1
|
Lázár I, Čelko L, Menelaou M. Aerogel-Based Materials in Bone and Cartilage Tissue Engineering-A Review with Future Implications. Gels 2023; 9:746. [PMID: 37754427 PMCID: PMC10530393 DOI: 10.3390/gels9090746] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Aerogels are fascinating solid materials known for their highly porous nanostructure and exceptional physical, chemical, and mechanical properties. They show great promise in various technological and biomedical applications, including tissue engineering, and bone and cartilage substitution. To evaluate the bioactivity of bone substitutes, researchers typically conduct in vitro tests using simulated body fluids and specific cell lines, while in vivo testing involves the study of materials in different animal species. In this context, our primary focus is to investigate the applications of different types of aerogels, considering their specific materials, microstructure, and porosity in the field of bone and cartilage tissue engineering. From clinically approved materials to experimental aerogels, we present a comprehensive list and summary of various aerogel building blocks and their biological activities. Additionally, we explore how the complexity of aerogel scaffolds influences their in vivo performance, ranging from simple single-component or hybrid aerogels to more intricate and organized structures. We also discuss commonly used formulation and drying methods in aerogel chemistry, including molding, freeze casting, supercritical foaming, freeze drying, subcritical, and supercritical drying techniques. These techniques play a crucial role in shaping aerogels for specific applications. Alongside the progress made, we acknowledge the challenges ahead and assess the near and far future of aerogel-based hard tissue engineering materials, as well as their potential connection with emerging healing techniques.
Collapse
Affiliation(s)
- István Lázár
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, 4032 Debrecen, Hungary
| | - Ladislav Čelko
- Central European Institute of Technology, Brno University of Technology, Purkynova 656/123, 612 00 Brno, Czech Republic;
| | - Melita Menelaou
- Department of Chemical Engineering, Cyprus University of Technology, 30 Arch. Kyprianos Str., Limassol 3036, Cyprus
| |
Collapse
|
2
|
Morimoto S, Kajiya M, Yoshii H, Yoshino M, Horikoshi S, Motoike S, Iwata T, Ouhara K, Ando T, Yoshimoto T, Shintani T, Mizuno N. A Cartilaginous Construct with Bone Collar Exerts Bone-Regenerative Property Via Rapid Endochondral Ossification. Stem Cell Rev Rep 2023; 19:1812-1827. [PMID: 37166558 DOI: 10.1007/s12015-023-10554-w] [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] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
Three-dimensional clumps of mesenchymal stem cells (MSCs)/extracellular matrix (ECM) complexes (C-MSCs) can be implanted into tissue defects with no artificial scaffolds. In addition, the cellular properties and characteristics of the ECM in C-MSCs can be regulated in vitro. Most bone formation in the developmental and healing process is due to endochondral ossification, which occurs after bone collar formation surrounding cartilage derived from MSCs. Thus, to develop a rapid and reliable bone-regenerative cell therapy, the present study aimed to generate cartilaginous tissue covered with a mineralized bone collar-like structure from human C-MSCs by combining chondrogenic and osteogenic induction. Human bone marrow-derived MSCs were cultured in xeno-free/serum-free (XF) growth medium. Confluent cells that formed cellular sheets were detached from the culture plate using a micropipette tip. The floating cellular sheet contracted to round clumps of cells (C-MSCs). C-MSCs were maintained in XF-chondro-inductive medium (CIM) and XF-osteo-inductive medium (OIM). The biological and bone-regenerative properties of the generated cellular constructs were assessed in vitro and in vivo. C-MSCs cultured in CIM/OIM formed cartilaginous tissue covered with a mineralized matrix layer, whereas CIM treatment alone induced cartilage with no mineralization. Transplantation of the cartilaginous tissue covered with a mineralized matrix induced more rapid bone reconstruction via endochondral ossification in the severe combined immunodeficiency mouse calvarial defect model than that of cartilage generated using only CIM. These results highlight the potential of C-MSC culture in combination with CIM/OIM to generate cartilage covered with a bone collar-like structure, which can be applied for novel bone-regenerative cell therapy.
Collapse
Affiliation(s)
- Shin Morimoto
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
- Department of Innovation and Precision Dentistry, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
| | - Hiroki Yoshii
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Mai Yoshino
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Susumu Horikoshi
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Souta Motoike
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Toshinori Ando
- Department of Innovation and Precision Dentistry, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Tetsuya Yoshimoto
- Department of Innovation and Precision Dentistry, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Tomoaki Shintani
- Department of Innovation and Precision Dentistry, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
3
|
Alfayez E, Veschini L, Dettin M, Zamuner A, Gaetani M, Carreca AP, Najman S, Ghanaati S, Coward T, Di Silvio L. DAR 16-II Primes Endothelial Cells for Angiogenesis Improving Bone Ingrowth in 3D-Printed BCP Scaffolds and Regeneration of Critically Sized Bone Defects. Biomolecules 2022; 12:biom12111619. [PMID: 36358970 PMCID: PMC9687468 DOI: 10.3390/biom12111619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Bone is a highly vascularized tissue and relies on the angiogenesis and response of cells in the immediate environmental niche at the defect site for regeneration. Hence, the ability to control angiogenesis and cellular responses during osteogenesis has important implications in tissue-engineered strategies. Self-assembling ionic-complementary peptides have received much interest as they mimic the natural extracellular matrix. Three-dimensional (3D)-printed biphasic calcium phosphate (BCP) scaffolds coated with self-assembling DAR 16-II peptide provide a support template with the ability to recruit and enhance the adhesion of cells. In vitro studies demonstrated prompt the adhesion of both human umbilical vein endothelial cells (HUVEC) and human mesenchymal stem cells (hMSC), favoring endothelial cell activation toward an angiogenic phenotype. The SEM-EDS and protein micro bicinchoninic acid (BCA) assays demonstrated the efficacy of the coating. Whole proteomic analysis of DAR 16-II-treated HUVECs demonstrated the upregulation of proteins involved in cell adhesion (HABP2), migration (AMOTL1), cytoskeletal re-arrangement (SHC1, TMOD2), immuno-modulation (AMBP, MIF), and morphogenesis (COL4A1). In vivo studies using DAR-16-II-coated scaffolds provided an architectural template, promoting cell colonization, osteogenesis, and angiogenesis. In conclusion, DAR 16-II acts as a proactive angiogenic factor when adsorbed onto BCP scaffolds and provides a simple and effective functionalization step to facilitate the translation of tailored 3D-printed BCP scaffolds for clinical applications.
Collapse
Affiliation(s)
- Eman Alfayez
- Faculty of Dentistry, Oral Biology Department, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Lorenzo Veschini
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, 35131 Padua, Italy
| | - Annj Zamuner
- Department of Civil, Environmental, and Architectural Engineering, University of Padua, 35131 Padua, Italy
| | - Massimiliano Gaetani
- Fondazione Ricerca nel Mediterraneo (Ri.MED) and Department of Laboratory Medicine and Advanced Biotechnologies, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, 90100 Palermo, Italy
- Chemical Proteomics, Department of Medical Biochemistry and Biophysics, Karolinska Institutet and SciLifeLab (Science for Life Laboratory), SE-17 177 Stockholm, Sweden
| | - Anna P. Carreca
- Fondazione Ricerca nel Mediterraneo (Ri.MED) and Department of Laboratory Medicine and Advanced Biotechnologies, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, 90100 Palermo, Italy
| | - Stevo Najman
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Shahram Ghanaati
- Department for Oral, Cranio-Maxillofacial and Facial Plastic Surgery, Medical Center of the Goethe University, 60323 Frankfurt, Germany
| | - Trevor Coward
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
| | - Lucy Di Silvio
- Faculty of Dentistry, Oral & Craniofacial Sciences King’s College London, London SE1 9RT, UK
- Correspondence: ; Tel.: +44-02-07848-8475
| |
Collapse
|
4
|
Ogawa T, Kajiya M, Horikoshi S, Yoshii H, Yoshino M, Motoike S, Morimoto S, Sone H, Iwata T, Ouhara K, Matsuda S, Mizuno N. Xenotransplantation of cryopreserved human clumps of mesenchymal stem cells/extracellular matrix complexes pretreated with IFN-γ induces rat calvarial bone regeneration. Regen Ther 2022; 20:117-125. [PMID: 35582709 PMCID: PMC9065482 DOI: 10.1016/j.reth.2022.04.003] [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: 01/04/2022] [Revised: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/10/2022] Open
Abstract
Introduction Three-dimensional (3D) clumps of mesenchymal stem cells (MSCs)/extracellular matrix (ECM) complexes, composed with cells and self-produced intact ECM, can be grafted into defect areas without artificial scaffold to induce successful bone regeneration. Moreover, C-MSCs pretreated with IFN-γ (C-MSCsγ) increased the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO) expression and thereby inhibited T cell activity. Xenotransplantation of human C-MSCsγ suppressed host T cell immune rejection and induced bone regeneration in mice. Besides, we have also reported that C-MSCs retain the 3D structure and bone regenerative property even after cryopreservation. To develop the "off-the-shelf" cell preparation for bone regenerative therapy that is promptly provided when needed, we investigated whether C-MSCsγ can retain the immunosuppressive and osteogenic properties after cryopreservation. Methods Confluent human MSCs that had formed on the cellular sheet were scratched using a micropipette tip and then torn off. The sheet was rolled to make a round clump of cells. The round cell clumps were incubated with a growth medium for 3 days, and then C-MSCs were obtained. To generate C-MSCsγ, after 2 days' culture, C-MSCs were stimulated with 50 ng/ml of IFN-γ. Both C-MSCs and C-MSCsγ were cryopreserved for 2 days and then thawed to obtain Cryo-C-MSCs and Cryo-C-MSCsγ, respectively. The biological properties of those cell clumps were assessed in vitro. In addition, to test whether human Cryo-C-MSCsγ attenuates immune rejection to induce bone regeneration, a xenograft study using a rat calvarial defect was performed. Results Both IFN-γ pretreatment and cryopreservation process did not affect the 3D structure and cell viability in all human cell clumps. Interestingly, Cryo-C-MSCsγ showed significantly increased IDO mRNA expression equivalent to C-MSCsγ. More importantly, xenotransplantation of human C-MSCsγ and Cryo-C-MSCsγ induced rat calvarial bone regeneration by suppressing rat T cells infiltration and the grafted human cells reduction in the grafted area. Finally, there were no human donor cells in the newly formed bone, implying that the bone reconstruction by C-MSCsγ and Cryo-C-MSCsγ can be due to indirect host osteogenesis. Conclusion These findings implied that Cryo-C-MSCsγ can be a promising bone regenerative allograft therapy that can be certainly and promptly supplied on demand.
Collapse
Affiliation(s)
- Tomoya Ogawa
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Susumu Horikoshi
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroki Yoshii
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mai Yoshino
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Souta Motoike
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Shin Morimoto
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hisakatsu Sone
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinji Matsuda
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
5
|
Nguyen HTN, Vu NB. A Simple Method to Produce Engineered Cartilage from Human Adipose-Derived Mesenchymal Stem Cells and Poly ε-Caprolactone Scaffolds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021:181-191. [PMID: 34739719 DOI: 10.1007/5584_2021_669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The damaged articular cartilage has limited self-regeneration capacity because of the absence of blood vessels, lymphatics, and nerves. Cartilage transplantation is, hence, a popular method used to treat this disease. However, sources of autograft and allogenic cartilage for transplantation are limited. Therefore, this study aims to suggest a simple method to produce engineered cartilage from human adipose-derived mesenchymal stem cells (ADSCs) and poly (ε-caprolactone) (PCL) scaffolds. METHODS ADSCs were isolated and expanded from fat tissues according to published protocols. PCL-porous scaffolds were produced from PCL with 5 × 5 × 0.6 mm3 with 200-400 μ m pore sizes. ADSCs were seeded on the PCL scaffolds at three different densities (104, 105, 106 cells per scaffold). The adherence of ADSCs on the surface of PCL scaffolds was evaluated based on an immunostaining assay to determine the presence of ADSCs. The cell proliferation on PCL scaffolds was determined by MTT assay. The complexity in ADSCs and PCL scaffolds was induced to cartilage using a chondrogenesis medium. The engineered cartilage was characterized by the accumulation of proteoglycan and aggrecan by Safranin O staining assay. Their structures were evaluated using an H-E staining assay. Finally, these engineered cartilage tissues were transplanted into mice to assess cartilage maturation when compared to natural cartilage. RESULTS The results showed that the engineered cartilage tissues could be successfully produced by cultures of ADSCs on poly ε-caprolactone scaffolds in combination with chondrogenesis medium. The suitable density of ADSCs was 106 cells/per scaffold of 5 × 6 × 0.6 mm3 with pore size from 200 to 400 μ m. CONCLUSION The results showed that an in vitro cartilage tissue was created from ADSCs and PCL scaffold. The cartilage tissue exists in the mice for 6 months.
Collapse
Affiliation(s)
- Hue Thi-Ngoc Nguyen
- Stem Cell Institute, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ngoc Bich Vu
- Stem Cell Institute, University of Science, Ho Chi Minh City, Vietnam.
- Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| |
Collapse
|
6
|
Wang G, Yan J, Zhang H, Massey P, Alexander JS, Kevil CG, Barton S, Dong Y. Transient activation of notch signaling enhances endogenous stromal cell expansion and subsequent bone defect repair. J Orthop Translat 2021; 31:26-32. [PMID: 34760622 PMCID: PMC8554104 DOI: 10.1016/j.jot.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Following traumatic bone loss or removal of bone tumors, the failure of bone allograft transplantation for large bone defect repair remains a significant problem in orthopedics. Therefore, new strategies that can efficiently enhance allograft healing and long-term incorporation are critically needed. METHOD In this study, we first injected Notch-activating Jagged1 peptide to mice and then isolated bone marrow tissues and cells for proliferation and differentiation assays. Femur bone allograft surgery was also performed in Jagged1 pre-treated mice, and bone defect healing process were monitored by histology, Micro-CT and biomechanical testing. RESULT Our results showed that Jagged1 therapeutic injection is sufficient to maximally activate Notch and promote bone marrow stromal cell proliferation in vivo, while no effects on bone structure were observed. More importantly, Jagged1 pre-treatment significantly promoted bone callus formation and increased bone mechanical strength during allograft healing in a femur bone defect mouse model. CONCLUSION This study reveals that Notch in vivo activation can be induced by injection of Jagged1 peptide for expansion of local native stromal cells that will significantly enhance bone callus formation. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE The clinical uses of this therapeutic strategy would be immediately applicable for chronic long bone defect repair. More importantly, this devised strategy for expansion of endogenous BMSCs can also be applied to enhance other tissue and organ repair.
Collapse
Affiliation(s)
- Guangxi Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Heilongjiang, China
- Department of Orthopedic Surgery, LSU Health Sciences Center, Shreveport, LA, USA
| | - Jinglong Yan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Harbin Medical University, Heilongjiang, China
| | - Hao Zhang
- Department of Orthopedic Surgery, LSU Health Sciences Center, Shreveport, LA, USA
| | - Patrick Massey
- Department of Orthopedic Surgery, LSU Health Sciences Center, Shreveport, LA, USA
| | - J. Steven Alexander
- Department of Molecular & Cellular Physiology, Medicine and Neurology, LSU Health Sciences Center, Shreveport, LA, USA
| | | | - Shane Barton
- Department of Orthopedic Surgery, LSU Health Sciences Center, Shreveport, LA, USA
| | - Yufeng Dong
- Department of Orthopedic Surgery, LSU Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
7
|
Clumps of Mesenchymal Stem Cells/Extracellular Matrix Complexes Generated with Xeno-Free Chondro-Inductive Medium Induce Bone Regeneration via Endochondral Ossification. Biomedicines 2021; 9:biomedicines9101408. [PMID: 34680525 PMCID: PMC8533314 DOI: 10.3390/biomedicines9101408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/04/2021] [Accepted: 09/28/2021] [Indexed: 01/14/2023] Open
Abstract
Three-dimensional clumps of mesenchymal stem cells (MSCs)/extracellular matrix (ECM) complexes (C-MSCs) can be transplanted into tissue defect site with no artificial scaffold. Importantly, most bone formation in the developing process or fracture healing proceeds via endochondral ossification. Accordingly, this present study investigated whether C-MSCs generated with chondro-inductive medium (CIM) can induce successful bone regeneration and assessed its healing process. Human bone marrow-derived MSCs were cultured with xeno-free/serum-free (XF) growth medium. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and then torn off. The sheet was rolled to make a round clump of cells. The cell clumps, i.e., C-MSCs, were maintained in XF-CIM. C-MSCs generated with XF-CIM showed enlarged round cells, cartilage matrix, and hypertrophic chondrocytes genes elevation in vitro. Transplantation of C-MSCs generated with XF-CIM induced successful bone regeneration in the SCID mouse calvaria defect model. Immunofluorescence staining for human-specific vimentin demonstrated that donor human and host mouse cells cooperatively contributed the bone formation. Besides, the replacement of the cartilage matrix into bone was observed in the early period. These findings suggested that cartilaginous C-MSCs generated with XF-CIM can induce bone regeneration via endochondral ossification.
Collapse
|
8
|
Rottensteiner-Brandl U, Bertram U, Lingens LF, Köhn K, Distel L, Fey T, Körner C, Horch RE, Arkudas A. Free Transplantation of a Tissue Engineered Bone Graft into an Irradiated, Critical-Size Femoral Defect in Rats. Cells 2021; 10:cells10092256. [PMID: 34571907 PMCID: PMC8467400 DOI: 10.3390/cells10092256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/09/2023] Open
Abstract
Healing of large bone defects remains a challenge in reconstructive surgery, especially with impaired healing potential due to severe trauma, infection or irradiation. In vivo studies are often performed in healthy animals, which might not accurately reflect the situation in clinical cases. In the present study, we successfully combined a critical-sized femoral defect model with an ionizing radiation protocol in rats. To support bone healing, tissue-engineered constructs were transferred into the defect after ectopic preossification and prevascularization. The combination of SiHA, MSCs and BMP-2 resulted in the significant ectopic formation of bone tissue, which can easily be transferred by means of our custom-made titanium chamber. Implanted osteogenic MSCs survived in vivo for a total of 18 weeks. The use of SiHA alone did not lead to bone formation after ectopic implantation. Analysis of gene expression showed early osteoblast differentiation and a hypoxic and inflammatory environment in implanted constructs. Irradiation led to impaired bone healing, decreased vascularization and lower short-term survival of implanted cells. We conclude that our model is highly valuable for the investigation of bone healing and tissue engineering in pre-damaged tissue and that healing of bone defects can be substantially supported by combining SiHA, MSCs and BMP-2.
Collapse
Affiliation(s)
- Ulrike Rottensteiner-Brandl
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
- Emil-Fischer Zentrum, Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ulf Bertram
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
- Department of Neurosurgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Lara F. Lingens
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
- Hand Surgery—Burn Center, Department of Plastic Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Katrin Köhn
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
| | - Luitpold Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Tobias Fey
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Frontier Research Institute for Materials Science, Nagoya Institute of Technology, Nagoya 466-8555, Japan
| | - Carolin Körner
- Department of Materials Science and Engineering, Institute of Science and Technology of Metals, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (U.R.-B.); (U.B.); (L.F.L.); (K.K.); (R.E.H.)
- Correspondence: ; Tel.: +49-9131-8533277
| |
Collapse
|
9
|
Zhao L, Zhao J, Tuo Z, Ren G. Repair of long bone defects of large size using a tissue-engineered periosteum in a rabbit model. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:105. [PMID: 34420103 PMCID: PMC8380237 DOI: 10.1007/s10856-021-06579-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 05/31/2021] [Indexed: 05/22/2023]
Abstract
Tissue engineering is a promising approach for bone regeneration. In this study, we aimed to investigate whether tissue engineered periosteum (TEP), which was fabricated by combining osteogenically-induced mesenchymal stem cells (MSCs) with porcine small intestinal submucosa (SIS), could restore long bone defects of large size in rabbits. Twenty-four adult New Zealand white rabbits (NZWRs) were used in the experiments. Long bone defects of large size (30 mm-50 mm; average, 40 mm) were established on both sides of NZWRs' radii. The defects were treated with TEP (Group A), allogeneic deproteinized bone (DPB, Group B), TEP combined with DPB (Group C), and pure SIS (Group D). The healing outcome was evaluated by radiography and histological examination at 4, 8, and 12 weeks post-treatment. The radiographical findings showed that bone defects of large size were all repaired in Groups A, B and C within 12 weeks, whereas Group D (pure SIS group) failed to result in defect healing at 4, 8, and 12 weeks. Although there was some new bone regeneration connecting the allografts and bone ends, as observed under radiographical and histological observations, bone defects of large sizes were restored primarily by structurally allografted DPB within 12 weeks. The TEP groups (Groups A and C) showed partial or total bone regeneration upon histological inspection. Based on 12-week histological examinations, significantly more bone was formed in Group A than Group C (P < 0.05), and both groups formed significantly more bone than in Groups B and D. The results indicated that long bone defects of a large size could be restored by TEP or TEP combined with the DPB scaffold, and such materials provide an alternative approach to resolving pathological bone defects in clinical settings.
Collapse
Affiliation(s)
- Lin Zhao
- Orthopedic Department of Guangming Traditional Chinese Medicine Hospital of Pudong New Area, Shanghai, China.
| | - Junli Zhao
- Department of Nephrology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, Shanghai, China
| | - Zhenhe Tuo
- Orthopaedic Department of Xianyang Central Hospital, Shaanxi Province, People's Republic of China
| | - Guangtie Ren
- Orthopaedic Department of Hanzhong Central Hospital, Shaanxi Province, People's Republic of China
| |
Collapse
|
10
|
Vascularization Strategies in Bone Tissue Engineering. Cells 2021; 10:cells10071749. [PMID: 34359919 PMCID: PMC8306064 DOI: 10.3390/cells10071749] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Bone is a highly vascularized tissue, and its development, maturation, remodeling, and regeneration are dependent on a tight regulation of blood vessel supply. This condition also has to be taken into consideration in the context of the development of artificial tissue substitutes. In classic tissue engineering, bone-forming cells such as primary osteoblasts or mesenchymal stem cells are introduced into suitable scaffolds and implanted in order to treat critical-size bone defects. However, such tissue substitutes are initially avascular. Because of the occurrence of hypoxic conditions, especially in larger tissue substitutes, this leads to the death of the implanted cells. Therefore, it is necessary to devise vascularization strategies aiming at fast and efficient vascularization of implanted artificial tissues. In this review article, we present and discuss the current vascularization strategies in bone tissue engineering. These are based on the use of angiogenic growth factors, the co-implantation of blood vessel forming cells, the ex vivo microfabrication of blood vessels by means of bioprinting, and surgical methods for creating surgically transferable composite tissues.
Collapse
|
11
|
Molecular Imprinting Strategies for Tissue Engineering Applications: A Review. Polymers (Basel) 2021; 13:polym13040548. [PMID: 33673361 PMCID: PMC7918123 DOI: 10.3390/polym13040548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/29/2022] Open
Abstract
Tissue Engineering (TE) represents a promising solution to fabricate engineered constructs able to restore tissue damage after implantation. In the classic TE approach, biomaterials are used alongside growth factors to create a scaffolding structure that supports cells during the construct maturation. A current challenge in TE is the creation of engineered constructs able to mimic the complex microenvironment found in the natural tissue, so as to promote and guide cell migration, proliferation, and differentiation. In this context, the introduction inside the scaffold of molecularly imprinted polymers (MIPs)—synthetic receptors able to reversibly bind to biomolecules—holds great promise to enhance the scaffold-cell interaction. In this review, we analyze the main strategies that have been used for MIP design and fabrication with a particular focus on biomedical research. Furthermore, to highlight the potential of MIPs for scaffold-based TE, we present recent examples on how MIPs have been used in TE to introduce biophysical cues as well as for drug delivery and sequestering.
Collapse
|
12
|
Acri TM, Laird NZ, Jaidev LR, Meyerholz DK, Salem AK, Shin K. Nonviral Gene Delivery Embedded in Biomimetically Mineralized Matrices for Bone Tissue Engineering. Tissue Eng Part A 2020; 27:1074-1083. [PMID: 33086991 DOI: 10.1089/ten.tea.2020.0206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Research in bone tissue engineering aims to design materials that are effective at generating bone without causing significant side effects. The osteogenic potential of combining matrices and protein growth factors has been well documented, however, improvements are necessary to achieve optimal therapeutic benefits upon clinical translation. In this article, rat calvarial defects were treated with gene-activated matrices (GAMs). The GAMs used were collagen sponges mineralized with a simulated body fluid (SBF) containing a nonviral gene delivery system. Both in vitro and in vivo studies were performed to determine the optimal mode of gene delivery. After 6 weeks, the defects were extracted to assess bone formation and tissue quality through histological and microcomputed tomography analyses. The optimal GAM consisted of a collagen sponge with polyethylenimine plasmid DNA (PEI-pDNA) complexes embedded in a calcium phosphate coating produced by SBF, which increased total bone formation by 39% compared with 19% for control samples. A follow-up in vivo study was performed to optimize the ratio of growth factors included in the GAM. The optimal ratio for supporting bone formation after 6 weeks of implantation was five parts of pBMP-2 to three parts pFGF-2. These studies demonstrated that collagen matrices biomimetically mineralized and activated with plasmids encoding fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) can optimally improve bone regeneration outcomes. Impact statement Bone tissue engineering has explored both nonviral gene delivery and the concept of biomimetic mineralization. In this study, we combined these two concepts to further enhance bone regeneration outcomes. We demonstrated that embedding polyethylenimine (PEI)-based gene delivery within a mineral layer formed from simulated body fluid (SBF) immersion can increase bone formation rates. We also demonstrated that the ratio of growth factors utilized for matrix fabrication can impact the amount of bone formed in the defect site. This research highlights a combined approach using SBF and nonviral gene delivery both in vitro and in vivo and prepares the way for future optimization of synthetic gene activated matrices.
Collapse
Affiliation(s)
- Timothy M Acri
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Noah Z Laird
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Leela R Jaidev
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa College of Pharmacy, Iowa City, Iowa, USA
| | - Kyungsup Shin
- Department of Orthodontics, University of Iowa College of Dentistry and Dental Clinics, Iowa City, Iowa, USA
| |
Collapse
|
13
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|
14
|
Papadogiannis F, Batsali A, Klontzas ME, Karabela M, Georgopoulou A, Mantalaris A, Zafeiropoulos NE, Chatzinikolaidou M, Pontikoglou C. Osteogenic differentiation of bone marrow mesenchymal stem cells on chitosan/gelatin scaffolds: gene expression profile and mechanical analysis. ACTA ACUST UNITED AC 2020; 15:064101. [PMID: 32629436 DOI: 10.1088/1748-605x/aba325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the present study we explore the extracellular matrix (ECM) produced by human bone marrow mesenchymal stem/stromal cells (BM-MSCs) induced to undergo osteogenic differentiation within porous chitosan/gelatin (CS:Gel) scaffolds by investigating their multiple gene expression profile and mechanical behavior. Initially, the efficiency of the BM-MSCs osteogenic differentiation within the constructs was confirmed by the significant rise in the expression of the osteogenesis associated genes DLX5, RUNX2, ALP and OSC. In line with these findings, OSC and Col1A1 protein expression was also detected in BM-MSCs on the CS:Gel scaffolds at day 14 of osteogenic differentiation. We then profiled, for the first time, the expression of 84 cell adhesion and ECM molecules using PCR arrays. The arrays, which were conducted at day 14 of osteogenic differentiation, demonstrated that 49 genes including collagens, integrins, laminins, ECM proteases, catenins, thrombospondins, ECM protease inhibitors and cell-cell adhesion molecules were differentially expressed in BM-MSCs seeded on scaffolds compared to tissue culture polystyrene control. Moreover, we performed dynamic mechanical analysis of the cell-loaded scaffolds on days 0, 7 and 14 to investigate the correlation between the biological results and the mechanical behavior of the constructs. Our data demonstrate a significant increase in the stiffness of the constructs with storage modulus values of 2 MPa on day 7, compared to 0.5 MPa on day 0, following a drop of the stiffness at 0.8 MPa on day 14, that may be attributed to the significant increase of specific ECM protease gene expression such as MMP1, MMP9, MMP11 and MMP16 at this time period.
Collapse
Affiliation(s)
- Fotios Papadogiannis
- Hemopoiesis Research Laboratory, School of Medicine, University of Crete, Heraklion, Greece. Department of Materials Science and Technology, University of Crete, Heraklion, Greece. All authors contributed equally to this work
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Esposito A, Wang L, Li T, Miranda M, Spagnoli A. Role of Prx1-expressing skeletal cells and Prx1-expression in fracture repair. Bone 2020; 139:115521. [PMID: 32629173 PMCID: PMC7484205 DOI: 10.1016/j.bone.2020.115521] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
The healing capacity of bones after fracture implies the existence of adult regenerative cells. However, information on identification and functional role of fracture-induced progenitors is still lacking. Paired-related homeobox 1 (Prx1) is expressed during skeletogenesis. We hypothesize that fracture recapitulates Prx1's expression, and Prx1 expressing cells are critical to induce repair. To address our hypothesis, we used a combination of in vivo and in vitro approaches, short and long-term cell tracking analyses of progenies and actively expressing cells, cell ablation studies, and rodent animal models for normal and defective fracture healing. We found that fracture elicits a periosteal and endosteal response of perivascular Prx1+ cells that participate in fracture healing and showed that Prx1-expressing cells have a functional role in the repair process. While Prx1-derived cells contribute to the callus, Prx1's expression decreases concurrently with differentiation into cartilaginous and bone cells, similarly to when Prx1+ cells are cultured in differentiating conditions. We determined that bone morphogenic protein 2 (BMP2), through C-X-C motif-ligand-12 (CXCL12) signaling, modulates the downregulation of Prx1. We demonstrated that fracture elicits an early increase in BMP2 expression, followed by a decrease in CXCL12 that in turn down-regulates Prx1, allowing cells to commit to osteochondrogenesis. In vivo and in vitro treatment with CXCR4 antagonist AMD3100 restored Prx1 expression by modulating the BMP2-CXCL12 axis. Our studies represent a shift in the current research that has primarily focused on the identification of markers for postnatal skeletal progenitors, and instead we characterized the function of a specific population (Prx1+ cells) and their expression marker (Prx1) as a crossroad in fracture repair. The identification of fracture-induced perivascular Prx1+ cells and regulation of Prx1's expression by BMP2 and in turn by CXCL12 in the orchestration of fracture repair, highlights a pathway in which to investigate defective mechanisms and therapeutic targets for fracture non-union.
Collapse
Affiliation(s)
- Alessandra Esposito
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Tieshi Li
- Department of Pediatrics, University of Nebraska Medical Center, Children's Hospital & Medical Center, Omaha, NE, USA
| | - Mariana Miranda
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Anna Spagnoli
- Department of Orthopaedic Surgery, Section of Molecular Medicine, Rush University Medical Center, Chicago, IL, USA; Department of Pediatrics, Division of Pediatric Endocrinology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
16
|
Babilotte J, Martin B, Guduric V, Bareille R, Agniel R, Roques S, Héroguez V, Dussauze M, Gaudon M, Le Nihouannen D, Catros S. Development and characterization of a PLGA-HA composite material to fabricate 3D-printed scaffolds for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111334. [PMID: 33254966 DOI: 10.1016/j.msec.2020.111334] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 02/03/2023]
Abstract
Additive manufacturing is a rising field in bone tissue engineering. Additive fabrication offers reproducibility, high precision and rapid manufacture of custom patient-specific scaffolds. The development of appropriate composite materials for biomedical applications is critical to reach clinical application of these novel biomaterials. In this work, medical grade poly(lactic-co-glycolic) acid (PLGA) was mixed with hydroxyapatite nanoparticles (nHA) to fabricate 3D porous scaffolds by Fused Deposition Modeling. We have first confirmed that the composite material could be printed in a reproductive manner. Physical characterization demonstrated a low degradation of the material during manufacturing steps and an expected loading and homogeneous distribution of nHA. In vitro biodegradation of the scaffolds showed modifications of morphological and physicochemical properties over time. The composite scaffolds were biocompatible and high cell viability was observed in vitro, as well as a maintain of cell proliferation. As expected, the addition of nHA displayed a positive impact on osteodifferentiation in vitro. Furthermore, a limited inflammatory reaction was observed after subcutaneous implantation of the materials in the rat. Overall, this study suggests that this composite material is suitable for bone tissue engineering applications.
Collapse
Affiliation(s)
| | - Benoit Martin
- Univ. Bordeaux, INSERM, BioTis U1026, 33076 Bordeaux, France
| | - Vera Guduric
- Univ. Bordeaux, INSERM, BioTis U1026, 33076 Bordeaux, France
| | - Reine Bareille
- Univ. Bordeaux, INSERM, BioTis U1026, 33076 Bordeaux, France
| | - Rémy Agniel
- CY Cergy Paris Université, Maison Internationale de la Recherche, Lab. ERRMECe, 95031 Neuville-Oise, France
| | - Samantha Roques
- Univ. Bordeaux, CIC1401, CHU Bordeaux, INSERM, 33076 Bordeaux, France
| | - Valérie Héroguez
- Laboratoire de Chimie des Polymères Organiques, CNRS, Bordeaux INP, UMR5629, 33607 Pessac, France
| | - Marc Dussauze
- Univ. Bordeaux, Institut des Sciences Moléculaires, UMR 5255 CNRS, 33405 Talence, France
| | - Manuel Gaudon
- CNRS, Univ. Bordeaux, Bordeaux INP, ICMCB, UMR5026, 33608 Pessac, France
| | | | - Sylvain Catros
- Univ. Bordeaux, INSERM, BioTis U1026, CHU Bordeaux, Dentistry and Oral Health Department, 33076 Bordeaux, France.
| |
Collapse
|
17
|
Turnbull G, Clarke J, Picard F, Zhang W, Riches P, Li B, Shu W. 3D biofabrication for soft tissue and cartilage engineering. Med Eng Phys 2020; 82:13-39. [PMID: 32709263 DOI: 10.1016/j.medengphy.2020.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
Soft tissue injuries (STIs) affect patients of all age groups and represent a common worldwide clinical problem, resulting from conditions including trauma, infection, cancer and burns. Within the spectrum of STIs a mixture of tissues can be injured, ranging from skin to underlying nerves, blood vessels, tendons and cartilaginous tissues. However, significant limitations affect current treatment options and clinical demand for soft tissue and cartilage regenerative therapies continues to rise. Improving the regeneration of soft tissues has therefore become a key area of focus within tissue engineering. As an emerging technology, 3D bioprinting can be used to build complex soft tissue constructs "from the bottom up," by depositing cells, growth factors, extracellular matrices and other biomaterials in a layer-by-layer fashion. In this way, regeneration of cartilage, skin, vasculature, nerves, tendons and other bodily tissues can be performed in a patient specific manner. This review will focus on recent use of 3D bioprinting and other biofabrication strategies in soft tissue repair and regeneration. Biofabrication of a variety of soft tissue types will be reviewed following an overview of available cell sources, bioinks and bioprinting techniques.
Collapse
Affiliation(s)
- Gareth Turnbull
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom; Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Jon Clarke
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Frédéric Picard
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom; Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank G81 4DY, United Kingdom
| | - Weidong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Philip Riches
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Wenmiao Shu
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow G4 0NW, United Kingdom.
| |
Collapse
|
18
|
Castaño IM, Raftery RM, Chen G, Cavanagh B, Quinn B, Duffy GP, O'Brien FJ, Curtin CM. Rapid bone repair with the recruitment of CD206 +M2-like macrophages using non-viral scaffold-mediated miR-133a inhibition of host cells. Acta Biomater 2020; 109:267-279. [PMID: 32251781 DOI: 10.1016/j.actbio.2020.03.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 01/01/2023]
Abstract
microRNAs offer vast therapeutic potential for multiple disciplines. From a bone perspective, inhibition of miR-133a may offer potential to enhance Runx2 activity and increase bone repair. This study aims to assess the therapeutic capability of antagomiR-133a delivery from collagen-nanohydroxyapatite (coll-nHA) scaffolds following cell-free implantation in rat calvarial defects (7 mm diameter). This is, to the best of our knowledge, the first report of successful in vivo antagomiR uptake in host cells of fully immunocompetent animals without distribution to other off-target tissues. Our results demonstrate the localized release of antagomiR-133a to the implant site at 1 week post-implantation with increased calcium deposits already evident in the antagomiR-133a loaded scaffolds at this early timepoint. This was followed by an approximate 2-fold increase in bone volume versus antagomiR-free scaffolds and a significant 10-fold increase over the empty defect controls, after just 4 weeks. An increase in host CD206+ cells suggests an accelerated pro-remodeling response by M2-like macrophages accompanying bone repair with this treatment. Overall, this non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the addition of exogenous cells - and underlines the role of M2 macrophage-like cells in directing accelerated bone repair. Expanding the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a variety of therapeutic indications. STATEMENT OF SIGNIFICANCE: microRNAs, small non-coding RNA molecules involved in gene regulation, may have potential as a new class of bone healing therapeutics as they can enhance the regenerative capacity of bone-forming cells. We developed a collagen-nanohydroxyapatite-microRNA scaffold system to investigate whether miR133a inhibition can enhance osteogenesis in rat MSCs and ultimately accelerate endogenous bone repair by host cells in vivo without pre-seeding cells prior to implantation. Overall, this off-the-shelf, non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the requirement of exogenous cells - and highlights the role of CD206+M2 macrophage-like cells in guiding accelerated bone repair. Translating the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a vast myriad of therapeutic indications.
Collapse
|
19
|
Zhang M, Matinlinna JP, Tsoi JK, Liu W, Cui X, Lu WW, Pan H. Recent developments in biomaterials for long-bone segmental defect reconstruction: A narrative overview. J Orthop Translat 2020; 22:26-33. [PMID: 32440496 PMCID: PMC7231954 DOI: 10.1016/j.jot.2019.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Reconstruction of long-bone segmental defects (LBSDs) has been one of the biggest challenges in orthopaedics. Biomaterials for the reconstruction are required to be strong, osteoinductive, osteoconductive, and allowing for fast angiogenesis, without causing any immune rejection or disease transmission. There are four main types of biomaterials including autograft, allograft, artificial material, and tissue-engineered bone. Remarkable progress has been made in LBSD reconstruction biomaterials in the last ten years. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Our aim is to summarize recent developments in the divided four biomaterials utilized in the LBSD reconstruction to provide the clinicians with new information and comprehension from the biomaterial point of view.
Collapse
Key Words
- ADSC, allogenic adipose-derived stem cells
- ALLO, partially demineralized allogeneic bone block
- ALP, alkaline phosphatase
- ASC, adipose-derived stem cell
- Allograft
- Artificial material
- Autograft
- BMP-2 & 4, bone morphogenetic protein-2 & 4
- BMSC, bone marrow–derived mesenchymal stem cell
- BV, baculovirus
- Biomaterial
- CS, chitosan
- DBM, decalcified bone matrix
- FGF-2, Fibroblast Growth Factor-2
- HDB, heterogeneous deproteinized bone
- LBSD, long-bone segmental defect
- Long-bone segmental defect reconstruction
- M-CSF, macrophage colony-stimulating factor
- MIC, fresh marrow-impregnated ceramic block
- MSC, autologous mesenchymal stem cells
- PCL, polycaprolactone
- PDGF, Platelet-Derived Growth Factor
- PDLLA, poly(DL-lactide)
- PET/CT, positron emission- and computed tomography
- PLA, poly(lactic acid)
- PPF, propylene fumarate
- SF, silk fibroin
- TCP, tricalcium phosphate
- TEB, combining ceramic block with osteogenic-induced mesenchymal stem cells and platelet-rich plasma
- TGF-β, Transforming Growth Factor-β
- Tissue engineering
- VEGF, Vascular Endothelial Growth Factor
- bFGF, basic Fibroblast Growth Factor
- htMSCs, human tubal mesenchymal stem cells
- nHA, nano-hydroxyapatite
- poly, (L-lactide-co-D,L-lactide)
- rADSC, rabbit adipose-derived mesenchymal stem cell
- rVEGF-A, recombinant vascular endothelial growth factor-A
- rhBMP-2, recombinant human bone morphogenetic protein-2
- rhBMP-7, recombinant human bone morphogenetic protein 7
- sRANKL, soluble RANKL
- β-TCP, β-tricalcium phosphate
Collapse
Affiliation(s)
- Meng Zhang
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| | - Jukka P. Matinlinna
- Applied Oral Sciences, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, China
| | - James K.H. Tsoi
- Applied Oral Sciences, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Sai Ying Pun, Hong Kong SAR, China
| | - Wenlong Liu
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| | - Xu Cui
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| | - William W. Lu
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
- Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, China
| | - Haobo Pan
- Research Center for Human Tissues and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China
| |
Collapse
|
20
|
Almeida ALG, Freitas GP, Lopes HB, Gimenes R, Siessere S, Sousa LG, Beloti MM, Rosa AL. Effect of stem cells combined with a polymer/ceramic membrane on osteoporotic bone repair. Braz Oral Res 2019; 33:e079. [PMID: 31531565 DOI: 10.1590/1807-3107bor-2019.vol33.0079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/06/2019] [Indexed: 11/22/2022] Open
Abstract
Cell therapy associated with guided bone regeneration (GBR) can be used to treat bone defects under challenging conditions such as osteoporosis. This study aimed to evaluate the effect of mesenchymal stem cells (MSCs) in combination with a poly(vinylidene-trifluoroethylene)/barium titanate (PVDF-TrFE/BT) membrane on bone repair in osteoporotic rats. Osteoporosis was induced in female rats by bilateral removal of the ovaries (OVX) or sham surgery (SHAM), and the osteoporotic condition was characterized after 5 months by microtomographic and morphometric analyses. Calvarial defects were created in osteoporotic rats that immediately received the PVDF-TrFE/BT membrane. After 2 weeks, bone marrow-derived MSCs from healthy rats, characterized by the expression of surface markers using flow cytometry, or phosphate-buffered saline (PBS) (Control) were injected into the defects and bone formation was evaluated 4 weeks post-injection by microtomographic, morphometric, and histological analyses. A reduction in the amount of bone tissue in the femurs of OVX compared with SHAM rats confirmed the osteoporotic condition of the experimental model. More bone formation was observed when the defects were injected with MSCs compared to that with PBS. The modification that we are proposing in this study for the classical GBR approach where cells are locally injected after a membrane implantation may be a promising therapeutic strategy to increase bone formation under osteoporotic condition.
Collapse
Affiliation(s)
| | - Gileade Pereira Freitas
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Bone Research Lab , Ribeirão Preto , SP , Brazil
| | - Helena Bacha Lopes
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Bone Research Lab , Ribeirão Preto , SP , Brazil
| | - Rossano Gimenes
- Universidade Federal de Itajubá - Unifei, Institute of Physics and Chemistry , Itajubá , MG , Brazil
| | - Selma Siessere
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Department of Basic and Oral Biology , Ribeirão Preto , SP , Brazil
| | - Luiz Gustavo Sousa
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Department of Basic and Oral Biology , Ribeirão Preto , SP , Brazil
| | - Marcio Mateus Beloti
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Bone Research Lab , Ribeirão Preto , SP , Brazil
| | - Adalberto Luiz Rosa
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto , Bone Research Lab , Ribeirão Preto , SP , Brazil
| |
Collapse
|
21
|
Aslankoohi N, Mondal D, Rizkalla AS, Mequanint K. Bone Repair and Regenerative Biomaterials: Towards Recapitulating the Microenvironment. Polymers (Basel) 2019; 11:E1437. [PMID: 31480693 PMCID: PMC6780693 DOI: 10.3390/polym11091437] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 08/25/2019] [Indexed: 02/07/2023] Open
Abstract
Biomaterials and tissue engineering scaffolds play a central role to repair bone defects. Although ceramic derivatives have been historically used to repair bone, hybrid materials have emerged as viable alternatives. The rationale for hybrid bone biomaterials is to recapitulate the native bone composition to which these materials are intended to replace. In addition to the mechanical and dimensional stability, bone repair scaffolds are needed to provide suitable microenvironments for cells. Therefore, scaffolds serve more than a mere structural template suggesting a need for better and interactive biomaterials. In this review article, we aim to provide a summary of the current materials used in bone tissue engineering. Due to the ever-increasing scientific publications on this topic, this review cannot be exhaustive; however, we attempted to provide readers with the latest advance without being redundant. Furthermore, every attempt is made to ensure that seminal works and significant research findings are included, with minimal bias. After a concise review of crystalline calcium phosphates and non-crystalline bioactive glasses, the remaining sections of the manuscript are focused on organic-inorganic hybrid materials.
Collapse
Affiliation(s)
- Neda Aslankoohi
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Dibakar Mondal
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Amin S Rizkalla
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Kibret Mequanint
- School of Biomedical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, ON N6A 5B9, Canada.
| |
Collapse
|
22
|
Clumps of Mesenchymal Stem Cell/Extracellular Matrix Complexes Generated with Xeno-Free Conditions Facilitate Bone Regeneration via Direct and Indirect Osteogenesis. Int J Mol Sci 2019; 20:ijms20163970. [PMID: 31443173 PMCID: PMC6720767 DOI: 10.3390/ijms20163970] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/09/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Three-dimensional clumps of mesenchymal stem cell (MSC)/extracellular matrix (ECM) complexes (C-MSCs) consist of cells and self-produced ECM. We demonstrated previously that C-MSCs can be transplanted into bone defect regions with no artificial scaffold to induce bone regeneration. To apply C-MSCs in a clinical setting as a reliable bone regenerative therapy, the present study aimed to generate C-MSCs in xeno-free/serum-free conditions that can exert successful bone regenerative properties and to monitor interactions between grafted cells and host cells during bone healing processes. Human bone marrow-derived MSCs were cultured in xeno-free/serum-free medium. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and then torn off. The sheet was rolled to make a round clump of cells. Then, C-MSCs were transplanted into an immunodeficient mouse calvarial defect model. Transplantation of C-MSCs induced bone regeneration in a time-dependent manner. Immunofluorescence staining showed that both donor human cells and host mice cells contributed to bone reconstruction. Decellularized C-MSCs implantation failed to induce bone regeneration, even though the host mice cells can infiltrate into the defect area. These findings suggested that C-MSCs generated in xeno-free/serum-free conditions can induce bone regeneration via direct and indirect osteogenesis.
Collapse
|
23
|
Hettwer W, Horstmann PF, Bischoff S, Güllmar D, Reichenbach JR, Poh PSP, van Griensven M, Gras F, Diefenbeck M. Establishment and effects of allograft and synthetic bone graft substitute treatment of a critical size metaphyseal bone defect model in the sheep femur. APMIS 2019; 127:53-63. [PMID: 30698307 PMCID: PMC6850422 DOI: 10.1111/apm.12918] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/10/2018] [Indexed: 01/17/2023]
Abstract
Assessment of bone graft material efficacy is difficult in humans, since invasive methods like staged CT scans or biopsies are ethically unjustifiable. Therefore, we developed a novel large animal model for the verification of a potential transformation of synthetic bone graft substitutes into vital bone. The model combines multiple imaging methods with corresponding histology in standardized critical sized cancellous bone defect. Cylindrical bone voids (10 ml) were created in the medial femoral condyles of both hind legs (first surgery at right hind leg, second surgery 3 months later at left hind leg) in three merino‐wool sheep and either (i) left empty, filled with (ii) cancellous allograft bone or (iii) a synthetic, gentamicin eluting bone graft substitute. All samples were analysed with radiographs, MRI, μCT, DEXA and histology after sacrifice at 6 months. Unfilled defects only showed ingrowth of fibrous tissue, whereas good integration of the cancellous graft was seen in the allograft group. The bone graft substitute showed centripetal biodegradation and new trabecular bone formation in the periphery of the void as early as 3 months. μCT gave excellent insight into the structural changes within the defects, particularly progressive allograft incorporation and the bone graft substitute biodegradation process. MRI completed the picture by clearly visualizing soft tissue ingrowth into unfilled bone voids and presence of fluid collections. Histology was essential for verification of trabecular bone and osteoid formation. Conventional radiographs and DEXA could not differentiate details of the ongoing transformation process. This model appears well suited for detailed in vivo and ex vivo evaluation of bone graft substitute behaviour within large bone defects.
Collapse
Affiliation(s)
- Werner Hettwer
- Musculoskeletal Tumor Section, Department of Orthopedic Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Peter F Horstmann
- Musculoskeletal Tumor Section, Department of Orthopedic Surgery, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sabine Bischoff
- Central Experimental Animal Facility, University Hospital Jena, Jena, Germany
| | - Daniel Güllmar
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Jena, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Jena, Germany
| | - Patrina S P Poh
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martijn van Griensven
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Florian Gras
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Jena, Jena, Germany
| | - Michael Diefenbeck
- BONESUPPORT AB, Lund, Sweden.,Scientific Consulting in Orthopaedic Surgery and Traumatology, Hamburg, Germany
| |
Collapse
|
24
|
Azadian E, Arjmand B, Khodaii Z, Ardeshirylajimi A. A comprehensive overview on utilizing electromagnetic fields in bone regenerative medicine. Electromagn Biol Med 2019; 38:1-20. [PMID: 30661411 DOI: 10.1080/15368378.2019.1567527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stem cells are one of the most important sources to develope a new strategy for repairing bone lesions through tissue engineering. Osteogenic differentiation of stem cells can be affected by various factors such as biological, chemical, physiological, and physical ones. The application of ELF-EMFs has been the subject of many research in bone tissue engineering and evidence suggests that this exogenous physical stimulus can promote osteogenic differentiation in several types of cells. The purpose of this paper is to review the current knowledge on the effects of EMFs on stem cells in bone tissue engineering studies. We recapitulated and analyzed 39 articles that were focused on the application of EMFs for bone tissue engineering purposes. We tabulated scattered information from these articles for easy use and tried to provide an overview of conducted research and identify the knowledge gaps in the field.
Collapse
Affiliation(s)
- Esmaeel Azadian
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Bahar Arjmand
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Zohreh Khodaii
- c Dietary supplements and Probiotics research center , Alborz University of Medical Sciences , Karaj , Iran.,d Department of Biochemistry, Genetics and Nutrition, Faculty of Medicine , Alborz University of Medical Sciences , Karaj , Iran
| | - Abdolreza Ardeshirylajimi
- a Urogenital Stem Cell Research Center , Shahid Beheshti University of Medical Sciences , Tehran , Iran.,b Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
25
|
Turnbull G, Clarke J, Picard F, Riches P, Jia L, Han F, Li B, Shu W. 3D bioactive composite scaffolds for bone tissue engineering. Bioact Mater 2018; 3:278-314. [PMID: 29744467 PMCID: PMC5935790 DOI: 10.1016/j.bioactmat.2017.10.001] [Citation(s) in RCA: 584] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022] Open
Abstract
Bone is the second most commonly transplanted tissue worldwide, with over four million operations using bone grafts or bone substitute materials annually to treat bone defects. However, significant limitations affect current treatment options and clinical demand for bone grafts continues to rise due to conditions such as trauma, cancer, infection and arthritis. Developing bioactive three-dimensional (3D) scaffolds to support bone regeneration has therefore become a key area of focus within bone tissue engineering (BTE). A variety of materials and manufacturing methods including 3D printing have been used to create novel alternatives to traditional bone grafts. However, individual groups of materials including polymers, ceramics and hydrogels have been unable to fully replicate the properties of bone when used alone. Favourable material properties can be combined and bioactivity improved when groups of materials are used together in composite 3D scaffolds. This review will therefore consider the ideal properties of bioactive composite 3D scaffolds and examine recent use of polymers, hydrogels, metals, ceramics and bio-glasses in BTE. Scaffold fabrication methodology, mechanical performance, biocompatibility, bioactivity, and potential clinical translations will be discussed.
Collapse
Affiliation(s)
- Gareth Turnbull
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Jon Clarke
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Frédéric Picard
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
- Department of Orthopaedic Surgery, Golden Jubilee National Hospital, Agamemnon St, Clydebank, G81 4DY, United Kingdom
| | - Philip Riches
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
| | - Luanluan Jia
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Fengxuan Han
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Bin Li
- Orthopaedic Institute, Department of Orthopaedic Surgery, The First Affiliated Hospital, Soochow University, Suzhou, Jiangsu, PR China
| | - Wenmiao Shu
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, 106 Rottenrow, Glasgow, G4 0NW, United Kingdom
| |
Collapse
|
26
|
Scheinpflug J, Pfeiffenberger M, Damerau A, Schwarz F, Textor M, Lang A, Schulze F. Journey into Bone Models: A Review. Genes (Basel) 2018; 9:E247. [PMID: 29748516 PMCID: PMC5977187 DOI: 10.3390/genes9050247] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/24/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
Bone is a complex tissue with a variety of functions, such as providing mechanical stability for locomotion, protection of the inner organs, mineral homeostasis and haematopoiesis. To fulfil these diverse roles in the human body, bone consists of a multitude of different cells and an extracellular matrix that is mechanically stable, yet flexible at the same time. Unlike most tissues, bone is under constant renewal facilitated by a coordinated interaction of bone-forming and bone-resorbing cells. It is thus challenging to recreate bone in its complexity in vitro and most current models rather focus on certain aspects of bone biology that are of relevance for the research question addressed. In addition, animal models are still regarded as the gold-standard in the context of bone biology and pathology, especially for the development of novel treatment strategies. However, species-specific differences impede the translation of findings from animal models to humans. The current review summarizes and discusses the latest developments in bone tissue engineering and organoid culture including suitable cell sources, extracellular matrices and microfluidic bioreactor systems. With available technology in mind, a best possible bone model will be hypothesized. Furthermore, the future need and application of such a complex model will be discussed.
Collapse
Affiliation(s)
- Julia Scheinpflug
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Moritz Pfeiffenberger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Alexandra Damerau
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Franziska Schwarz
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Martin Textor
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| | - Annemarie Lang
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany.
| | - Frank Schulze
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R),10589 Berlin, Germany.
| |
Collapse
|
27
|
Motoike S, Kajiya M, Komatsu N, Takewaki M, Horikoshi S, Matsuda S, Ouhara K, Iwata T, Takeda K, Fujita T, Kurihara H. Cryopreserved clumps of mesenchymal stem cell/extracellular matrix complexes retain osteogenic capacity and induce bone regeneration. Stem Cell Res Ther 2018; 9:73. [PMID: 29562931 PMCID: PMC5863484 DOI: 10.1186/s13287-018-0826-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/16/2018] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Three-dimensional (3D) cultured clumps of mesenchymal stem cell (MSC)/extracellular matrix (ECM) complexes (C-MSCs) consist of cells and self-produced ECM. C-MSCs can regulate cellular functions in vitro and can be grafted into a defect site without an artificial scaffold to induce bone regeneration. Long-term cryopreservation of C-MSCs, which can enable them to serve as a ready-to-use cell preparation, may be helpful in developing beneficial cell therapy for bone regeneration. Therefore, the aim of this study was to investigate the effect of cryopreservation on C-MSCs. METHODS MSCs isolated from rat femurs were cultured in growth medium supplemented with ascorbic acid. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and were then torn off. The sheet was rolled to make a round clumps of cells. The C-MSCs were cryopreserved in cryomedium including 10% dimethyl sulfoxide. RESULTS Cryopreserved C-MSCs retained their 3D structure and did not exhibit a decrease in cell viability. In addition, stem cell marker expression levels and the osteogenic differentiation properties of C-MSCs were not reduced by cryopreservation. However, C-MSCs pretreated with collagenase before cryopreservation showed a lower level of type I collagen and could not retain their 3D structure, and their rates of cell death increased during cryopreservation. Both C-MSC and cryopreserved C-MSC transplantation into rat calvarial defects induced successful bone regeneration. CONCLUSION These data indicate that cryopreservation does not reduce the biological properties of C-MSCs because of its abundant type I collagen. More specifically, cryopreserved C-MSCs could be applicable for novel bone regenerative therapies.
Collapse
Affiliation(s)
- Souta Motoike
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan.
| | - Nao Komatsu
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Manabu Takewaki
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Susumu Horikoshi
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Shinji Matsuda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Katsuhiro Takeda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Tsuyoshi Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| |
Collapse
|
28
|
Lam J, Lee EJ, Clark EC, Mikos AG. Honing Cell and Tissue Culture Conditions for Bone and Cartilage Tissue Engineering. Cold Spring Harb Perspect Med 2017; 7:a025734. [PMID: 28348176 PMCID: PMC5710100 DOI: 10.1101/cshperspect.a025734] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
An avenue of tremendous interest and need in health care encompasses the regeneration of bone and cartilage. Over the years, numerous tissue engineering strategies have contributed substantial progress toward the realization of clinically relevant therapies. Cell and tissue culture protocols, however, show many variations that make experimental results among different publications challenging to compare. This collection surveys prevalent cell sources, soluble factors, culture medium formulations, environmental factors, and genetic modification approaches in the literature. The intent of consolidating this information is to provide a starting resource for scientists considering how to optimize the parameters for cell differentiation and tissue culture procedures within the context of bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Johnny Lam
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Esther J Lee
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Elisa C Clark
- Department of Bioengineering, Rice University, Houston, Texas 77251
| | - Antonios G Mikos
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77251
| |
Collapse
|
29
|
Effect of low-intensity pulsed ultrasound on osteogenic human mesenchymal stem cells commitment in a new bone scaffold. J Appl Biomater Funct Mater 2017; 15:e215-e222. [PMID: 28478615 PMCID: PMC6379883 DOI: 10.5301/jabfm.5000342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose Bone tissue engineering is helpful in finding alternatives to overcome
surgery limitations. Bone growth and repair are under the control of
biochemical and mechanical signals; therefore, in recent years several
approaches to improve bone regeneration have been evaluated. Osteo-inductive
biomaterials, stem cells, specific growth factors and biophysical stimuli
are among those. The aim of the present study was to evaluate if
low-intensity pulsed ultrasound stimulation (LIPUS) treatment would improve
the colonization of an MgHA/Coll hybrid composite scaffold by human
mesenchymal stem cells (hMSCs) and their osteogenic differentiation. LIPUS
stimulation was applied to hMSCs cultured on MgHA/Coll hybrid composite
scaffold in osteogenic medium, mimicking the microenvironment of a bone
fracture. Methods hMSCs were seeded on MgHA/Coll hybrid composite scaffold in an
osteo-inductive medium and exposed to LIPUS treatment for 20 min/day for
different experimental times (7 days, 14 days). The investigation was
focused on (i) the improvement of hMSCs to colonize the MgHA/Coll hybrid
composite scaffold by LIPUS, in terms of cell viability and ultrastructural
analysis; (ii) the activation of MAPK/ERK, osteogenic
(ALPL, COL1A1, BGLAP,
SPP1) and angiogenetic (VEGF, IL8)
pathways, through gene expression and protein release analysis, after LIPUS
stimuli. Results LIPUS exposure improved MgHA/Coll hybrid composite scaffold colonization and
induced in vitro osteogenic differentiation of hMSCs seeded on the
scaffold. Conclusions This work shows that the combined use of new biomimetic osteo-inductive
composite and LIPUS treatment could be a useful therapeutic approach in
order to accelerate bone regeneration pathways.
Collapse
|
30
|
Zhao L, Zhao J, Yu J, Sun R, Zhang X, Hu S. In vivo investigation of tissue-engineered periosteum for the repair of allogeneic critical size bone defects in rabbits. Regen Med 2017. [PMID: 28621175 DOI: 10.2217/rme-2016-0157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aim: The aim of the study was to evaluate the efficacy of tissue-engineered periosteum (TEP) in repairing allogenic bone defects in the long term. Materials & methods: TEP was biofabricated with osteoinduced rabbit bone marrow mesenchymal stem cells and porcine small intestinal submucosa (SIS). A total of 24 critical sized defects were created bilaterally in radii of 12 New Zealand White rabbits. TEP/SIS was implanted into the defect site. Bone defect repair was evaluated with radiographic and histological examination at 4, 8 and 12 weeks. Results: Bone defects were structurally reconstructed in the TEP group with mature cortical bone and medullary canals, however this was not observed in the SIS group at 12 weeks. Conclusion: The TEP approach can effectively restore allogenic critical sized defects, and achieve maturity of long-bone structure in 12 weeks in rabbit models.
Collapse
Affiliation(s)
- Lin Zhao
- Orthopaedic Department, Jinshan Branch of the Sixth People’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai 201500, China
| | - Junli Zhao
- Department of Nephrology, Shanghai ZhouPu Hospital, Shanghai 201318, China
| | - Jiajia Yu
- Orthopaedic Institute, the Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Rui Sun
- Orthopaedic Institute, the Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Xiaofeng Zhang
- Orthopaedic Department, Jinshan Branch of the Sixth People’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai 201500, China
| | - Shuhua Hu
- Orthopaedic Department, Jinshan Branch of the Sixth People’s Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai 201500, China
| |
Collapse
|
31
|
Orciani M, Fini M, Di Primio R, Mattioli-Belmonte M. Biofabrication and Bone Tissue Regeneration: Cell Source, Approaches, and Challenges. Front Bioeng Biotechnol 2017; 5:17. [PMID: 28386538 PMCID: PMC5362636 DOI: 10.3389/fbioe.2017.00017] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/22/2017] [Indexed: 01/06/2023] Open
Abstract
The growing occurrence of bone disorders and the increase in aging population have resulted in the need for more effective therapies to meet this request. Bone tissue engineering strategies, by combining biomaterials, cells, and signaling factors, are seen as alternatives to conventional bone grafts for repairing or rebuilding bone defects. Indeed, skeletal tissue engineering has not yet achieved full translation into clinical practice because of several challenges. Bone biofabrication by additive manufacturing techniques may represent a possible solution, with its intrinsic capability for accuracy, reproducibility, and customization of scaffolds as well as cell and signaling molecule delivery. This review examines the existing research in bone biofabrication and the appropriate cells and factors selection for successful bone regeneration as well as limitations affecting these approaches. Challenges that need to be tackled with the highest priority are the obtainment of appropriate vascularized scaffolds with an accurate spatiotemporal biochemical and mechanical stimuli release, in order to improve osseointegration as well as osteogenesis.
Collapse
Affiliation(s)
- Monia Orciani
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| | - Milena Fini
- Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute , Bologna , Italy
| | - Roberto Di Primio
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| | - Monica Mattioli-Belmonte
- Department of Molecular and Clinical Sciences, Università Politenica delle Marche , Ancona , Italy
| |
Collapse
|
32
|
Ye Y, Jing X, Li N, Wu Y, Li B, Xu T. Icariin promotes proliferation and osteogenic differentiation of rat adipose-derived stem cells by activating the RhoA-TAZ signaling pathway. Biomed Pharmacother 2017; 88:384-394. [PMID: 28122303 DOI: 10.1016/j.biopha.2017.01.075] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/02/2017] [Accepted: 01/12/2017] [Indexed: 12/18/2022] Open
Abstract
Icariin, the main active flavonoid glucoside isolated from Herba epimedii, has been demonstrated to be a potential alternative therapy to prevent postmenopausal osteoporosis. Icariin has also been shown to regulate the proliferation and osteogenic differentiation of rat bone marrow stromal cells (rBMSCs). However, the detailed molecular mechanism of icariin has remained largely unknown. Besides, no investigation has focused on the relevance of icariin in the regulation of rat adipose-derived stem cells (rASCs) proliferation and osteogenic differentiation. In the present study, we detected that icariin promotes proliferation and osteogenic differentiation of rASCs in a concentration range from 10-8M to 10-6M, with 10-7M to be the optimal concentration. We found that 10-7M icariin significantly increased active RhoA protein expression and ROCK substrate molecule p-MYPT1 expression in rASCs. C3 (the RhoA inhibitor) treatment abrogated the increased proliferation and osteogenic differentiation of rASCs induced by icariin. Interestingly, we also found that C3 abrogated the activation of TAZ induced by icariin. Depletion of TAZ by siRNA transfection significantly blocked icariin promoted proliferation and osteogenic differentiation of rASCs. However, icariin induced active RhoA protein expression was not affected by TAZ specific siRNA transfection, suggesting that RhoA lies upstream of TAZ. Taken together, our data indicate that icariin promotes proliferation and osteogenic differentiation of rASCs by activating the RhoA-TAZ signaling pathway.
Collapse
Affiliation(s)
- Yaping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xingzhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Na Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yingxing Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bingbing Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
33
|
Mobini S, Leppik L, Thottakkattumana Parameswaran V, Barker JH. In vitro effect of direct current electrical stimulation on rat mesenchymal stem cells. PeerJ 2017; 5:e2821. [PMID: 28097053 PMCID: PMC5237370 DOI: 10.7717/peerj.2821] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/22/2016] [Indexed: 12/11/2022] Open
Abstract
Background Electrical stimulation (ES) has been successfully used to treat bone defects clinically. Recently, both cellular and molecular approaches have demonstrated that ES can change cell behavior such as migration, proliferation and differentiation. Methods In the present study we exposed rat bone marrow- (BM-) and adipose tissue- (AT-) derived mesenchymal stem cells (MSCs) to direct current electrical stimulation (DC ES) and assessed temporal changes in osteogenic differentiation. We applied 100 mV/mm of DC ES for 1 h per day for three, seven and 14 days to cells cultivated in osteogenic differentiation medium and assessed viability and calcium deposition at the different time points. In addition, expression of osteogenic genes, Runx2, Osteopontin, and Col1A2 was assessed in BM- and AT-derived MSCs at the different time points. Results Results showed that ES changed osteogenic gene expression patterns in both BM- and AT-MSCs, and these changes differed between the two groups. In BM-MSCs, ES caused a significant increase in mRNA levels of Runx2, Osteopontin and Col1A2 at day 7, while in AT-MSCs, the increase in Runx2 and Osteopontin expression were observed after 14 days of ES. Discussion This study shows that rat bone marrow- and adipose tissue-derived stem cells react differently to electrical stimuli, an observation that could be important for application of electrical stimulation in tissue engineering.
Collapse
Affiliation(s)
- Sahba Mobini
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany.,School of Materials, Faculty of Engineering and Physical Sciences, University of Manchester, Manchester, United Kingdom
| | - Liudmila Leppik
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Vishnu Thottakkattumana Parameswaran
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - John Howard Barker
- Frankfurt Initiative for Regenerative Medicine, Experimental Orthopedics and Trauma Surgery, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
34
|
Zhang H, Yu N, Zhou Y, Ma H, Wang J, Ma X, Liu J, Huang J, An Y. Construction and characterization of osteogenic and vascular endothelial cell sheets from rat adipose-derived mesenchymal stem cells. Tissue Cell 2016; 48:488-95. [DOI: 10.1016/j.tice.2016.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 12/31/2022]
|
35
|
Wu Q, Yang B, Hu K, Cao C, Man Y, Wang P. Deriving Osteogenic Cells from Induced Pluripotent Stem Cells for Bone Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:1-8. [PMID: 27392674 DOI: 10.1089/ten.teb.2015.0559] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Induced pluripotent stem cells (iPSCs), reprogrammed from adult somatic cells using defined transcription factors, are regarded as a promising cell source for tissue engineering. For the purpose of bone tissue regeneration, efficient in vitro differentiation of iPSCs into downstream cells, such as mesenchymal stem cells (MSCs), osteoblasts, or osteocyte-like cells, before use is necessary to limit undesired tumorogenesis associated with the pluripotency of iPSCs. Until recently numerous techniques on the production of iPSC-derived osteogenic progenitors have been introduced. We reviewed these protocols and provided a perspective on the comparisons of osteogenic potentials of (1) iPSC-derived osteogenic cells produced by different protocols, (2) iPSCs from different somatic origins, and (3) iPSC-derived MSC-like cells and bone marrow stem cells. Finally, we discussed the potential application of the diseased iPSCs for systematic bone disorders.
Collapse
Affiliation(s)
- Qingqing Wu
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Bo Yang
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Kevin Hu
- 2 University of Maryland Dental School , Baltimore, Maryland
| | - Cong Cao
- 3 Department of Stomatology, China-Japan Friendship Hospital , Beijing, China
| | - Yi Man
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China
| | - Ping Wang
- 1 State Key Laboratory of Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University , Chengdu, China .,2 University of Maryland Dental School , Baltimore, Maryland
| |
Collapse
|
36
|
Collette NM, Yee CS, Hum NR, Murugesh DK, Christiansen BA, Xie L, Economides AN, Manilay JO, Robling AG, Loots GG. Sostdc1 deficiency accelerates fracture healing by promoting the expansion of periosteal mesenchymal stem cells. Bone 2016; 88:20-30. [PMID: 27102547 PMCID: PMC6277141 DOI: 10.1016/j.bone.2016.04.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/16/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Loss of Sostdc1, a growth factor paralogous to Sost, causes the formation of ectopic incisors, fused molars, abnormal hair follicles, and resistance to kidney disease. Sostdc1 is expressed in the periosteum, a source of osteoblasts, fibroblasts and mesenchymal progenitor cells, which are critically important for fracture repair. Here, we investigated the role of Sostdc1 in bone metabolism and fracture repair. Mice lacking Sostdc1 (Sostdc1(-/-)) had a low bone mass phenotype associated with loss of trabecular bone in both lumbar vertebrae and in the appendicular skeleton. In contrast, Sostdc1(-/-) cortical bone measurements revealed larger bones with higher BMD, suggesting that Sostdc1 exerts differential effects on cortical and trabecular bone. Mid-diaphyseal femoral fractures induced in Sostdc1(-/-) mice showed that the periosteal population normally positive for Sostdc1 rapidly expands during periosteal thickening and these cells migrate into the fracture callus at 3days post fracture. Quantitative analysis of mesenchymal stem cell (MSC) and osteoblast populations determined that MSCs express Sostdc1, and that Sostdc1(-/-) 5day calluses harbor >2-fold more MSCs than fractured wildtype controls. Histologically a fraction of Sostdc1-positive cells also expressed nestin and α-smooth muscle actin, suggesting that Sostdc1 marks a population of osteochondral progenitor cells that actively participate in callus formation and bone repair. Elevated numbers of MSCs in D5 calluses resulted in a larger, more vascularized cartilage callus at day 7, and a more rapid turnover of cartilage with significantly more remodeled bone and a thicker cortical shell at 21days post fracture. These data support accelerated or enhanced bone formation/remodeling of the callus in Sostdc1(-/-) mice, suggesting that Sostdc1 may promote and maintain mesenchymal stem cell quiescence in the periosteum.
Collapse
Affiliation(s)
- Nicole M Collette
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | - Cristal S Yee
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA; Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA
| | - Nicholas R Hum
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | - Deepa K Murugesh
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA
| | | | - LiQin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Jennifer O Manilay
- Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA
| | | | - Gabriela G Loots
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550, USA; Molecular and Cell Biology Unit, School of Natural Sciences, University of California at Merced, Merced, CA, USA.
| |
Collapse
|
37
|
Khojasteh A, Nazeman P, Rad MR. Dental Stem Cells in Oral, Maxillofacial and Craniofacial Regeneration. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-28947-2_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Teng S, Liu C, Guenther D, Omar M, Neunaber C, Krettek C, Jagodzinski M. Influence of biomechanical and biochemical stimulation on the proliferation and differentiation of bone marrow stromal cells seeded on polyurethane scaffolds. Exp Ther Med 2016; 11:2086-2094. [PMID: 27284290 PMCID: PMC4888012 DOI: 10.3892/etm.2016.3206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 02/19/2016] [Indexed: 02/06/2023] Open
Abstract
The aim of the present investigation was to compare the effects of cyclic compression, perfusion, dexamethasone (DEX) and bone morphogenetic protein-7 (BMP-7) on the proliferation and differentiation of human bone marrow stromal cells (hBMSCs) in polyurethane scaffolds in a perfusion bioreactor. Polyurethane scaffolds seeded with hBMSCs were cultured under six different conditions, as follows: 10% Cyclic compression at 0.5 and 5 Hz; 10 ml/min perfusion; 100 nM DEX; 100 ng/ml BMP-7; and 1 ml/min perfusion without mechanical and biochemical stimulation (control). On days 7 and 14, samples were tested for the following data: Cell proliferation; mRNA expression of Runx2, COL1A1 and osteocalcin; osteocalcin content; calcium deposition; and the equilibrium modulus of the tissue specimen. The results indicated that BMP-7 and 10 ml/min perfusion promoted cell proliferation, which was inhibited by 5 Hz cyclic compression and DEX. On day 7, the 5 Hz cyclic compression inhibited Runx2 expression, whereas the 0.5 Hz cyclic compression and BMP-7 upregulated the COL1A1 mRNA levels on day 7 and enhanced the osteocalcin expression on day 14. The DEX-treated hBMSCs exhibited downregulated osteocalcin expression. After 14 days, the BMP-7 group exhibited the highest calcium deposition, followed by the 0.5 Hz cyclic compression and the DEX groups. The equilibrium modulus of the engineered constructs significantly increased in the BMP-7, 0.5 Hz cyclic compression and DEX groups. In conclusion, the present results suggest that BMP-7 and perfusion enhance cell proliferation, whereas high frequency cyclic compression inhibits the proliferation and osteogenic differentiation of hBMSCs. Low frequency cyclic compression is more effective than DEX, but less effective compared with BMP-7 on the osteogenic differentiation of hBMSCs seeded on polyurethane scaffolds.
Collapse
Affiliation(s)
- Songsong Teng
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Chaoxu Liu
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Daniel Guenther
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Mohamed Omar
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Claudia Neunaber
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Krettek
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Jagodzinski
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
39
|
Gohil SV, Kuo C, Adams DJ, Maye P, Rowe DW, Nair LS. Evaluation of the donor cell contribution in rh
BMP
‐2 mediated bone formation with chitosan thermogels using fluorescent protein reporter mice. J Biomed Mater Res A 2016; 104:928-41. [DOI: 10.1002/jbm.a.35634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/18/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Shalini V. Gohil
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
| | - Chia‐Ling Kuo
- Connecticut Institute for Clinical and Translational Science, Institute for Systems Genomics, University of ConnecticutFarmington Connecticut06030
| | - Douglas J. Adams
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
| | - Peter Maye
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - David W. Rowe
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - Lakshmi S. Nair
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
- Departments of Material Science and Engineering, Biomedical Engineering and Institute of Material ScienceUniversity of ConnecticutStorrs Connecticut06269
| |
Collapse
|
40
|
Pizzute T, Lynch K, Pei M. Impact of tissue-specific stem cells on lineage-specific differentiation: a focus on the musculoskeletal system. Stem Cell Rev Rep 2015; 11:119-32. [PMID: 25113801 DOI: 10.1007/s12015-014-9546-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tissue-specific stem cells are found throughout the body and, with proper intervention and environmental cues, these stem cells exercise their capabilities for differentiation into several lineages to form cartilage, bone, muscle, and adipose tissue in vitro and in vivo. Interestingly, it has been widely demonstrated that they do not differentiate with the same efficacy during lineage-specific differentiation studies, as the tissue-specific stem cells are generally more effective when differentiating toward the tissues from which they were derived. This review focuses on four mesodermal lineages for tissue-specific stem cell differentiation: adipogenesis, chondrogenesis, myogenesis, and osteogenesis. It is intended to give insight into current multilineage differentiation and comparative research, highlight and contrast known trends regarding differentiation, and introduce supporting evidence which demonstrates particular tissue-specific stem cells' superiority in lineage-specific differentiation, along with their resident tissue origins and natural roles. In addition, some epigenetic and transcriptomic differences between stem cells which may explain the observed trends are discussed.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
| | | | | |
Collapse
|
41
|
Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 2015; 48:62. [PMID: 26530042 PMCID: PMC4630918 DOI: 10.1186/s40659-015-0053-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.
Collapse
Affiliation(s)
- Jin Shao
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tieyi Yang
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
42
|
Improving vascularization of engineered bone through the generation of pro-angiogenic effects in co-culture systems. Adv Drug Deliv Rev 2015; 94:116-25. [PMID: 25817732 DOI: 10.1016/j.addr.2015.03.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/27/2015] [Accepted: 03/20/2015] [Indexed: 01/31/2023]
Abstract
One of the major problems with bone tissue engineering is the development of a rapid vascularization after implantation to supply the growing osteoblast cells with the nutrients to grow and survive as well as to remove waste products. It has been demonstrated that capillary-like structures produced in vitro will anastomose rapidly after implantation and become functioning blood vessels. For this reason, in recent years many studies have examined a variety of human osteoblast and endothelial cell co-culture systems in order to distribute osteoblasts on all parts of the bone scaffold and at the same time provide conditions for the endothelial cells to migrate to form a network of capillary-like structures throughout the osteoblast-colonized scaffold. The movement and proliferation of endothelial cells to form capillary-like structures is known as angiogenesis and is dependent on a variety of pro-angiogenic factors. This review summarizes human 2- and 3-D co-culture models to date, the types and origins of cells used in the co-cultures and the proangiogenic factors that have been identified in the co-culture models.
Collapse
|
43
|
A preclinical large animal study on a novel intervertebral fusion cage covered with high porosity titanium sheets with a triple pore structure used for spinal fusion. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2015; 24:2530-7. [DOI: 10.1007/s00586-015-4047-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/16/2015] [Accepted: 05/23/2015] [Indexed: 11/30/2022]
|
44
|
Roux BM, Cheng MH, Brey EM. Engineering clinically relevant volumes of vascularized bone. J Cell Mol Med 2015; 19:903-14. [PMID: 25877690 PMCID: PMC4420594 DOI: 10.1111/jcmm.12569] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/29/2015] [Indexed: 12/15/2022] Open
Abstract
Vascularization remains one of the most important challenges that must be overcome for tissue engineering to be consistently implemented for reconstruction of large volume bone defects. An extensive vascular network is needed for transport of nutrients, waste and progenitor cells required for remodelling and repair. A variety of tissue engineering strategies have been investigated in an attempt to vascularize tissues, including those applying cells, soluble factor delivery strategies, novel design and optimization of bio-active materials, vascular assembly pre-implantation and surgical techniques. However, many of these strategies face substantial barriers that must be overcome prior to their ultimate translation into clinical application. In this review recent progress in engineering vascularized bone will be presented with an emphasis on clinical feasibility.
Collapse
Affiliation(s)
- Brianna M Roux
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA; Research Service, Edward Hines Jr. V.A. Hospital, Hines, IL, USA
| | | | | |
Collapse
|
45
|
Nanoparticle delivery of stable miR-199a-5p agomir improves the osteogenesis of human mesenchymal stem cells via the HIF1a pathway. Biomaterials 2015; 53:239-50. [PMID: 25890723 DOI: 10.1016/j.biomaterials.2015.02.071] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 02/10/2015] [Accepted: 02/15/2015] [Indexed: 12/22/2022]
Abstract
Elucidating the regulatory mechanisms of osteogenesis of human mesenchymal stem cell (hMSC) is important for the development of cell therapies for bone loss and regeneration. Here we showed that hsa-miR-199a-5p modulated osteogenic differentiation of hMSCs at both early and late stages through HIF1a pathway. hsa-miR-199a expression was up-regulated during osteogenesis for both of two mature forms, miR-199a-5p and -3p. Over-expression of miR-199a-5p but not -3p enhanced differentiation of hMSCs in vitro, whereas inhibition of miR-199a-5p reduced the expression of osteoblast-specific genes, alkaline phosphatase (ALP) activity, and mineralization. Furthermore, over-expression of miR-199a enhanced ectopic bone formation in vivo. Chitosan nanoparticles were used for delivery of stable modified hsa-miR-199a-5p (agomir) both in vitro and in vivo, as a proof-of-concept for stable agomir delivery on bone regeneration. The hsa-mir199a-5p agomir were mixed with Chitosan nanoparticles to form nanoparticle/hsa-mir199a-5p agomir plasmid (nanoparticle/agomir) complexes, and nanoparticle/agomir complexes could improve the in vivo regeneration of bone. Further mechanism studies revealed that hypoxia enhanced osteogenesis at early stage and inhibited osteogenesis maturation at late stage through HIF1a-Twist1 pathway. At early stage of differentiation, hypoxia induced HIF1a-Twist1 pathway to enhance osteogenesis by up-regulating miR-199a-5p, while at late stage of differentiation, miR-199a-5p enhanced osteogenesis maturation by inhibiting HIF1α-Twist1 pathway.
Collapse
|
46
|
Kittaka M, Kajiya M, Shiba H, Takewaki M, Takeshita K, Khung R, Fujita T, Iwata T, Nguyen TQ, Ouhara K, Takeda K, Fujita T, Kurihara H. Clumps of a mesenchymal stromal cell/extracellular matrix complex can be a novel tissue engineering therapy for bone regeneration. Cytotherapy 2015; 17:860-73. [PMID: 25743634 DOI: 10.1016/j.jcyt.2015.01.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/17/2015] [Accepted: 01/27/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND AIMS The transplantation of mesenchymal stromal cells (MSCs) to damaged tissue has attracted attention in scientific and medical fields as an effective regenerative therapy. Nevertheless, additional studies are required to develop an MSC transplant method for bone regeneration because the use of an artificial scaffold restricts the number of transplanted cells and their function. Furthermore, regulating the degree of cell differentiation in vitro is desirable for a more effective regenerative therapy. To address these unresolved issues, with the use of a self-produced extracellular matrix (ECM), we developed clumps of an MSC/ECM complex (C-MSCs). METHODS MSCs isolated from rat femur were cultured in growth medium supplemented with 50 μg/mL of ascorbic acid for 7 days. To obtain C-MSCs, confluent cells were scratched with the use of a micropipette tip to roll up the cellular sheet, which consisted of ECM produced by the MSCs. The biological properties of C-MSCs were assessed in vitro and their bone regenerative activity was tested by use of a rat calvarial defect model. RESULTS Immunofluorescent confocal microscopic analysis revealed that type I collagen formed C-MSCs. Osteopontin messenger RNA expression and amount of calcium content were higher in C-MSCs cultured in osteo-inductive medium than those of untreated C-MSCs. The transplantation of osteogenic-differentiated C-MSCs led to rapid bone regeneration in the rat calvarial defect model. CONCLUSIONS These results suggest that the use of C-MSCs refined by self-produced ECM, which contain no artificial scaffold and can be processed in vitro, may represent a novel tissue engineering therapy.
Collapse
Affiliation(s)
- Mizuho Kittaka
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Shiba
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Manabu Takewaki
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kei Takeshita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Rathvisal Khung
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takako Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Truong Quoc Nguyen
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsuhiro Takeda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tsuyoshi Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
47
|
Lewallen EA, Riester SM, Bonin CA, Kremers HM, Dudakovic A, Kakar S, Cohen RC, Westendorf JJ, Lewallen DG, van Wijnen AJ. Biological strategies for improved osseointegration and osteoinduction of porous metal orthopedic implants. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:218-30. [PMID: 25348836 DOI: 10.1089/ten.teb.2014.0333] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The biological interface between an orthopedic implant and the surrounding host tissue may have a dramatic effect upon clinical outcome. Desired effects include bony ingrowth (osseointegration), stimulation of osteogenesis (osteoinduction), increased vascularization, and improved mechanical stability. Implant loosening, fibrous encapsulation, corrosion, infection, and inflammation, as well as physical mismatch may have deleterious clinical effects. This is particularly true of implants used in the reconstruction of load-bearing synovial joints such as the knee, hip, and the shoulder. The surfaces of orthopedic implants have evolved from solid-smooth to roughened-coarse and most recently, to porous in an effort to create a three-dimensional architecture for bone apposition and osseointegration. Total joint surgeries are increasingly performed in younger individuals with a longer life expectancy, and therefore, the postimplantation lifespan of devices must increase commensurately. This review discusses advancements in biomaterials science and cell-based therapies that may further improve orthopedic success rates. We focus on material and biological properties of orthopedic implants fabricated from porous metal and highlight some relevant developments in stem-cell research. We posit that the ideal primary and revision orthopedic load-bearing metal implants are highly porous and may be chemically modified to induce stem cell growth and osteogenic differentiation, while minimizing inflammation and infection. We conclude that integration of new biological, chemical, and mechanical methods is likely to yield more effective strategies to control and modify the implant-bone interface and thereby improve long-term clinical outcomes.
Collapse
|
48
|
Wang J, Wang L, Yang M, Zhu Y, Tomsia A, Mao C. Untangling the effects of peptide sequences and nanotopographies in a biomimetic niche for directed differentiation of iPSCs by assemblies of genetically engineered viral nanofibers. NANO LETTERS 2014; 14:6850-6856. [PMID: 25456151 PMCID: PMC4291459 DOI: 10.1021/nl504358j] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Here we report the design of a unique matrix, assembled from engineered M13 phage bionanofibers with specific cues of nanotopographies and versatile signal peptides to simulate native niche for directing the fate of induced pluripotent stem cells (iPSCs). By independently varying the peptide sequences and nanotopographies, we find that the resident iPSCs on the phage matrix are first differentiated into mesenchymal progenitor cells (MPCs), which are further differentiated into osteoblasts in the absence of osteogenic supplements due to the elongation induced by phage nanofibers. The phage-based matrix represents not only a biomimetic stem cell niche enabling independently varying biochemical and biophysical cues in one system but also a substrate for generating a safe and efficient cell source for tissue engineering.
Collapse
Affiliation(s)
- Jianglin Wang
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Lin Wang
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, China
- Corresponding Authors M.Y.: . Fax: +86-571-88982185. C.M.: . Fax: +1-405-325-6111
| | - Ye Zhu
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Antoni Tomsia
- Materials Science Division, Lawrence Berkeley National Laboratory, One Cyclotron Road, Berkeley, California 94720, United States
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
- Corresponding Authors M.Y.: . Fax: +86-571-88982185. C.M.: . Fax: +1-405-325-6111
| |
Collapse
|
49
|
Malhotra A, Pelletier MH, Yu Y, Christou C, Walsh WR. A sheep model for cancellous bone healing. Front Surg 2014; 1:37. [PMID: 25593961 PMCID: PMC4286987 DOI: 10.3389/fsurg.2014.00037] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/25/2014] [Indexed: 11/23/2022] Open
Abstract
Appropriate well-characterized bone defect animal models remain essential for preclinical research. This pilot study demonstrates a relevant animal model for cancellous bone defect healing. Three different defect diameters (8, 11, 14 mm) of fixed depth (25 mm) were compared in both skeletally immature (18-month-old) and aged sheep (5-year-old). In each animal, four defects were surgically created and placed in the cancellous bone of the medial distal femoral and proximal tibial epiphyses bilaterally. Animals were euthanized at 4 weeks post-operatively to assess early healing and any biological response. Defect sites were graded radiographically, and new bone formation quantified using μCT and histomorphometry. Fibrous tissue was found within the central region in most of the defects with woven bone normally forming near the periphery of the defect. Bone volume fraction [bone volume (BV)/TV] significantly decreased with an increasing defect diameter. Actual BV, however, increased with defect diameter. Bone ingrowth was lower for all defect diameters in the aged group. This pilot study proposes that the surgical creation of 11 mm diameter defects in the proximal tibial and distal femoral epiphyses of aged sheep is a suitable large animal model to study early healing of cancellous bone defects. The refined model allows for the placement of four separate bone defects per animal and encourages a reduction in animal numbers required for preclinical research.
Collapse
Affiliation(s)
- Angad Malhotra
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales , Sydney, NSW , Australia
| | - Matthew Henry Pelletier
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales , Sydney, NSW , Australia
| | - Yan Yu
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales , Sydney, NSW , Australia
| | - Chris Christou
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales , Sydney, NSW , Australia
| | - William Robert Walsh
- Surgical and Orthopedic Research Laboratories, Prince of Wales Clinical School, University of New South Wales , Sydney, NSW , Australia
| |
Collapse
|
50
|
Ferretti C, Vozzi G, Falconi M, Orciani M, Gesi M, Di Primio R, Mattioli-Belmonte M. Role of IGF1 and IGF1/VEGF on Human Mesenchymal Stromal Cells in Bone Healing: Two Sources and Two Fates. Tissue Eng Part A 2014; 20:2473-82. [DOI: 10.1089/ten.tea.2013.0453] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Concetta Ferretti
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Giovanni Vozzi
- Faculty of Engineering, Research Centre “E. Piaggio,” University of Pisa, Pisa, Italy
| | - Mirella Falconi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Gesi
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberto Di Primio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|