Journal of Peking University (Health Sciences) ›› 2021, Vol. 53 ›› Issue (6): 1171-1177. doi: 10.19723/j.issn.1671-167X.2021.06.027

Previous Articles     Next Articles

Early constant observation of the effect of deferoxamine mesylate on improvement of vascularized bone regeneration in SD rat skull critical size defect model

DU Wen-yu,YANG Jing-wen(),JIANG Ting()   

  1. Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
  • Received:2019-12-12 Online:2021-12-18 Published:2021-12-13
  • Contact: Jing-wen YANG,Ting JIANG E-mail:jingwen.yang@foxmail.com;jt_ketizu@163.com
  • Supported by:
    National Natural Science Foundation of China(81771045);Research Foundation of Peking University School and Hospital of Stomatology(PKUSS20180106)

Abstract:

Objective: To investigate the effect of local administration of deferoxamine mesylate (DFO) on vascularization and osteogenesis and its ability to maintain the activity of hypoxia inducible factor-1α (HIF-1α), by constantly observing early changes of vessel-like structures and bone tissues during bone defects healing. Methods: Skull critical bone defect models were constructed on a total of thirty male SD rats (6-8 weeks old). The rats were randomly divided into experimental group (DFO group) or control group (normal saline group). 300 μL 200 μmol/L DFO solution or normal saline was locally injected on the 4th day after the defect was made. On the 5th, 7th, 10th, 14th, and 28th days after surgery, three rats in each group were sacrificed respectively. HE staining and Masson staining were performed to observe new bone formation and mineralization. HIF-1α immunohistochemistry staining was performed to examine relative expression of protein. Qualitative analysis and comparation were performed by t-tests on relative expression of HIF-1α, numbers of blood vessels and percentages of mineralization tissues of new bone areas. Results: On the 5th, 7th, 10th, 14th and 28th days after surgery, the average numbers of blood vessels were 30.40±12.15, 62.00±17.87, 73.43±15.63, 40.00±7.84, 48.71±11.64 in the DFO group, and 18.75±6.63, 19.13±2.80, 51.35±16.21, 27.18±7.32, 30.88±13.43 in the control group. The number of blood vessels in the DFO group was significantly higher than that of the control group at each time point (P<0.05). The mass of new bone in the DFO group was higher than that in the control group on the 14th and 28th days after surgery. The percentage of mineralization tissues of new bone area on the 14th and 28th days after injection were (27.73±5.93)% and (46.53±3.66)% in the DFO group, and (11.99±2.02)% and (31.98±4.22)% in the control group. The percentage of mineralization tissues in the DFO group was significantly higher than that of the control group at each time point (P<0.001). The relative expression of HIF-1α in the DFO group compared with the control group was 2.86±0.48, 1.32±0.26, 1.32±0.32, 1.28±0.38 and 1.05±0.34 on the 5th, 7th, 10th, 14th and 28th days, with significant expression difference on the 5th day (P<0.01). Conclusion: Use of DFO in bone defects promotes vascularization and osteogenesis in the defect area, and maintains the protein activity of HIF-1α temporarily.

Key words: Deferoxamine, Vascularization, Osteogenesis, Hypoxia inducible factor 1, alpha subunit

CLC Number: 

  • R318.08

Figure 1

SD rats skull critical size defect modelA critical size defect of skull was made and the periosteum was kept complete."

Figure 2

Immunohistochemistry staining images of the vessels marker CD31 (n=3) Yellow arrow indicates the examples of vascular tissue, “B” marks the defect border or the edge of new bone. DFO, deferoxamine mesylate."

Table 1

Counting of vessel numbers in the DFO group and control group"

Post-operation days/d Control group ($\overline{x}$±s) DFO group ($\overline{x}$±s) t P
5 18.75±6.63 30.40±12.15 2.439 0.027
7 19.13±2.80 62.00±17.87 6.671 <0.001
10 51.35±16.21 73.43±15.63 4.349 <0.001
14 27.18±7.32 40.00±7.84 3.660 0.002
28 30.88±13.43 48.71±11.64 2.727 0.017

Figure 3

HE staining (A) and Masson staining (B) images of the bone defect area (n=3) New bone is circulated by yellow and marked with “NB”. DFO, deferoxamine mesylate."

Table 2

Measuring of percentages of mineralized tissue area"

Post-operation days/d Control group ($\overline{x}$±s) DFO group ($\overline{x}$±s) t P
14 (11.99±2.02)% (27.73±5.93)% 5.967 <0.001
28 (31.98±4.22)% (46.53±3.66)% 6.045 <0.001

Figure 4

Expression of HIF-1α (DAB ×200, n=3) Examples of HIF-1α protein expression positively stained in brown is indicated with yellow arrow. DFO, deferoxamine mesylate; HIF-1α, hypoxia inducible factor 1α."

Table 3

Relative expression of HIF-1α (fold change)"

Post-operation
days/d
Relative expression of
HIF-1α (fold change, $\overline{x}$±s)
t P
5 2.86±0.48 7.660 0.002
7 1.32±0.26 2.131 0.083
10 1.32±0.32 2.007 0.069
14 1.28±0.38 1.454 0.121
28 1.05±0.34 0.196 0.438
[1] Yellowley CE, Genetos DC. Hypoxia signaling in the skeleton: Implications for bone health[J]. Curr Osteoporos Rep, 2019, 17(1):26-35.
doi: 10.1007/s11914-019-00500-6 pmid: 30725321
[2] Holden P, Nair L. Deferoxamine: An angiogenic and antioxidant molecule for tissue regeneration[J]. Tissue Eng Part B Rev, 2019, 25(6):461-470.
doi: 10.1089/ten.teb.2019.0111
[3] Temiz G, Sirinoglu H, Yesiloglu N, et al. Effects of deferoxamine on fat graft survival[J]. Facial Plast Surg, 2016, 32(4):438-443.
doi: 10.1055/s-0036-1584236
[4] Farzan R, Moeinian M, Abdollahi A, et al. Effects of amniotic membrane extract and deferoxamine on angiogenesis in wound healing: An in vivo model[J]. J Wound Care, 2018, 27(Suppl 6):s26-s32.
[5] Ram M, Singh V, Kumawat S, et al. Deferoxamine modulates cytokines and growth factors to accelerate cutaneous wound healing in diabetic rats[J]. Eur J Pharmacol, 2015, 764:9-21.
doi: 10.1016/j.ejphar.2015.06.029
[6] Gao SQ, Chang C, Li JJ, et al. Co-delivery of deferoxamine and hydroxysafflor yellow A to accelerate diabetic wound healing via enhanced angiogenesis[J]. Drug Deliv, 2018, 25(1):1779-1789.
doi: 10.1080/10717544.2018.1513608
[7] Yang Q, He GW, Underwood MJ, et al. Cellular and molecular mechanisms of endothelial ischemia/reperfusion injury: Perspectives and implications for postischemic myocardial protection[J]. Am J Transl Res, 2016, 8(2):765-777.
pmid: 27158368
[8] Jia P, Chen H, Kang H, et al. Deferoxamine released from poly(lactic-co-glycolic acid) promotes healing of osteoporotic bone defect via enhanced angiogenesis and osteogenesis[J]. J Biomed Mater Res A, 2016, 104(10):2515-2527.
doi: 10.1002/jbm.a.35793
[9] 姚洋, 杜宇, 古霞, 等. 局部注射外源性神经生长因子促进小鼠钛种植体周骨胶原早期成熟的研究[J]. 华西口腔医学杂志, 2018, 36(2):128-132.
[10] Koivunen P, Serpi R, Dimova EY. Hypoxia-inducible factor prolyl 4-hydroxylase inhibition in cardiometabolic diseases[J]. Pharmacol Res, 2016, 114:265-273.
doi: S1043-6618(16)31160-4 pmid: 27832958
[11] Lanigan S, Corcoran AE, Wall A, et al. Acute hypoxic exposure and prolyl-hydroxylase inhibition improves synaptic transmission recovery time from a subsequent hypoxic insult in rat hippocampus[J]. Brain Res, 2018, 1701:212-218.
[12] Farberg AS, Sarhaddi D, Donneys A, et al. Deferoxamine enhances bone regeneration in mandibular distraction osteogenesis[J]. Plast Reconstr Surg, 2014, 133(3):666-671.
doi: 10.1097/01.prs.0000438050.36881.a9 pmid: 24572857
[13] Zhang J, Zheng L, Wang Z, et al. Lowering iron level protects against bone loss in focally irradiated and contralateral femurs through distinct mechanisms[J]. Bone, 2019, 120:50-60.
doi: S8756-3282(18)30369-7 pmid: 30304704
[14] Einhorn TA, Gerstenfeld LC. Fracture healing: mechanisms and interventions[J]. Nat Rev Rheumatol, 2015, 11(1):45-54.
doi: 10.1038/nrrheum.2014.164 pmid: 25266456
[15] Morgan EF, Giacomo A, Gerstenfeld LC. Overview of skeletal repair (fracture healing and its assessment)[J]. Methods Mol Biol, 2021, 2230:17-37.
[16] Donneys A, Deshpande SS, Tchanque-Fossuo CN, et al. Deferoxa-mine expedites consolidation during mandibular distraction osteogenesis[J]. Bone, 2013, 55(2):384-390.
doi: 10.1016/j.bone.2013.04.005 pmid: 23598047
[17] Zimna A, Kurpisz M. Hypoxia-inducible factor-1 in physiological and pathophysiological angiogenesis: Applications and therapies[J]. Biomed Res Int, 2015, 2015:549412.
[18] Drager J, Harvey EJ, Barralet J. Hypoxia signalling manipulation for bone regeneration[J]. Expert Rev Mol Med, 2015, 17:e6.
doi: 10.1017/erm.2015.4
[19] Matsubara H, Hogan DE, Morgan EF, et al. Vascular tissues are a primary source of BMP2 expression during bone formation induced by distraction osteogenesis[J]. Bone, 2012, 51(1):168-180.
doi: 10.1016/j.bone.2012.02.017 pmid: 22391215
[20] Bouletreau PJ, Warren SM, Spector JA, et al. Hypoxia and VEGF up-regulate BMP-2 mRNA and protein expression in microvascular endothelial cells: Implications for fracture healing[J]. Plast Reconstr Surg, 2002, 109(7):2384-2397.
pmid: 12045566
[1] WANG Jing-qi,WANG Xiao. In vivo study of strontium-doped calcium phosphate cement for biological properties [J]. Journal of Peking University (Health Sciences), 2021, 53(2): 378-383.
[2] ZHANG Sheng-nan,AN Na,OUYANG Xiang-ying,LIU Ying-jun,WANG Xue-kui. Role of growth arrest-specific protein 6 in migration and osteogenic differentiation of human periodontal ligament cells [J]. Journal of Peking University (Health Sciences), 2021, 53(1): 9-15.
[3] Mei WANG, Bo-wen LI, Si-wen WANG, Yu-hua LIU. Preparation and osteogenic effect study of small intestinal submucosa sponge [J]. Journal of Peking University (Health Sciences), 2020, 52(5): 952-958.
[4] Ying CHEN,Zhong-ning LIU,Bo LI,Ting JIANG. Preparation of aspirin sustained-release microsphere and its in vitro releasing [J]. Journal of Peking University(Health Sciences), 2019, 51(5): 907-912.
[5] SUI Hua-xin, LV Pei-jun, WANG Yong, FENG Yu-chi. Effects of low level laser irradiation on the osteogenic capacity of sodium alginate/gelatin/human adipose-derived stem cells 3D bio-printing construct [J]. Journal of Peking University(Health Sciences), 2018, 50(5): 868-875.
[6] LU Jin-hui, QIAN Jun, LIU He, ZHU Jun-xia. Clinical study on autologus platelet-rich fibrin aided revascularization of immature permanent teeth [J]. Journal of Peking University(Health Sciences), 2018, 50(4): 672-679.
[7] LIU Jing-yin, CHEN Fei, GE Yan-jun, WEI Ling, PAN Shao-xia, FENG Hai-lan. Influence of implants prepared by selective laser melting on early bone healing [J]. Journal of Peking University(Health Sciences), 2018, 50(1): 117-122.
[8] ZHANG Lu-feng, LING Yun-peng, YANG Hang, GONG Yi-chen, SONG Zhi-ming, WAN Feng. Comparison of outcomes of two minimally invasive approaches for multi-vessel coronary revascularization [J]. Journal of Peking University(Health Sciences), 2017, 49(6): 1066-1070.
[9] CHEN Fei, PAN Shao-xia, FENG Hai-lan. Distribution and content of transforming growth factor-β1 and vascular endothelial growth factor in each layer of concentrated growth factors [J]. Journal of Peking University(Health Sciences), 2016, 48(5): 860-865.
[10] QIN Xue-yan, ZHAO Hua-xiang, ZHANG Qian, CHEN Feng, LIN Jiu-xiang. NELL-1: a novel highly efficient and specific growth factor [J]. Journal of Peking University(Health Sciences), 2016, 48(2): 380-383.
[11] GE Wen-shu, TANG Yi-man, ZHANG Xiao, LIU Yun-song, ZHOU Yong-sheng. Establishing a luciferase reporter system to evaluate osteogenic differentiation potential of human adipose-derived stem cells [J]. Journal of Peking University(Health Sciences), 2016, 48(1): 170-174.
[12] SONG Yang, WANG Xiao-fei, WANG Yu-guang, SUN Yu-chun, LV Pei-jun. Osteogenesis of human adipose-derived mesenchymal stem cells-biomaterial mixture in vivo after 3D bio-printing [J]. Journal of Peking University(Health Sciences), 2016, 48(1): 45-50.
[13] 欧Meng-恩 , ZHANG Xiao, LIU Yun-Song, GE Yan-Jun, ZHOU Yong-Sheng. Ectopic osteogenesis of stromal cell-derived factor 1 combined with simvastatin loaded collagen scaffold in vivo [J]. Journal of Peking University(Health Sciences), 2015, 47(1): 47-51.
[14] DING Qian, ZHANG Feng-qiu, MA Yu-shi. Effect of icariin on osteoblastic differentiation gene expression of human periodontal ligament cells [J]. Journal of Peking University(Health Sciences), 2013, 45(6): 975-978.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
[1] . [J]. Journal of Peking University(Health Sciences), 2009, 41(4): 456 -458 .
[2] . [J]. Journal of Peking University(Health Sciences), 2009, 41(2): 125 -128 .
[3] . [J]. Journal of Peking University(Health Sciences), 2009, 41(2): 135 -140 .
[4] . [J]. Journal of Peking University(Health Sciences), 2009, 41(2): 158 -161 .
[5] . [J]. Journal of Peking University(Health Sciences), 2009, 41(2): 217 -220 .
[6] . [J]. Journal of Peking University(Health Sciences), 2009, 41(1): 52 -55 .
[7] . [J]. Journal of Peking University(Health Sciences), 2009, 41(1): 109 -111 .
[8] . [J]. Journal of Peking University(Health Sciences), 2009, 41(3): 297 -301 .
[9] . [J]. Journal of Peking University(Health Sciences), 2009, 41(5): 505 -515 .
[10] . [J]. Journal of Peking University(Health Sciences), 2009, 41(5): 599 -601 .