Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2024, Vol. 18 Issue (3) : 516-537    https://doi.org/10.1007/s11684-023-1024-8
A distinct “repair” role of regulatory T cells in fracture healing
Tingting Wu1,2, Lulu Wang1,2, Chen Jian1,2, Zhenhe Zhang3, Ruiyin Zeng3, Bobin Mi3, Guohui Liu3, Yu Zhang1,2, Chen Shi1,2()
1. Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
2. Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
3. Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
 Download: PDF(3994 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Regulatory T cells (Tregs) suppress immune responses and inflammation. Here, we described the distinct nonimmunological role of Tregs in fracture healing. The recruitment from the circulation pool, peripheral induction, and local expansion rapidly enriched Tregs in the injured bone. The Tregs in the injured bone displayed superiority in direct osteogenesis over Tregs from lymphoid organs. Punctual depletion of Tregs compromised the fracture healing process, which leads to increased bone nonunion. In addition, bone callus Tregs showed unique T-cell receptor repertoires. Amphiregulin was the most overexpressed protein in bone callus Tregs, and it can directly facilitate the proliferation and differentiation of osteogenic precursor cells by activation of phosphatidylinositol 3-kinase/protein kinase B signaling pathways. The results of loss- and gain-function studies further evidenced that amphiregulin can reverse the compromised healing caused by Treg dysfunction. Tregs also enriched in patient bone callus and amphiregulin can promote the osteogenesis of human pre-osteoblastic cells. Our findings indicate the distinct and nonredundant role of Tregs in fracture healing, which will provide a new therapeutic target and strategy in the clinical treatment of fractures.

Keywords regulatory T cells      fracture healing      amphiregulin      non-union      osteogenesis     
Corresponding Author(s): Chen Shi   
Just Accepted Date: 28 November 2023   Online First Date: 15 March 2024    Issue Date: 17 June 2024
 Cite this article:   
Tingting Wu,Lulu Wang,Chen Jian, et al. A distinct “repair” role of regulatory T cells in fracture healing[J]. Front. Med., 2024, 18(3): 516-537.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-023-1024-8
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I3/516
Fig.1  Treg accumulation in fractured bone. (A) Flow scatter diagram of CD4+ T cells in the spleen, peripheral blood, and bone callus before fracture (day 0) and days 1, 3, 5, 14, and 21 after fracture. Numbers indicate the proportion of cells in the frame. (B–D) Quantitative analysis of CD4+ T cells in the spleen (B), peripheral blood (C), and bone callus (D) at different time points after fracture (n = 3). (E) Flow scatter diagram of Tregs in the spleen, peripheral blood, and bone callus before fracture (day 0) and on days 1, 3, 5, 14, and day 21 after. Numbers indicate the proportion of cells in the frame. (F–H) Proportions of Tregs in the spleen (F), peripheral blood (G), and bone callus (H) at different time points after fracture. Five mice were set as one group for all tissues. Data are shown as means ± SD. *P < 0.05, ***P < 0.001, ****P < 0.0001.
Fig.2  Sources of Tregs in fractured bone. (A, B) Percentages of CD4+ T cells (A) and Tregs (B) in the bone callus (n = 4). FTY720 or solvents were administrated on day 1 before fracture and days 1, 2, 3, and 4 after fracture. Bone callus was collected on day 5 after fracture with the extraction of lymphocytes. (C) Histograms depict the expression of neuropilin-1 and Helios in Tregs from the bone callus and spleen on day 5 post fracture. (D) FCM graphs of Ki67 expression in spleen and bone callus Tregs and Tconvs. Bone callus and spleen were dissected for the analysis 5 days post fracture. Numbers indicate the proportion of cells in the frame. (E) Quantitative analysis of Ki67 in the spleen and bone callus Treg and Tconv cell subsets (n = 3). (F) FCM graphs of EdU expression in spleen and bone callus Tregs and Tconvs. Numbers indicate the proportion of cells in the frame. A total of 1 mg EdU was intraperitoneally injected to the mice, and FCM analysis was performed on 24 h post EdU administration. (G) Quantitative analysis of EdU in spleen and bone callus Tregs and Tconvs (n = 3). Data are shown as means ± SD. *P < 0.05, **P < 0.01, and ***P < 0.001.
Fig.3  Tregs from bone callus promoted BMSC proliferation and osteogenesis. (A) Proliferation of BMSCs cultured with different sources of Tregs was tested via EdU assay. The higher the proportion of red dots, the more proliferative the cells. Scale bar = 200 µm. (B) ALP staining of BMSCs in different treatments after culturing with osteogenic induction medium for 14 days. Scale bar = 200 µm. (C) Quantification analysis of the percentage of EdU+ cells (n = 5). (D) Quantitative analysis of ALP-positive area calculated using ImageJ software (n = 5). Data are shown as means ± SD. *, P < 0.05.
Fig.4  Depletion of Tregs compromised fracture healing. (A) X-Ray image of fractured mice. On days 1, 3, 5, 7, 14, and 21, the mice were anesthetized using 0.3% pentobarbital (w/v) and scanned via X-ray. (B, C) Micro-CT three-dimensional reconstructed images of the fractured femur on day 21 post fracture. The mice were sacrificed, and the femur was separated for micro-CT scanning. Four mice were included in each group. (D) Calculated BMD, TV, BV, and BV/TV for each group (n = 5). The ROIs were selected as areas 1 mm above and below the fracture gap. (E) mRNA levels of osteogenesis-related proteins (collagen I, OCN, and RUNX2) in bone callus (n = 5). Mice were sacrificed on day 21 post fracture, and the bone callus was analyzed via qRT-PCT. Data are shown as means ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig.5  Bone callus Tregs displayed distinct TCR repertoire and transcriptome. (A) Left, t-SNE projection of spleen CD4+ T cells displaying the main cell clusters. The purple dots denote the Treg clusters. Each dot represents a single cell, and each color indicates a different cluster. CD4+ T cells were sorted via FCM on day 5 post fracture, with the cell number reaching 10591. Right, t-SNE projection of bone callus CD4+ T cells and Foxp3+ Tregs, with the cell number reaching 9916. (B) Identical paired CDR3 α and CDR3 β sequences in bone callus Tregs. (C) GSEA revealing the Gene Ontology enrichment of the biological process category in bone callus Tregs vs spleen Tregs. The top 20 pathways are listed and ranked by normalized enrichment score. (D) Heat map of selected differential genes between bone callus and spleen Tregs. Averaged from three independent experiments. (E) Cytofluorometric analyses of amphiregulin in bone callus and spleen Tregs (n = 4). Data are shown as means ± SD. *, P < 0.05. Areg indicated as amphiregulin in the graphs.
Fig.6  Amphiregulin promoted proliferation and osteogenesis of BMSCs. (A) After the treatment with different concentrations of amphiregulin from 0 ng/mL to 2.5 ng/mL, EdU assay was performed in 48 h. Scale bar = 200 µm. (B) Quantification analysis of the percentage of EdU-positive cells (n = 5). (C) ALP staining of BMSCs treated with different concentrations of amphiregulin from 0 ng/mL to 100 ng/mL after culturing with osteogenic induction medium for two weeks. (D) Alizarin Red staining of BMSCs treated with different concentrations of Areg from 0 ng/mL to 100 ng/mL after culturing with osteogenic induction medium for six weeks. (E) Quantitative analysis of ALP-positive area calculated using ImageJ software (n = 5). (F) Quantitative analysis of Alizarin Red-positive area calculated using ImageJ software (n = 5). (G) Protein levels of AKT and p-AKT in BMSCs treated with amphiregulin with or without the PI3K inhibitor LY294002 and AKT inhibitor deguelin at different time points (0, 30, 60, and 90 min). (H) Protein expressions of osteogenic markers RUNX2 and BMP-2 (0, 3, and 7 days). Data are shown as means ± SD. *, P < 0.05; **, P < 0.01. Areg indicated as amphiregulin in the graphs.
Fig.7  Amphiregulin reversed the compromised healing caused by Treg depletion. (A) Mouse model and administration protocol. (B) X-Rays images of fracture healing process among different groups on days 7, 14, and 21 post fracture. The mice were anesthetized via injection of sodium pentobarbital to capture X-ray images. (C) Micro-CT three-dimensional reconstructed images of fractured femur on day 21 post fracture. The mice were sacrificed, and the femur was separated for micro-CT scanning. Four mice were included in each group. (D) Calculated BMD, TV, BV, and BV/TV in each group (n = 5). The ROIs were selected as areas 1 mm above and below the fracture gap. (E–G) Serum levels of OCN (E), ALP (F), and B-ALP (G) in different groups (n = 3). (H) Representative histological evaluation of callus formation on day 21. The calluses were stained with H&E/TRAP/AB. Scale bar for HE and TRAP = 200 µm; scale bar for AB = 100 µm. Data are shown as means ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; and ****, P < 0.0001. Areg indicated as amphiregulin in the graphs.
Fig.8  Exploration of amphiregulin for human fracture healing. (A) Flow scatter diagrams of human CD4+ T (hCD4+ T) cells in peripheral blood and bone callus of fracture patients. Numbers indicate the proportion of cells in the frame. (B) Proportions of hCD4+ T cells in peripheral blood and bone callus of fracture patients (n = 15). (C) Flow scatter diagram of Tregs (hTregs) in peripheral blood and bone callus of fracture patients. Numbers indicate the proportion of cells in the frame. (D) Proportions of hTregs of fracture patients (n = 15). (E) mRNA levels of amphiregulin in blood and bone callus Tregs (n = 8). (F) EdU assay of hUC-MSCs treated with different concentrations of amphiregulin for 48 h. Scale bar = 200 µm. (G) Quantification analysis of the percentage of EdU-positive cells (n = 5). (H) ALP and Alizarin Red staining of hUC-MSCs treated with different concentrations of amphiregulin. Scale bar = 100 µm. (I,J) Quantitative analysis of ALP and Alizarin Red-positive area calculated using ImageJ software (n = 5). Data are shown as means ± SD. *, P < 0.05; ***, P < 0.001. Areg indicated as amphiregulin in the graphs.
Fig.9  Proposed mechanism of Treg-mediated fracture healing process. PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate; BGLAP, osteocalcin.
1 SZ Josefowicz, LF Lu, AY Rudensky. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012; 30(1): 531–564
https://doi.org/10.1146/annurev.immunol.25.022106.141623
2 H Lei, K Schmidt-Bleek, A Dienelt, P Reinke, HD Volk. Regulatory T cell-mediated anti-inflammatory effects promote successful tissue repair in both indirect and direct manners. Front Pharmacol 2015; 6: 184
https://doi.org/10.3389/fphar.2015.00184
3 DM Rothstein, G Camirand. New insights into the mechanisms of Treg function. Curr Opin Organ Transplant 2015; 20(4): 376–384
https://doi.org/10.1097/MOT.0000000000000212
4 CT Luo, MO Li. Transcriptional control of regulatory T cell development and function. Trends Immunol 2013; 34(11): 531–539
https://doi.org/10.1016/j.it.2013.08.003
5 G Roncador, PJ Brown, L Maestre, S Hue, JL Martínez-Torrecuadrada, KL Ling, S Pratap, C Toms, BC Fox, V Cerundolo, F Powrie, AH Banham. Analysis of FOXP3 protein expression in human CD4+CD25+ regulatory T cells at the single-cell level. Eur J Immunol 2005; 35(6): 1681–1691
https://doi.org/10.1002/eji.200526189
6 M Kanamori, H Nakatsukasa, M Okada, Q Lu, A Yoshimura. Induced regulatory T cells: their development, stability, and applications. Trends Immunol 2016; 37(11): 803–811
https://doi.org/10.1016/j.it.2016.08.012
7 D Burzyn, C Benoist, D Mathis. Regulatory T cells in nonlymphoid tissues. Nat Immunol 2013; 14(10): 1007–1013
https://doi.org/10.1038/ni.2683
8 M Feuerer, L Herrero, D Cipolletta, A Naaz, J Wong, A Nayer, J Lee, AB Goldfine, C Benoist, S Shoelson, D Mathis. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med 2009; 15(8): 930–939
https://doi.org/10.1038/nm.2002
9 D Cipolletta, M Feuerer, A Li, N Kamei, J Lee, SE Shoelson, C Benoist, D Mathis. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 2012; 486(7404): 549–553
https://doi.org/10.1038/nature11132
10 D Burzyn, W Kuswanto, D Kolodin, JL Shadrach, M Cerletti, Y Jang, E Sefik, TG Tan, AJ Wagers, C Benoist, D Mathis. A special population of regulatory T cells potentiates muscle repair. Cell 2013; 155(6): 1282–1295
https://doi.org/10.1016/j.cell.2013.10.054
11 N Arpaia, JA Green, B Moltedo, A Arvey, S Hemmers, S Yuan, PM Treuting, AY Rudensky. A distinct function of regulatory T Cells in tissue protection. Cell 2015; 162(5): 1078–1089
https://doi.org/10.1016/j.cell.2015.08.021
12 N Xia, Y Lu, M Gu, N Li, M Liu, J Jiao, Z Zhu, J Li, D Li, T Tang, B Lv, S Nie, M Zhang, M Liao, Y Liao, X Yang, X Cheng. A unique population of regulatory T cells in heart potentiates cardiac protection from myocardial infarction. Circulation 2020; 142(20): 1956–1973
https://doi.org/10.1161/CIRCULATIONAHA.120.046789
13 D Deliyanti, DM Talia, T Zhu, MJ Maxwell, A Agrotis, JR Jerome, EM Hargreaves, S Gerondakis, ML Hibbs, F Mackay, JL Wilkinson-Berka. Foxp3+ Tregs are recruited to the retina to repair pathological angiogenesis. Nat Commun 2017; 8(1): 748
https://doi.org/10.1038/s41467-017-00751-w
14 N Ali, B Zirak, RS Rodriguez, ML Pauli, HA Truong, K Lai, R Ahn, K Corbin, MM Lowe, TC Scharschmidt, K Taravati, MR Tan, RR Ricardo-Gonzalez, A Nosbaum, M Bertolini, W Liao, FO Nestle, R Paus, G Cotsarelis, AK Abbas, MD Rosenblum. Regulatory T cells in skin facilitate epithelial stem cell differentiation. Cell 2017; 169(6): 1119–1129.e11
https://doi.org/10.1016/j.cell.2017.05.002
15 GS Baht, L Vi, BA Alman. The role of the immune cells in fracture healing. Curr Osteoporos Rep 2018; 16(2): 138–145
https://doi.org/10.1007/s11914-018-0423-2
16 B Wildemann, A Ignatius, F Leung, LA Taitsman, RM Smith, R Pesántez, MJ Stoddart, RG Richards, JB Jupiter. Non-union bone fractures. Nat Rev Dis Primers 2021; 7(1): 57
https://doi.org/10.1038/s41572-021-00289-8
17 R Zura, Z Xiong, T Einhorn, JT Watson, RF Ostrum, MJ Prayson, GJ Della Rocca, S Mehta, T McKinley, Z Wang, RG Steen. Epidemiology of fracture nonunion in 18 human bones. JAMA Surg 2016; 151(11): e162775
https://doi.org/10.1001/jamasurg.2016.2775
18 JE Horton, LG Raisz, HA Simmons, JJ Oppenheim, SE Mergenhagen. Bone resorbing activity in supernatant fluid from cultured human peripheral blood leukocytes. Science 1972; 177(4051): 793–795
https://doi.org/10.1126/science.177.4051.793
19 JR Arron, Y Choi. Bone versus immune system. Nature 2000; 408(6812): 535–536
https://doi.org/10.1038/35046196
20 K Wing, Y Onishi, P Prieto-Martin, T Yamaguchi, M Miyara, Z Fehervari, T Nomura, S Sakaguchi. CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008; 322(5899): 271–275
https://doi.org/10.1126/science.1160062
21 A Bozec, MM Zaiss. T regulatory cells in bone remodelling. Curr Osteoporos Rep 2017; 15(3): 121–125
https://doi.org/10.1007/s11914-017-0356-1
22 E Cano-Gamez, B Soskic, TI Roumeliotis, E So, DJ Smyth, M Baldrighi, D Willé, N Nakic, J Esparza-Gordillo, CGC Larminie, PG Bronson, DF Tough, WC Rowan, JS Choudhary, G Trynka. Single-cell transcriptomics identifies an effectorness gradient shaping the response of CD4+ T cells to cytokines. Nat Commun 2020; 11(1): 1801
https://doi.org/10.1038/s41467-020-15543-y
23 A Butler, P Hoffman, P Smibert, E Papalexi, R Satija. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol 2018; 36(5): 411–420
https://doi.org/10.1038/nbt.4096
24 GX Zheng, JM Terry, P Belgrader, P Ryvkin, ZW Bent, R Wilson, SB Ziraldo, TD Wheeler, GP McDermott, J Zhu, MT Gregory, J Shuga, L Montesclaros, JG Underwood, DA Masquelier, SY Nishimura, M Schnall-Levin, PW Wyatt, CM Hindson, R Bharadwaj, A Wong, KD Ness, LW Beppu, HJ Deeg, C McFarland, KR Loeb, WJ Valente, NG Ericson, EA Stevens, JP Radich, TS Mikkelsen, BJ Hindson, JH Bielas. Massively parallel digital transcriptional profiling of single cells. Nat Commun 2017; 8(1): 14049
https://doi.org/10.1038/ncomms14049
25 X Fan, J Dong, S Zhong, Y Wei, Q Wu, L Yan, J Yong, L Sun, X Wang, Y Zhao, W Wang, J Yan, X Wang, J Qiao, F Tang. Spatial transcriptomic survey of human embryonic cerebral cortex by single-cell RNA-seq analysis. Cell Res 2018; 28(7): 730–745
https://doi.org/10.1038/s41422-018-0053-3
26 J Li, J Tan, MM Martino, KO Lui. Regulatory T-cells: potential regulator of tissue repair and regeneration. Front Immunol 2018; 9: 585
https://doi.org/10.3389/fimmu.2018.00585
27 RV Luckheeram, R Zhou, AD Verma, B Xia. CD4+ T cells: differentiation and functions. Clin Dev Immunol 2012; 2012: 925135
https://doi.org/10.1155/2012/925135
28 C Zhang, L Li, K Feng, D Fan, W Xue, J Lu. ‘Repair’ Treg cells in tissue injury. Cell Physiol Biochem 2017; 43(6): 2155–2169
https://doi.org/10.1159/000484295
29 A Saxena, M Dobaczewski, V Rai, Z Haque, W Chen, N Li, NG Frangogiannis. Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function. Am J Physiol Heart Circ Physiol 2014; 307(8): H1233–H1242
https://doi.org/10.1152/ajpheart.00328.2014
30 GT Kunkel, M Maceyka, S Milstien, S Spiegel. Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond. Nat Rev Drug Discov 2013; 12(9): 688–702
https://doi.org/10.1038/nrd4099
31 M Yadav, C Louvet, D Davini, JM Gardner, M Martinez-Llordella, S Bailey-Bucktrout, BA Anthony, FM Sverdrup, R Head, DJ Kuster, P Ruminski, D Weiss, D Von Schack, JA Bluestone. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med 2012; 209(10): 1713–1722, s1–19
https://doi.org/10.1084/jem.20120822
32 AM Thornton, PE Korty, DQ Tran, EA Wohlfert, PE Murray, Y Belkaid, EM Shevach. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol 2010; 184(7): 3433–3441
https://doi.org/10.4049/jimmunol.0904028
33 E Nikolouli, Y Elfaki, S Herppich, C Schelmbauer, M Delacher, C Falk, IA Mufazalov, A Waisman, M Feuerer, J Huehn. Recirculating IL-1R2+ Tregs fine-tune intrathymic Treg development under inflammatory conditions. Cell Mol Immunol 2021; 18(1): 182–193
https://doi.org/10.1038/s41423-019-0352-8
34 S Cuylen, C Blaukopf, AZ Politi, T Müller-Reichert, B Neumann, I Poser, J Ellenberg, AA Hyman, DW Gerlich. Ki-67 acts as a biological surfactant to disperse mitotic chromosomes. Nature 2016; 535(7611): 308–312
https://doi.org/10.1038/nature18610
35 C Zeng, F Pan, LA Jones, MM Lim, EA Griffin, YI Sheline, MA Mintun, DM Holtzman, RH Mach. Evaluation of 5-ethynyl-2′-deoxyuridine staining as a sensitive and reliable method for studying cell proliferation in the adult nervous system. Brain Res 2010; 1319: 21–32
https://doi.org/10.1016/j.brainres.2009.12.092
36 L Yin, D Huang, X Liu, Y Wang, J Liu, F Liu, B Yu. Omentin-1 effects on mesenchymal stem cells: proliferation, apoptosis, and angiogenesis in vitro. Stem Cell Res Ther 2017; 8(1): 224
https://doi.org/10.1186/s13287-017-0676-1
37 S Suvas, AK Azkur, BS Kim, U Kumaraguru, BT Rouse. CD4+CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J Immunol 2004; 172(7): 4123–4132
https://doi.org/10.4049/jimmunol.172.7.4123
38 C Shi, T Wu, Y He, Y Zhang, D Fu. Recent advances in bone-targeted therapy. Pharmacol Ther 2020; 207: 107473
https://doi.org/10.1016/j.pharmthera.2020.107473
39 Muris Consortium; Overall coordination; Logistical coordination; Organ collection Tabula, Library preparation processing;, Computational data analysis; Cell type annotation; Writing group; Supplemental text writing group; Principal investigators sequencing;. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 2018; 562(7727): 367–372
https://doi.org/10.1038/s41586-018-0590-4
40 J Li, Z Wang, J Wang, Q Guo, Y Fu, Z Dai, M Wang, Y Bai, X Liu, PR Cooper, J Wu, W He. Amphiregulin regulates odontogenic differentiation of dental pulp stem cells by activation of mitogen-activated protein kinase and the phosphatidylinositol 3-kinase signaling pathways. Stem Cell Res Ther 2022; 13(1): 304
https://doi.org/10.1186/s13287-022-02971-4
41 T Wang, X Zhang, DD Bikle. Osteogenic differentiation of periosteal cells during fracture healing. J Cell Physiol 2017; 232(5): 913–921
https://doi.org/10.1002/jcp.25641
42 C Yang, X Liu, K Zhao, Y Zhu, B Hu, Y Zhou, M Wang, Y Wu, C Zhang, J Xu, Y Ning, D Zou. miRNA-21 promotes osteogenesis via the PTEN/PI3K/Akt/HIF-1α pathway and enhances bone regeneration in critical size defects. Stem Cell Res Ther 2019; 10(1): 65
https://doi.org/10.1186/s13287-019-1168-2
43 T Komori. Functions of osteocalcin in bone, pancreas, testis, and muscle. Int J Mol Sci 2020; 21(20): 7513
https://doi.org/10.3390/ijms21207513
44 S Vimalraj. Alkaline phosphatase: structure, expression and its function in bone mineralization. Gene 2020; 754: 144855
https://doi.org/10.1016/j.gene.2020.144855
45 T Furuta, S Miyaki, H Ishitobi, T Ogura, Y Kato, N Kamei, K Miyado, Y Higashi, M Ochi. Mesenchymal stem cell-derived exosomes promote fracture healing in a mouse model. Stem Cells Transl Med 2016; 5(12): 1620–1630
https://doi.org/10.5966/sctm.2015-0285
46 L Claes, S Recknagel, A Ignatius. Fracture healing under healthy and inflammatory conditions. Nat Rev Rheumatol 2012; 8(3): 133–143
https://doi.org/10.1038/nrrheum.2012.1
47 P Nowak-Sliwinska, K Alitalo, E Allen, A Anisimov, AC Aplin, R Auerbach, HG Augustin, DO Bates, Beijnum JR van, RHF Bender, G Bergers, A Bikfalvi, J Bischoff, BC Böck, PC Brooks, F Bussolino, B Cakir, P Carmeliet, D Castranova, AM Cimpean, O Cleaver, G Coukos, GE Davis, Palma M De, A Dimberg, RPM Dings, V Djonov, AC Dudley, NP Dufton, SM Fendt, N Ferrara, M Fruttiger, D Fukumura, B Ghesquière, Y Gong, RJ Griffin, AL Harris, CCW Hughes, NW Hultgren, ML Iruela-Arispe, M Irving, RK Jain, R Kalluri, J Kalucka, RS Kerbel, J Kitajewski, I Klaassen, HK Kleinmann, P Koolwijk, E Kuczynski, BR Kwak, K Marien, JM Melero-Martin, LL Munn, RF Nicosia, A Noel, J Nurro, AK Olsson, TV Petrova, K Pietras, R Pili, JW Pollard, MJ Post, PHA Quax, GA Rabinovich, M Raica, AM Randi, D Ribatti, C Ruegg, RO Schlingemann, S Schulte-Merker, LEH Smith, JW Song, SA Stacker, J Stalin, AN Stratman, de Velde M Van, Hinsbergh VWM van, PB Vermeulen, J Waltenberger, BM Weinstein, H Xin, B Yetkin-Arik, S Yla-Herttuala, MC Yoder, AW Griffioen. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018; 21(3): 425–532
https://doi.org/10.1007/s10456-018-9613-x
48 NJ Protopsaltis, W Liang, E Nudleman, N Ferrara. Interleukin-22 promotes tumor angiogenesis. Angiogenesis 2019; 22(2): 311–323
https://doi.org/10.1007/s10456-018-9658-x
49 L Zhang, G Jiao, S Ren, X Zhang, C Li, W Wu, H Wang, H Liu, H Zhou, Y Chen. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res Ther 2020; 11(1): 38
https://doi.org/10.1186/s13287-020-1562-9
50 D Taylor. Fracture and repair of bone: a multiscale problem. J Mater Sci 2007; 42(21): 8911–8918
https://doi.org/10.1007/s10853-007-1698-3
51 SAT Mitchell, LA Majuta, PW Mantyh. New insights in understanding and treating bone fracture pain. Curr Osteoporos Rep 2018; 16(4): 325–332
https://doi.org/10.1007/s11914-018-0446-8
52 D Holmes. Non-union bone fracture: a quicker fix. Nature 2017; 550(7677): S193
https://doi.org/10.1038/550S193a
53 I Marzi. Focus on non-union of fractures. Eur J Trauma Emerg Surg 2019; 45(1): 1–2
https://doi.org/10.1007/s00068-019-01079-4
54 C Schlundt, CH Bucher, S Tsitsilonis, H Schell, GN Duda, K Schmidt-Bleek. Clinical and research approaches to treat non-union fracture. Curr Osteoporos Rep 2018; 16(2): 155–168
https://doi.org/10.1007/s11914-018-0432-1
55 TC Dülgeroglu, H Metineren. Evaluation of the effect of platelet-rich fibrin on long bone healing: an experimental rat model. Orthopedics 2017; 40(3): e479–e484
https://doi.org/10.3928/01477447-20170308-02
56 OW Bastian, L Koenderman, J Alblas, LP Leenen, TJ Blokhuis. Neutrophils contribute to fracture healing by synthesizing fibronectin+ extracellular matrix rapidly after injury. Clin Immunol 2016; 164: 78–84
https://doi.org/10.1016/j.clim.2016.02.001
57 M Timlin, D Toomey, C Condron, C Power, J Street, P Murray, D Bouchier-Hayes. Fracture hematoma is a potent proinflammatory mediator of neutrophil function. J Trauma 2005; 58(6): 1223–1229
https://doi.org/10.1097/01.TA.0000169866.88781.F1
58 EA Ross, A Devitt, JR Johnson. Macrophages: the good, the bad, and the gluttony. Front Immunol 2021; 12: 708186
https://doi.org/10.3389/fimmu.2021.708186
59 S Arora, K Dev, B Agarwal, P Das, MA Syed. Macrophages: their role, activation and polarization in pulmonary diseases. Immunobiology 2018; 223(4–5): 383–396
https://doi.org/10.1016/j.imbio.2017.11.001
60 I Könnecke, A Serra, Khassawna T El, C Schlundt, H Schell, A Hauser, A Ellinghaus, HD Volk, A Radbruch, GN Duda, K Schmidt-Bleek. T and B cells participate in bone repair by infiltrating the fracture callus in a two-wave fashion. Bone 2014; 64: 155–165
https://doi.org/10.1016/j.bone.2014.03.052
61 CY Luo, L Wang, C Sun, DJ Li. Estrogen enhances the functions of CD4+CD25+Foxp3+ regulatory T cells that suppress osteoclast differentiation and bone resorption in vitro. Cell Mol Immunol 2011; 8(1): 50–58
https://doi.org/10.1038/cmi.2010.54
62 H Kelchtermans, L Geboes, T Mitera, D Huskens, G Leclercq, P Matthys. Activated CD4+CD25+ regulatory T cells inhibit osteoclastogenesis and collagen-induced arthritis. Ann Rheum Dis 2009; 68(5): 744–750
https://doi.org/10.1136/ard.2007.086066
63 F Xu, C Guanghao, Y Liang, W Jun, W Wei, H Baorong. Treg-promoted new bone formation through suppressing TH17 by secreting Interleukin-10 in ankylosing spondylitis. Spine 2019; 44(23): E1349–E1355
https://doi.org/10.1097/BRS.0000000000003169
64 L Wang, DL Simons, X Lu, TY Tu, S Solomon, R Wang, A Rosario, C Avalos, D Schmolze, J Yim, J Waisman, PP Lee. Connecting blood and intratumoral Treg cell activity in predicting future relapse in breast cancer. Nat Immunol 2019; 20(9): 1220–1230
https://doi.org/10.1038/s41590-019-0429-7
65 FR D’Alessio, K Tsushima, NR Aggarwal, EE West, MH Willett, MF Britos, MR Pipeling, RG Brower, RM Tuder, JF McDyer, LS King. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J Clin Invest 2009; 119(10): 2898–2913
https://doi.org/10.1172/JCI36498
66 B Mahata, X Zhang, AA Kolodziejczyk, V Proserpio, L Haim-Vilmovsky, AE Taylor, D Hebenstreit, FA Dingler, V Moignard, B Göttgens, W Arlt, AN McKenzie, SA Teichmann. Single-cell RNA sequencing reveals T helper cells synthesizing steroids de novo to contribute to immune homeostasis. Cell Rep 2014; 7(4): 1130–1142
https://doi.org/10.1016/j.celrep.2014.04.011
67 DE Rothem, L Rothem, A Dahan, R Eliakim, M Soudry. Nicotinic modulation of gene expression in osteoblast cells, MG-63. Bone 2011; 48(4): 903–909
https://doi.org/10.1016/j.bone.2010.12.007
68 DMW Zaiss, WC Gause, LC Osborne, D Artis. Emerging functions of amphiregulin in orchestrating immunity, inflammation, and tissue repair. Immunity 2015; 42(2): 216–226
https://doi.org/10.1016/j.immuni.2015.01.020
69 K Tamama, H Kawasaki, A Wells. Epidermal growth factor (EGF) treatment on multipotential stromal cells (MSCs). Possible enhancement of therapeutic potential of MSC. J Biomed Biotechnol 2010; 2010: 795385
https://doi.org/10.1155/2010/795385
[1] FMD-23043-OF-SC_suppl_1 Download
[1] Xiaoyu Wang,Yuxuan Gao,Haigang Shi,Na Liu,Wei Zhang,Hongbo Li. Influence of the intensity and loading time of direct current electric field on the directional migration of rat bone marrow mesenchymal stem cells[J]. Front. Med., 2016, 10(3): 286-296.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed