1
|
Hali K, Gagnon S, Raleigh M, Ali I, Sniderman J, Halai M, Hall J, Schemitsch EH, Nauth A. The Effect of Cryopreservation on the Bone Healing Capacity of Endothelial Progenitor Cells in a Bone Defect Model. J Orthop Res 2025; 43:904-911. [PMID: 39888074 DOI: 10.1002/jor.26051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 02/01/2025]
Abstract
Endothelial progenitor cells (EPCs) have proven to be a highly effective cell therapy for critical-sized bone defects. Cryopreservation can enable long-term storage of EPCs, allowing their immediate availability on demand. This study compares the therapeutic potential of EPCs before and after cryopreservation in a small animal critical-sized bone defect model. Five-millimeter segmental defects were created in the right femora of Fischer 344 rats, followed by stabilization with a miniplate and screws. The animals received 2 × 106 fresh EPCs (n = 7) or 2 × 106 cryopreserved EPCs (n = 9) delivered on a gelatin scaffold. Cryopreserved EPCs were stored for 7 days at -80°C prior to thawing and loading onto the gelatin scaffold. Biweekly radiographs were taken until the animals were euthanized 10 weeks after surgery. The operated femora were then evaluated using microscopic-computed tomography (micro-CT) and biomechanical testing. All animals treated with fresh (n = 7/7) or cryopreserved (n = 9/9) EPCs achieved radiographic union at 10 weeks. Animals treated with fresh EPCs had statistically significant higher radiographic scores at 2 weeks (p < 0.05) but showed no statistically significant differences thereafter (p > 0.05). Micro-CT analysis showed no statistically significant differences between the groups in bone volume (BV) or BV normalized to total volume (p > 0.05), with excellent bone formation in both groups. Finally, there were no differences in biomechanical outcomes between the groups (p > 0.05). These results demonstrate that cryopreserved EPCs are highly effective and equivalent to fresh EPCs for healing critical-sized bone defects in a rat model of nonunion.
Collapse
Affiliation(s)
- Kalter Hali
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Stéphane Gagnon
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Matthew Raleigh
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Division of Orthopaedic Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Ikran Ali
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Jhase Sniderman
- Department of Surgery, Division of Orthopaedic Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Mansur Halai
- Department of Surgery, Division of Orthopaedic Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Jeremy Hall
- Department of Surgery, Division of Orthopaedic Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | | | - Aaron Nauth
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Division of Orthopaedic Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Godbout C, Ryan G, Ramnaraign DJ, Hegner C, Desjardins S, Gagnon S, Bates BD, Whatley I, Schemitsch EH, Nauth A. Optimal delivery of endothelial progenitor cells in a rat model of critical-size bone defects. J Orthop Res 2024; 42:193-201. [PMID: 37416978 DOI: 10.1002/jor.25658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/21/2023] [Accepted: 07/01/2023] [Indexed: 07/08/2023]
Abstract
Nonunion and segmental bone defects are complex issues in orthopedic trauma. The use of endothelial progenitor cells (EPCs), as part of a cell-based therapy for bone healing is a promising approach. In preclinical studies, culture medium (CM) is commonly used to deliver EPCs to the defect site, which has the potential for immunogenicity in humans. The goal of this study was to find an effective and clinically translatable delivery medium for EPCs. Accordingly, this study compared EPCs delivered in CM, phosphate-buffered saline (PBS), platelet-poor plasma (PPP), and platelet-rich plasma (PRP) in a rat model of femoral critical-size defects. Fischer 344 rats (n = 35) were divided into six groups: EPC+CM, EPC+PBS, EPC+PPP, EPC+PRP, PPP alone, and PRP alone. A 5 mm mid-diaphyseal defect was created in the right femur and stabilized with a miniplate. The defect was filled with a gelatin scaffold impregnated with the corresponding treatment. Radiographic, microcomputed tomography and biomechanical analyses were performed. Overall, regardless of the delivery medium, groups that received EPCs had higher radiographic scores and union rates, higher bone volume, and superior biomechanical properties compared to groups treated with PPP or PRP alone. There were no significant differences in any outcomes between EPC subgroups or between PPP and PRP alone. These results suggest that EPCs are effective in treating segmental defects in a rat model of critical-size defects regardless of the delivery medium used. Consequently, PBS could be the optimal medium for delivering EPCs, given its low cost, ease of preparation, accessibility, noninvasiveness, and nonimmunogenic properties.
Collapse
Affiliation(s)
- Charles Godbout
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Gareth Ryan
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - David J Ramnaraign
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Christian Hegner
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Sarah Desjardins
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Stéphane Gagnon
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Brent D Bates
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Ian Whatley
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| | - Emil H Schemitsch
- Department of Surgery, Division of Orthopaedic Surgery, University of Western Ontario, London, Ontario, Canada
| | - Aaron Nauth
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Division of Orthopaedic Surgery, St. Michael's Hospital-Unity Health Toronto, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Ramnaraign DJ, Godbout C, Hali K, Hegner C, Bates BD, Desjardins S, Peck J, Schemitsch EH, Nauth A. Endothelial Progenitor Cell Therapy for Fracture Healing: A Dose-Response Study in a Rat Femoral Defect Model. J Tissue Eng Regen Med 2023; 2023:8105599. [PMID: 40226398 PMCID: PMC11918885 DOI: 10.1155/2023/8105599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 04/15/2025]
Abstract
Endothelial progenitor cell (EPC) therapy has been successfully used in orthopaedic preclinical models to heal bone defects. However, no previous studies have investigated the dose-response relationship between EPC therapy and bone healing. This study aimed to assess the effect of different EPC doses on bone healing in a rat model to define an optimal dose. Five-millimeter segmental defects were created in the right femora of Fischer 344 rats, followed by stabilization with a miniplate and screws. Rats were assigned to one of six groups (control, 0.1 M, 0.5 M, 1.0 M, 2.0 M, and 4.0 M; n = 6), receiving 0, 1 × 105, 5 × 105, 1 × 106, 2 × 106, and 4 × 106 EPCs, respectively, delivered into the defect on a gelatin scaffold. Radiographs were taken every two weeks until the animals were euthanized 10 weeks after surgery. The operated femora were then evaluated using micro-computed tomography and biomechanical testing. Overall, the groups that received higher doses of EPCs (0.5 M, 1.0 M, 2.0 M, and 4.0 M) reached better outcomes. At 10 weeks, full radiographic union was observed in 67% of animals in the 0.5 M group, 83% of animals in the 1.0 M group, and 100% of the animals in the 2.0 M and 4.0 M groups, but none in the control and 0.1 M groups. The 2.0 M group also displayed the strongest biomechanical properties, which significantly improved relative to the control and 0.1 M groups. In summary, this study defined a dose-response relationship between EPC therapy and bone healing, with 2 × 106 EPCs being the optimal dose in this model. Our findings emphasize the importance of dosing considerations in the application of cell therapies aimed at tissue regeneration and will help guide future investigations and clinical translation of EPC therapy.
Collapse
Affiliation(s)
- David J. Ramnaraign
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Charles Godbout
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Kalter Hali
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Christian Hegner
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Brent D. Bates
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Sarah Desjardins
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Peck
- Division of Orthopaedic Surgery, Department of Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| | | | - Aaron Nauth
- Keenan Research Centre for Biomedical Science, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
- Division of Orthopaedic Surgery, Department of Surgery, Unity Health Toronto (St. Michael's Hospital), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Khosravipour A, Mostafavinia A, Amini A, Gazor R, Zare F, Fallahnezhad S, Rezaei F, Asgari M, Mohammadian F, Mohsenifar Z, Chien S, Bayat M. Different Protocols of Combined Application of Photobiomodulation In Vitro and In Vivo Plus Adipose-Derived Stem Cells Improve the Healing of Bones in Critical Size Defects in Rat Models. J Lasers Med Sci 2022; 13:e10. [PMID: 35996492 PMCID: PMC9392890 DOI: 10.34172/jlms.2022.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/12/2021] [Indexed: 10/05/2023]
Abstract
Introduction: Long bone segmental deficiencies are challenging complications to treat. Hereby, the effects of the scaffold derived from the human demineralized bone matrix (hDBMS) plus human adipose stem cells (hADSs) plus photobiomodulation (PBM) (in vitro and or in vivo) on the catabolic step of femoral bone repair in rats with critical size femoral defects (CDFDs) were evaluated with stereology and high stress load (HSL) assessment methods. Methods: hADSs were exposed to PBM in vitro; then, the mixed influences of hDBMS+hADS+PBM on CSFDs were evaluated. CSFDs were made on both femurs; then hDBMSs were engrafted into both CSFDs of all rats. There were 6 groups (G)s: G1 was the control; in G2 (hADS), hADSs only were engrafted into hDBMS of CSFD; in G3 (PBM) only PBM therapy for CSFD was provided; in G4 (hADS+PBM in vivo), seeded hADSs on hDBMS of CSFDs were radiated with a laser in vivo; in G5 (hADSs+PBM under in vitro condition), hADSs in a culture system were radiated with a laser, then transferred on hDBMS of CSFDs; and in G6 (hADS+PBM in conditions of in vivo and in vitro), laser-exposed hADSs were transplanted on hDBMS of CSFDs, and then CSFDs were exposed to a laser in vivo. Results: Groups 4, 5, and 6 meaningfully improved HSLs of CSFD in comparison with groups 3, 1, and 2 (all, P=0.001). HSL of G5 was significantly more than G4 and G6 (both, P=0.000). Gs 6 and 4 significantly increased new bone volumes of CSFD compared to Gs 2 (all, P=0.000) and 1 (P=0.001 & P=0.003 respectively). HSL of G 1 was significantly lower than G5 (P=0.026). Conclusion: HSLs of CSFD in rats that received treatments of hDBMS plus hADS plus PBM were significantly higher than treatments with hADS and PBM alone and control groups.
Collapse
Affiliation(s)
- Armin Khosravipour
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atarodalsadat Mostafavinia
- Department of Anatomy, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Abdollah Amini
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rouhallah Gazor
- Department of Anatomy, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Zare
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somaye Fallahnezhad
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemehalsadat Rezaei
- University of Kentucky, College of Pharmacy, 789 South Limestone, Lexington, Kentucky 40536, USA
| | - Mehrdad Asgari
- Department of Anatomy, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Mohammadian
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhaleh Mohsenifar
- Department of Pathology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC, Louisville, Kentucky, USA
| | - Mohammad Bayat
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC, Louisville, Kentucky, USA
| |
Collapse
|
5
|
Zhao H, Shen S, Zhao L, Xu Y, Li Y, Zhuo N. 3D printing of dual-cell delivery titanium alloy scaffolds for improving osseointegration through enhancing angiogenesis and osteogenesis. BMC Musculoskelet Disord 2021; 22:734. [PMID: 34452607 PMCID: PMC8401189 DOI: 10.1186/s12891-021-04617-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/11/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The repair of large bone defects is a great challenge for orthopedics. Although the development of three-dimensional (3D) printed titanium alloy (Ti6Al4V) implants with optimized the pore structure have effectively promoted the osseointegration. However, due to the biological inertia of Ti6Al4Vsurface and the neglect of angiogenesis, some patients still suffer from postoperative complications such as dislocation or loosening of the prosthesis. METHODS The purpose of this study was to construct 3D printed porous Ti6Al4V scaffolds filled with bone marrow mesenchymal stem cells (BMSC) and endothelial progenitor cells (EPC) loaded hydrogel and evaluate the efficacy of this composite implants on osteogenesis and angiogenesis, thus promoting osseointegration. RESULTS The porosity and pore size of prepared 3D printed porous Ti6Al4V scaffolds were 69.2 ± 0.9 % and 593.4 ± 16.9 μm, respectively, which parameters were beneficial to bone ingrowth and blood vessel formation. The BMSC and EPC filled into the pores of the scaffolds after being encapsulated by hydrogels can maintain high viability. As a cell containing composite implant, BMSC and EPC loaded hydrogel incorporated into 3D printed porous Ti6Al4V scaffolds enhancing osteogenesis and angiogenesis to repair bone defects efficiently. At the transcriptional level, the composite implant up-regulated the expression levels of the osteogenesis-related genes alkaline phosphatase (ALP) and osteocalcin (OCN), and angiogenesis-related genes hypoxia-inducible factor 1 alpha (HIF-1α), and vascular endothelial growth factor (VEGF). CONCLUSIONS Overall, the strategy of loading porous Ti6Al4V scaffolds to incorporate cells is a promising treatment for improving osseointegration.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Shi Shen
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Lu Zhao
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Yulin Xu
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Yang Li
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Naiqiang Zhuo
- Department of Department of Bone and Joint, Affiliated Hospital of Southwest Medical University, 646000, Luzhou, People's Republic of China.
| |
Collapse
|
6
|
Liang W, Chen X, Dong Y, Zhou P, Xu F. Recent advances in biomaterials as instructive scaffolds for stem cells in tissue repair and regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1848832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People’s Hospital, Shaoxing, P. R. China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| |
Collapse
|
7
|
Menger MM, Laschke MW, Orth M, Pohlemann T, Menger MD, Histing T. Vascularization Strategies in the Prevention of Nonunion Formation. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:107-132. [PMID: 32635857 DOI: 10.1089/ten.teb.2020.0111] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Delayed healing and nonunion formation are major challenges in orthopedic surgery, which require the development of novel treatment strategies. Vascularization is considered one of the major prerequisites for successful bone healing, providing an adequate nutrient supply and allowing the infiltration of progenitor cells to the fracture site. Hence, during the last decade, a considerable number of studies have focused on the evaluation of vascularization strategies to prevent or to treat nonunion formation. These involve (1) biophysical applications, (2) systemic pharmacological interventions, and (3) tissue engineering, including sophisticated scaffold materials, local growth factor delivery systems, cell-based techniques, and surgical vascularization approaches. Accumulating evidence indicates that in nonunions, these strategies are indeed capable of improving the process of bone healing. The major challenge for the future will now be the translation of these strategies into clinical practice to make them accessible for the majority of patients. If this succeeds, these vascularization strategies may markedly reduce the incidence of nonunion formation. Impact statement Delayed healing and nonunion formation are a major clinical problem in orthopedic surgery. This review provides an overview of vascularization strategies for the prevention and treatment of nonunions. The successful translation of these strategies in clinical practice is of major importance to achieve adequate bone healing.
Collapse
Affiliation(s)
- Maximilian M Menger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Marcel Orth
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Tim Pohlemann
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Tina Histing
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany
| |
Collapse
|
8
|
Wang N, Liu X, Shi L, Liu Y, Guo S, Liu W, Li X, Meng J, Ma X, Guo Z. Identification of a prolonged action molecular GLP-1R agonist for the treatment of femoral defects. Biomater Sci 2020; 8:1604-1614. [PMID: 31967113 DOI: 10.1039/c9bm01426h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Poly-GLP-1 promotes angiogenesis to accelerate bone formationviaBMSC differentiation and M2 polarization.
Collapse
|
9
|
He J, Han X, Wang S, Zhang Y, Dai X, Liu B, Liu L, Zhao X. Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro. Exp Ther Med 2019; 18:3333-3340. [PMID: 31602206 PMCID: PMC6777308 DOI: 10.3892/etm.2019.7982] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 05/02/2019] [Indexed: 12/20/2022] Open
Abstract
Bone tissue engineering provides a substitute for bone transplantation to address various bone defects. However, bone regeneration involves a large number of cellular events. In addition, obtaining sufficient source material for autogenous bone or alloplastic bone substitutes remains an unsolved issue. In previous studies, it was confirmed that bone marrow stromal cells (BMSCs) and endothelial progenitor cells (EPCs) had the capacity to promote bone regeneration. Additionally, bone morphogenetic protein-2 (BMP-2) has been demonstrated to be an active inducer of osteoblast differentiation. Therefore, the aim of the present study was to produce an effective integration system, including a scaffold, reparative cells and growth factors, that may enhance bone regeneration. Firstly, bone marrow-derived BMSCs and EPCs were isolated and identified by flow cytometry. Cell proliferation ability, secreted BMP-2 levels and alkaline phosphatase (ALP) activity were highest in the cell sheets containing BMP-2-modified BMSCs and EPCs. In addition, the expression levels of osteogenesis-associated genes, including runt related transcription factor 2 (Runx2), distal-less homeobox 5 (Dlx5), ALP and integrin-binding sialoprotein (Ibsp), and osteogenesis-associated proteins, including Runx2, Dlx, ALP, Ibsp, vascular endothelial growth factor, osteonectin, osteopontin and type I collagen, gradually increased during the co-culture of ad-BMP-2-BMSCs/EPCs. The levels of these genes and proteins were increased compared with those observed in the BMSC, EPC and BMP-2-modified BMSC groups. Finally, scanning electron microscopy observation also demonstrated that the BMP2-modified BMSCs were able to combine well with EPCs to construct a cell sheet for bone formation. Collectively, these results describe an adenovirus (ad)-BMP2-BMSCs/EPCs co-culture system that may significantly accelerate bone regeneration compared with a BMSCs/EPCs co-culture system or ad-BMP2-BMSCs alone.
Collapse
Affiliation(s)
- Jia He
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xuesong Han
- Department of Obstetrics and Gynecology, Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Songmei Wang
- School of Public Health, Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Ying Zhang
- Department of Cardiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Xiaoming Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Boyan Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Liu Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xian Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
10
|
Wu J, Liu S, Wang Z, Ma S, Meng H, Hu J. Calcitonin gene-related peptide promotes proliferation and inhibits apoptosis in endothelial progenitor cells via inhibiting MAPK signaling. Proteome Sci 2018; 16:18. [PMID: 30473635 PMCID: PMC6236989 DOI: 10.1186/s12953-018-0146-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/23/2018] [Indexed: 11/20/2022] Open
Abstract
Background Calcitonin gene-related peptide (CGRP) contributes to bone formation by stimulating bone marrow stromal cell (BMSC) proliferation and differentiation. However, the proliferative and apoptotic effects of CGRP on bone marrow-derived endothelial progenitor cells (EPCs) have not been investigated. Methods We tested the effects of CGRP on EPC proliferation and apoptosis by Cell Counting Kit-8, flow cytometry, and studied the effects of CGRP on the expression of proliferation- and apoptosis-related markers in EPCs and the underlying mitogen-activated protein kinase (MAPK) signalling pathway by quantitative polymerase chain reaction and western blotting. Results We detected EPC markers (CD34, CD133 and VEGFR-2) in 7-day cultures and found that CGRP (10− 10–10− 12 M) promoted the proliferation of cultured EPCs, with a peak increase of 30% at 10− 10 M CGRP. CGRP also upregulated the expression of proliferation-associated genes, including cyclin D1 and cyclin E, and increased the percentages of G2/M-phase and S-phase cells after incubation 72 h. CGRP inhibited serum deprivation (SD)-induced apoptosis in EPCs after 24 and 48 h and downregulated the expression of apoptosis-related genes, including caspase-3, caspase-8, caspase-9 and Bax. Phosphorylated (p-)ERK1/2, p-p38 and p-JNK protein levels in EPCs treated with CGRP were significantly lower than those in untreated EPCs. Pre-treatment with the calcitonin receptor-like receptor (CRLR) antagonist CGRP8–37 or a MAPK pathway inhibitor (PD98059, SB203580 or SP600125) completely or partially reversed the pro-proliferative and anti-apoptotic effects and the reduced p-ERK1/2, p-p38 and p-JNK expression induced by CGRP. Conclusion Our results show that CGRP exerts pro-proliferative and anti-apoptotic effects on EPCs and may act by inhibiting MAPK pathways. Electronic supplementary material The online version of this article (10.1186/s12953-018-0146-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianqun Wu
- Department of Spine Surgery, Huadu District People's Hospital, Guangzhou, Guangzhou, 510800 Guangdong Province China
| | - Song Liu
- 2Department of Orthopedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou City, 510150 Guangdong Province China.,3Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou City, 510515 Guangdong Province China
| | - Zhao Wang
- 2Department of Orthopedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou City, 510150 Guangdong Province China
| | - Shenghui Ma
- 3Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou City, 510515 Guangdong Province China
| | - Huan Meng
- 3Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou City, 510515 Guangdong Province China
| | - Jijie Hu
- 3Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou City, 510515 Guangdong Province China
| |
Collapse
|
11
|
Fiset S, Godbout C, Crookshank MC, Zdero R, Nauth A, Schemitsch EH. Experimental Validation of the Radiographic Union Score for Tibial Fractures (RUST) Using Micro-Computed Tomography Scanning and Biomechanical Testing in an in-Vivo Rat Model. J Bone Joint Surg Am 2018; 100:1871-1878. [PMID: 30399082 DOI: 10.2106/jbjs.18.00035] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The Radiographic Union Score for Tibial fractures (RUST) and the modified version of the system, mRUST, are popular standards for assessing fracture-healing progress with use of radiographs. To our knowledge, this is the first study to experimentally validate the ability of RUST and mRUST to accurately assess bone-healing progression with use of both micro-computed tomography (micro-CT) scanning and biomechanical testing. METHODS Adult male rats (n = 29) underwent osteotomy with a midshaft fracture gap repaired with use of a polyetheretherketone plate. Anteroposterior and lateral radiographs were made of the repaired femora prior to rat death at end points of 5, 6, 7, 8, 9, and 17 weeks, and 2 fellowship-trained orthopaedic trauma surgeons independently assigned RUST and mRUST scores to repaired femora. The repaired and intact contralateral femora were then dissected. Bones underwent dissection, micro-CT scanning, and biomechanical torsion testing at the end points. RESULTS RUST scores ranged from 5 to 12 and mRUST scores ranged from 5 to 16. Intraclass correlation coefficients (ICCs) were 0.89 (95% confidence interval [CI]: 0.78 to 0.94) for RUST and 0.86 (95% CI: 0.74 to 0.93) for mRUST, which fall within the "almost perfect agreement" category for ICCs. Spearman rank correlation coefficients (RS) showed correlation of RUST (RS range, 0.456 to 0.818) and mRUST (RS range, 0.519 to 0.862) with micro-CT measurements of mineralized callus volume (BV), total callus volume (TV), and BV/TV ratio, but less so with bone mineral density (BMD). Additionally, RUST (RS range, 0.524 to 0.863) and mRUST (RS range, 0.434 to 0.850) were correlated with some biomechanical properties. A RUST score of 10 or an mRUST score of 15 may be considered the threshold above which a plated bone is "healed" because, at these scores, 120% or 140% of failure torque, respectively, was achieved by the repaired femora as compared with the intact contralateral femora. CONCLUSIONS RUST and mRUST both show strong statistical correlations with micro-CT and biomechanical parameters. CLINICAL RELEVANCE RUST and mRUST scoring systems provide clinicians with validated, reliable, and available tools to assess the progress of fracture-healing.
Collapse
Affiliation(s)
| | | | | | - Radovan Zdero
- London Health Sciences Centre, London, Ontario, Canada.,Department of Surgery, Western University, London, Ontario, Canada.,Department of Mechanical and Materials Engineering, Western University, London, Ontario, Canada
| | - Aaron Nauth
- University of Toronto, Toronto, Ontario, Canada.,St. Michael's Hospital, Toronto, Ontario, Canada
| | - Emil H Schemitsch
- University of Toronto, Toronto, Ontario, Canada.,St. Michael's Hospital, Toronto, Ontario, Canada.,London Health Sciences Centre, London, Ontario, Canada.,Department of Surgery, Western University, London, Ontario, Canada
| |
Collapse
|
12
|
Pirosa A, Gottardi R, Alexander PG, Tuan RS. Engineering in-vitro stem cell-based vascularized bone models for drug screening and predictive toxicology. Stem Cell Res Ther 2018; 9:112. [PMID: 29678192 PMCID: PMC5910611 DOI: 10.1186/s13287-018-0847-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The production of veritable in-vitro models of bone tissue is essential to understand the biology of bone and its surrounding environment, to analyze the pathogenesis of bone diseases (e.g., osteoporosis, osteoarthritis, osteomyelitis, etc.), to develop effective therapeutic drug screening, and to test potential therapeutic strategies. Dysregulated interactions between vasculature and bone cells are often related to the aforementioned pathologies, underscoring the need for a bone model that contains engineered vasculature. Due to ethical restraints and limited prediction power of animal models, human stem cell-based tissue engineering has gained increasing relevance as a candidate approach to overcome the limitations of animals and to serve as preclinical models for drug testing. Since bone is a highly vascularized tissue, the concomitant development of vasculature and mineralized matrix requires a synergistic interaction between osteogenic and endothelial precursors. A number of experimental approaches have been used to achieve this goal, such as the combination of angiogenic factors and three-dimensional scaffolds, prevascularization strategies, and coculture systems. In this review, we present an overview of the current models and approaches to generate in-vitro stem cell-based vascularized bone, with emphasis on the main challenges of vasculature engineering. These challenges are related to the choice of biomaterials, scaffold fabrication techniques, and cells, as well as the type of culturing conditions required, and specifically the application of dynamic culture systems using bioreactors.
Collapse
Affiliation(s)
- Alessandro Pirosa
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
- Ri.MED Foundation, Via Bandiera 11, Palermo, 90133 Italy
| | - Peter G. Alexander
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219 USA
| |
Collapse
|
13
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
14
|
Zigdon-Giladi H, Elimelech R, Michaeli-Geller G, Rudich U, Machtei EE. Safety profile and long-term engraftment of human CD31 + blood progenitors in bone tissue engineering. Cytotherapy 2017; 19:895-908. [PMID: 28495397 DOI: 10.1016/j.jcyt.2017.03.079] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) participate in angiogenesis and induce favorable micro-environments for tissue regeneration. The efficacy of EPCs in regenerative medicine is extensively studied; however, their safety profile remains unknown. Therefore, our aims were to evaluate the safety profile of human peripheral blood-derived EPCs (hEPCs) and to assess the long-term efficacy of hEPCs in bone tissue engineering. METHODS hEPCs were isolated from peripheral blood, cultured and characterized. β tricalcium phosphate scaffold (βTCP, control) or 106 hEPCs loaded onto βTCP were transplanted in a nude rat calvaria model. New bone formation and blood vessel density were analyzed using histomorphometry and micro-computed tomography (CT). Safety of hEPCs using karyotype analysis, tumorigenecity and biodistribution to target organs was evaluated. RESULTS On the cellular level, hEPCs retained their karyotype during cell expansion (seven passages). Five months following local hEPC transplantation, on the tissue and organ level, no inflammatory reaction or dysplastic change was evident at the transplanted site or in distant organs. Direct engraftment was evident as CD31 human antigens were detected lining vessel walls in the transplanted site. In distant organs human antigens were absent, negating biodistribution. Bone area fraction and bone height were doubled by hEPC transplantation without affecting mineral density and bone architecture. Additionally, local transplantation of hEPCs increased blood vessel density by nine-fold. CONCLUSIONS Local transplantation of hEPCs showed a positive safety profile. Furthermore, enhanced angiogenesis and osteogenesis without mineral density change was found. These results bring us one step closer to first-in-human trials using hEPCs for bone regeneration.
Collapse
Affiliation(s)
- Hadar Zigdon-Giladi
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Rina Elimelech
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Gal Michaeli-Geller
- Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel
| | - Utai Rudich
- Orthopedic Department, Rambam Health Care Campus, Haifa, Israel
| | - Eli E Machtei
- Department of Periodontology, School of Graduate Dentistry, Rambam Health Care Campus, Haifa, Israel; Research Institute for Bone Repair, Rambam Health Care Campus, Haifa, Israel; The Rappaport Family Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
Delayed Endothelial Progenitor Cell Therapy Promotes Bone Defect Repair in a Clinically Relevant Rat Model. Stem Cells Int 2017; 2017:7923826. [PMID: 28523072 PMCID: PMC5390645 DOI: 10.1155/2017/7923826] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 02/14/2017] [Indexed: 11/18/2022] Open
Abstract
The repair of segmental bone defects remains a significant challenge for orthopaedic surgeons. Endothelial progenitor cells (EPCs) have successfully promoted the repair of acute defects in animal models; however, the ability of EPCs to induce the repair of chronic nonhealing defects, such as those often encountered clinically, has not been investigated. Therefore, the purpose of this study was to investigate the ability of EPCs delivered in delayed fashion to induce the repair of nonhealing defects in a clinically relevant model. In order to simulate delayed treatment, 5 mm segmental defects in Fischer 344 rat femora were treated with bone marrow-derived EPCs on a Gelfoam scaffold at 3 weeks post creation of the defect. At ten weeks posttreatment, 100% of EPC-treated defects achieved union, whereas complete union was only achieved in 37.5% of defects treated with Gelfoam alone. Furthermore, significant increases in ultimate torque (p = 0.022) and torsional stiffness (p = 0.003) were found in EPC-treated defects compared to controls. Critically, no differences in outcomes were observed between acute and delayed EPC treatments. These results suggest that EPCs can enhance bone healing when applied in an acute or delayed fashion and that their use may represent a clinically translatable therapy for bone healing in humans.
Collapse
|
16
|
The Use of Endothelial Progenitor Cells for the Regeneration of Musculoskeletal and Neural Tissues. Stem Cells Int 2017; 2017:1960804. [PMID: 28458693 PMCID: PMC5387841 DOI: 10.1155/2017/1960804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 03/12/2017] [Indexed: 12/18/2022] Open
Abstract
Endothelial progenitor cells (EPCs) derived from bone marrow and blood can differentiate into endothelial cells and promote neovascularization. In addition, EPCs are a promising cell source for the repair of various types of vascularized tissues and have been used in animal experiments and clinical trials for tissue repair. In this review, we focused on the kinetics of endogenous EPCs during tissue repair and the application of EPCs or stem cell populations containing EPCs for tissue regeneration in musculoskeletal and neural tissues including the bone, skeletal muscle, ligaments, spinal cord, and peripheral nerves. EPCs can be mobilized from bone marrow and recruited to injured tissue to contribute to neovascularization and tissue repair. In addition, EPCs or stem cell populations containing EPCs promote neovascularization and tissue repair through their differentiation to endothelial cells or tissue-specific cells, the upregulation of growth factors, and the induction and activation of endogenous stem cells. Human peripheral blood CD34(+) cells containing EPCs have been used in clinical trials of bone repair. Thus, EPCs are a promising cell source for the treatment of musculoskeletal and neural tissue injury.
Collapse
|
17
|
Zhou J, Rogers JH, Lee SH, Sun D, Yao H, Mao JJ, Kong KY. Oral Mucosa Harbors a High Frequency of Endothelial Cells: A Novel Postnatal Cell Source for Angiogenic Regeneration. Stem Cells Dev 2016; 26:91-101. [PMID: 27832737 DOI: 10.1089/scd.2016.0175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Endothelial progenitor cells/endothelial cells (EPCs/ECs) have great potential to treat pathological conditions such as cardiac infarction, muscle ischemia, and bone fractures, but isolation of EPC/ECs from existing cell sources is challenging due to their low EC frequency. We have isolated endothelial progenitor (EP)-like cells from rat oral mucosa and characterized their yield, immunophenotype, growth, and in vivo angiogenic potential. The frequency of EP-like cells derived from oral mucosa is thousands of folds higher than EPCs derived from donor-match bone marrow samples. EP-like cells from oral mucosa were positive for EC markers CD31, VE-Cadherin, and VEGFR2. Oral mucosa-derived EP-like cells displayed robust uptake of acetylated low-density lipoprotein and formed stable capillary networks in Matrigel. Subcutaneously implanted oral mucosa-derived EP-like cells anastomosed with host blood vessels, implicating their ability to elicit angiogenesis. Similar to endothelial colony-forming cells, EP-like cells from oral mucosa have a significantly higher proliferative rate than human umbilical vein endothelial cells. These findings identify a putative EPC source that is easily accessible in the oral cavity, potentially from discarded tissue specimens, and yet with robust yield and potency for angiogenesis in tissue and organ regeneration.
Collapse
Affiliation(s)
- Jian Zhou
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York.,2 Department of General Dentistry, Capital Medical University School of Stomatology , Beijing, China
| | - Jason H Rogers
- 3 Department of Internal Medicine and the Cancer Research and Treatment Center, University of New Mexico Health Science Center , Albuquerque, New Mexico
| | - Scott H Lee
- 4 Pratt School of Engineering, Duke University , Durham, North Carolina
| | - DongMing Sun
- 5 W. M. Keck Center for Collaborative Neuroscience, Rutgers University , New Brunswick, New Jersey
| | - Hai Yao
- 6 Clemson-MUSC Bioengineering Program , Department of Craniofacial Biology, Charleston, South Carolina
| | - Jeremy J Mao
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York
| | - Kimi Y Kong
- 1 Center for Craniofacial Regeneration, Columbia University Medical Center , New York, New York.,7 Hematology/Oncology Division, Department of Medicine, University of Florida , Gainesville, Florida
| |
Collapse
|
18
|
Abstract
Unlike many other postnatal tissues, bone can regenerate and repair itself; nevertheless, this capacity can be overcome. Traditionally, surgical reconstructive strategies have implemented autologous, allogeneic, and prosthetic materials. Autologous bone--the best option--is limited in supply and also mandates an additional surgical procedure. In regenerative tissue engineering, there are myriad issues to consider in the creation of a functional, implantable replacement tissue. Importantly, there must exist an easily accessible, abundant cell source with the capacity to express the phenotype of the desired tissue, and a biocompatible scaffold to deliver the cells to the damaged region. A literature review was performed using PubMed; peer-reviewed publications were screened for relevance in order to identify key advances in stem and progenitor cell contribution to the field of bone tissue engineering. In this review, we briefly introduce various adult stem cells implemented in bone tissue engineering such as mesenchymal stem cells (including bone marrow- and adipose-derived stem cells), endothelial progenitor cells, and induced pluripotent stem cells. We then discuss numerous advances associated with their application and subsequently focus on technological advances in the field, before addressing key regenerative strategies currently used in clinical practice. Stem and progenitor cell implementation in bone tissue engineering strategies have the ability to make a major impact on regenerative medicine and reduce patient morbidity. As the field of regenerative medicine endeavors to harness the body's own cells for treatment, scientific innovation has led to great advances in stem cell-based therapies in the past decade.
Collapse
|
19
|
Almubarak S, Nethercott H, Freeberg M, Beaudon C, Jha A, Jackson W, Marcucio R, Miclau T, Healy K, Bahney C. Tissue engineering strategies for promoting vascularized bone regeneration. Bone 2016; 83:197-209. [PMID: 26608518 PMCID: PMC4911893 DOI: 10.1016/j.bone.2015.11.011] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/06/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
This review focuses on current tissue engineering strategies for promoting vascularized bone regeneration. We review the role of angiogenic growth factors in promoting vascularized bone regeneration and discuss the different therapeutic strategies for controlled/sustained growth factor delivery. Next, we address the therapeutic uses of stem cells in vascularized bone regeneration. Specifically, this review addresses the concept of co-culture using osteogenic and vasculogenic stem cells, and how adipose derived stem cells compare to bone marrow derived mesenchymal stem cells in the promotion of angiogenesis. We conclude this review with a discussion of a novel approach to bone regeneration through a cartilage intermediate, and discuss why it has the potential to be more effective than traditional bone grafting methods.
Collapse
Affiliation(s)
- Sarah Almubarak
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Hubert Nethercott
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Marie Freeberg
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Caroline Beaudon
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; UCSF-UCB Masters of Translational Medicine Program, Berkeley and San Francisco, CA, United States
| | - Amit Jha
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Wesley Jackson
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Ralph Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin Healy
- Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States
| | - Chelsea Bahney
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco, San Francisco, CA, United States; Departments of Bioengineering, and Material Science and Engineering, University of California, Berkeley (UCB), Berkeley, CA, United States.
| |
Collapse
|
20
|
Kolind M, Bobyn JD, Matthews BG, Mikulec K, Aiken A, Little DG, Kalajzic I, Schindeler A. Lineage tracking of mesenchymal and endothelial progenitors in BMP-induced bone formation. Bone 2015; 81:53-59. [PMID: 26141839 PMCID: PMC4844190 DOI: 10.1016/j.bone.2015.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/26/2015] [Accepted: 06/29/2015] [Indexed: 11/29/2022]
Abstract
To better understand the relative contributions of mesenchymal and endothelial progenitor cells to rhBMP-2 induced bone formation, we examined the distribution of lineage-labeled cells in Tie2-Cre:Ai9 and αSMA-creERT2:Col2.3-GFP:Ai9 reporter mice. Established orthopedic models of ectopic bone formation in the hind limb and spine fusion were employed. Tie2-lineage cells were found extensively in the ectopic bone and spine fusion masses, but co-staining was only seen with tartrate-resistant acid phosphatase (TRAP) activity (osteoclasts) and CD31 immunohistochemistry (vascular endothelial cells), and not alkaline phosphatase (AP) activity (osteoblasts). To further confirm the lack of a functional contribution of Tie2-lineage cells to BMP-induced bone, we developed conditional knockout mice where Tie2-lineage cells are rendered null for key bone transcription factor osterix (Tie2-cre:Osx(fx/fx) mice). Conditional knockout mice showed no difference in BMP-induced bone formation compared to littermate controls. Pulse labeling of mesenchymal cells with Tamoxifen in mice undergoing spine fusion revealed that αSMA-lineage cells contributed to the osteoblastic lineage (Col2.3-GFP), but not to endothelial cells or osteoclast populations. These data indicate that the αSMA+ and Tie2+ progenitor lineages make distinct cellular contributions to bone formation, angiogenesis, and resorption/remodeling.
Collapse
Affiliation(s)
- Mille Kolind
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Justin D Bobyn
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Brya G Matthews
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Kathy Mikulec
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Alastair Aiken
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - David G Little
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmington, CT, USA
| | - Aaron Schindeler
- Centre for Children's Bone Health, The Children's Hospital at Westmead, Westmead, NSW, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
21
|
Zigdon-Giladi H, Rudich U, Michaeli Geller G, Evron A. Recent advances in bone regeneration using adult stem cells. World J Stem Cells 2015; 7:630-640. [PMID: 25914769 PMCID: PMC4404397 DOI: 10.4252/wjsc.v7.i3.630] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/30/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023] Open
Abstract
Bone is a highly vascularized tissue reliant on the close spatial and temporal association between blood vessels and bone cells. Therefore, cells that participate in vasculogenesis and osteogenesis play a pivotal role in bone formation during prenatal and postnatal periods. Nevertheless, spontaneous healing of bone fracture is occasionally impaired due to insufficient blood and cellular supply to the site of injury. In these cases, bone regeneration process is interrupted, which might result in delayed union or even nonunion of the fracture. Nonunion fracture is difficult to treat and have a high financial impact. In the last decade, numerous technological advancements in bone tissue engineering and cell-therapy opened new horizon in the field of bone regeneration. This review starts with presentation of the biological processes involved in bone development, bone remodeling, fracture healing process and the microenvironment at bone healing sites. Then, we discuss the rationale for using adult stem cells and listed the characteristics of the available cells for bone regeneration. The mechanism of action and epigenetic regulations for osteogenic differentiation are also described. Finally, we review the literature for translational and clinical trials that investigated the use of adult stem cells (mesenchymal stem cells, endothelial progenitor cells and CD34+ blood progenitors) for bone regeneration.
Collapse
|
22
|
Piatkowski A, Grieb G, Simons D, Bernhagen J, van der Hulst RR. Endothelial progenitor cells--potential new avenues to improve neoangiogenesis and reendothelialization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 306:43-81. [PMID: 24016523 DOI: 10.1016/b978-0-12-407694-5.00002-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term endothelial progenitor cell (EPC) was established more than 10 years ago and is used to refer to a group of circulating cells that display endothelial lineage qualities and are able to home to areas of ischemia or vascular injury and to facilitate the repair of damaged blood vessels or develop new vessels as needed. This chapter reviews the current lineage relationships among all the cells called EPC and will clear the terminology used in EPC research. Furthermore, an overview of the clinical and in vitro research, as well as cytokine and drug interactions and potential EPC applications, is given.
Collapse
Affiliation(s)
- Andrzej Piatkowski
- Department of Plastic Surgery, academisch ziekenhuis Maastricht, MUMC+, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
23
|
Abstract
PURPOSE Endothelial progenitor cells (EPCs) represent a population of novel precursor cells with known ability to participate in angiogenesis. Our previous studies have shown that local EPC therapy significantly increased angiogenesis and osteogenesis to promote fracture healing in an animal bone defect model. However, the cellular and molecular mechanisms by which EPC therapy promotes fracture healing remain largely unknown. The purpose of this study was to quantify local bone morphogenetic protein (BMP-2) expression after EPC therapy for a rat segmental bone defect, in hopes of further defining the potential mechanisms by which EPCs promote fracture healing. METHOD EPCs were isolated from the bone marrow of syngeneic rats and cultured ex vivo for 7-10 days before transfer to the bone defect. A total of 56 rats were studied. The treatment group received 1 × 10 EPCs on a gelfoam scaffold at the bone defect, and control animals received gelfoam/saline only. Before euthanasia, radiographs of the femur were performed. Animals were euthanized at 1, 2, 3, and 10 weeks, and specimens from the fracture gap area were collected, pulverized, and total messenger RNA (mRNA) was extracted. BMP-2 mRNA was measured by reverse transcriptase-polymerase chain reaction and quantified by VisionWorksLS. All measurements were performed in triplicate. RESULTS All EPC-treated bone defects healed radiographically by 10 weeks, whereas control-treated defects developed a nonunion. The expression of BMP-2 mRNA was significantly elevated in EPC-treated defects relative to controls at week 1 (EPC, 0.59 ± 0.10; control, 0.31 ± 0.08; P = 0.05), week 2 (EPC, 0.40 ± 0.06; control, 0.23 ± 0.04; P = 0.04), and week 3 (EPC, 0.33 ± 0.06; control, 0.18 ± 0.03; P = 0.04), but not at week 10 (EPC, 0.31 ± 0.06; control, 0.21 ± 0.04, P = 0.15). The highest mean expression of BMP-2 in EPC-treated defects was observed at 1 week, with a progressive decline in BMP-2 expression noted thereafter. CONCLUSIONS These findings demonstrate that EPC-treated bone defects demonstrate both radiographic healing and elevated expression of BMP-2 relative to control-treated defects. These results provide further insight into the potential mechanisms by which EPC therapy may promote fracture healing and provide further evidence to suggest that the trophic actions of EPC therapy may be a critical factor in their contribution to fracture healing.
Collapse
|
24
|
Kuroda R, Matsumoto T, Kawakami Y, Fukui T, Mifune Y, Kurosaka M. Clinical impact of circulating CD34-positive cells on bone regeneration and healing. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:190-9. [PMID: 24372338 DOI: 10.1089/ten.teb.2013.0511] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Failures in fracture healing after conventional autologous and allogenic bone grafting are mainly due to poor vascularization. To meet the clinical demand, recent attentions in the regeneration and repair of bone have been focused on the use of stem cells such as bone marrow mesenchymal stem cells and circulating skeletal stem cells. Circulating stem cells are currently paid a lot of attention due to their ease of clinical setting and high potential for osteogenesis and angiogenesis. In this report, we focus on the first proof-of-principle experiments demonstrating the collaborative characteristics of circulating CD34(+) cells, known as endothelial and hematopoietic progenitor cell-rich population, which are capable to differentiate into both endothelial cells and osteoblasts. Transplantation of circulating CD34(+) cells provides a favorable environment for fracture healing via angiogenesis/vasculogenesis and osteogenesis, finally leading to functional recovery from fracture. Based on a series of basic studies, we performed a phase 1/2 clinical trial of autologous CD34(+) cell transplantation in patients with tibial or femoral nonunions and reported the safety and efficacy of this novel therapy. In this review, the current concepts and strategies in circulating CD34(+) cell-based therapy and its potential applications for bone repair will be highlighted.
Collapse
Affiliation(s)
- Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine , Kobe, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Schindeler A, Kolind M, Little DG. Cellular transitions and tissue engineering. Cell Reprogram 2013; 15:101-6. [PMID: 23550730 DOI: 10.1089/cell.2012.0054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) describe complex changes in progenitor lineage, cell morphology, and gene expression. Stimulated by environmental cues, these cellular transitions are essential for elements of embryonic development and can be pathologically dysregulated in disease states. EMT occurs in biological processes such as gastrulation, cardiogenesis, and fibrosis. EndMT is involved in development and tissue fibrosis, but recent studies have implicated this process in musculoskeletal biology and pathology. Tissue engineering and regenerative medicine typically rely on endogenous progenitors or progenitors expanded ex vivo to repair damaged or impaired tissues or organs. The processes of EMT and EndMT may aid in elucidating new methods for reducing fibrosis and identifying novel plastic progenitor populations for tissue repair. This review will discuss the potential for EMT and EndMT to impact on tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Aaron Schindeler
- Department of Orthopaedic Research & Biotechnology, the Children's Hospital at Westmead, Sydney, Australia.
| | | | | |
Collapse
|
26
|
An emerging cell-based strategy in orthopaedics: endothelial progenitor cells. Knee Surg Sports Traumatol Arthrosc 2012; 20:1366-77. [PMID: 22402606 DOI: 10.1007/s00167-012-1940-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/15/2011] [Indexed: 12/17/2022]
Abstract
PURPOSE The purpose of this article was to analyze the results of studies in the literature, which evaluated the use of endothelial progenitor cells (EPCs) as a cell-based tissue engineering strategy. METHODS EPCs have been successfully used in regenerative medicine to augment neovascularization in patients after myocardial infarction and limb ischemia. EPCs' important role as vasculogenic progenitors presents them as a potential source for cell-based therapies to promote bone healing. RESULTS EPCs have been shown to have prominent effects in promoting bone regeneration in several animal models. Evidence indicates that EPCs promote bone regeneration by stimulating both angiogenesis and osteogenesis through a differentiation process toward endothelial cell lineage and formation of osteoblasts. Moreover, EPCs increase vascularization and osteogenesis by increased secretion of growth factors and cytokines through paracrine mechanisms. CONCLUSION EPCs offer the potential to emerge as a new strategy among other cell-based therapies to promote bone regeneration. Further investigations and human trials are required to address current questions with regard to biology and mechanisms of action of EPCs in bone tissue engineering.
Collapse
|