makhdom AM, Rauch F, Lauzier D, Hamdy RC. The effect of systemic administration of sclerostin antibody in a mouse model of distraction osteogenesis. J Musculoskelet Neuronal Interact. 2014;14:124–130.PubMed
19.
Zurück zum Zitat Marsh D. Concepts of fracture union, delayed union, and nonunion. Clin Orthop Relat Res. 1998;355(suppl):S22–30.CrossRefPubMed Marsh D. Concepts of fracture union, delayed union, and nonunion. Clin Orthop Relat Res. 1998;355(suppl):S22–30.CrossRefPubMed
20.
Zurück zum Zitat Morse A, McDonald M, Kelly N, Melville K, Schindeler A, Kramer I, Kneissel M, van der Meulen M, Little D. Mechanical load increases in bone formation via a dclerostin-independent pathway. J Bone Miner Res. 2014;29:2456–2467.CrossRefPubMedPubMedCentral Morse A, McDonald M, Kelly N, Melville K, Schindeler A, Kramer I, Kneissel M, van der Meulen M, Little D. Mechanical load increases in bone formation via a dclerostin-independent pathway. J Bone Miner Res. 2014;29:2456–2467.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Morse A, Yu NY, Peacock L, Mikulec K, Kramer I, Kneissel M, McDonald MM, Little DG. Endochondral fracture healing with external fixation in the Sost knockout mouse results in earlier fibrocartilage callus removal and increased bone volume fraction and strength. Bone. 2015;71:155–163.CrossRefPubMed Morse A, Yu NY, Peacock L, Mikulec K, Kramer I, Kneissel M, McDonald MM, Little DG. Endochondral fracture healing with external fixation in the Sost knockout mouse results in earlier fibrocartilage callus removal and increased bone volume fraction and strength. Bone. 2015;71:155–163.CrossRefPubMed
22.
Zurück zum Zitat Ominsky MS, Li C, Li X, Tan HL, Lee E, Barrero M, Asuncion FJ, Dwyer D, Han CY, Vlasseros F, Samadfam R, Jolette J, Smith SY, Stolina M, Lacey DL, Simonet WS, Paszty C, Li G, Ke HZ. Inhibition of sclerostin by monoclonal antibody enhances bone healing and improves bone density and strength of nonfractured bones. J Bone Miner Res. 2011;26:1012–1021.CrossRefPubMed Ominsky MS, Li C, Li X, Tan HL, Lee E, Barrero M, Asuncion FJ, Dwyer D, Han CY, Vlasseros F, Samadfam R, Jolette J, Smith SY, Stolina M, Lacey DL, Simonet WS, Paszty C, Li G, Ke HZ. Inhibition of sclerostin by monoclonal antibody enhances bone healing and improves bone density and strength of nonfractured bones. J Bone Miner Res. 2011;26:1012–1021.CrossRefPubMed
23.
Zurück zum Zitat Phieffer LS, Goulet JA. Delayed unions of the tibia. J Bone Joint Surg Am. 2006;88:206–216.PubMed Phieffer LS, Goulet JA. Delayed unions of the tibia. J Bone Joint Surg Am. 2006;88:206–216.PubMed
24.
Zurück zum Zitat Poynton AR, Lane JM. Safety profile for the clinical use of bone morphogenetic proteins in the spine. Spine (Phila Pa 1976). 2002;27(16 suppl 1):S40–48. Poynton AR, Lane JM. Safety profile for the clinical use of bone morphogenetic proteins in the spine. Spine (Phila Pa 1976). 2002;27(16 suppl 1):S40–48.
25.
Zurück zum Zitat Sarahrudi K, Thomas A, Albrecht C, Aharinejad S. Strongly enhanced levels of sclerostin during human fracture healing. J Orthop Res. 2012;30:1549–1555.CrossRefPubMed Sarahrudi K, Thomas A, Albrecht C, Aharinejad S. Strongly enhanced levels of sclerostin during human fracture healing. J Orthop Res. 2012;30:1549–1555.CrossRefPubMed
26.
Zurück zum Zitat ten Dijke P, Krause C, de Gorter DJ, Lowik CW, van Bezooijen RL. Osteocyte-derived sclerostin inhibits bone formation: its role in bone morphogenetic protein and Wnt signaling. J Bone Joint Surg Am. 2008;90(suppl 1):31–35.CrossRefPubMed ten Dijke P, Krause C, de Gorter DJ, Lowik CW, van Bezooijen RL. Osteocyte-derived sclerostin inhibits bone formation: its role in bone morphogenetic protein and Wnt signaling. J Bone Joint Surg Am. 2008;90(suppl 1):31–35.CrossRefPubMed
27.
Zurück zum Zitat U.S. Department of Health and Human Services. The Guide for the Care and Use of Laboratory Animals. (NIH Publication No. 85–23). Bethesda, MD: National Institutes of Health; 1985. U.S. Department of Health and Human Services. The Guide for the Care and Use of Laboratory Animals. (NIH Publication No. 85–23). Bethesda, MD: National Institutes of Health; 1985.
28.
Zurück zum Zitat Winkler DG, Sutherland MK, Geoghegan JC, Yu C, Hayes T, Skonier JE, Shpektor D, Jonas M, Kovacevich BE, Staehling-Hampton K, Appleby M, Brunkow ME, Latham JA. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. EMBO J. 2003;22:6267–6276.CrossRefPubMedPubMedCentral Winkler DG, Sutherland MK, Geoghegan JC, Yu C, Hayes T, Skonier JE, Shpektor D, Jonas M, Kovacevich BE, Staehling-Hampton K, Appleby M, Brunkow ME, Latham JA. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. EMBO J. 2003;22:6267–6276.CrossRefPubMedPubMedCentral
Metadaten
Titel
Does Sclerostin Depletion Stimulate Fracture Healing in a Mouse Model?
verfasst von
Mohammad M. Alzahrani, MD, MSc
Frank Rauch, MD
Reggie C. Hamdy, MB, MSc(Ortho), FRCS(C)
Publikationsdatum
25.11.2015
Verlag
Springer US
Erschienen in
Clinical Orthopaedics and Related Research® / Ausgabe 5/2016
Print ISSN: 0009-921X
Elektronische ISSN: 1528-1132
DOI
https://doi.org/10.1007/s11999-015-4640-z

Weitere Artikel der Ausgabe 5/2016

Clinical Orthopaedics and Related Research® 5/2016 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.