1
|
Lucia U, Bergandi L, Grisolia G, Fino D, Mareschi K, Marini E, Santa Banche Niclot AG, Tirtei E, Asaftei SD, Fagioli F, Ponzetto A, Silvagno F. The exposure to extremely low frequency electromagnetic-fields inhibits the growth and potentiates the sensitivity to chemotherapy of bidimensional and tridimensional human osteosarcoma models. Biomed Pharmacother 2024; 177:117162. [PMID: 39024997 DOI: 10.1016/j.biopha.2024.117162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
We previously established a thermodynamical model to calculate the specific frequencies of extremely low frequency-electromagnetic field (ELF-EMF) able to arrest the growth of cancer cells. In the present study, for the first time, we investigated the efficacy of this technology on osteosarcoma, and we applied a precise frequency of the electromagnetic field on three human osteosarcoma cell lines, grown as adherent cells and spheroids. We evaluated the antitumour efficacy of irradiation in terms of response to chemotherapeutic treatments, which is usually poor in this type of cancer. Importantly, the results of this novel combinatorial approach revealed that the specific exposure can potentiate the efficacy of several chemotherapeutic drugs, both on bidimensional and tridimensional cancer models. The effectiveness of cisplatinum, methotrexate, ifosfamide and doxorubicin was greatly increased by the concomitant application of the specific ELF-EMF. Moreover, our experiments confirmed that ELF-EMF inhibited the proliferation and modulated the mitochondrial metabolism of all cancer models tested, whereas mesenchymal cells were not affected. The latter finding is extremely valuable, given the importance of preserving the cell reservoir necessary for tissue regeneration after chemotherapy. Altogether, this novel evidence opens new avenues to the clinical applications of ELF-EMF in oncology.
Collapse
Affiliation(s)
- Umberto Lucia
- Dipartimento Energia "Galileo Ferraris", Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy.
| | - Loredana Bergandi
- Department of Oncology, University of Torino, Via Santena 5 bis, Torino 10126, Italy
| | - Giulia Grisolia
- Dipartimento di Ingegneria dell'Ambiente, del Territorio e delle Infrastrutture, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Debora Fino
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Elena Marini
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy
| | | | - Elisa Tirtei
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Sebastian Dorin Asaftei
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Franca Fagioli
- Department of Public Health and Paediatrics, The University of Turin, Piazza Polonia 94, Torino 10126, Italy; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco - Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin, Torino 10126, Italy
| | - Antonio Ponzetto
- Dipartimento di Scienze Mediche, Università di Torino, Corso Dogliotti 14, Torino 10126, Italy
| | - Francesca Silvagno
- Department of Oncology, University of Torino, Via Santena 5 bis, Torino 10126, Italy.
| |
Collapse
|
2
|
Jiang Y, Zhou D, Jiang Y. Three-dimensional bioprinted GelMA/GO composite hydrogel for stem cell osteogenic differentiation both in vitro and in vivo. J Biomater Appl 2024; 38:1087-1099. [PMID: 38561006 DOI: 10.1177/08853282241243337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In this study, we evaluated the use of graphene oxide (GO) mixed with methyl methacrylate gelatin (GelMA) for the construction of a microenvironmental implant to repair bone defects in orthopedic surgery. A scaffold containing a GelMA/GO composite with mesenchymal stem cells (MSCs) was constructed using three-dimensional bioprinting. The survival and osteogenic capacity of MSCs in the composite bioink were evaluated using cell viability and proliferation assays, osteogenesis-related gene expression analysis, and implantation under the skin of nude mice. The printing process had little effect on cell viability. We found that GO enhanced cell proliferation but had no significant effect on cell viability. In vitro experiments suggested that GO promoted material-cell interactions and the expression of osteogenesis-related genes. In vivo experiments showed that GO decreased the degradation time of the material and increased calcium nodule deposition. In contrast to pure GelMA, the addition of GO created a suitable microenvironment to promote the differentiation of loaded exogenous MSCs in vitro and in vivo, providing a basis for the repair of bone defects.
Collapse
Affiliation(s)
- Yerong Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Yanan Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
3
|
The Osteogenic Potential of Falciform Ligament-Derived Stromal Cells-A Comparative Analysis between Two Osteogenic Induction Programs. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120810. [PMID: 36551016 PMCID: PMC9774535 DOI: 10.3390/bioengineering9120810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Mesenchymal stromal cells (MSCs) have gained special relevance in bone tissue regenerative applications. MSCs have been isolated from different depots, with adipose tissue being acknowledged as one of the most convenient sources, given the wide availability, high cellular yield, and obtainability. Recently, the falciform ligament (FL) has been regarded as a potential depot for adipose tissue-derived stromal cells (FL-ADSCs) isolation. Nonetheless, the osteogenic capability of FL-ADSCs has not been previously characterized. Thus, the present study aimed the detailed characterization of FL-ADSCs' functionality upon osteogenic induction through a classic (dexamethasone-based-DEX) or an innovative strategy with retinoic acid (RA) in a comparative approach with ADSCs from a control visceral region. Cultures were characterized for cell proliferation, metabolic activity, cellular morphology, fluorescent cytoskeletal and mitochondrial organization, and osteogenic activity-gene expression analysis and cytochemical staining. FL-derived populations expressed significantly higher levels of osteogenic genes and cytochemical markers, particularly with DEX induction, as compared to control ADSCs that were more responsive to RA. FL-ADSCs were identified as a potential source for bone regenerative applications, given the heightened osteogenic functionality. Furthermore, data highlighted the importance of the selection of the most adequate osteogenic-inducing program concerning the specificities of the basal cell population.
Collapse
|
4
|
Wang W, Liang X, Zheng K, Ge G, Chen X, Xu Y, Bai J, Pan G, Geng D. Horizon of exosome-mediated bone tissue regeneration: The all-rounder role in biomaterial engineering. Mater Today Bio 2022; 16:100355. [PMID: 35875196 PMCID: PMC9304878 DOI: 10.1016/j.mtbio.2022.100355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/02/2022] Open
Abstract
Bone injury repair has always been a tricky problem in clinic, the recent emergence of bone tissue engineering provides a new direction for the repair of bone injury. However, some bone tissue processes fail to achieve satisfactory results mainly due to insufficient vascularization or cellular immune rejection. Exosomes with the ability of vesicle-mediated intercellular signal transmission have gained worldwide attention and can achieve cell-free therapy. Exosomes are small vesicles that are secreted by cells, which contain genetic material, lipids, proteins and other substances. It has been found to play the function of material exchange between cells. It is widely used in bone tissue engineering to achieve cell-free therapy because it not only does not produce some immune rejection like cells, but also can play a cell-like function. Exosomes from different sources can bind to scaffolds in various ways and affect osteoblast, angioblast, and macrophage polarization in vivo to promote bone regeneration. This article reviews the recent research progress of exosome-loaded tissue engineering, focusing on the mechanism of exosomes from different sources and the application of exosome-loaded scaffolds in promoting bone regeneration. Finally, the existing deficiencies and challenges, future development directions and prospects are summarized.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xiaolong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Kai Zheng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| |
Collapse
|
5
|
Dorozhkin SV. Calcium Orthophosphate (CaPO4)-Based Bioceramics: Preparation, Properties, and Applications. COATINGS 2022; 12:1380. [DOI: 10.3390/coatings12101380] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Various types of materials have been traditionally used to restore damaged bones. In the late 1960s, a strong interest was raised in studying ceramics as potential bone grafts due to their biomechanical properties. A short time later, such synthetic biomaterials were called bioceramics. Bioceramics can be prepared from diverse inorganic substances, but this review is limited to calcium orthophosphate (CaPO4)-based formulations only, due to its chemical similarity to mammalian bones and teeth. During the past 50 years, there have been a number of important achievements in this field. Namely, after the initial development of bioceramics that was just tolerated in the physiological environment, an emphasis was shifted towards the formulations able to form direct chemical bonds with the adjacent bones. Afterwards, by the structural and compositional controls, it became possible to choose whether the CaPO4-based implants would remain biologically stable once incorporated into the skeletal structure or whether they would be resorbed over time. At the turn of the millennium, a new concept of regenerative bioceramics was developed, and such formulations became an integrated part of the tissue engineering approach. Now, CaPO4-based scaffolds are designed to induce bone formation and vascularization. These scaffolds are usually porous and harbor various biomolecules and/or cells. Therefore, current biomedical applications of CaPO4-based bioceramics include artificial bone grafts, bone augmentations, maxillofacial reconstruction, spinal fusion, and periodontal disease repairs, as well as bone fillers after tumor surgery. Prospective future applications comprise drug delivery and tissue engineering purposes because CaPO4 appear to be promising carriers of growth factors, bioactive peptides, and various types of cells.
Collapse
|
6
|
Venkataiah VS, Yahata Y, Kitagawa A, Inagaki M, Kakiuchi Y, Nakano M, Suzuki S, Handa K, Saito M. Clinical Applications of Cell-Scaffold Constructs for Bone Regeneration Therapy. Cells 2021; 10:2687. [PMID: 34685667 PMCID: PMC8534498 DOI: 10.3390/cells10102687] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/20/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Bone tissue engineering (BTE) is a process of combining live osteoblast progenitors with a biocompatible scaffold to produce a biological substitute that can integrate into host bone tissue and recover its function. Mesenchymal stem cells (MSCs) are the most researched post-natal stem cells because they have self-renewal properties and a multi-differentiation capacity that can give rise to various cell lineages, including osteoblasts. BTE technology utilizes a combination of MSCs and biodegradable scaffold material, which provides a suitable environment for functional bone recovery and has been developed as a therapeutic approach to bone regeneration. Although prior clinical trials of BTE approaches have shown promising results, the regeneration of large bone defects is still an unmet medical need in patients that have suffered a significant loss of bone function. In this present review, we discuss the osteogenic potential of MSCs in bone tissue engineering and propose the use of immature osteoblasts, which can differentiate into osteoblasts upon transplantation, as an alternative cell source for regeneration in large bone defects.
Collapse
Affiliation(s)
- Venkata Suresh Venkataiah
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Yoshio Yahata
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Akira Kitagawa
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- OsteRenatos Ltd., Sendai Capital Tower 2F, 4-10-3 Central, Aoba-ku, Sendai 980-0021, Japan
| | - Masahiko Inagaki
- National Institute of Advanced Industrial Science and Technology, 2266-98 Anagahora, Nagoya 463-8560, Japan;
| | - Yusuke Kakiuchi
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Masato Nakano
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Shigeto Suzuki
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
| | - Keisuke Handa
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- Department of Oral Science, Division of Oral Biochemistry, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka 238-8580, Japan
| | - Masahiro Saito
- Department of Restorative Dentistry, Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai 980-8575, Japan; (Y.Y.); (A.K.); (Y.K.); (M.N.); (S.S.); (K.H.); (M.S.)
- OsteRenatos Ltd., Sendai Capital Tower 2F, 4-10-3 Central, Aoba-ku, Sendai 980-0021, Japan
| |
Collapse
|
7
|
Yang P, Zhou J, Ai Q, Yu B, Deng M, Luo F, Xie Z, Xing J, Hou T. Comparison of Individual Tissue-Engineered Bones and Allogeneic Bone in Treating Bone Defects: A Long-Term Follow-Up Study. Cell Transplant 2021; 29:963689720940722. [PMID: 32731815 PMCID: PMC7563814 DOI: 10.1177/0963689720940722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The treatment of bone defects has always been a challenge for orthopedic surgeons. The development of tissue engineering technology provides a novel method for repairing bone defects and has been used in animal experiments and clinical trials. However, there are few clinical studies on comparing the long-term outcomes of tissue-engineered bones (TEBs) and other bone grafts in treating bone defects, and the long-term efficiency of TEBs remains controversial. Therefore, a study designed by us was aimed to compare the long-term efficacy and safety of individual tissue-engineered bones (iTEBs) and allogeneic bone granules (ABGs) in treating bone defects caused by curettage of benign bone tumors and tumor-like lesions. From September 2003 to November 2009, 48 patients who received tumor curettage and bone grafting were analyzed with a mean follow-up of 122 mo (range 60 to 173 mo). Based on implant style, patients were divided into groups of iTEBs (n = 23) and ABGs (n = 25). Postoperatively, the healing time, healing quality, incidence of complications, and functional scores were compared between the two groups. The Musculoskeletal Tumor Society functional evaluation system and Activities of Daily Living Scale scores were significantly improved in both groups with no significant difference. The average healing time of ABGs was longer than that of iTEBs (P < 0.05). At the final follow-up, iTEBs had a better performance in the bone healing quality evaluated by modified Neer classification (P < 0.05). In the group of iTEBs, the complication and reoperation rate was lower than that in the group of ABGs, with no tumorigenesis or immune rejection observed. In summary, for treating bone defects caused by tumor curettage, iTEBs were safe, effective, and tagged with more rapid healing speed, better healing outcome, and lower complication and reoperation rate, in comparison with ABGs.
Collapse
Affiliation(s)
- Peng Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Jiangling Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Qiuchi Ai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Bo Yu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Moyuan Deng
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Zhao Xie
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Junchao Xing
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| | - Tianyong Hou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China.,Tissue Engineering Laboratory of Chongqing City, Chongqing, China.,Key Lab of Military Bone Tissue Engineering, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
Bouland C, Philippart P, Dequanter D, Corrillon F, Loeb I, Bron D, Lagneaux L, Meuleman N. Cross-Talk Between Mesenchymal Stromal Cells (MSCs) and Endothelial Progenitor Cells (EPCs) in Bone Regeneration. Front Cell Dev Biol 2021; 9:674084. [PMID: 34079804 PMCID: PMC8166285 DOI: 10.3389/fcell.2021.674084] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Bone regeneration is a complex, well-orchestrated process based on the interactions between osteogenesis and angiogenesis, observed in both physiological and pathological situations. However, specific conditions (e.g., bone regeneration in large quantity, immunocompromised regenerative process) require additional support. Tissue engineering offers novel strategies. Bone regeneration requires a cell source, a matrix, growth factors and mechanical stimulation. Regenerative cells, endowed with proliferation and differentiation capacities, aim to recover, maintain, and improve bone functions. Vascularization is mandatory for bone formation, skeletal development, and different osseointegration processes. The latter delivers nutrients, growth factors, oxygen, minerals, etc. The development of mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) cocultures has shown synergy between the two cell populations. The phenomena of osteogenesis and angiogenesis are intimately intertwined. Thus, cells of the endothelial line indirectly foster osteogenesis, and conversely, MSCs promote angiogenesis through different interaction mechanisms. In addition, various studies have highlighted the importance of the microenvironment via the release of extracellular vesicles (EVs). These EVs stimulate bone regeneration and angiogenesis. In this review, we describe (1) the phenomenon of bone regeneration by different sources of MSCs. We assess (2) the input of EPCs in coculture in bone regeneration and describe their contribution to the osteogenic potential of MSCs. We discuss (3) the interaction mechanisms between MSCs and EPCs in the context of osteogenesis: direct or indirect contact, production of growth factors, and the importance of the microenvironment via the release of EVs.
Collapse
Affiliation(s)
- Cyril Bouland
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Pierre Philippart
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Stomatology and Maxillofacial Surgery, IRIS South Hospital, Brussels, Belgium
| | - Didier Dequanter
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Florent Corrillon
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Loeb
- Department of Stomatology and Maxillofacial Surgery, Saint-Pierre Hospital, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Dominique Bron
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium.,Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
9
|
Jiang S, Wang M, He J. A review of biomimetic scaffolds for bone regeneration: Toward a cell-free strategy. Bioeng Transl Med 2021; 6:e10206. [PMID: 34027093 PMCID: PMC8126827 DOI: 10.1002/btm2.10206] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 12/20/2022] Open
Abstract
In clinical terms, bone grafting currently involves the application of autogenous, allogeneic, or xenogeneic bone grafts, as well as natural or artificially synthesized materials, such as polymers, bioceramics, and other composites. Many of these are associated with limitations. The ideal scaffold for bone tissue engineering should provide mechanical support while promoting osteogenesis, osteoconduction, and even osteoinduction. There are various structural complications and engineering difficulties to be considered. Here, we describe the biomimetic possibilities of the modification of natural or synthetic materials through physical and chemical design to facilitate bone tissue repair. This review summarizes recent progresses in the strategies for constructing biomimetic scaffolds, including ion-functionalized scaffolds, decellularized extracellular matrix scaffolds, and micro- and nano-scale biomimetic scaffold structures, as well as reactive scaffolds induced by physical factors, and other acellular scaffolds. The fabrication techniques for these scaffolds, along with current strategies in clinical bone repair, are described. The developments in each category are discussed in terms of the connection between the scaffold materials and tissue repair, as well as the interactions with endogenous cells. As the advances in bone tissue engineering move toward application in the clinical setting, the demonstration of the therapeutic efficacy of these novel scaffold designs is critical.
Collapse
Affiliation(s)
- Sijing Jiang
- Department of Plastic SurgeryFirst Affiliated Hospital of Anhui Medical University, Anhui Medical UniversityHefeiChina
| | - Mohan Wang
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| | - Jiacai He
- Stomatologic Hospital & College, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui ProvinceHefeiChina
| |
Collapse
|
10
|
Mukherjee S, Agarwal M, Bakshi A, Sawant S, Thomas L, Fujii N, Nair P, Kode J. Chemokine SDF1 Mediated Bone Regeneration Using Biodegradable Poly(D,L-lactide- co-glycolide) 3D Scaffolds and Bone Marrow-Derived Mesenchymal Stem Cells: Implication for the Development of an "Off-the-Shelf" Pharmacologically Active Construct. Biomacromolecules 2020; 21:4888-4903. [PMID: 33136384 DOI: 10.1021/acs.biomac.0c01134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is an increasing need for bone substitutes for reconstructive orthopedic surgery following removal of bone tumors. Despite the advances in bone regeneration, the use of autologous mesenchymal stem cells (MSC) presents a significant challenge, particularly for the treatment of large bone defects in cancer patients. This study aims at developing new chemokine-based technology to generate biodegradable scaffolds that bind pharmacologically active proteins for regeneration/repair of target injured tissues in patients. Primary MSC were cultured from the uninvolved bone marrow (BM) of cancer patients and further characterized for "stemness". Their ability to differentiate into an osteogenic lineage was studied in 2D cultures as well as on 3D macroporous PLGA scaffolds incorporated with biomacromolecules bFGF and homing factor chemokine stromal-cell derived factor-1 (SDF1). MSC from the uninvolved BM of cancer patients exhibited properties similar to that reported for MSC from BM of healthy individuals. Macroporous PLGA discs were prepared and characterized for pore size, architecture, functional groups, thermostability, and cytocompatibility by ESEM, FTIR, DSC, and CCK-8 dye proliferation assay, respectively. It was observed that the MSC+PLGA+bFGF+SDF1 construct cultured for 14 days supported significant cell growth, osteo-lineage differentiation with increased osteocalcin expression, alkaline phosphatase secretion, calcium mineralization, bone volume, and soluble IL6 compared to unseeded PLGA and PLGA+MSC, as analyzed by confocal microscopy, biochemistry, ESEM, microCT imaging, flow cytometry, and EDS. Thus, chemotactic biomacromolecule SDF1-guided tissue repair/regeneration ability of MSC from cancer patients opens up the avenues for development of "off-the-shelf" pharmacologically active construct for optimal repair of the target injured tissue in postsurgery cancer patients, bone defects, damaged bladder tissue, and radiation-induced skin/mucosal lesions.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC Australia 3168
| | - Manish Agarwal
- Department of Orthopaedic Oncology, Tata Memorial Hospital, TMC, Parel, Mumbai 400012, India
- Department of Orthopedic Oncology, P.D. Hinduja National Hospital & Medical Research Centre, Mumbai, India
| | - Ashish Bakshi
- Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- Department of Bone Marrow Transplantation, Department of Medical Oncology, Hiranandani Hospital, Powai, Mumbai 400076, India
| | - Sharada Sawant
- Electron Microscopy Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
| | - Lynda Thomas
- Laboratory for Polymer Analysis, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology Poojappura, Trivandrum, India
| | - Nobutaka Fujii
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Prabha Nair
- Laboratory for Polymer Analysis, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology Poojappura, Trivandrum, India
| | - Jyoti Kode
- Tumor Immunology and Immunotherapy Group, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre (TMC), Kharghar, Navi Mumbai 410210, India
- Homi Bhabha National Institute (HBNI), Training School Complex, Anushakti Nagar, Mumbai 400094, India
| |
Collapse
|
11
|
Metz C, Duda GN, Checa S. Towards multi-dynamic mechano-biological optimization of 3D-printed scaffolds to foster bone regeneration. Acta Biomater 2020; 101:117-127. [PMID: 31669697 DOI: 10.1016/j.actbio.2019.10.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/26/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
Substantial tissue loss, such as in large bone defects, represents a clinical challenge for which regenerative therapies and tissue engineering strategies aim at offering treatment alternatives to conventional replacement approaches by metallic implants. 3D printing technologies provide endless opportunities to shape scaffold structures that could support endogenous regeneration. However, it remains unclear which of the numerous parameters at hand eventually enhance tissue regeneration. In the last decades, a significant effort has been made in the development of computer tools to optimize scaffold designs. Here, we aim at giving a more comprehensive overview summarizing current computer optimization framework technologies. We confront these with the most recent advances in scaffold mechano-biological optimization, discuss their limitations and provide suggestions for future development. We conclude that the field needs to move forward to not only optimize scaffolds to avoid implant failures but to improve their mechano-biological behaviour: providing an initial stimulus for fast tissue organisation and healing and accounting for remodelling, scaffold degradation and consecutive filling with host tissue. So far, modelling approaches fall short in including the various scales of tissue dynamics. With this review, we wish to stimulate a move towards multi-dynamic mechano-biological optimization of 3D-printed scaffolds. STATEMENT OF SIGNIFICANCE: Large bone defects represent a clinical challenge for which tissue engineering strategies aim at offering alternatives to conventional treatment strategies. 3D printing technologies provide endless opportunities to shape scaffold structures that could support endogenous regeneration. However, it remains unclear which of the numerous parameters at hand eventually enhance tissue regeneration. In the last decades, a significant effort has been made in the development of computer tools to optimize scaffold designs. This review summarizes current computer optimization frameworks and most recent advances in mechano-biological optimization of bone scaffolds to better stimulate bone regeneration. We wish to stimulate a move towards multi-dynamic mechano-biological optimization of 3D-printed scaffolds.
Collapse
Affiliation(s)
- Camille Metz
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany; MINES ParisTech - PSL Research University, Paris, France
| | - Georg N Duda
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
12
|
Stamatopoulos A, Stamatopoulos T, Gamie Z, Kenanidis E, Ribeiro RDC, Rankin KS, Gerrand C, Dalgarno K, Tsiridis E. Mesenchymal stromal cells for bone sarcoma treatment: Roadmap to clinical practice. J Bone Oncol 2019; 16:100231. [PMID: 30956944 PMCID: PMC6434099 DOI: 10.1016/j.jbo.2019.100231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, there has been growing interest in understanding the molecular mechanisms of cancer pathogenesis and progression, as it is still associated with high morbidity and mortality. Current management of large bone sarcomas typically includes the complex therapeutic approach of limb salvage or sacrifice combined with pre- and postoperative multidrug chemotherapy and/or radiotherapy, and is still associated with high recurrence rates. The development of cellular strategies against specific characteristics of tumour cells appears to be promising, as they can target cancer cells selectively. Recently, Mesenchymal Stromal Cells (MSCs) have been the subject of significant research in orthopaedic clinical practice through their use in regenerative medicine. Further research has been directed at the use of MSCs for more personalized bone sarcoma treatments, taking advantage of their wide range of potential biological functions, which can be augmented by using tissue engineering approaches to promote healing of large defects. In this review, we explore the use of MSCs in bone sarcoma treatment, by analyzing MSCs and tumour cell interactions, transduction of MSCs to target sarcoma, and their clinical applications on humans concerning bone regeneration after bone sarcoma extraction.
Collapse
Key Words
- 5-FC, 5-fluorocytosine
- AAT, a1-antitrypsin
- APCs, antigen presenting cells
- ASC, adipose-derived stromal/stem cells
- Abs, antibodies
- Ang1, angiopoietin-1
- BD, bone defect
- BMMSCs, bone marrow-derived mesenchymal stromal cells
- Biology
- Bone
- CAM, cell adhesion molecules
- CCL5, chemokine ligand 5
- CCR2, chemokine receptor 2
- CD, classification determinants
- CD, cytosine deaminase
- CLUAP1, clusterin associated protein 1
- CSPG4, Chondroitin sulfate proteoglycan 4
- CX3CL1, chemokine (C-X3-C motif) ligand 1
- CXCL12/CXCR4, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 4
- CXCL12/CXCR7, C-X-C chemokine ligand 12/ C-X-C chemokine receptor 7
- CXCR4, chemokine receptor type 4
- Cell
- DBM, Demineralized Bone Marrow
- DKK1, dickkopf-related protein 1
- ECM, extracellular matrix
- EMT, epithelial-mesenchymal transition
- FGF-2, fibroblast growth factors-2
- FGF-7, fibroblast growth factors-7
- GD2, disialoganglioside 2
- HER2, human epidermal growth factor receptor 2
- HGF, hepatocyte growth factor
- HMGB1/RACE, high mobility group box-1 protein/ receptor for advanced glycation end-products
- IDO, indoleamine 2,3-dioxygenase
- IFN-α, interferon alpha
- IFN-β, interferon beta
- IFN-γ, interferon gamma
- IGF-1R, insulin-like growth factor 1 receptor
- IL-10, interleukin-10
- IL-12, interleukin-12
- IL-18, interleukin-18
- IL-1b, interleukin-1b
- IL-21, interleukin-21
- IL-2a, interleukin-2a
- IL-6, interleukin-6
- IL-8, interleukin-8
- IL11RA, Interleukin 11 Receptor Subunit Alpha
- MAGE, melanoma antigen gene
- MCP-1, monocyte chemoattractant protein-1
- MMP-2, matrix metalloproteinase-2
- MMP2/9, matrix metalloproteinase-2/9
- MRP, multidrug resistance protein
- MSCs, mesenchymal stem/stromal cells
- Mesenchymal
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- OPG, osteoprotegerin
- Orthopaedic
- PBS, phosphate-buffered saline
- PDGF, platelet-derived growth factor
- PDX, patient derived xenograft
- PEDF, pigment epithelium-derived factor
- PGE2, prostaglandin E2
- PI3K/Akt, phosphoinositide 3-kinase/protein kinase B
- PTX, paclitaxel
- RANK, receptor activator of nuclear factor kappa-B
- RANKL, receptor activator of nuclear factor kappa-B ligand
- RBCs, red blood cells
- RES, reticuloendothelial system
- RNA, ribonucleic acid
- Regeneration
- SC, stem cells
- SCF, stem cells factor
- SDF-1, stromal cell-derived factor 1
- STAT-3, signal transducer and activator of transcription 3
- Sarcoma
- Stromal
- TAAs, tumour-associated antigens
- TCR, T cell receptor
- TGF-b, transforming growth factor beta
- TGF-b1, transforming growth factor beta 1
- TNF, tumour necrosis factor
- TNF-a, tumour necrosis factor alpha
- TRAIL, tumour necrosis factor related apoptosis-inducing ligand
- Tissue
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- WBCs, white blood cell
- hMSCs, human mesenchymal stromal cells
- rh-TRAIL, recombinant human tumour necrosis factor related apoptosis-inducing ligand
Collapse
Affiliation(s)
- Alexandros Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Theodosios Stamatopoulos
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Zakareya Gamie
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Eustathios Kenanidis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| | - Ricardo Da Conceicao Ribeiro
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Kenneth Samora Rankin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Craig Gerrand
- Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, HA7 4LP, UK
| | - Kenneth Dalgarno
- School of Mechanical and Systems Engineering, Stephenson Building, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | - Eleftherios Tsiridis
- Academic Orthopaedic Unit, Papageorgiou General Hospital, Aristotle University Medical School, West Ring Road of Thessaloniki, Pavlos Melas Area, N. Efkarpia, 56403 Thessaloniki, Greece
- Center of Orthopaedics and Regenerative Medicine (C.O.RE.), Center for Interdisciplinary Research and Innovation (C.I.R.I.), Aristotle University Thessaloniki, Greece
| |
Collapse
|
13
|
Mandibular Regeneration With Autologous Human Bone Marrow Derived Mesenchymal Stem Cells to Treat Unicystic Ameloblastoma. J Craniofac Surg 2019; 30:e187-e189. [DOI: 10.1097/scs.0000000000004917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
14
|
Abstract
Limb salvage is widely practiced as standard of care in most cases of extremity bone sarcoma. Allograft and endoprosthesis reconstructions are the most widely utilized modalities for the reconstruction of large segment defects, however complication rates remain high. Aseptic loosening and infection remain the most common modes of failure. Implant integration, soft-tissue function, and infection prevention are crucial for implant longevity and function. Macro and micro alterations in implant design are reviewed in this manuscript. Tissue engineering principles using nanoparticles, cell-based, and biological augments have been utilized to develop implant coatings that improve osseointegration and decrease infection. Similar techniques have been used to improve the interaction between soft tissues and implants. Tissue engineered constructs (TEC) used in combination with, or in place of, traditional reconstructive techniques may represent the next major advancement in orthopaedic oncology reconstructive science, although preclinical results have yet to achieve durable translation to the bedside.
Collapse
|
15
|
Zeng JH, Liu SW, Xiong L, Qiu P, Ding LH, Xiong SL, Li JT, Liao XG, Tang ZM. Scaffolds for the repair of bone defects in clinical studies: a systematic review. J Orthop Surg Res 2018; 13:33. [PMID: 29433544 PMCID: PMC5809923 DOI: 10.1186/s13018-018-0724-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/17/2018] [Indexed: 01/06/2023] Open
Abstract
Background This systematic review aims to summarize the clinical studies on the use of scaffolds in the repair of bony defects. Methods The relevant articles were searched through PubMed database. The following keywords and search terms were used: “scaffolds,” “patient,” “clinic,” “bone repair,” “bone regeneration,” “repairing bone defect,” “repair of bone,” “osteanagenesis,” “osteanaphysis,” and “osteoanagenesis.” The articles were screened according to inclusion and exclusion criteria, performed by two reviewers. Results A total of 373 articles were obtained using PubMed database. After screening, 20 articles were identified as relevant for the purpose of this systematic review. We collected the data of biological scaffolds and synthetic scaffolds. There are eight clinical studies of biological scaffolds included collagen, gelatin, and cellular scaffolds for bone healing. In addition, 12 clinical studies of synthetic scaffolds on HAp, TCP, bonelike, and their complex scaffolds for repairing bone defects were involved in this systematic review. Conclusions There are a lot of clinical evidences showed that application of scaffolds had a good ability to facilitate bone repair and osteogenesis. However, the ideal and reliable guidelines are insufficiently applied and the number and quality of studies in this field remain to be improved.
Collapse
Affiliation(s)
- Jian-Hua Zeng
- Department of Orthopaedics, Jiangxi People's Hospital, No.152, Ai guo Road, Nanchang, 330006, China
| | - Shi-Wei Liu
- Department of Orthopaedics, Jiangxi medical college, Nanchang university, Nanchang, China
| | - Long Xiong
- Department of Orthopaedics, Jiangxi People's Hospital, No.152, Ai guo Road, Nanchang, 330006, China.
| | - Peng Qiu
- Department of Orthopaedics, Jiangxi medical college, Nanchang university, Nanchang, China
| | - Ling-Hua Ding
- Department of Orthopaedics, Jiangxi medical college, Nanchang university, Nanchang, China
| | | | - Jing-Tang Li
- Department of Orthopaedics, Jiangxi People's Hospital, No.152, Ai guo Road, Nanchang, 330006, China
| | - Xin-Gen Liao
- Department of Orthopaedics, Jiangxi People's Hospital, No.152, Ai guo Road, Nanchang, 330006, China
| | - Zhi-Ming Tang
- Department of Orthopaedics, Jiangxi People's Hospital, No.152, Ai guo Road, Nanchang, 330006, China
| |
Collapse
|
16
|
Daly AC, Pitacco P, Nulty J, Cunniffe GM, Kelly DJ. 3D printed microchannel networks to direct vascularisation during endochondral bone repair. Biomaterials 2018; 162:34-46. [PMID: 29432987 DOI: 10.1016/j.biomaterials.2018.01.057] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/16/2018] [Accepted: 01/30/2018] [Indexed: 01/02/2023]
Abstract
Bone tissue engineering strategies that recapitulate the developmental process of endochondral ossification offer a promising route to bone repair. Clinical translation of such endochondral tissue engineering strategies will require overcoming a number of challenges, including the engineering of large and often anatomically complex cartilage grafts, as well as the persistence of core regions of avascular cartilage following their implantation into large bone defects. Here 3D printing technology is utilized to develop a versatile and scalable approach to guide vascularisation during endochondral bone repair. First, a sacrificial pluronic ink was used to 3D print interconnected microchannel networks in a mesenchymal stem cell (MSC) laden gelatin-methacryloyl (GelMA) hydrogel. These constructs (with and without microchannels) were next chondrogenically primed in vitro and then implanted into critically sized femoral bone defects in rats. The solid and microchanneled cartilage templates enhanced bone repair compared to untreated controls, with the solid cartilage templates (without microchannels) supporting the highest levels of total bone formation. However, the inclusion of 3D printed microchannels was found to promote osteoclast/immune cell invasion, hydrogel degradation, and vascularisation following implantation. In addition, the endochondral bone tissue engineering strategy was found to support comparable levels of bone healing to BMP-2 delivery, whilst promoting lower levels of heterotopic bone formation, with the microchanneled templates supporting the lowest levels of heterotopic bone formation. Taken together, these results demonstrate that 3D printed hypertrophic cartilage grafts represent a promising approach for the repair of complex bone fractures, particularly for larger defects where vascularisation will be a key challenge.
Collapse
Affiliation(s)
- Andrew C Daly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Pierluca Pitacco
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Jessica Nulty
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Gráinne M Cunniffe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
17
|
Lin J, Shao J, Juan L, Yu W, Song X, Liu P, Weng W, Xu J, Mehl C. Enhancing bone regeneration by combining mesenchymal stem cell sheets with β-TCP/COL-I scaffolds. J Biomed Mater Res B Appl Biomater 2017; 106:2037-2045. [PMID: 29098765 DOI: 10.1002/jbm.b.34003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Jun Lin
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Jiaqi Shao
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Li Juan
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Wenke Yu
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Xiaojia Song
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Pengruofeng Liu
- Department of Stomatology; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Wenjian Weng
- School of Materials Science and Engineering; Zhejiang University; 310027 Hangzhou China
| | - Jinghong Xu
- Department of Plastic Surgery; First Affiliated Hospital of Zhejiang University; 310003 Hangzhou China
| | - Christian Mehl
- Department of Prosthodontics, Propaedeutics and Dental Materials; Christian-Albrechts University at Kiel; 2415 Kiel Germany
| |
Collapse
|
18
|
Sladkova M, Palmer M, Öhman C, Cheng J, Al-Ansari S, Saad M, Engqvist H, de Peppo GM. Engineering human bone grafts with new macroporous calcium phosphate cement scaffolds. J Tissue Eng Regen Med 2017. [PMID: 28635177 DOI: 10.1002/term.2491] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone engineering opens the possibility to grow large amounts of tissue products by combining patient-specific cells with compliant biomaterials. Decellularized tissue matrices represent suitable biomaterials, but availability, long processing time, excessive cost, and concerns on pathogen transmission have led to the development of biomimetic synthetic alternatives. We recently fabricated calcium phosphate cement (CPC) scaffolds with variable macroporosity using a facile synthesis method with minimal manufacturing steps and demonstrated long-term biocompatibility in vitro. However, there is no knowledge on the potential use of these scaffolds for bone engineering and whether the porosity of the scaffolds affects osteogenic differentiation and tissue formation in vitro. In this study, we explored the bone engineering potential of CPC scaffolds with two different macroporosities using human mesenchymal progenitors derived from induced pluripotent stem cells (iPSC-MP) or isolated from bone marrow (BMSC). Biomimetic decellularized bone scaffolds were used as reference material in all experiments. The results demonstrate that, irrespective of their macroporosity, the CPC scaffolds tested in this study support attachment, viability, and growth of iPSC-MP and BMSC cells similarly to decellularized bone. Importantly, the tested materials sustained differentiation of the cells as evidenced by increased expression of osteogenic markers and formation of a mineralized tissue. In conclusion, the results of this study suggest that the CPC scaffolds fabricated using our method are suitable to engineer bone grafts from different cell sources and could lead to the development of safe and more affordable tissue grafts for reconstructive dentistry and orthopaedics and in vitro models for basic and applied research.
Collapse
Affiliation(s)
- Martina Sladkova
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Michael Palmer
- Division of Applied Material Sciences, Uppsala University, Uppsala, Sweden
| | - Caroline Öhman
- Division of Applied Material Sciences, Uppsala University, Uppsala, Sweden
| | - Jiayi Cheng
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Shoug Al-Ansari
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Munerah Saad
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Håkan Engqvist
- Division of Applied Material Sciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
19
|
Li X, Wang M, Deng Y, Chen X, Xiao Y, Zhang X. Fabrication and Properties of Ca-P Bioceramic Spherical Granules with Interconnected Porous Structure. ACS Biomater Sci Eng 2017; 3:1557-1566. [DOI: 10.1021/acsbiomaterials.7b00232] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xiangfeng Li
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Menglu Wang
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yanglong Deng
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xuening Chen
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yumei Xiao
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research
Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
20
|
Takebe Y, Tatehara S, Fukushima T, Tokuyama-Toda R, Yasuhara R, Mishima K, Satomura K. Cryopreservation Method for the Effective Collection of Dental Pulp Stem Cells. Tissue Eng Part C Methods 2017; 23:251-261. [DOI: 10.1089/ten.tec.2016.0519] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yusuke Takebe
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Seiko Tatehara
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Tatsuhiro Fukushima
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Reiko Tokuyama-Toda
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Rika Yasuhara
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan
| | - Kazuhito Satomura
- Department of Oral Medicine and Stomatology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
21
|
Arahira T, Maruta M, Matsuya S. Characterization and in vitro evaluation of biphasic α-tricalcium phosphate/β-tricalcium phosphate cement. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 74:478-484. [DOI: 10.1016/j.msec.2016.12.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 11/25/2016] [Accepted: 12/11/2016] [Indexed: 12/30/2022]
|
22
|
Hosseini S, Shamekhi MA, Jahangir S, Bagheri F, Eslaminejad MB. The Robust Potential of Mesenchymal Stem Cell-Loaded Constructs for Hard Tissue Regeneration After Cancer Removal. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1084:17-43. [DOI: 10.1007/5584_2017_131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
23
|
Promotion of Osteogenesis and Angiogenesis in Vascularized Tissue-Engineered Bone Using Osteogenic Matrix Cell Sheets. Plast Reconstr Surg 2016; 137:1476-1484. [PMID: 27119922 DOI: 10.1097/prs.0000000000002079] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The regeneration of large, poorly vascularized bone defects remains a significant challenge. Although vascularized bone grafts promote osteogenesis, the required tissue harvesting causes problematic donor-site morbidity. Artificial bone substitutes are promising alternatives for regenerative medicine applications, but the incorporation of suitable cells and/or growth factors is necessary for their successful clinical application. The inclusion of vascular bundles can further enhance the bone-forming capability of bone substitutes by promoting tissue neovascularization. Little is known about how neovascularization occurs and how new bone extends within vascularized tissue-engineered bone, because no previous studies have used tissue-engineered bone to treat large, poorly vascularized defects. METHODS In this study, the authors developed a novel vascularized tissue-engineered bone scaffold composed of osteogenic matrix cell sheets wrapped around vascular bundles within β-tricalcium phosphate ceramics. RESULTS Four weeks after subcutaneous transplantation in rats, making use of the femoral vascular bundle, vascularized tissue-engineered bone demonstrated more angiogenesis and higher osteogenic potential than the controls. After vascularized tissue-engineered bone implantation, abundant vascularization and new bone formation were observed radially from the vascular bundle, with increased mRNA expression of alkaline phosphatase, bone morphogenetic protein-2, osteocalcin, and vascular endothelial growth factor-A. CONCLUSION This novel method for preparing vascularized tissue-engineered bone scaffolds may promote the regeneration of large bone defects, particularly where vascularization has been compromised.
Collapse
|
24
|
Nasef A, Fouillard L, El-Taguri A, Lopez M. Human bone marrow-derived mesenchymal stem cells. Libyan J Med 2016. [DOI: 10.3402/ljm.v2i4.4729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- A. Nasef
- EA 1638 –Hématologie, Faculté de Médicine Saint-Antoine, Université de Pierre et Marie Curie, Paris VI, 27 Rue de Chaligny, 75012 Paris, France and
| | - L. Fouillard
- EA 1638 –Hématologie, Faculté de Médicine Saint-Antoine, Université de Pierre et Marie Curie, Paris VI, 27 Rue de Chaligny, 75012 Paris, France and
| | | | - M. Lopez
- EA 1638 –Hématologie, Faculté de Médicine Saint-Antoine, Université de Pierre et Marie Curie, Paris VI, 27 Rue de Chaligny, 75012 Paris, France and
| |
Collapse
|
25
|
Ueyama Y, Yagyuu T, Maeda M, Imada M, Akahane M, Kawate K, Tanaka Y, Kirita T. Maxillofacial bone regeneration with osteogenic matrix cell sheets: An experimental study in rats. Arch Oral Biol 2016; 72:138-145. [DOI: 10.1016/j.archoralbio.2016.08.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
|
26
|
Freeman FE, McNamara LM. Endochondral Priming: A Developmental Engineering Strategy for Bone Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:128-141. [PMID: 27758156 DOI: 10.1089/ten.teb.2016.0197] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue engineering and regenerative medicine have significant potential to treat bone pathologies by exploiting the capacity for bone progenitors to grow and produce tissue constituents under specific biochemical and physical conditions. However, conventional tissue engineering approaches, which combine stem cells with biomaterial scaffolds, are limited as the constructs often degrade, due to a lack of vascularization, and lack the mechanical integrity to fulfill load bearing functions, and as such are not yet widely used for clinical treatment of large bone defects. Recent studies have proposed that in vitro tissue engineering approaches should strive to simulate in vivo bone developmental processes and, thereby, imitate natural factors governing cell differentiation and matrix production, following the paradigm recently defined as "developmental engineering." Although developmental engineering strategies have been recently developed that mimic specific aspects of the endochondral ossification bone formation process, these findings are not widely understood. Moreover, a critical comparison of these approaches to standard biomaterial-based bone tissue engineering has not yet been undertaken. For that reason, this article presents noteworthy experimental findings from researchers focusing on developing an endochondral-based developmental engineering strategy for bone tissue regeneration. These studies have established that in vitro approaches, which mimic certain aspects of the endochondral ossification process, namely the formation of the cartilage template and the vascularization of the cartilage template, can promote mineralization and vascularization to a certain extent both in vitro and in vivo. Finally, this article outlines specific experimental challenges that must be overcome to further exploit the biology of endochondral ossification and provide a tissue engineering construct for clinical treatment of large bone/nonunion defects and obviate the need for bone tissue graft.
Collapse
Affiliation(s)
- Fiona E Freeman
- Centre for Biomechanics Research (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway , Galway, Ireland
| | - Laoise M McNamara
- Centre for Biomechanics Research (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway , Galway, Ireland
| |
Collapse
|
27
|
|
28
|
Shikata H, Kaku M, Kojima SI, Sumi H, Kojima ST, Yamamoto T, Yashima Y, Kawata T, Tanne K, Tanimoto K. The effect of magnetic field during freezing and thawing of rat bone marrow-derived mesenchymal stem cells. Cryobiology 2016; 73:15-9. [DOI: 10.1016/j.cryobiol.2016.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/22/2022]
|
29
|
Up-and-Coming Mandibular Reconstruction Technique With Autologous Human Bone Marrow Stem Cells and Iliac Bone Graft in Patients With Large Bony Defect. J Craniofac Surg 2016; 26:e718-20. [PMID: 26594984 DOI: 10.1097/scs.0000000000002224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Large bony defects followed by resection of the mandible need to be reconstructed by various surgical techniques such as the fibular flap. In this article, we report the case of mandibular reconstruction with autologous human bone marrow mesenchymal stem cells and autogenous bone graft, followed by placement of dental implants and prosthodontic treatment in a patient who has been failed to reconstruct mandibular bone defect after resection of mandible.
Collapse
|
30
|
Banks TA, Luckman PSB, Frith JE, Cooper-White JJ. Effects of electric fields on human mesenchymal stem cell behaviour and morphology using a novel multichannel device. Integr Biol (Camb) 2016; 7:693-712. [PMID: 25988194 DOI: 10.1039/c4ib00297k] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The intrinsic piezoelectric nature of collagenous-rich tissues, such as bone and cartilage, can result in the production of small, endogenous electric fields (EFs) during applied mechanical stresses. In vivo, these EFs may influence cell migration, a vital component of wound healing. As a result, the application of small external EFs to bone fractures and cutaneous wounds is actively practiced clinically. Due to the significant regenerative potential of stem cells in bone and cartilage healing, and their potential role in the observed improved healing in vivo post applied EFs, using a novel medium throughput device, we investigated the impacts of physiological and aphysiological EFs on human bone marrow-derived mesenchymal stem cells (hBM-MSCs) for up to 15 hours. The applied EFs had significant impacts on hBM-MSC morphology and migration; cells displayed varying degrees of conversion to a highly elongated phenotype dependent on the EF strength, consistent perpendicular alignment to the EF vector, and definitive cathodal migration in response to EF strengths ≥0.5 V cm(-1), with the fastest migration speeds observed at between 1.7 and 3 V cm(-1). We observed variability in hBM-MSC donor-to-donor responses and overall tolerances to applied EFs. This study thus confirms hBM-MSCs are responsive to applied EFs, and their rate of migration towards the cathode is controllable depending on the EF strength, providing new insight into the physiology of hBM-MSCs and possibly a significant opportunity for the utilisation of EFs in directed scaffold colonisation in vitro for tissue engineering applications or in vivo post implantation.
Collapse
Affiliation(s)
- T A Banks
- Tissue Engineering and Microfluidics Laboratory, Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, St. Lucia, Qld 4072, Australia.
| | | | | | | |
Collapse
|
31
|
Abstract
Stimulating bone growth and regeneration, especially in patients with delayed union or non-union of bone, is a challenge for orthopaedic surgeons. Treatments employed for bone regeneration are based on the use of cells, biomaterials and factors. Among these therapies, cell treatment with mesenchymal stem cells (MSCs) has a number of advantages as MSCs: (1) are multipotent cells that can migrate to sites of injury; (2) are capable of suppressing the local immune response; and (3) are available in large quantities from the patients themselves. MSC therapies have been used for stimulating bone regeneration in animal models and in patients. Methods of application range from direct MSC injection, seeding MSCs on synthetic scaffolds, the use of gene-modified MSCs, and hetero-MSCs application. However, only a small number of these cell-based strategies are in clinical use, and none of these treatments has become the gold standard treatment for delayed or non-union of bone.
Collapse
Affiliation(s)
- Yunhao Qin
- Shanghai Sixth People's Hospital affiliated to Department of Orthopaedic, Shanghai Jiaotong University, Shanghai, China
| | - Junjie Guan
- Shanghai Sixth People's Hospital affiliated to Department of Orthopaedic, Shanghai Jiaotong University, Shanghai, China
| | - Changqing Zhang
- Shanghai Sixth People's Hospital affiliated to Department of Orthopaedic, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
32
|
Kura T, Akahane M, Shimizu T, Uchihara Y, Tohma Y, Morita Y, Koizumi M, Kawate K, Tanaka Y. Use of Cryopreserved Osteogenic Matrix Cell Sheets for Bone Reconstruction. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/scd.2016.61002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
|
34
|
Kim H, Nam K, Nam JP, Kim HS, Kim YM, Joo WS, Kim SW. VEGF therapeutic gene delivery using dendrimer type bio-reducible polymer into human mesenchymal stem cells (hMSCs). J Control Release 2015; 220:222-228. [DOI: 10.1016/j.jconrel.2015.09.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 09/02/2015] [Accepted: 09/10/2015] [Indexed: 01/10/2023]
|
35
|
Review of various treatment options and potential therapies for osteonecrosis of the femoral head. J Orthop Translat 2015; 4:57-70. [PMID: 30035066 PMCID: PMC5987013 DOI: 10.1016/j.jot.2015.09.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/05/2015] [Accepted: 09/21/2015] [Indexed: 12/20/2022] Open
Abstract
Size and location of the lesion, subchondral collapse occurrence, and articular cartilage involvement are general disease progression criteria for direct osteonecrosis of the femoral head (ONFH) classifications. Treatment options for ONFH are usually based on individual factors and lesion characteristics. Although spontaneous repair of ONFH occurs in some cases, untreated ONFH is unlikely to escape the fate of subchondral collapse and usually ends up with total hip arthroplasty. Operations to preserve the femoral head, e.g., core decompression and bone grafting, are usually recommended in younger patients. They are helpful to relieve pain and improve function in the affected femoral head without subchondral collapse, however, poor prognosis after surgical procedures remains the major problem for ONFH. Pharmacological and physical therapies only work in the early stage of ONFH and have also been recommended as a supplement or prevention treatment for osteonecrosis. Following advances in basic science, many new insights focus on bone tissue engineering to optimize therapies and facilitate prognosis of ONFH. In this review, disease classifications, current treatment options, potential therapies, and the relevant translational barriers are reviewed in the context of clinical application and preclinical exploration, which would provide guidance for preferable treatment options and translation into novel therapies.
Collapse
|
36
|
Corre P, Merceron C, Longis J, Khonsari R, Pilet P, thi TN, Battaglia S, Sourice S, Masson M, Sohier J, Espitalier F, Guicheux J, Weiss P. Direct comparison of current cell-based and cell-free approaches towards the repair of craniofacial bone defects - A preclinical study. Acta Biomater 2015; 26:306-17. [PMID: 26283163 DOI: 10.1016/j.actbio.2015.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/09/2015] [Accepted: 08/13/2015] [Indexed: 12/27/2022]
Abstract
For craniofacial bone defect repair, several alternatives to bone graft (BG) exist, including the combination of biphasic calcium phosphate (BCP) biomaterials with total bone marrow (TBM) and bone marrow-derived mesenchymal stromal cells (MSCs), or the use of growth factors like recombinant human bone morphogenic protein-2 (RhBMP-2) and various scaffolds. Therefore, clinicians might be unsure as to which approach will offer their patients the most benefit. Here, we aimed to compare different clinically relevant bone tissue engineering methods in an "all-in-one" study in rat calvarial defects. TBM, and MSCs committed or not, and cultured in two- or three-dimensions were mixed with BCP and implanted in bilateral parietal bone defects in rats. RhBMP-2 and BG were used as positive controls. After 7 weeks, significant de novo bone formation was observed in rhBMP-2 and BG groups, and in a lesser amount, when BCP biomaterials were mixed with TBM or committed MSCs cultured in three-dimensions. Due to the efficacy and safety of the TBM/BCP combination approach, we recommend this one-step procedure for further clinical investigation. STATEMENT OF SIGNIFICANCE For craniofacial repair, total bone marrow (BM) and BM mesenchymal stem cell (MSC)-based regenerative medicine have shown to be promising in alternative to bone grafting (BG). Therefore, clinicians might be unsure as to which approach will offer the most benefit. Here, BM and MSCs committed or not were mixed with calcium phosphate ceramics (CaP) and implanted in bone defects in rats. RhBMP-2 and BG were used as positive controls. After 7 weeks, significant bone formation was observed in rhBMP-2 and BG groups, and when CaP were mixed with BM or committed MSCs. Since the BM-based procedure does not require bone harvest or cell culture, but provides de novo bone formation, we recommend consideration of this strategy for craniofacial applications.
Collapse
|
37
|
Gamie Z, MacFarlane RJ, Tomkinson A, Moniakis A, Tran GT, Gamie Y, Mantalaris A, Tsiridis E. Skeletal tissue engineering using mesenchymal or embryonic stem cells: clinical and experimental data. Expert Opin Biol Ther 2015; 14:1611-39. [PMID: 25303322 DOI: 10.1517/14712598.2014.945414] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) can be obtained from a wide variety of tissues for bone tissue engineering such as bone marrow, adipose, birth-associated, peripheral blood, periosteum, dental and muscle. MSCs from human fetal bone marrow and embryonic stem cells (ESCs) are also promising cell sources. AREAS COVERED In vitro, in vivo and clinical evidence was collected using MEDLINE® (1950 to January 2014), EMBASE (1980 to January 2014) and Google Scholar (1980 to January 2014) databases. EXPERT OPINION Enhanced results have been found when combining bone marrow-derived mesenchymal stem cells (BMMSCs) with recently developed scaffolds such as glass ceramics and starch-based polymeric scaffolds. Preclinical studies investigating adipose tissue-derived stem cells and umbilical cord tissue-derived stem cells suggest that they are likely to become promising alternatives. Stem cells derived from periosteum and dental tissues such as the periodontal ligament have an osteogenic potential similar to BMMSCs. Stem cells from human fetal bone marrow have demonstrated superior proliferation and osteogenic differentiation than perinatal and postnatal tissues. Despite ethical concerns and potential for teratoma formation, developments have also been made for the use of ESCs in terms of culture and ideal scaffold.
Collapse
Affiliation(s)
- Zakareya Gamie
- Aristotle University Medical School, 'PapaGeorgiou' Hospital, Academic Orthopaedic Unit , Thessaloniki , Greece
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Maxhimer JB, Bradley JP, Lee JC. Signaling pathways in osteogenesis and osteoclastogenesis: Lessons from cranial sutures and applications to regenerative medicine. Genes Dis 2015; 2:57-68. [PMID: 25961069 PMCID: PMC4425620 DOI: 10.1016/j.gendis.2014.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
One of the simplest models for examining the interplay between bone formation and resorption is the junction between the cranial bones. Although only roughly a quarter of patients diagnosed with craniosynostosis have been linked to known genetic disturbances, the molecular mechanisms elucidated from these studies have provided basic knowledge of bone homeostasis. This work has translated to methods and advances in bone tissue engineering. In this review, we examine the current knowledge of cranial suture biology derived from human craniosynostosis syndromes and discuss its application to regenerative medicine.
Collapse
Affiliation(s)
- Justin B. Maxhimer
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
| | - James P. Bradley
- Division of Plastic and Reconstructive Surgery, Temple University/St. Christopher's Hospital for Children, PA, USA
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
- Division of Plastic and Reconstructive Surgery, Greater Los Angeles VA Healthcare System, USA
- Corresponding author. UCLA Division of Plastic and Reconstructive Surgery, 200 UCLA Medical Plaza, Suite 465, Los Angeles, CA 90095, USA. Tel.: +1 310 794 7616; fax: +1 310 206 6833.
| |
Collapse
|
39
|
Bléry P, Corre P, Malard O, Sourice S, Pilet P, Amouriq Y, Guicheux J, Weiss P, Espitalier F. Evaluation of new bone formation in irradiated areas using association of mesenchymal stem cells and total fresh bone marrow mixed with calcium phosphate scaffold. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:2711-2720. [PMID: 25081644 DOI: 10.1007/s10856-014-5282-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 07/16/2014] [Indexed: 06/03/2023]
Abstract
The consequences of the treatment of the squamous cell carcinomas of the upper aerodigestive tract (bone removal and external radiation therapy) are constant. Tissue engineering using biphasic calcium phosphate (BCP) and mesenchymal stem cells (MSC) is considered as a promising alternative. We previously demonstrated the efficacy of BCP and total fresh bone marrow (TBM) in regenerating irradiated bone defect. The aim of this study was to know if adding MSC to BCP + TBM mixture could improve the bone formation in irradiated bone defects. Twenty-four Lewis 1A rats received a single dose of 20 Gy to the hind limbs. MSC were sampled from non-irradiated donors and amplified in proliferative, and a part in osteogenic, medium. 3 weeks after, defects were created on femurs and tibias, which were filled with BCP alone, BCP + TBM, BCP + TBM + uncommitted MSC, or BCP + TBM + committed MSC. 3 weeks after, samples were removed and prepared for qualitative and quantitative analysis. The rate of bone ingrowth was significantly higher after implantation of BCP + TBM mixture. The adding of a high concentration of MSC, committed or not, didn't improve the bone regeneration. The association BCP + TBM remains the most efficient material for bone substitution in irradiated areas.
Collapse
Affiliation(s)
- P Bléry
- INSERM, UMR-S 791, Laboratoire d'Ingénierie Ostéo-Articulaire et Dentaire, LIOAD, 1 Place Alexis Ricordeau, 44042, Nantes Cedex 1, France,
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Spelzini F, Manodoro S, Frigerio M, Nicolini G, Maggioni D, Donzelli E, Altomare L, Farè S, Veneziano F, Avezza F, Tredici G, Milani R. Stem cell augmented mesh materials: an in vitro and in vivo study. Int Urogynecol J 2014; 26:675-83. [PMID: 25416022 DOI: 10.1007/s00192-014-2570-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 11/04/2014] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND HYPOTHESIS To test in vitro and in vivo the capability of mesh materials to act as scaffolds for rat-derived mesenchymal stem cells (rMSCs) and to compare inflammatory response and collagen characteristics of implant materials, either seeded or not with rMSCs. METHODS rMSCs isolated from rat bone marrow were seeded and cultured in vitro on four different implant materials. Implants showing the best rMSC proliferation rate were selected for the in vivo experiment. Forty-eight adult female Sprague-Dawley rats were randomly divided into two treatment groups. The implant of interest-either seeded or not with rMSCs-was laid and fixed over the muscular abdominal wall. Main outcome measures were: in vitro, proliferation of rMSCs on selected materials; in vivo, the occurrence of topical complications, the evaluation of systemic and local inflammatory response and examination of the biomechanical properties of explants. RESULTS Surgisis and Pelvitex displayed the best cell growth in vitro. At 90 days in the rat model, rMSCs were related to a lower count of neutrophil cells for Pelvitex and a greater organisation and collagen amount for Surgisis. At 7 days Surgisis samples seeded with rMSCs displayed higher breaking force and stiffness. CONCLUSIONS The presence of rMSCs reduced the systemic inflammatory response on synthetic implants and improved collagen characteristics at the interface between biological grafts and native tissues. rMSCs enhanced the stripping force on biological explants.
Collapse
Affiliation(s)
- Federico Spelzini
- Department of Obstetrics and Gynaecology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
WANG CHENGGONG, ZHONG DA, ZHOU XING, YIN KE, LIAO QIANDE, KONG LINGYU, LIU ANSONG. Preparation of a new composite combining strengthened β-tricalcium phosphate with platelet-rich plasma as a potential scaffold for the repair of bone defects. Exp Ther Med 2014; 8:1081-1086. [PMID: 25187800 PMCID: PMC4151786 DOI: 10.3892/etm.2014.1912] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/18/2014] [Indexed: 01/25/2023] Open
Abstract
β-tricalcium phosphate (β-TCP) and platelet-rich plasma (PRP) are commonly used in bone tissue engineering. In the present study, a new composite combining strengthened β-TCP and PRP was prepared and its morphological and mechanical properties were investigated by scanning electron microscopy (SEM) and material testing. The biocompatibility was evaluated by measuring the adhesion rate and cytotoxicity of bone marrow stromal cells (BMSCs). The strengthened β-TCP/PRP composite had an appearance like the fungus Boletus kermesinus with the PRP gel distributed on the surface of the micropores. The maximum load and load intensity were 945.6±86.4 N and 13.1±0.5 MPa, which were significantly higher than those of β-TCP (110.1±14.3 N and 1.6±0.2 MPa; P<0.05). The BMSC adhesion rate on the strengthened β-TCP/PRP composite was >96% after 24 h, with a cell cytotoxicity value of zero. SEM micrographs revealed that following seeding of BMSCs onto the composite in high-glucose Dulbecco's modified Eagle's medium culture for two weeks, the cells grew well and exhibited fusiform, spherical and polygonal morphologies, as well as pseudopodial connections. The strengthened β-TCP/PRP composite has the potential to be used as a scaffold in bone tissue engineering due to its effective biocompatibility and mechanical properties.
Collapse
Affiliation(s)
| | - DA ZHONG
- Correspondence to: Dr Da Zhong, Department of Orthopedics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, P.R. China, E-mail:
| | | | | | | | | | | |
Collapse
|
42
|
Li JJ, Kaplan DL, Zreiqat H. Scaffold-based regeneration of skeletal tissues to meet clinical challenges. J Mater Chem B 2014; 2:7272-7306. [PMID: 32261954 DOI: 10.1039/c4tb01073f] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The management and reconstruction of damaged or diseased skeletal tissues have remained a significant global healthcare challenge. The limited efficacy of conventional treatment strategies for large bone, cartilage and osteochondral defects has inspired the development of scaffold-based tissue engineering solutions, with the aim of achieving complete biological and functional restoration of the affected tissue in the presence of a supporting matrix. Nevertheless, significant regulatory hurdles have rendered the clinical translation of novel scaffold designs to be an inefficient process, mainly due to the difficulties of arriving at a simple, reproducible and effective solution that does not rely on the incorporation of cells and/or bioactive molecules. In the context of the current clinical situation and recent research advances, this review will discuss scaffold-based strategies for the regeneration of skeletal tissues, with focus on the contribution of bioactive ceramic scaffolds and silk fibroin, and combinations thereof, towards the development of clinically viable solutions.
Collapse
Affiliation(s)
- Jiao Jiao Li
- Biomaterials and Tissue Engineering Research Unit, School of AMME, University of Sydney, Sydney, NSW 2006, Australia.
| | | | | |
Collapse
|
43
|
Takitoh T, Bessho M, Hirose M, Ohgushi H, Mori H, Hara M. Gamma-cross-linked nonfibrillar collagen gel as a scaffold for osteogenic differentiation of mesenchymal stem cells. J Biosci Bioeng 2014; 119:217-25. [PMID: 25176637 DOI: 10.1016/j.jbiosc.2014.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 02/04/2023]
Abstract
We fabricated a transparent nonfibrillar collagen gel using gamma irradiation (5 kGy) and cultured rat mesenchymal stem cells (MSCs) on both the gamma-irradiated collagen gel and on unirradiated fibrillar collagen gel. Cells attached well and proliferated with high viability on the surface of both gels. The cells cultured on the gamma-irradiated nonfibrillar gel had a unique elongated shape and adhered to each other in culture. After 21 days of culture in dexamethasone-containing culture medium, the contents of bone-specific osteocalcin and calcium on the gamma-irradiated nonfibrillar gel were 1.4 and 1.9 times higher than those on fibrillar collagen gel, respectively. These data show that osteogenic differentiation of MSCs was promoted more efficiently on the gamma-cross-linked nonfibrillar gel than on the fibrillar gel and demonstrate the potential of the gamma-irradiated collagen gel for use in bone tissue engineering.
Collapse
Affiliation(s)
- Takako Takitoh
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Masahiko Bessho
- International Center for Research and Education on Mineral and Energy Resources, Akita University, 1-1 Tegata, Gakuen-machi, Akita, Akita 010-8502, Japan
| | - Motohiro Hirose
- Human Life Technology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Hajime Ohgushi
- Health Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 3-11-46 Nakouji, Amagasaki, Hyogo 661-0974, Japan
| | - Hideki Mori
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Masayuki Hara
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan.
| |
Collapse
|
44
|
Mandibular reconstruction with autologous human bone marrow stem cells and autogenous bone graft in a patient with plexiform ameloblastoma. J Craniofac Surg 2014; 24:e409-11. [PMID: 23851884 DOI: 10.1097/scs.0b013e318292c91d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Ameloblastoma is a histologically benign tumor, but it shows a tendency of locally aggressive behavior. To our knowledge, this is the first report of a successful reconstruction performed for treating a mandibular defect by using autologous human bone marrow mesenchymal stem cells in a patient with plexiform ameloblastoma. In this article, we report the result of the mandibular reconstruction with autologous human bone marrow mesenchymal stem cells and autogenous bone graft, followed by the placement of osteointegrated dental implant and prosthodontic treatment in a patient with plexiform ameloblastoma.
Collapse
|
45
|
Regenerative therapy with mesenchymal stem cells at the site of malignant primary bone tumour resection: what are the risks of early or late local recurrence? INTERNATIONAL ORTHOPAEDICS 2014; 38:1825-35. [PMID: 24906983 DOI: 10.1007/s00264-014-2384-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 12/11/2022]
Abstract
PURPOSE There is concern that regenerative cell-based therapies at the site of malignant primary bone tumours could result in increased risk of local tumour recurrence. We therefore investigated the long-term risks for site-specific recurrences in patients who had received an autologous bone marrow derived mesenchymal stem cell suspension to improve healing at the host-to-allograft bone junction of the reconstruction after bone tumour resection. METHODS A total of 92 patients were treated from 1993 to 2003 with bone marrow-derived mesenchymal stem cells after bone tumour resection. Patients were monitored for cancer incidence from the date of first operation (1993) until death, or until 31 December 2013. The mean follow-up time was 15.4 years (range ten to 20 years). The average number of MSCs returned to the patient was 234,000 MSCs ± 215,000. The primary outcome was to evaluate the risk of tumorigenesis recurrence at the cell therapy treatment sites with radiographs and/or MRIs. The relative risk of cancer recurrence was expressed as the ratio of observed and expected number of cases according to three different control populations. RESULTS Thirteen recurrences were found at the treatment sites among the 92 patients. The expected number of recurrences based on incidence in the three cohort populations was between 15 and 20 for the same cancer, age and sex distribution. The standardized incidence ratio (equal to observed cancers divided by expected cancers) for the entire follow-up period and for all recurrences was between 0.65 and 0.86 (95 % CI 0.60-1.20). CONCLUSION This study found no increased cancer local recurrence risk in patients after application of autologous cell-based therapy using bone marrow-derived mesenchymal stem cells at the treatment site after an average follow-up period of 15.4 years, ranging from ten to 20 years.
Collapse
|
46
|
Watson L, Elliman SJ, Coleman CM. From isolation to implantation: a concise review of mesenchymal stem cell therapy in bone fracture repair. Stem Cell Res Ther 2014; 5:51. [PMID: 25099622 PMCID: PMC4055164 DOI: 10.1186/scrt439] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Compromised bone-regenerating capability following a long bone fracture is often the result of reduced host bone marrow (BM) progenitor cell numbers and efficacy. Without surgical intervention, these malunions result in mobility restrictions, deformities, and disability. The clinical application of BM-derived mesenchymal stem cells (MSCs) is a feasible, minimally invasive therapeutic option to treat non-union fractures. This review focuses on novel, newly identified cell surface markers in both the mouse and human enabling the isolation and purification of osteogenic progenitor cells as well as their direct and indirect contributions to fracture repair upon administration. Furthermore, clinical success to date is summarized with commentary on autologous versus allogeneic cell sources and the methodology of cell administration. Given our clinical success to date in combination with recent advances in the identification, isolation, and mechanism of action of MSCs, there is a significant opportunity to develop improved technologies for defining therapeutic MSCs and potential to critically inform future clinical strategies for MSC-based bone regeneration.
Collapse
|
47
|
Lucarelli E, Bellotti C, Mantelli M, Avanzini MA, Maccario R, Novara F, Arrigo G, Zuffardi O, Zuntini M, Pandolfi M, Sangiorgi L, Lisini D, Donati D, Duchi S. In vitro biosafety profile evaluation of multipotent mesenchymal stem cells derived from the bone marrow of sarcoma patients. J Transl Med 2014; 12:95. [PMID: 24716831 PMCID: PMC4022272 DOI: 10.1186/1479-5876-12-95] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/31/2014] [Indexed: 11/30/2022] Open
Abstract
Background In osteosarcoma (OS) and most Ewing sarcoma (EWS) patients, the primary tumor originates in the bone. Although tumor resection surgery is commonly used to treat these diseases, it frequently leaves massive bone defects that are particularly difficult to be treated. Due to the therapeutic potential of mesenchymal stem cells (MSCs), OS and EWS patients could benefit from an autologous MSCs-based bone reconstruction. However, safety concerns regarding the in vitro expansion of bone marrow-derived MSCs have been raised. To investigate the possible oncogenic potential of MSCs from OS or EWS patients (MSC-SAR) after expansion, this study focused on a biosafety assessment of MSC-SAR obtained after short- and long-term cultivation compared with MSCs from healthy donors (MSC-CTRL). Methods We initially characterized the morphology, immunophenotype, and differentiation multipotency of isolated MSC-SAR. MSC-SAR and MSC-CTRL were subsequently expanded under identical culture conditions. Cells at the early (P3/P4) and late (P10) passages were collected for the in vitro analyses including: sequencing of genes frequently mutated in OS and EWS, evaluation of telomerase activity, assessment of the gene expression profile and activity of major cancer pathways, cytogenetic analysis on synchronous MSCs, and molecular karyotyping using a comparative genomic hybridization (CGH) array. Results MSC-SAR displayed comparable morphology, immunophenotype, proliferation rate, differentiation potential, and telomerase activity to MSC-CTRL. Both cell types displayed signs of senescence in the late stages of culture with no relevant changes in cancer gene expression. However, cytogenetic analysis detected chromosomal anomalies in the early and late stages of MSC-SAR and MSC-CTRL after culture. Conclusions Our results demonstrated that the in vitro expansion of MSCs does not influence or favor malignant transformation since MSC-SAR were not more prone than MSC-CTRL to deleterious changes during culture. However, the presence of chromosomal aberrations supports rigorous phenotypic, functional and genetic evaluation of the biosafety of MSCs, which is important for clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Serena Duchi
- Osteoarticolar Regeneration Laboratory, Rizzoli Orthopaedic Institute, Bologna, Italy.
| |
Collapse
|
48
|
Fekete N, Rojewski MT, Lotfi R, Schrezenmeier H. Essential Components for Ex Vivo Proliferation of Mesenchymal Stromal Cells. Tissue Eng Part C Methods 2014; 20:129-39. [DOI: 10.1089/ten.tec.2013.0061] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Natalie Fekete
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, University Hospital Ulm, Ulm, Germany
| | - Markus Thomas Rojewski
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, University Hospital Ulm, Ulm, Germany
| | - Ramin Lotfi
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, University Hospital Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
49
|
Rosset P, Deschaseaux F, Layrolle P. Cell therapy for bone repair. Orthop Traumatol Surg Res 2014; 100:S107-12. [PMID: 24411717 DOI: 10.1016/j.otsr.2013.11.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 11/08/2013] [Accepted: 11/15/2013] [Indexed: 02/07/2023]
Abstract
When natural bone repair mechanisms fail, autologous bone grafting is the current standard of care. The osteogenic cells and bone matrix in the graft provide the osteo-inductive and osteo-conductive properties required for successful bone repair. Bone marrow (BM) mesenchymal stem cells (MSCs) can differentiate into osteogenic cells. MSC-based cell therapy holds promise for promoting bone repair. The amount of MSCs available from iliac-crest aspirates is too small to be clinically useful, and either concentration or culture must therefore be used to expand the MSC population. MSCs can be administered alone via percutaneous injection or implanted during open surgery with a biomaterial, usually biphasic hydroxyapatite/β-calcium-triphosphate granules. Encouraging preliminary results have been obtained in patients with delayed healing of long bone fractures or avascular necrosis of the femoral head. Bone tissue engineering involves in vitro MSC culturing on biomaterials to obtain colonisation of the biomaterial and differentiation of the cells. The biomaterial-cell construct is then implanted into the zone to be treated. Few published data are available on bone tissue engineering. Much work remains to be done before determining whether this method is suitable for the routine filling of bone tissue defects. Increasing cell survival and promoting implant vascularisation are major challenges. Improved expertise with culturing techniques, together with the incorporation of regulatory requirements, will open the way to high-quality clinical trials investigating the usefulness of cell therapy as a method for achieving bone repair. Cell therapy avoids the drawbacks of autologous bone grafting, preserving the bone stock and diminishing treatment invasiveness.
Collapse
Affiliation(s)
- P Rosset
- Service de chirurgie orthopédique 2, hôpital Trousseau, Université François-Rabelais de Tours, CHU de Tours, 37044 Tours cedex 09, France; Inserm U957, Laboratoire de physiopathologie de la résorption osseuse et thérapie des tumeurs osseuses primitives (LPRO), Faculté de Médecine, Université de Nantes, Nantes, France.
| | - F Deschaseaux
- StromaLab, UMR CNRS 5273, U1031 Inserm, Établissement Français du Sang Pyrénées-Méditerranée, Université P.-Sabatier, Toulouse, France
| | - P Layrolle
- Inserm U957, Laboratoire de physiopathologie de la résorption osseuse et thérapie des tumeurs osseuses primitives (LPRO), Faculté de Médecine, Université de Nantes, Nantes, France
| |
Collapse
|
50
|
Hodgkinson T, Yuan XF, Bayat A. Adult stem cells in tissue engineering. Expert Rev Med Devices 2014; 6:621-40. [DOI: 10.1586/erd.09.48] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|