1
|
Onoki T, Kanczler J, Rawlings A, Smith M, Kim YH, Hashimoto K, Aizawa T, Oreffo ROC. Modulation of osteoblastogenesis by NRF2: NRF2 activation suppresses osteogenic differentiation and enhances mineralization in human bone marrow-derived mesenchymal stromal cells. FASEB J 2024; 38:e23892. [PMID: 39230563 DOI: 10.1096/fj.202400602r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024]
Abstract
Mesenchymal stromal stem cells (MSCs) or skeletal stem cells (SSCs) play a major role in tissue repair due to their robust ability to differentiate into osteoblasts, chondrocytes, and adipocytes. Complex cell signaling cascades tightly regulate this differentiation. In osteogenic differentiation, Runt-related transcription factor 2 (RUNX2) and ALP activity are essential. Furthermore, during the latter stages of osteogenic differentiation, mineral formation mediated by the osteoblast occurs with the secretion of a collagenous extracellular matrix and calcium deposition. Activation of nuclear factor erythroid 2-related factor 2 (NRF2), an important transcription factor against oxidative stress, inhibits osteogenic differentiation and mineralization via modulation of RUNX2 function; however, the exact role of NRF2 in osteoblastogenesis remains unclear. Here, we demonstrate that NRF2 activation in human bone marrow-derived stromal cells (HBMSCs) suppressed osteogenic differentiation. NRF2 activation increased the expression of STRO-1 and KITLG (stem cell markers), indicating NRF2 protects HBMSCs stemness against osteogenic differentiation. In contrast, NRF2 activation enhanced mineralization, which is typically linked to osteogenic differentiation. We determined that these divergent results were due in part to the modulation of cellular calcium flux genes by NRF2 activation. The current findings demonstrate a dual role for NRF2 as a HBMSC maintenance factor as well as a central factor in mineralization, with implications therein for elucidation of bone formation and cellular Ca2+ kinetics, dystrophic calcification and, potentially, application in the modulation of bone formation.
Collapse
Affiliation(s)
- Takahiro Onoki
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Andrew Rawlings
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Melanie Smith
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Yang-Hee Kim
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Ko Hashimoto
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Toshimi Aizawa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Hussein N, Meade J, Pandit H, Jones E, El-Gendy R. Characterisation and Expression of Osteogenic and Periodontal Markers of Bone Marrow Mesenchymal Stem Cells (BM-MSCs) from Diabetic Knee Joints. Int J Mol Sci 2024; 25:2851. [PMID: 38474098 DOI: 10.3390/ijms25052851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents a significant health problem globally and is linked to a number of complications such as cardiovascular disease, bone fragility and periodontitis. Autologous bone marrow mesenchymal stem cells (BM-MSCs) are a promising therapeutic approach for bone and periodontal regeneration; however, the effect of T2DM on the expression of osteogenic and periodontal markers in BM-MSCs is not fully established. Furthermore, the effect of the presence of comorbidities such as diabetes and osteoarthritis on BM-MSCs is also yet to be investigated. In the present study, BM-MSCs were isolated from osteoarthritic knee joints of diabetic and nondiabetic donors. Both cell groups were compared for their clonogenicity, proliferation rates, MSC enumeration and expression of surface markers. Formation of calcified deposits and expression of osteogenic and periodontal markers were assessed after 1, 2 and 3 weeks of basal and osteogenic culture. Diabetic and nondiabetic BM-MSCs showed similar clonogenic and growth potentials along with comparable numbers of MSCs. However, diabetic BM-MSCs displayed lower expression of periostin (POSTN) and cementum protein 1 (CEMP-1) at Wk3 osteogenic and Wk1 basal cultures, respectively. BM-MSCs from T2DM patients might be suitable candidates for stem cell-based therapeutics. However, further investigations into these cells' behaviours in vitro and in vivo under inflammatory environments and hyperglycaemic conditions are still required.
Collapse
Affiliation(s)
- Nancy Hussein
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds LS9 7TF, UK
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mansoura University, Mansoura 35516, Egypt
| | - Josephine Meade
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds LS9 7TF, UK
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Reem El-Gendy
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds LS9 7TF, UK
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
3
|
Buss LF, de Martin GS, Martinez EF, Filgueiras IADAAP, Magnabosco JL, Alves BF, de Macedo Almeida B, Kotaka T, Teixeira ML, Ferreira JRM, da Rocha DN, Canal R, Aloise AC, Holliday LS, Pelegrine AA. Conditioned Media from Human Pulp Stem Cell Cultures Improve Bone Regeneration in Rat Calvarial Critical-Size Defects. J Funct Biomater 2023; 14:396. [PMID: 37623641 PMCID: PMC10455841 DOI: 10.3390/jfb14080396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
The aim of this study was to test whether lyophilized conditioned media from human dental pulp mesenchymal stem cell cultures promote the healing of critical-size defects created in the calvaria of rats. Prior to the surgical procedure, the medium in which dental pulp stem cells were cultured was frozen and lyophilized. After general anesthesia, an 8 mm diameter bone defect was created in the calvaria of twenty-four rats. The defects were filled with the following materials: xenograft alone (G1) or xenograft associated with lyophilized conditioned medium (G2). After 14 or 42 days, the animals were euthanized, and the specimens processed for histologic and immunohistochemical analysis. Bone formation at the center of the defect was observed only in the G2 at 42 days. At both timepoints, increased staining for VEGF, a marker for angiogenesis, was observed in G2. Consistent with this, at 14 days, G2 also had a higher number of blood vessels detected by immunostaining with an anti-CD34 antibody. In conclusion, conditioned media from human dental pulp mesenchymal stem cell cultures had a positive effect on the regenerative process in rat critical-size bone defects. Both the formation of bone and enhancement of vascularization were stimulated by the conditioned media.
Collapse
Affiliation(s)
- Leonardo Fernandes Buss
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | - Gustavo Sigrist de Martin
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | | | | | - José Luiz Magnabosco
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | - Bruno Frenhan Alves
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | - Bruno de Macedo Almeida
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | - Tatiana Kotaka
- Faculdade de Odontologia São Leopoldo Mandic, Campinas 13045-755, SP, Brazil; (L.F.B.); (G.S.d.M.); (I.A.d.A.A.P.F.); (J.L.M.); (B.F.A.); (B.d.M.A.); (T.K.)
| | | | | | | | | | - Antonio Carlos Aloise
- Division of Oral Implantology, Faculdade São Leopoldo Mandic, Campinas 13045-755, SP, Brazil;
| | | | - André Antonio Pelegrine
- Division of Oral Implantology, Faculdade São Leopoldo Mandic, Campinas 13045-755, SP, Brazil;
| |
Collapse
|
4
|
Ross EA, Turner LA, Donnelly H, Saeed A, Tsimbouri MP, Burgess KV, Blackburn G, Jayawarna V, Xiao Y, Oliva MAG, Willis J, Bansal J, Reynolds P, Wells JA, Mountford J, Vassalli M, Gadegaard N, Oreffo ROC, Salmeron-Sanchez M, Dalby MJ. Nanotopography reveals metabolites that maintain the immunomodulatory phenotype of mesenchymal stromal cells. Nat Commun 2023; 14:753. [PMID: 36765065 PMCID: PMC9918539 DOI: 10.1038/s41467-023-36293-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent progenitor cells that are of considerable clinical potential in transplantation and anti-inflammatory therapies due to their capacity for tissue repair and immunomodulation. However, MSCs rapidly differentiate once in culture, making their large-scale expansion for use in immunomodulatory therapies challenging. Although the differentiation mechanisms of MSCs have been extensively investigated using materials, little is known about how materials can influence paracrine activities of MSCs. Here, we show that nanotopography can control the immunomodulatory capacity of MSCs through decreased intracellular tension and increasing oxidative glycolysis. We use nanotopography to identify bioactive metabolites that modulate intracellular tension, growth and immunomodulatory phenotype of MSCs in standard culture and during larger scale cell manufacture. Our findings demonstrate an effective route to support large-scale expansion of functional MSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Ewan A Ross
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Lesley-Anne Turner
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Hannah Donnelly
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Anwer Saeed
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Monica P Tsimbouri
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Karl V Burgess
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, Bearsden, Glasgow, G61 1QH, UK
| | - Gavin Blackburn
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, Garscube Campus, Bearsden, Glasgow, G61 1QH, UK
| | - Vineetha Jayawarna
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Mariana A G Oliva
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Jennifer Willis
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Jaspreet Bansal
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - Paul Reynolds
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Julia A Wells
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK
| | - Joanne Mountford
- Scottish National Blood Transfusion Service, Advanced Therapeutics, Jack Copland Centre, 52 Research Avenue North, Heriot Watt Research Park, Edinburgh, EH14 4BE, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, UK.
| |
Collapse
|
5
|
Tompkins YH, Liu G, Kim WK. Impact of exogenous hydrogen peroxide on osteogenic differentiation of broiler chicken compact bones derived mesenchymal stem cells. Front Physiol 2023; 14:1124355. [PMID: 36776980 PMCID: PMC9909420 DOI: 10.3389/fphys.2023.1124355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
The effects of hydrogen peroxide (H2O2) on the osteogenic differentiation of primary chicken mesenchymal stem cells (MSCs) were investigated. MSCs were subjected to an osteogenic program and exposed to various concentrations of H2O2 for 14 days. Results showed that high concentrations of H2O2 (200 and 400 nM) significantly increased pro-apoptotic marker CASP8 expression and impaired osteogenic differentiation, as indicated by decreased mRNA expression levels of osteogenesis-related genes and reduced in vitro mineralization. In contrast, long-term H2O2 exposure promoted basal expression of adipogenic markers at the expense of osteogenesis in MSCs during osteogenic differentiation, and increased intracellular reactive oxygen species (ROS) production, as well as altered antioxidant enzyme gene expression. These findings suggest that long-term H2O2-induced ROS production impairs osteogenic differentiation in chicken MSCs under an osteogenic program.
Collapse
|
6
|
Matthews EZ, Lanham S, White K, Kyriazi ME, Alexaki K, El-Sagheer AH, Brown T, Kanaras AG, J West J, MacArthur BD, Stumpf PS, Oreffo ROC. Single-cell RNA-sequence analysis of human bone marrow reveals new targets for isolation of skeletal stem cells using spherical nucleic acids. J Tissue Eng 2023; 14:20417314231169375. [PMID: 37216034 PMCID: PMC10192814 DOI: 10.1177/20417314231169375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/24/2023] [Indexed: 05/24/2023] Open
Abstract
There is a wealth of data indicating human bone marrow contains skeletal stem cells (SSC) with the capacity for osteogenic, chondrogenic and adipogenic differentiation. However, current methods to isolate SSCs are restricted by the lack of a defined marker, limiting understanding of SSC fate, immunophenotype, function and clinical application. The current study applied single-cell RNA-sequencing to profile human adult bone marrow populations from 11 donors and identified novel targets for SSC enrichment. Spherical nucleic acids were used to detect these mRNA targets in SSCs. This methodology was able to rapidly isolate potential SSCs found at a frequency of <1 in 1,000,000 in human bone marrow, with the capacity for tri-lineage differentiation in vitro and ectopic bone formation in vivo. The current studies detail the development of a platform to advance SSC enrichment from human bone marrow, offering an invaluable resource for further SSC characterisation, with significant therapeutic impact therein.
Collapse
Affiliation(s)
- Elloise Z Matthews
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Stuart Lanham
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| | - Kate White
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
| | - Maria-Eleni Kyriazi
- College of Engineering and Technology,
American University of the Middle East, Kuwait
| | - Konstantina Alexaki
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Afaf H El-Sagheer
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
- Chemistry Branch, Department of Science
and Mathematics, Faculty of Petroleum and Mining Engineering, Suez University, Suez,
Egypt
| | - Tom Brown
- Department of Chemistry, Chemistry
Research Laboratory, University of Oxford, Oxford, UK
| | - Antonios G Kanaras
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
| | - Jonathan J West
- Cancer Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
- Physics and Astronomy, Faculty of
Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Ben D MacArthur
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- Mathematical Sciences, University of
Southampton, Southampton, UK
| | - Patrick S Stumpf
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Joint Research Center for Computational
Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Richard OC Oreffo
- Faculty of Medicine, Centre for Human
Development, Stem Cells and Regeneration, Human Development and Health, Institute of
Developmental Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, University
of Southampton, Southampton, UK
- College of Biomedical Engineering,
China Medical University, Taichung, Taiwan
| |
Collapse
|
7
|
Walther AR, Ditzel N, Kassem M, Andersen MØ, Hedegaard MAB. In vivo non-invasive monitoring of tissue development in 3D printed subcutaneous bone scaffolds using fibre-optic Raman spectroscopy. BIOMATERIALS AND BIOSYSTEMS 2022; 7:100059. [PMID: 36824488 PMCID: PMC9934492 DOI: 10.1016/j.bbiosy.2022.100059] [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: 02/24/2022] [Revised: 07/04/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022] Open
Abstract
The development of novel biomaterials for regenerative therapy relies on the ability to assess tissue development, quality, and similarity with native tissue types in in vivo experiments. Non-invasive imaging modalities such as X-ray computed tomography offer high spatial resolution but limited biochemical information while histology and biochemical assays are destructive. Raman spectroscopy is a non-invasive, label-free and non-destructive technique widely applied for biochemical characterization. Here we demonstrate the use of fibre-optic Raman spectroscopy for in vivo quantitative monitoring of tissue development in subcutaneous calcium phosphate scaffolds in mice over 16 weeks. Raman spectroscopy was able to quantify the time dependency of different tissue components related to the presence, absence, and quantity of mesenchymal stem cells. Scaffolds seeded with stem cells produced 3-5 times higher amount of collagen-rich extracellular matrix after 16 weeks implantation compared to scaffolds without. These however, showed a 2.5 times higher amount of lipid-rich tissue compared to implants with stem cells. Ex vivo micro-computed tomography and histology showed stem cell mediated collagen and bone development. Histological measures of collagen correlated well with Raman derived quantifications (correlation coefficient in vivo 0.74, ex vivo 0.93). In the absence of stem cells, the scaffolds were largely occupied by adipocytes. The technique developed here could potentially be adapted for a range of small animal experiments for assessing tissue engineering strategies at the biochemical level.
Collapse
Affiliation(s)
- Anders Runge Walther
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Nicholas Ditzel
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Moustapha Kassem
- Endocrine Research (KMEB), Department of Endocrinology, Odense University Hospital and University of Southern Denmark, J.B. Winsløws Vej 25, DK-5000 Odense, Denmark
| | - Morten Østergaard Andersen
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| | - Martin Aage Barsøe Hedegaard
- SDU Biotechnology, Department of Green Technology, University of Southern Denmark, Campusvej 55, DK-5230 Odense, Denmark
| |
Collapse
|
8
|
Chen C, Yan S, Geng Z, Wang Z. Fracture repair by IOX2: Regulation of the hypoxia inducible factor-1α signaling pathway and BMSCs. Eur J Pharmacol 2022; 921:174864. [PMID: 35219731 DOI: 10.1016/j.ejphar.2022.174864] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
The treatment of fracture delayed union and nonunion has become a challenging problem. Hypoxia inducible factor-1α (HIF-1α) is reported to be a key factor in fracture healing, and is degraded by hydroxylation of prolyl hydroxylase (PHDs) under normal oxygen. Small molecules could inhibit the activity of PHDs, stabilize HIF-1α protein, regulate the expression of downstream target genes of HIF-1α, and make the body adapt to hypoxia. The migration and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is the most promising candidate for the treatment of fracture nonunion. Here we reported that IOX2, an HIF-1α PHD inhibitor, markedly improved the proliferation and migration of BMSCs by upregulating intracellular Ca2+ and concomitant decreasing reactive oxygen species (ROS) in vitro, and facilitated the repair of bone fracture by increasing the number of BMSCs and cartilage formation in vivo. No significant influence of IOX2 on the proliferation and migration of BMSCs after silencing of the HIF-1α. Together, our findings indicated that IOX2 promoted the proliferation and migration of BMSCs via the HIF-1α pathway and further accelerated fracture healing. These results provide a deeper understanding of the mechanism by which HIF promotes fracture healing.
Collapse
Affiliation(s)
- Chunxia Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China; Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, Henan University, Kaifeng, 475004, China
| | - Shihai Yan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China; Department of Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Zhirong Geng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
9
|
The Effect of Diabetes Mellitus on IGF Axis and Stem Cell Mediated Regeneration of the Periodontium. Bioengineering (Basel) 2021; 8:bioengineering8120202. [PMID: 34940355 PMCID: PMC8698546 DOI: 10.3390/bioengineering8120202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Periodontitis and diabetes mellitus (DM) are two of the most common and challenging health problems worldwide and they affect each other mutually and adversely. Current periodontal therapies have unpredictable outcome in diabetic patients. Periodontal tissue engineering is a challenging but promising approach that aims at restoring periodontal tissues using one or all of the following: stem cells, signalling molecules and scaffolds. Mesenchymal stem cells (MSCs) and insulin-like growth factor (IGF) represent ideal examples of stem cells and signalling molecules. This review outlines the most recent updates in characterizing MSCs isolated from diabetics to fully understand why diabetics are more prone to periodontitis that theoretically reflect the impaired regenerative capabilities of their native stem cells. This characterisation is of utmost importance to enhance autologous stem cells based tissue regeneration in diabetic patients using both MSCs and members of IGF axis.
Collapse
|
10
|
Alghfeli L, Parambath D, Manzoor S, Roach HI, Oreffo RO, El-Serafi AT. Synthesis of scaffold-free, three dimensional, osteogenic constructs following culture of skeletal osteoprogenitor cells on glass surfaces. Bone Rep 2021; 15:101143. [PMID: 34746337 PMCID: PMC8554168 DOI: 10.1016/j.bonr.2021.101143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Efficient differentiation of stem cells into three-dimensional (3D) osteogenic construct is still an unmet challenge. These constructs can be crucial for patients with bone defects due to congenital or traumatic reasons. The modulation of cell fate and function as a consequence of interaction with the physical and chemical properties of materials is well known. METHODS The current study has examined the osteogenic differentiation potential of human skeletal populations following culture on glass surfaces, as a monolayer, or in glass tubes as a pellet culture. The 3D prosperities were assessed morphometrically and the differentiation was evaluated through molecular characterization as well as matrix formation. RESULTS Early temporal expression of alkaline phosphatase expression of skeletal populations was observed following culture on glass surfaces. Skeletal populations seeded on glass tubes, adhered as a monolayer to the tube base and subsequently formed 3D pellets at the air -media interface. The pellets cultured on glass displayed 4.9 ± 1.3 times the weight and 2.9 ± 0.1 the diameter of their counterpart cultured in plastic tubes and displayed enhanced production of osteogenic matrix proteins, such a collagen I and osteonectin. The size and weight of the pellets correlated with surface area in contrast to cell numbers seeded. Global DNA methylation level was decreased in pellets cultured on glass. In contrast, gene expression analysis confirmed upregulation extracellular matrix proteins and osteogenesis-related growth factors. CONCLUSION This simple approach to the culture of skeletal cells on glass tubes provides a scaffold-free, 3D construct platform for generating pellets enabling analysis and evaluation of tissue development and integration of multiple constructs with implications for tissue repair and regenerative application on scale-up.
Collapse
Key Words
- 3D culture
- 3D, three-dimensional
- A/S, Alcian blue/Sirius red/Weigert's haematoxylin
- ALP, Alkaline Phosphatase
- BMP, bone morphogenic protein
- BMSC, human bone marrow stromal
- CSF, colony stimulating factor
- Ct, Cycle threshold
- Differentiation
- EGF, epidermal growth factor
- FC, Fetal bone cells
- FCS, Fetal Calf Serum
- FGF, fibroblast growth factor
- FN1, fibronectin
- GLI, GLI family zinc finger 1
- HIPPIE, Human Integrated Protein Interaction Reference
- ITGA3, integrin A3
- MMP, matrix metalloprotease
- Osteogenesis
- Osteoprogenitor cells
- P/S, penicillin and streptomycin
- Pellets
- R, receptor
- TGF, β transforming growth factor beta
- TGFBR2 transforming growth factor beta receptor 2 VDR, vitamin D receptor
- gDNA, genomic DNA
- iMSC, immortalized human bone marrow derived, mesenchymal stem cells
- vWF, von Willebrand factor
Collapse
Affiliation(s)
- Latifa Alghfeli
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Divyasree Parambath
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Shaista Manzoor
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
| | - Helmtrud I. Roach
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Richard O.C. Oreffo
- Bone and Joint Research Group, Institute of Developmental Sciences, University of Southampton, School of Medicine, UK
| | - Ahmed T. El-Serafi
- Research Institute for Medical and Health Sciences, University of Sharjah, United Arab Emirates
- Medical Biochemistry Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Sweden
| |
Collapse
|
11
|
Flück M, Kasper S, Benn MC, Clement Frey F, von Rechenberg B, Giraud MN, Meyer DC, Wieser K, Gerber C. Transplant of Autologous Mesenchymal Stem Cells Halts Fatty Atrophy of Detached Rotator Cuff Muscle After Tendon Repair: Molecular, Microscopic, and Macroscopic Results From an Ovine Model. Am J Sports Med 2021; 49:3970-3980. [PMID: 34714701 PMCID: PMC8649427 DOI: 10.1177/03635465211052566] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/13/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND The injection of mesenchymal stem cells (MSCs) mitigates fat accumulation in released rotator cuff muscle after tendon repair in rodents. PURPOSE To investigate whether the injection of autologous MSCs halts muscle-to-fat conversion after tendon repair in a large animal model for rotator cuff tendon release via regional effects on extracellular fat tissue and muscle fiber regeneration. STUDY DESIGN Controlled laboratory study. METHODS Infraspinatus (ISP) muscles of the right shoulder of Swiss Alpine sheep (n = 14) were released by osteotomy and reattached 16 weeks later without (group T; n = 6) or with (group T-MSC; n = 8) electropulse-assisted injection of 0.9 Mio fluorescently labeled MSCs as microtissues with media in demarcated regions; animals were allowed 6 weeks of recovery. ISP volume and composition were documented with computed tomography and magnetic resonance imaging. Area percentages of muscle fiber types, fat, extracellular ground substance, and fluorescence-positive tissue; mean cross-sectional area (MCSA) of muscle fibers; and expression of myogenic (myogenin), regeneration (tenascin-C), and adipogenic markers (peroxisome proliferator-activated receptor gamma [PPARG2]) were quantified in injected and noninjected regions after recovery. RESULTS At 16 weeks after tendon release, the ISP volume was reduced and the fat fraction of ISP muscle was increased in group T (137 vs 185 mL; 49% vs 7%) and group T-MSC (130 vs 166 mL; 53% vs 10%). In group T-MSC versus group T, changes during recovery after tendon reattachment were abrogated for fat-free mass (-5% vs -29%, respectively; P = .018) and fat fraction (+1% vs +24%, respectively; P = .009%). The area percentage of fat was lower (9% vs 20%; P = .018) and the percentage of the extracellular ground substance was higher (26% vs 20%; P = .007) in the noninjected ISP region for group T-MSC versus group T, respectively. Regionally, MCS injection increased tenascin-C levels (+59%) and the water fraction, maintaining the reduced PPARG2 levels but not the 29% increased fiber MCSA, with media injection. CONCLUSION In a sheep model, injection of autologous MSCs in degenerated rotator cuff muscle halted muscle-to-fat conversion during recovery from tendon repair by preserving fat-free mass in association with extracellular reactions and stopping adjuvant-induced muscle fiber hypertrophy. CLINICAL RELEVANCE A relatively small dose of MSCs is therapeutically effective to halt fatty atrophy in a large animal model.
Collapse
Affiliation(s)
- Martin Flück
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
| | - Stephanie Kasper
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
| | - Mario C. Benn
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Flurina Clement Frey
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Marie-Noëlle Giraud
- Cardiology, Faculty of Sciences and
Medicine, University of Fribourg, Fribourg, Switzerland
| | - Dominik C. Meyer
- Author deceased
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| | - Karl Wieser
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| | - Christian Gerber
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| |
Collapse
|
12
|
Kirankumar S, Gurusamy N, Rajasingh S, Sigamani V, Vasanthan J, Perales SG, Rajasingh J. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Exp Biol Med (Maywood) 2021; 247:433-445. [PMID: 34648374 DOI: 10.1177/15353702211052927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The process of bone repair has always been a natural mystery. Although bones do repair themselves, supplemental treatment is required for the initiation of the self-regeneration process. Predominantly, surgical procedures are employed for bone regeneration. Recently, cell-based therapy for bone regeneration has proven to be more effective than traditional methods, as it eliminates the immune risk and painful surgeries. In clinical trials, various stem cells, especially mesenchymal stem cells, have shown to be more efficient for the treatment of several bone-related diseases, such as non-union fracture, osteogenesis imperfecta, osteosarcoma, and osteoporosis. Furthermore, the stem cells grown in a suitable three-dimensional scaffold support were found to be more efficient for osteogenesis. It has been shown that the three-dimensional bioscaffolds support and simulate an in vivo environment, which helps in differentiation of stem cells into bone cells. Bone regeneration in patients with bone disorders can be improved through modification of stem cells with several osteogenic factors or using stem cells as carriers for osteogenic factors. In this review, we focused on the various types of stem cells and scaffolds that are being used for bone regeneration. In addition, the molecular mechanisms of various transcription factors, signaling pathways that support bone regeneration and the senescence of the stem cells, which limits bone regeneration, have been discussed.
Collapse
Affiliation(s)
- Shivaani Kirankumar
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sheeja Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Vinoth Sigamani
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jayavardini Vasanthan
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Genetic Engineering, 93104SRM Institute of Science and Technology, Chennai 603203, India
| | - Selene G Perales
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johnson Rajasingh
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
13
|
Mahmoud E, Sayed M, El-Kady AM, Elsayed H, Naga S. In vitro and in vivo study of naturally derived alginate/hydroxyapatite bio composite scaffolds. Int J Biol Macromol 2020; 165:1346-1360. [DOI: 10.1016/j.ijbiomac.2020.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/19/2020] [Accepted: 10/01/2020] [Indexed: 12/25/2022]
|
14
|
Elangovan S, Gajendrareddy P, Ravindran S, Salem AK. Emerging local delivery strategies to enhance bone regeneration. ACTA ACUST UNITED AC 2020; 15:062001. [PMID: 32647095 PMCID: PMC10148649 DOI: 10.1088/1748-605x/aba446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In orthopedics and dentistry there is an increasing need for novel biomaterials and clinical strategies to achieve predictable bone regeneration. These novel molecular strategies have the potential to eliminate the limitations of currently available approaches. Specifically, they have the potential to reduce or eliminate the need to harvest autogenous bone, and the overall complexity of the clinical procedures. In this review, emerging tissue engineering strategies that have been, or are currently being, developed based on the current understanding of bone biology, development and wound healing will be discussed. In particular, protein/peptide based approaches, DNA/RNA therapeutics, cell therapy, and the use of exosomes will be briefly covered. The review ends with a summary of the current status of these approaches, their clinical translational potentials and their challenges.
Collapse
Affiliation(s)
- Satheesh Elangovan
- Department of Periodontics, The University of Iowa College of Dentistry, Iowa City, IA 52242, United States of America
| | | | | | | |
Collapse
|
15
|
Chen C, Yan S, Qiu S, Geng Z, Wang Z. HIF/Ca 2+/NO/ROS is critical in roxadustat treating bone fracture by stimulating the proliferation and migration of BMSCs. Life Sci 2020; 264:118684. [PMID: 33129877 DOI: 10.1016/j.lfs.2020.118684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
AIMS Fracture site is regionally hypoxic resulting from vasculature disruption. HIF-1αplays an essential role in fracture repair. This study aims to investigate the influence of FG4592 on the femur fracture of SD rats and the proliferation, migration of BMSCs. MATERIALS AND METHODS After the femoral fracture model was established, computed tomography imaging and histological analyses were used to quantify bone healing and the expression of CD90, HIF-1α, VEGF were observed by means of immunohistochemistry method on Day 10 and Day 20. In addition, CCK-8 assay, transwell, flow cytometric analysis, laser confocal microscopy assay, western blot and rT-PCR were performed to text the proliferation and migration of BMSCs using FG4592. KEY FINDINGS In vivo, FG4592 facilitated the repair of bone fracture by increasing the number of BMSCs and cartilage formation. In vitro, FG4592 markedly improved the proliferation, migration of BMSCs via upregulation of intracellular Ca2+, NO and concomitant decrease of ROS. Gene silencing of HIF-1α resulted in the opposite phenomenon in BMSCs with the treatment of FG4592. SIGNIFICANCE The transplantation of BMSCs is the most promising candidate for the treatment of fracture non-union. We illustrated that FG4592 promoted the proliferation, migration of BMSCs via the HIF/Ca2+/NO/ROS pathway and further accelerated fracture healing. These results provide a deeper understanding for the mechanism of HIF in promoting fracture healing.
Collapse
Affiliation(s)
- Chunxia Chen
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210023, PR China
| | - Shihai Yan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210023, PR China; Department of Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China
| | - Shuang Qiu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210023, PR China
| | - Zhirong Geng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210023, PR China.
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructure, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
16
|
Guided Bone Regeneration with Ammoniomethacrylate-Based Barrier Membranes in a Radial Defect Model. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5905740. [PMID: 33150177 PMCID: PMC7603551 DOI: 10.1155/2020/5905740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/24/2020] [Indexed: 11/17/2022]
Abstract
Large bone defects pose an unsolved challenge for orthopedic surgeons. Our group has previously reported the construction of a barrier membrane made of ammoniomethacrylate copolymer USP (AMCA), which supports the adhesion, proliferation, and osteoblastic differentiation of human mesenchymal stem cells (hMSCs). In this study, we report the use of AMCA membranes to seclude critical segmental defect (~1.0 cm) created in the middle third of rabbit radius and test the efficiency of bone regeneration. Bone regeneration was assessed by radiography, biweekly for 8 weeks. The results were verified by histology and micro-CT at the end of the follow-up. The AMCA membranes were found superior to no treatment in terms of new bone formation in the defect, bone volume, callus surface area normalized to total volume, and the number of bone trabeculae, after eight weeks. Additional factors were then assessed, and these included the addition of simvastatin to the membrane, coating the membrane with human MSC, and a combination of those. The addition of simvastatin to the membranes demonstrated a stronger effect at a similar radiological follow-up. We conclude that AMCA barrier membranes per se and simvastatin delivered in a controlled manner improve bone regeneration outcome.
Collapse
|
17
|
Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater 2020; 6:666-683. [PMID: 33005830 PMCID: PMC7509590 DOI: 10.1016/j.bioactmat.2020.08.014] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022] Open
Abstract
Reconstruction of bone defects, especially the critical-sized defects, with mechanical integrity to the skeleton is important for a patient's rehabilitation, however, it still remains challenge. Utilizing biomaterials of human origin bone tissue for therapeutic purposes has provided a facilitated approach that closely mimics the critical aspects of natural bone tissue with regard to its properties. However, not only efficacious and safe but also cost-effective and convenient are important for regenerative biomaterials to achieve clinical translation and commercial success. Advances in our understanding of regenerative biomaterials and their roles in new bone formation potentially opened a new frontier in the fast-growing field of regenerative medicine. Taking inspiration from the role and multicomponent construction of native extracellular matrix (ECM) for cell accommodation, the ECM-mimicking biomaterials and the naturally decellularized ECM scaffolds were used to create new tissues for bone restoration. On the other hand, with the going deep in understanding of mesenchymal stem cells (MSCs), they have shown great promise to jumpstart and facilitate bone healing even in diseased microenvironments with pharmacology-based endogenous MSCs rescue/mobilization, systemic/local infusion of MSCs for cytotherapy, biomaterials-based approaches, cell-sheets/-aggregates technology and usage of subcellular vesicles of MSCs to achieve scaffolds-free or cell-free delivery system, all of them have been shown can improve MSCs-mediated regeneration in preclinical studies and several clinical trials. Here, following an overview discussed autogenous/allogenic and ECM-based bone biomaterials for reconstructive surgery and applications of MSCs-mediated bone healing and tissue engineering to further offer principles and effective strategies to optimize MSCs-based bone regeneration. Focusing on MSCs based bone regeneration. Discussed cytotherapy, cell-free therapies and cell-aggregates technology in detail. Stating the approaches of MSCs in diseased microenvironments.
Collapse
Affiliation(s)
- Fengqing Shang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Stomatology, The 306th Hospital of PLA, Beijing, 100101, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leiguo Ming
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongjie Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, 850000, China
| | - Jiayu Zhao
- Bureau of Service for Veteran Cadres of PLA in Beijing, Beijing, 100001, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Corresponding author.
| |
Collapse
|
18
|
Dilogo IH, Rahmatika D, Pawitan JA, Liem IK, Kurniawati T, Kispa T, Mujadid F. Allogeneic umbilical cord-derived mesenchymal stem cells for treating critical-sized bone defects: a translational study. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2020; 31:265-273. [PMID: 32804289 DOI: 10.1007/s00590-020-02765-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION The current 'gold-standard' treatment of critical-sized bone defects (CSBDs) is autografts; however, they have drawbacks including lack of massive bone source donor site morbidity, incomplete remodeling, and the risk of infection. One potential treatment for treating CSBDs is bone marrow-derived mesenchymal stem cells (BM-MSCs). Previously, there were no studies regarding the use of human umbilical cord-mesenchymal stem cells (hUC-MSCs) for treating BDs. We aim to investigate the use of allogeneic hUC-MSCs for treating CSBDs. METHOD We included subjects who were diagnosed with non-union fracture with CSBDs who agreed to undergo hUC-MSCs implantation. All patients were given allogeneic hUC-MSCs. All MSCs were obtained and cultured using the multiple-harvest explant method. Subjects were evaluated functionally using the Lower Extremity Functional Scale (LEFS) and radiologically by volume defect reduction. RESULT A total of seven (3 male, 4 female) subjects were recruited for this study. The subjects age ranged from 14 to 62 years. All seven subjects had increased LEFS during the end of the follow-up period, indicating improved functional ability. The follow-up period ranged from 12 to 36 months. One subject had wound dehiscence and infection, and two subjects developed partial union. CONCLUSION Umbilical cord mesenchymal stem cells are a potential new treatment for CSBDs. Additional studies with larger samples and control groups are required to further investigate the safety and efficacy of umbilical cord-derived mesenchymal stem cells for treating CSBDs.
Collapse
Affiliation(s)
- Ismail Hadisoebroto Dilogo
- Department of Orthopaedic and Traumatology, Dr. Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia.
- Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
| | - Dina Rahmatika
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
| | - Jeanne Adiwinata Pawitan
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
- Department Histology, Faculty of Medicine, Universitas Indonesia, Jl. Salemba 6, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Isabella Kurnia Liem
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
- Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jl. Salemba 6, Jakarta, Indonesia
| | - Tri Kurniawati
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
- Stem Cell and Tissue Engineering Research Center, IMERI, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Tera Kispa
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
| | - Fajar Mujadid
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, CMU 2 Building 5th Floor, Jl. Diponegoro 71, Jakarta Pusat, Indonesia
| |
Collapse
|
19
|
Xu GP, Zhang XF, Sun L, Chen EM. Current and future uses of skeletal stem cells for bone regeneration. World J Stem Cells 2020; 12:339-350. [PMID: 32547682 PMCID: PMC7280866 DOI: 10.4252/wjsc.v12.i5.339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.
Collapse
Affiliation(s)
- Guo-Ping Xu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiang-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Lu Sun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, United States
| | - Er-Man Chen
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
20
|
Rostami F, Tamjid E, Behmanesh M. Drug-eluting PCL/graphene oxide nanocomposite scaffolds for enhanced osteogenic differentiation of mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111102. [PMID: 32600706 DOI: 10.1016/j.msec.2020.111102] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
Recently, drug-eluting nanofibrous scaffolds have attracted a great attention to enhance the cell differentiation through biomimicking the extracellular matrix (ECM) in regenerative medicine. In this study, electrospun nanocomposite polycaprolactone (PCL)-based scaffolds containing synthesized graphene oxide (GO) nanosheets and osteogenic drugs, i.e. dexamethasone and simvastatin were fabricated. The physicochemical and surface properties of the scaffolds were investigated through FTIR, wettability, pH, and drug release studies. The cell viability, differentiation, and biomineralization were studied on mesenchymal stem cells (MSCs) by Alamar Blue, alkaline phosphatase (ALP) activity, and Alizarin Red-S staining, respectively. Uniformly distributed GO (thickness < 1 nm) in PCL nanofibers was observed by electron microscopy. It was revealed that the addition of GO and the drugs improved the hydrophilicity, cell viability, and osteogenic differentiation, in addition to pH changes, in comparison with PCL scaffolds. Despite the notable reduction in the cell viability, significant differentiation was revealed by ALP assay on PCL/GO-Dex scaffolds. Noteworthy, a twofold increase in the osteogenic differentiation was observed in comparison with the cells cultured in osteogenic differentiation medium, while a significant biomineralization was observed. The results of this study indicate the synergistic effect of GO and dexamethasone on improving osteogenic differentiation of drug-eluting nanocomposite scaffolds in bone tissue engineering applications.
Collapse
Affiliation(s)
- Fatemeh Rostami
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elnaz Tamjid
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mehrdad Behmanesh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran; Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
21
|
Padilla S, Benito-Garzón L, Enciso Sanz S, Garzón-Gutiérrez A, García Carrodeguas R, Rodríguez MA, Garcia de Castro A, Canillas M. Novel Osteoinductive and Osteogenic Scaffolds of Monetite, Amorphous Calcium Phosphate, Hydroxyapatite, and Silica Gel: Influence of the Hydroxyapatite/Monetite Ratio on Their In Vivo Behavior and on Their Physical and Chemical Properties. ACS Biomater Sci Eng 2020; 6:3440-3453. [DOI: 10.1021/acsbiomaterials.9b01689] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Sussette Padilla
- Departamento de Química en Ciencias Farmacéuticas, Facultad Farmacia, Universidad Complutense, Madrid 28040, Spain
- AzureBio SL, Tres Cantos, Madrid 28760, Spain
| | - Lorena Benito-Garzón
- Departamento de Cirugía, Facultad de Medicina, Universidad de Salamanca, Salamanca 37008, Spain
| | | | | | | | | | | | - María Canillas
- Instituto de Cerámica y Vidrio, CSIC, Cantoblanco, Madrid 28049, Spain
| |
Collapse
|
22
|
López-Fernández A, Barro V, Ortiz-Hernández M, Manzanares MC, Vivas D, Vives J, Vélez R, Ginebra MP, Aguirre M. Effect of Allogeneic Cell-Based Tissue-Engineered Treatments in a Sheep Osteonecrosis Model. Tissue Eng Part A 2020; 26:993-1004. [PMID: 32122263 DOI: 10.1089/ten.tea.2019.0339] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is defined as a tissue disorder and successive subchondral bone collapse resulting from an ischemic process, which may progress to hip osteoarthritis. Cell therapy with multipotent bone marrow mesenchymal stromal cells (BM-MSC) of autologous origin appears to be safe and has shown regenerative potential in previous preclinical and clinical studies. The use of allogeneic cells is far more challenging, but may be a promising alternative to use of autologous cells. Moreover, an optimized dosage of cells from an allogeneic source is needed to obtain off-the-shelf tissue engineering products (TEPs). The purpose of this study was to evaluate the efficacy of a TEP composed of undifferentiated ex vivo expanded BM-MSC of allogeneic origin, combined with bone matrix particles in variable doses. A comparative analysis of TEP's bone regenerative properties against its autologous counterpart was performed in an early-stage ONFH preclinical model in mature sheep. Allogeneic BM-MSC groups demonstrated bone regeneration capacity in osteonecrotic lesions equivalent to autologous BM-MSC groups 6 weeks after treatment. Likewise, stimulation of bone regeneration by a low cell dose of 0.5 × 106 BM-MSC/cm3 was equivalent to that of a high cell dose, 5 × 106 BM-MSC/cm3. Neither local nor systemic immunological reactions nor tumorigenesis were reported, strengthening the safety profile of allogeneic BM-MSC therapy in this model. Our results suggest that low-dose allogeneic BM-MSC is sufficient to promote bone regeneration in femoral head osteonecrotic lesions, and should be considered in translation of new allogeneic cell-based TEPs to human clinics. Impact statement Cell therapy and tissue engineering hold promise as novel regenerative therapies for musculoskeletal diseases, and particularly in bone regeneration strategies. In this article, we report the evaluation of the efficacy of an allogeneic cell-based tissue engineering product (TEP) in an early-stage osteonecrosis of the femoral head preclinical model in skeletally mature sheep. Moreover, we demonstrate its bone regeneration capacity and safety in vivo and its equivalence to autologous counterparts. These findings have important implications for the translation of new allogeneic cell-based TEPs to human clinics.
Collapse
Affiliation(s)
- Alba López-Fernández
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Víctor Barro
- Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Mònica Ortiz-Hernández
- Biomaterials, Biomechanics and Tissue Engineering Group (BBT), Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain
| | - Maria Cristina Manzanares
- Human Anatomy and Embryology Unit, Department of Pathology and Experimental Therapeutics, Universitat de Barcelona, Barcelona, Spain
| | - Daniel Vivas
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Barcelona, Spain
| | - Joaquim Vives
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Servei de Teràpia Cel·lular, Banc de Sang i Teixits, Barcelona, Spain.,Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Roberto Vélez
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Maria Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group (BBT), Department of Materials Science and Metallurgical Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona, Spain.,Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Màrius Aguirre
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Orthopedic Surgery Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
23
|
hCTLA4-Gene-Modified Human Bone Marrow-Derived Mesenchymal Stem Cells (hBMMSCs) Maintain POSTN Secretion to Enhance the Migration Capability of Allogeneic hBMMSCs through the Integrin αv β3/FAK/ERK Signaling Pathway. Stem Cells Int 2020; 2020:3608284. [PMID: 32269594 PMCID: PMC7128042 DOI: 10.1155/2020/3608284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 02/04/2023] Open
Abstract
Cytotoxic T-lymphocyte-associated protein 4- (CTLA4-) modified human bone marrow-derived mesenchymal stem cells (hBMMSCs) might be promising seed cells for bone tissue engineering. However, the underlying mechanism is not clear. In the present study, we investigated whether CTLA4-modified hBMMSCs are involved in the migration of allogeneic hBMMSCs (allo-hBMMSCs) by maintaining POSTN secretion. hBMMSCs were isolated from different groups, named hBMMSCs and allo-hBMMSCs. hBMMSCs that were infected with the negative control (NC), empty adenovirus- or recombinant adenovirus-expressing CTLA4, POSTN, or CTLA4 plus the shRNA of POSTN were named NC hBMMSCs, CTLA4-modified hBMMSCs, POSTN-modified hBMMSCs, or CTLA4+shPOSTN-modified hBMMSCs, respectively. They were then cocultured with PBMCs in a 1 : 5 ratio with 2.5 μg/mL phytohemagglutinin (PHA). The coculture supernatant was collected to treat allo-hBMMSCs with anti-integrin αvβ3 IgG, or negative control IgG, as a control. Following this, ELISA, Transwell assays, wound healing assays, and western blotting were performed. We found that the POSTN level was higher in the culture supernatant of CTLA4- and POSTN-modified hBMMSCs than in NC hBMMSCs cocultured with PBMCs treated with PHA. The migration capability of allo-hBMMSCs was enhanced, and the integrin αvβ3/FAK/ERK signaling pathway in allo-hBMMSCs was activated by the culture supernatant of CTLA4- and POSTN-modified hBMMSCs cocultured with PBMCs treated with PHA. Additionally, these induced effects can be weakened by POSTN knockdown, and the migration capability of allo-hBMMSCs was blocked by anti-integrin αvβ3 IgG. In conclusion, hCTLA4-gene-modified hBMMSCs maintain POSTN secretion to enhance the migration capability of allogeneic hBMMSCs through the integrin αvβ3/FAK/ERK signaling pathway in the T cell immune activation environment.
Collapse
|
24
|
Kim JW, Park JH, Muthukumar T, Shin EY, Shin ME, Song JE, Khang G. Accelerating bone defects healing in calvarial defect model using 3D cultured bone marrow-derived mesenchymal stem cells on demineralized bone particle scaffold. J Tissue Eng Regen Med 2020; 14:563-574. [PMID: 32061025 DOI: 10.1002/term.3020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 11/10/2022]
Abstract
Bone defects are usually difficult to be regenerated due to pathological states or the size of the injury. Researchers are focusing on tissue engineering approaches in order to drive the regenerative events, using stem cells to regenerate bone. The purpose of this study is to evaluate the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) on biologically derived Gallus gallus domesticus-derived demineralized bone particle (GDD) sponge. The sponges were prepared by freeze-drying method using 1, 2, and 3 wt% GDD and cross-linked with glutaraldehyde. The GDD sponge was characterized using scanning electron microscopy, compressive strength, porosity, and Fourier transform infrared. The potential bioactivity of the sponge was evaluated by osteogenic differentiation of BMSCs using 3(4, dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay and quantifying alkaline phosphatase (ALP) activity. in vivo experiments were evaluated through a micro-computerized tomography (μ-CT) and histological assays. The analysis confirmed that an increase in the concentration of the GDD in the sponge leads to a higher bone formation and deposition in rat calvarial defects. Histological assay results were in line with μ-CT. The results reported in this study demonstrated the potential application of GDD sponges as osteoinductor in bone tissue engineering in pathological or nonunion bone defects.
Collapse
Affiliation(s)
- Jin Woo Kim
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Jong Ho Park
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Thangavelu Muthukumar
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Eun Yeong Shin
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Myeong Eun Shin
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Jeong Eun Song
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Gilson Khang
- Department of BIN Convergence Technology, Department of PolymerNano Science & Technology and Polymer BIN Research Center, Jeonbuk National University, Jeonju-si, Republic of Korea
| |
Collapse
|
25
|
Lee YC, Chan YH, Hsieh SC, Lew WZ, Feng SW. Comparing the Osteogenic Potentials and Bone Regeneration Capacities of Bone Marrow and Dental Pulp Mesenchymal Stem Cells in a Rabbit Calvarial Bone Defect Model. Int J Mol Sci 2019; 20:ijms20205015. [PMID: 31658685 PMCID: PMC6834129 DOI: 10.3390/ijms20205015] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
The bone regeneration efficiency of bone marrow mesenchymal stem cells (BMSCs) and dental pulp mesenchymal stem cells (DPSCs) combined with xenografts in the craniofacial region remains unclear. Accordingly, this study commenced by comparing the cell morphology, cell proliferation, trilineage differentiation, mineral synthesis, and osteogenic gene expression of BMSCs and DPSCs in vitro. Four experimental groups (empty control, Bio-Oss only, Bio-Oss+BMSCs, and Bio-Oss+DPSCs) were then designed and implanted in rabbit calvarial defects. The BMSCs and DPSCs showed a similar morphology, proliferative ability, surface marker profile, and trilineage-differentiation potential in vitro. However, the BMSCs exhibited a higher mineral deposition and expression levels of osteogenic marker genes, including alkaline phosphatase (ALP), runt related transcription factor 2 (RUNX2), and osteocalcin (OCN). In the in vivo studies, the bone volume density in both MSC groups was significantly greater than that in the empty control or Bio-Oss only group. Moreover, the new bone formation and Collagen I / osteoprotegerin protein expressions of the scaffold+MSC groups were higher than those of the Bio-Oss only group. Finally, the Bio-Oss+BMSC and Bio-Oss+DPSC groups had a similar bone mineral density, new bone formation, and osteogenesis-related protein expression. Overall, the DPSCs seeded on Bio-Oss matched the bone regeneration efficacy of BMSCs in vivo and hence appear to be a promising strategy for craniofacial defect repair in future clinical applications.
Collapse
Affiliation(s)
- Yu-Chieh Lee
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Ya-Hui Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Sung-Chih Hsieh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Wei-Zhen Lew
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Sheng-Wei Feng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Division of Prosthodontics, Department of Dentistry, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
26
|
Barbosa WT, de Almeida KV, de Lima GG, Rodriguez MA, Lia Fook MV, García-Carrodeguas R, Amaro da Silva Junior V, de Sousa Segundo FA, de Sá MJC. Synthesis and in vivo evaluation of a scaffold containing wollastonite/β-TCP for bone repair in a rabbit tibial defect model. J Biomed Mater Res B Appl Biomater 2019; 108:1107-1116. [PMID: 31393675 DOI: 10.1002/jbm.b.34462] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 11/09/2022]
Abstract
Scaffolds are models designed to aid the interaction between cells and extracellular bone matrix, providing structural support for newly formed bone tissue. In this work, wollastonite with β-TCP porous ceramic scaffolds was developed by the polymer sponge replication. Their microstructure, cell viability and bioactivity were tested. in vivo was performed to evaluate the use of a calcium silicate-based implant in the repair of rabbit tibias. Holes were made in the both proximal and distal tibial metaphysis of each animal and filled with calcium silicate-based implant, and in the left tibia, no implant were used, serving as control group. Animals underwent euthanasia after 30 and 60 days of study. The animals were submitted to clinical-radiographic evaluations and their histology was analyzed by optical and scanning electron microscope. The studied calcium silicate implant provided biocompatibility and promoted bone formation, stimulating the process of bone repair in rabbits, features observed by gradual radiopacity shown in the radiographic evaluations.
Collapse
Affiliation(s)
- Willams T Barbosa
- Certbio, Universidade Federal de Campina Grande (UFCG), Campina Grande, Paraíba, Brazil
| | | | - Gabriel G de Lima
- Programa de Pós-Graduação em Engenharia e Ciência dos Materiais - PIPE, Universidade Federal do Paraná, Curitiba, Paraná, Brazil.,Materials Research Institute, Athlone Institute of Technology, Athlone, Ireland
| | | | - Marcos V Lia Fook
- Certbio, Universidade Federal de Campina Grande (UFCG), Campina Grande, Paraíba, Brazil
| | | | | | - Francisco A de Sousa Segundo
- Programa de Pós-Graduação em Medicina Veterinária (PPGMV), Universidade Federal de Campina Grande (UFCG), Campina Grande, Paraíba, Brazil
| | - Marcelo J C de Sá
- Materials Research Institute, Athlone Institute of Technology, Athlone, Ireland.,Programa de Pós-Graduação em Medicina Veterinária (PPGMV), Universidade Federal de Campina Grande (UFCG), Campina Grande, Paraíba, Brazil
| |
Collapse
|
27
|
Loozen LD, Kruyt MC, Kragten AHM, Schoenfeldt T, Croes M, Oner CF, Dhert WJA, Alblas J. BMP-2 gene delivery in cell-loaded and cell-free constructs for bone regeneration. PLoS One 2019; 14:e0220028. [PMID: 31365542 PMCID: PMC6668905 DOI: 10.1371/journal.pone.0220028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/08/2019] [Indexed: 11/18/2022] Open
Abstract
To induce osteogenicity in bone graft substitutes, plasmid-based expression of BMP-2 (pBMP-2) has been successfully applied in gene activated matrices based on alginate polymer constructs. Here, we investigated whether cell seeding is necessary for non-viral BMP-2 gene expression in vivo. Furthermore, to gain insight in the role of BMP-producing cells, we compared inclusion of bone progenitor cells with non-osteogenic target cells in gene delivery constructs. Plasmid DNA encoding GFP (pGFP) was used to trace transfection of host tissue cells and seeded cells in a rat model. Transgene expression was followed in both cell-free alginate-ceramic constructs as well as constructs seeded with syngeneic fibroblasts or multipotent mesenchymal stromal cells (MSCs). Titration of pGFP revealed that the highest pGFP dose resulted in frequent presence of positive host cells in the constructs. Both cell-loaded groups were associated with transgene expression, most effectively in the MSC-loaded constructs. Subsequently, we investigated effectiveness of cell-free and cell-loaded alginate-ceramic constructs with pBMP-2 to induce bone formation. Local BMP-2 production was found in all groups containing BMP-2 plasmid DNA, and was most pronounced in the groups with MSCs transfected with high concentration pBMP-2. Bone formation was only apparent in the recombinant protein BMP-2 group. In conclusion, we show that non-viral gene delivery of BMP-2 is a potentially effective way to induce transgene expression in vivo, both in cell-seeded as well as cell-free conditions. However, alginate-based gene delivery of BMP-2 to host cells or seeded cells did not result in protein levels adequate for bone formation in this setting, calling for more reliable scaffold compatible transfection methods.
Collapse
Affiliation(s)
- Loek D. Loozen
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Moyo C. Kruyt
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Ted Schoenfeldt
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michiel Croes
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cumhur F. Oner
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Wouter J. A. Dhert
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jacqueline Alblas
- Dept. Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
28
|
Zeng Y, Hoque J, Varghese S. Biomaterial-assisted local and systemic delivery of bioactive agents for bone repair. Acta Biomater 2019; 93:152-168. [PMID: 30711659 PMCID: PMC6615988 DOI: 10.1016/j.actbio.2019.01.060] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 01/05/2023]
Abstract
Although bone tissues possess an intrinsic capacity for repair, there are cases where bone healing is either impaired or insufficient, such as fracture non-union, osteoporosis, osteomyelitis, and cancers. In these cases, treatments like surgical interventions are used, either alone or in combination with bioactive agents, to promote tissue repair and manage associated clinical complications. Improving the efficacy of bioactive agents often requires carriers, with biomaterials being a pivotal player. In this review, we discuss the role of biomaterials in realizing the local and systemic delivery of biomolecules to the bone tissue. The versatility of biomaterials enables design of carriers with the desired loading efficiency, release profile, and on-demand delivery. Besides local administration, systemic administration of drugs is necessary to combat diseases like osteoporosis, warranting bone-targeting drug delivery systems. Thus, chemical moieties with the affinity towards bone extracellular matrix components like apatite minerals have been widely utilized to create bone-targeting carriers with better biodistribution, which cannot be achieved by the drugs alone. Bone-targeting carriers combined with the desired drugs or bioactive agents have been extensively investigated to enhance bone healing while minimizing off-target effects. Herein, these advancements in the field have been systematically reviewed. STATEMENT OF SIGNIFICANCE: Drug delivery is imperative when surgical interventions are not sufficient to address various bone diseases/defects. Biomaterial-assisted delivery systems have been designed to provide drugs with the desired loading efficiency, sustained release, and on-demand delivery to enhance bone healing. By surveying recent advances in the field, this review outlines the design of biomaterials as carriers for the local and systemic delivery of bioactive agents to the bone tissue. Particularly, biomaterials that bear chemical moieties with affinity to bone are attractive, as they can present the desired bioactive agents to the bone tissue efficiently and thus enhance the drug efficacy for bone repair.
Collapse
Affiliation(s)
- Yuze Zeng
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA
| | - Jiaul Hoque
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
29
|
Costa Moura C, Lanham SA, Monfort T, Bourdakos KN, Tare RS, Oreffo ROC, Mahajan S. Quantitative temporal interrogation in 3D of bioengineered human cartilage using multimodal label-free imaging. Integr Biol (Camb) 2019; 10:635-645. [PMID: 30225469 DOI: 10.1039/c8ib00050f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The unique properties of skeletal stem cells have attracted significant attention in the development of strategies for skeletal regeneration. However, there remains a crucial unmet need to develop quantitative tools to elucidate skeletal cell development and monitor the formation of regenerated tissues using non-destructive techniques in 3D. Label-free methods such as coherent anti-Stokes Raman scattering (CARS), second harmonic generation (SHG) and two-photon excited auto-fluorescence (TPEAF) microscopy are minimally invasive, non-destructive, and present new powerful alternatives to conventional imaging techniques. Here we report a combination of these techniques in a single multimodal system for the temporal assessment of cartilage formation by human skeletal cells. The evaluation of bioengineered cartilage, with a new parameter measuring the amount of collagen per cell, collagen fibre structure and chondrocyte distribution, was performed using the 3D non-destructive platform. Such 3D label-free temporal quantification paves the way for tracking skeletal cell development in real-time and offers a paradigm shift in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Catarina Costa Moura
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK. and Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Stuart A Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Tual Monfort
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| | - Konstantinos N Bourdakos
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| | - Rahul S Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK. and Mechanical Engineering Department, Faculty of Engineering and the Environment, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Sumeet Mahajan
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| |
Collapse
|
30
|
Zhai W, Yong D, El-Jawhari JJ, Cuthbert R, McGonagle D, Win Naing M, Jones E. Identification of senescent cells in multipotent mesenchymal stromal cell cultures: Current methods and future directions. Cytotherapy 2019; 21:803-819. [PMID: 31138507 DOI: 10.1016/j.jcyt.2019.05.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/30/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022]
Abstract
Regardless of their tissue of origin, multipotent mesenchymal stromal cells (MSCs) are commonly expanded in vitro for several population doublings to achieve a sufficient number of cells for therapy. Prolonged MSC expansion has been shown to result in phenotypical, morphological and gene expression changes in MSCs, which ultimately lead to the state of senescence. The presence of senescent cells in therapeutic MSC batches is undesirable because it reduces their viability, differentiation potential and trophic capabilities. Additionally, senescent cells acquire senescence-activated secretory phenotype, which may not only induce apoptosis in the neighboring host cells following MSC transplantation, but also trigger local inflammatory reactions. This review outlines the current and promising new methodologies for the identification of senescent cells in MSC cultures, with a particular emphasis on non-destructive and label-free methodologies. Technologies allowing identification of individual senescent cells, based on new surface markers, offer potential advantage for targeted senescent cell removal using new-generation senolytic agents, and subsequent production of therapeutic MSC batches fully devoid of senescent cells. Methods or a combination of methods that are non-destructive and label-free, for example, involving cell size and spectroscopic measurements, could be the best way forward because they do not modify the cells of interest, thus maximizing the final output of therapeutic-grade MSC cultures. The further incorporation of machine learning methods has also recently shown promise in facilitating, automating and enhancing the analysis of these measured data.
Collapse
Affiliation(s)
- Weichao Zhai
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Derrick Yong
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Jehan Jomaa El-Jawhari
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK; Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Richard Cuthbert
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK
| | - May Win Naing
- Singapore Institute of Manufacturing Technology, A*STAR, Innovis, Singapore
| | - Elena Jones
- Leeds Institute of Rheumatic and musculoskeletal Medicine, Leeds, UK.
| |
Collapse
|
31
|
Asperosaponin VI stimulates osteogenic differentiation of rat adipose-derived stem cells. Regen Ther 2019; 11:17-24. [PMID: 31193169 PMCID: PMC6518317 DOI: 10.1016/j.reth.2019.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 02/14/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022] Open
Abstract
In the aging population, the decrease on osteogenic differentiation resulted into a significant reduction in bone formation. Bone tissue engineering has been a successful technique for treatment of bone defects. It is reported that adipose-derived stem cells (ADSCs) have pluripotency to differentiate into adipocytes and osteoblasts. However little is revealed about the effect of the herbal medicine Asperosaponin VI (ASA VI) on ADSCs differentiation. In our study, we isolated and identified ADSCs from rats. We examined the effect of different concentrations of ASA VI in ADSCs on alkaline phosphatase (ALP) activity, calcium deposition, the expression of bone-related proteins and the release of inflammatory cytokines. Flowcytometry assay showed ADSCs were highly expressed CD44 and CD105, but hardly expressed CD34 and CD45, suggesting ADSCs were successfully isolated for follow-up experiments. ALP activity examination and Alizarin red (AR) stain showed that ASA VI enhanced the ALP activity and promoted matrix mineralization in ADSCs. In addition, bone-related protein OCN and RUNX2, and Smad2/3 phosphorylation was upregulated after ASA VI treatment in ADSCs. ELISA results showed that ASA VI blocked the release of TNF-α, IL-6 and IL-1β in ADSCs. Considering this results, we concluded that ASA VI promotes osteogenic differentiation of ADSCs through inducing the expression of bone-related proteins. These findings enriched the function of ASA VI as a regenerative medicine and shed new light for the treatment of bone defects in clinical research.
Collapse
|
32
|
Moura CC, Bourdakos KN, Tare RS, Oreffo ROC, Mahajan S. Live-imaging of Bioengineered Cartilage Tissue using Multimodal Non-linear Molecular Imaging. Sci Rep 2019; 9:5561. [PMID: 30944358 PMCID: PMC6447547 DOI: 10.1038/s41598-019-41466-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 03/04/2019] [Indexed: 12/15/2022] Open
Abstract
Coherent anti-Stokes Raman scattering (CARS) and second harmonic generation (SHG) are non-linear techniques that allow label-free, non-destructive and non-invasive imaging for cellular and tissue analysis. Although live-imaging studies have been performed previously, concerns that they do not cause any changes at the molecular level in sensitive biological samples have not been addressed. This is important especially for stem cell differentiation and tissue engineering, if CARS/SHG microscopy is to be used as a non-invasive, label-free tool for assessment of the developing neo-tissue. In this work, we monitored the differentiation of human fetal-femur derived skeletal cells into cartilage in three-dimensional cultures using CARS and SHG microscopy and demonstrate the live-imaging of the same developing neo-tissue over time. Our work conclusively establishes that non-linear label-free imaging does not alter the phenotype or the gene expression at the different stages of differentiation and has no adverse effect on human skeletal cell growth and behaviour. Additionally, we show that CARS microscopy allows imaging of different molecules of interest, including lipids, proteins and glycosaminoglycans, in the bioengineered neo-cartilage. These studies demonstrate the label-free and truly non-invasive nature of live CARS and SHG imaging and their value and translation potential in skeletal research, regeneration medicine and tissue engineering.
Collapse
Affiliation(s)
- Catarina Costa Moura
- Institute for Life Sciences and Department of Chemistry, Highfield Campus, University of Southampton, SO17 1BJ, Southampton, UK.,Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, UK
| | - Konstantinos N Bourdakos
- Institute for Life Sciences and Department of Chemistry, Highfield Campus, University of Southampton, SO17 1BJ, Southampton, UK
| | - Rahul S Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, UK.,Mechanical Engineering Department, Faculty of Engineering and the Environment, Highfield Campus, University of Southampton, SO17 1BJ, Southampton, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, UK.
| | - Sumeet Mahajan
- Institute for Life Sciences and Department of Chemistry, Highfield Campus, University of Southampton, SO17 1BJ, Southampton, UK.
| |
Collapse
|
33
|
Srinivasaiah S, Musumeci G, Mohan T, Castrogiovanni P, Absenger-Novak M, Zefferer U, Mostofi S, Bonyadi Rad E, Grün NG, Weinberg AM, Schäfer U. A 300 μm Organotypic Bone Slice Culture Model for Temporal Investigation of Endochondral Osteogenesis. Tissue Eng Part C Methods 2019; 25:197-212. [DOI: 10.1089/ten.tec.2018.0368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Sriveena Srinivasaiah
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Giuseppe Musumeci
- Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Tamilselvan Mohan
- Institute of Chemistry, University of Graz, Graz, Austria
- Laboratory for Characterization and Processing, Faculty of Mechanical Engineering, University of Maribor, Maribor, Slovenia
| | - Paola Castrogiovanni
- Human Anatomy and Histology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Ulrike Zefferer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| | - Sepideh Mostofi
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
| | - Ehsan Bonyadi Rad
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
- Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nicole Gabriele Grün
- Department of Orthopedics and Trauma Surgery, Medical University of Graz, Graz, Austria
| | | | - Ute Schäfer
- Research Unit for Experimental Neurotraumatology, Department of Neurosurgery, Medical University of Graz, Graz, Austria
| |
Collapse
|
34
|
Yoshida Y, Matsubara H, Fang X, Hayashi K, Nomura I, Ugaji S, Hamada T, Tsuchiya H. Adipose-derived stem cell sheets accelerate bone healing in rat femoral defects. PLoS One 2019; 14:e0214488. [PMID: 30921414 PMCID: PMC6438603 DOI: 10.1371/journal.pone.0214488] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 03/13/2019] [Indexed: 12/15/2022] Open
Abstract
In the present study, we investigated whether both adipose-derived stem cell (ADSC) and osteogenic-induced ADSC sheets could promote bone healing in a rat distal femoral metaphysis bone defect model. A through-hole defect of 1 mm diameter was drilled into each distal femur of 12 week old rats. Forty-five rats were randomly assigned to three groups: (1) control group; (2) ADSC sheet group; or (3) osteogenic-induced ADSC sheet group. We evaluated each group by analysis of computerized tomography scans every week after the surgery, histological analysis, and DiI labeling (a method of membrane staining for post implant cell tracing). Radiological and histological evaluations showed that a part of the hole persisted in the control group at four weeks after surgery, whereas the hole was restored almost completely by new bone formation in both sheet groups. The mean value of bone density (in Houndsfield units) for the bone defect area was significantly higher in both sheet groups than that in the control group (p = 0.05) at four weeks postoperative. A large number of osteocalcin positive osteoblasts were observed at the area of bone defect, especially in the osteogenic-induced ADCS sheet group. DiI labeling in the newly formed bone showed that each sheet had differentiated into bone tissue at four weeks after surgery. The ADSC and the osteogenic-induced ADSC sheets promoted significantly quicker bone healing in the bone defect. Moreover, the osteogenic-induced ADSC sheet may be more advantageous for bone healing than the ADSC sheet because of the higher number of osteocalcin positive osteoblasts via the transplantation.
Collapse
Affiliation(s)
- Yasuhisa Yoshida
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hidenori Matsubara
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Xiang Fang
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Issei Nomura
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Shuhei Ugaji
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tomo Hamada
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
35
|
The paracrine effects of human induced pluripotent stem cells promote bone-like structures via the upregulation of BMP expression in a mouse ectopic model. Sci Rep 2018; 8:17106. [PMID: 30459360 PMCID: PMC6244408 DOI: 10.1038/s41598-018-35546-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/26/2018] [Indexed: 01/20/2023] Open
Abstract
Use of human induced pluripotent stem cells (h-iPSCs) for bone tissue engineering is most appealing, because h-iPSCs are an inexhaustible source of osteocompetent cells. The present study investigated the contribution of undifferentiated h-iPSCs and elucidated aspects of the underlying mechanism(s) of the involvement of these cells to new bone formation. Implantation of undifferentiated h-iPSCs seeded on coral particles in ectopic sites of mice resulted in expression of osteocalcin and DMP-1, and in mineral content similar to that of the murine bone. The number of the implanted h-iPSCs decreased with time and disappeared by 30 days post-implantation. In contrast, expression of the murine osteogenic genes at day 15 and 30 post-implantation provided, for the first time, evidence that the implanted h-iPSCs affected the observed outcomes via paracrine mechanisms. Supporting evidence was provided because supernatant conditioned media from h-iPSCs (h-iPSC CM), promoted the osteogenic differentiation of human mesenchymal stem cells (h-MSCs) in vitro. Specifically, h-iPSC CM induced upregulation of the BMP-2, BMP-4 and BMP-6 genes, and promoted mineralization of the extracellular matrix. Given the current interest in the use of h-iPSCs for regenerative medicine applications, our study contributes new insights into aspects of the mechanism underlying the bone promoting capability of h-iPSCs.
Collapse
|
36
|
McEwan JK, Tribe HC, Jacobs N, Hancock N, Qureshi AA, Dunlop DG, Oreffo RO. Regenerative medicine in lower limb reconstruction. Regen Med 2018; 13:477-490. [PMID: 29985779 DOI: 10.2217/rme-2018-0011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bone is a highly specialized connective tissue and has a rare quality as one of the few tissues that can repair without a scar to regain pre-injury structure and function. Despite the excellent healing capacity of bone, tumor, infection, trauma and surgery can lead to significant bone loss requiring skeletal augmentation. Bone loss in the lower limb poses a complex clinical problem, requiring reconstructive techniques to restore form and function. In the past, amputation may have been the only option; however, there is now an array of reconstructive possibilities and cellular therapies available to salvage a limb. In this review, we will evaluate current applications of bone tissue engineering techniques in limb reconstruction and identify potential strategies for future work.
Collapse
Affiliation(s)
- Josephine K McEwan
- Bone & Joint Research Group, Centre for Human Development, Stem Cell & Regeneration, Institute of Developmental Sciences, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Howard C Tribe
- Bone & Joint Research Group, Centre for Human Development, Stem Cell & Regeneration, Institute of Developmental Sciences, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Neal Jacobs
- Salisbury NHS Foundation Trust, Salisbury, Wiltshire, UK
| | - Nicholas Hancock
- Trauma & Orthopaedic Department, University Hospital Southampton, Southampton, UK
| | - Amir A Qureshi
- Trauma & Orthopaedic Department, University Hospital Southampton, Southampton, UK
| | - Douglas G Dunlop
- Trauma & Orthopaedic Department, University Hospital Southampton, Southampton, UK
| | - Richard Oc Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cell & Regeneration, Institute of Developmental Sciences, University of Southampton, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
37
|
Piuzzi NS, Dominici M, Long M, Pascual-Garrido C, Rodeo S, Huard J, Guicheux J, McFarland R, Goodrich LR, Maddens S, Robey PG, Bauer TW, Barrett J, Barry F, Karli D, Chu CR, Weiss DJ, Martin I, Jorgensen C, Muschler GF. Proceedings of the signature series symposium "cellular therapies for orthopaedics and musculoskeletal disease proven and unproven therapies-promise, facts and fantasy," international society for cellular therapies, montreal, canada, may 2, 2018. Cytotherapy 2018; 20:1381-1400. [PMID: 30316562 PMCID: PMC8487641 DOI: 10.1016/j.jcyt.2018.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/06/2018] [Indexed: 12/17/2022]
Abstract
The Signature Series Symposium "Cellular Therapies for Orthopaedics and Musculoskeletal Disease Proven and Unproven Therapies-Promise, Facts and Fantasy" was held as a pre-meeting of the 26th International Society for Cellular Therapy (ISCT) annual congress in Montreal, Canada, May 2, 2018. This was the first ISCT program that was entirely dedicated to the advancement of cell-based therapies for musculoskeletal diseases. Cellular therapies in musculoskeletal medicine are a source of great promise and opportunity. They are also the source of public controversy, confusion and misinformation. Patients, clinicians, scientists, industry and government share a commitment to clear communication and responsible development of the field. Therefore, this symposium convened thought leaders from around the world in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value to patients with musculoskeletal conditions.
Collapse
Affiliation(s)
- Nicolas S Piuzzi
- Department of Orthopedic Surgery and Biomedical Engineering Cleveland Clinic, Cleveland, Ohio, USA; Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Marc Long
- MTF Biologics, Edison, New Jersey, USA
| | - Cecilia Pascual-Garrido
- Adult Reconstruction-Adolescent and Young Adult Hip Service, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Scott Rodeo
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, UTHealth Medical School, Houston, Texas, USA; Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Jérome Guicheux
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Nantes University School of Dental Medicine, ONIRIS, Nantes, France; CHU Nantes, PHU4 OTONN, Nantes, France
| | - Richard McFarland
- Advanced Regenerative Manufacturing Institute, Manchester, New Hampshire, USA, and Standards Coordinating Body, Gaithersburg, Maryland, USA
| | - Laurie R Goodrich
- Orthopaedic Research Center and Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | | | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA
| | - Thomas W Bauer
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, New York, USA
| | - John Barrett
- Stem Cell Allogeneic Transplant Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Frank Barry
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - David Karli
- Steadman Philippon Research Institute, Vail, Colorado, USA; Greyledge Technologies, LLC, Vail, Colorado, USA
| | - Constance R Chu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA; Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Daniel J Weiss
- University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Christian Jorgensen
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - George F Muschler
- Department of Orthopedic Surgery and Biomedical Engineering Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
38
|
Eyvazi M, Farahzadi R, Karimian Fathi N, Karimipour M, Soleimani Rad J, Montaseri A. Mummy Material Can Promote Differentiation of Adipose Derived Stem Cells into Osteoblast through Enhancement of Bone Specific Transcription Factors Expression. Adv Pharm Bull 2018; 8:457-464. [PMID: 30276142 PMCID: PMC6156472 DOI: 10.15171/apb.2018.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022] Open
Abstract
Purpose: Application of Mummy material for treatment of different diseases such as bone fracture, cutaneous wounds and joint inflammation has been advised since hundred years ago in Persian traditional medicine. Due to the claims of indigenous people and advice of traditional medicine for application of this material in healing of bone fractures, this study has been designed to evaluate whether Mummy material can promote the differentiation of mesenchymal stem cells into osteoblasts and enhance the expression of bone specific genes and proteins. Methods: Adipose derived stem cells (ASCs) at fourth cell passage were divided into control, osteogenesis group (received osteogenic medium), Mummy group (received Mummy at concentration of 500 µg/ml). ASCs in the fourth group were treated with both osteogenic medium and Mummy (500µg/ml). Cells in all groups were harvested on days 7, 14 and 21 days for further evaluation through Real time RT-PCR, Von kossa staining, Immunocytochemistry and flowcytometery. Results: Treatment of ASCs with Mummy at concentration of 500µg/ml promotes the expression level of Osteocalcin, RUNX-2 and β1-integrin genes in different time points but that of the Osterix did not changed. Furthermore the expression of Osteocalcin protein enhanced significantly in ASCs treated with Mummy detected by Immunocytochemistry and flowcytometery technique compared to the control groups. The results of this study also showed that treatment of ASCs with Mummy resulted in formation of mineral deposits which was evaluated by Von Kossa staining method. Conclusion: Obtained data from this study reveals that Mummy is a potent enhancer for differentiation of ASCs into osteoblasts in in vitro system, probably through increasing the level of bone specific genes and proteins.
Collapse
Affiliation(s)
- Maryam Eyvazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Karimian Fathi
- Biochemistry Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Anatomical Sciences Department, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimani Rad
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azadeh Montaseri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
Loozen LD, Kruyt MC, Vandersteen A, Kragten AHM, Croes M, Öner FC, Alblas J. Osteoinduction by Ex Vivo Nonviral Bone Morphogenetic Protein Gene Delivery Is Independent of Cell Type. Tissue Eng Part A 2018; 24:1423-1431. [PMID: 29766760 DOI: 10.1089/ten.tea.2018.0032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Ex vivo nonviral gene delivery of bone inductive factors has the potential to heal bone defects. Due to their inherent role in new bone formation, multipotent stromal cells (MSCs) have been studied as the primary target cell for gene delivery in a preclinical setting. The relative contribution of autocrine and paracrine mechanisms, and the need of osteogenic cells, remains unclear. This study investigates the contribution of MSCs as producer of transgenic bone morphogenetic proteins (BMPs) and to what extent the seeded MSCs participate in actual osteogenesis. Rat-derived MSCs or fibroblasts (FBs) were cotransfected with pBMP-2 and pBMP-6 or pBMP-7 via nucleofection. The bioactivity of BMP products was shown through in vitro osteogenic differentiation assays. To investigate their role in new bone formation, transfected cells were seeded on ceramic scaffolds and implanted subcutaneously in rats. Bone formation was assessed by histomorphometry after 8 weeks. As a proof of principle, we also investigated the suitability of bone marrow-derived mononuclear cells and the stromal vascular fraction isolated from adipose tissue for a one-stage gene delivery strategy. Bone formation was induced in all conditions containing cells overexpressing BMP heterodimers. Constructs seeded with FBs transfected with BMP-2/6 and MSCs transfected with BMP-2/6 showed comparable bone volumes, both significantly higher than controls. Single-stage gene delivery proved possible and resulted in some bone formation. We conclude that bone formation as a result of ex vivo BMP gene delivery can be achieved even without direct osteogenic potential of the transfected cell type, suggesting that transfected cells mainly function as a production facility for osteoinductive proteins. In addition, single-stage transfection and reimplantation of cells appeared feasible, thus facilitating future clinical translation of the method.
Collapse
Affiliation(s)
- Loek D Loozen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Moyo C Kruyt
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela Vandersteen
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Angela H M Kragten
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Michiel Croes
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - F Cumhur Öner
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| | - Jacqueline Alblas
- Department of Orthopaedics, University Medical Center Utrecht , Utrecht, The Netherlands
| |
Collapse
|
40
|
Xavier M, de Andrés MC, Spencer D, Oreffo ROC, Morgan H. Size and dielectric properties of skeletal stem cells change critically after enrichment and expansion from human bone marrow: consequences for microfluidic cell sorting. J R Soc Interface 2018; 14:rsif.2017.0233. [PMID: 28835540 PMCID: PMC5582119 DOI: 10.1098/rsif.2017.0233] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/27/2017] [Indexed: 12/14/2022] Open
Abstract
The capacity of bone and cartilage to regenerate can be attributed to skeletal stem cells (SSCs) that reside within the bone marrow (BM). Given SSCs are rare and lack specific surface markers, antibody-based sorting has failed to deliver the cell purity required for clinical translation. Microfluidics offers new methods of isolating cells based on biophysical features including, but not limited to, size, electrical properties and stiffness. Here we report the characterization of the dielectric properties of unexpanded SSCs using single-cell microfluidic impedance cytometry (MIC). Unexpanded SSCs had a mean size of 9.0 µm; larger than the majority of BM cells. During expansion, often used to purify and increase the number of SSCs, cell size and membrane capacitance increased significantly, highlighting the importance of characterizing unaltered SSCs. In addition, MIC was used to track the osteogenic differentiation of SSCs and showed an increased membrane capacitance with differentiation. The electrical properties of primary SSCs were indistinct from other BM cells precluding its use as an isolation method. However, the current studies indicate that cell size in combination with another biophysical parameter, such as stiffness, could be used to design label-free devices for sorting SSCs with significant clinical impact.
Collapse
Affiliation(s)
- Miguel Xavier
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.,Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - María C de Andrés
- Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - Daniel Spencer
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Southampton General Hospital, Tremona Road, SO16 6YD Southampton, UK
| | - Hywel Morgan
- Faculty of Physical Sciences and Engineering, and Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
41
|
Abstract
Craniofacial surgery, since its inauguration, has been the culmination of collaborative efforts to solve complex congenital, dysplastic, oncological, and traumatic cranial bone defects. Now, 50 years on from the first craniofacial meeting, the collaborative efforts between surgeons, scientists, and bioengineers are further advancing craniofacial surgery with new discoveries in tissue regeneration. Recent advances in regenerative medicine and stem cell biology have transformed the authors' understanding of bone healing, the role of stem cells governing bone healing, and the effects of the niche environment and extracellular matrix on stem cell fate. This review aims at summarizing the advances within each of these fields.
Collapse
|
42
|
Wang Y, Newman MR, Benoit DSW. Development of controlled drug delivery systems for bone fracture-targeted therapeutic delivery: A review. Eur J Pharm Biopharm 2018; 127:223-236. [PMID: 29471078 DOI: 10.1016/j.ejpb.2018.02.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/17/2018] [Accepted: 02/17/2018] [Indexed: 01/09/2023]
Abstract
Impaired fracture healing is a major clinical problem that can lead to patient disability, prolonged hospitalization, and significant financial burden. Although the majority of fractures heal using standard clinical practices, approximately 10% suffer from delayed unions or non-unions. A wide range of factors contribute to the risk for nonunions including internal factors, such as patient age, gender, and comorbidities, and external factors, such as the location and extent of injury. Current clinical approaches to treat nonunions include bone grafts and low-intensity pulsed ultrasound (LIPUS), which realizes clinical success only to select patients due to limitations including donor morbidities (grafts) and necessity of fracture reduction (LIPUS), respectively. To date, therapeutic approaches for bone regeneration rely heavily on protein-based growth factors such as INFUSE, an FDA-approved scaffold for delivery of bone morphogenetic protein 2 (BMP-2). Small molecule modulators and RNAi therapeutics are under development to circumvent challenges associated with traditional growth factors. While preclinical studies has shown promise, drug delivery has become a major hurdle stalling clinical translation. Therefore, this review overviews current therapies employed to stimulate fracture healing pre-clinically and clinically, including a focus on drug delivery systems for growth factors, parathyroid hormone (PTH), small molecules, and RNAi therapeutics, as well as recent advances and future promise of fracture-targeted drug delivery.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Maureen R Newman
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, 308 Robert B. Goergen Hall, University of Rochester, Rochester, NY 14627, USA; Center for Musculoskeletal Research, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Chemical Engineering, 4517 Wegmans Hall, University of Rochester, Rochester, NY 14627, USA; Department of Orthopaedics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Department of Biomedical Genetics, 601 Elmwood Ave, University of Rochester, Rochester, NY 14642, USA; Center for Oral Biology, 601 Elmwood Ave, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
43
|
Pajarinen J, Lin T, Gibon E, Kohno Y, Maruyama M, Nathan K, Lu L, Yao Z, Goodman SB. Mesenchymal stem cell-macrophage crosstalk and bone healing. Biomaterials 2018; 196:80-89. [PMID: 29329642 DOI: 10.1016/j.biomaterials.2017.12.025] [Citation(s) in RCA: 539] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/25/2017] [Accepted: 12/31/2017] [Indexed: 12/12/2022]
Abstract
Recent research has brought about a clear understanding that successful fracture healing is based on carefully coordinated cross-talk between inflammatory and bone forming cells. In particular, the key role that macrophages play in the recruitment and regulation of the differentiation of mesenchymal stem cells (MSCs) during bone regeneration has been brought to focus. Indeed, animal studies have comprehensively demonstrated that fractures do not heal without the direct involvement of macrophages. Yet the exact mechanisms by which macrophages contribute to bone regeneration remain to be elucidated. Macrophage-derived paracrine signaling molecules such as Oncostatin M, Prostaglandin E2 (PGE2), and Bone Morphogenetic Protein-2 (BMP2) have been shown to play critical roles; however the relative importance of inflammatory (M1) and tissue regenerative (M2) macrophages in guiding MSC differentiation along the osteogenic pathway remains poorly understood. In this review, we summarize the current understanding of the interaction of macrophages and MSCs during bone regeneration, with the emphasis on the role of macrophages in regulating bone formation. The potential implications of aging to this cellular cross-talk are reviewed. Emerging treatment options to improve facture healing by utilizing or targeting MSC-macrophage crosstalk are also discussed.
Collapse
Affiliation(s)
- Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tzuhua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Emmanuel Gibon
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yusuke Kohno
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masahiro Maruyama
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Karthik Nathan
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Lu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
44
|
Waddell SJ, de Andrés MC, Tsimbouri PM, Alakpa EV, Cusack M, Dalby MJ, Oreffo ROC. Biomimetic oyster shell-replicated topography alters the behaviour of human skeletal stem cells. J Tissue Eng 2018; 9:2041731418794007. [PMID: 30202512 PMCID: PMC6124183 DOI: 10.1177/2041731418794007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022] Open
Abstract
The regenerative potential of skeletal stem cells provides an attractive prospect to generate bone tissue needed for musculoskeletal reparation. A central issue remains efficacious, controlled cell differentiation strategies to aid progression of cell therapies to the clinic. The nacre surface from Pinctada maxima shells is known to enhance bone formation. However, to date, there is a paucity of information on the role of the topography of P. maxima surfaces, nacre and prism. To investigate this, nacre and prism topographical features were replicated onto polycaprolactone and skeletal stem cell behaviour on the surfaces studied. Skeletal stem cells on nacre surfaces exhibited an increase in cell area, increase in expression of osteogenic markers ALP (p < 0.05) and OCN (p < 0.01) and increased metabolite intensity (p < 0.05), indicating a role of nacre surface to induce osteogenic differentiation, while on prism surfaces, skeletal stem cells did not show alterations in cell area or osteogenic marker expression and a decrease in metabolite intensity (p < 0.05), demonstrating a distinct role for the prism surface, with the potential to maintain the skeletal stem cell phenotype.
Collapse
Affiliation(s)
- Shona J Waddell
- Centre for Human Development, Stem Cells
and Regeneration, Institute of Developmental Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| | - María C de Andrés
- Centre for Human Development, Stem Cells
and Regeneration, Institute of Developmental Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| | - Penelope M Tsimbouri
- Centre for Cell Engineering, Institute
of Molecular, Cell and Systems Biology, CMVLS, University of Glasgow, Glasgow,
UK
| | - Enateri V Alakpa
- Department of Integrative Medical
Biology, Umeå University, Umeå, Sweden
| | - Maggie Cusack
- Division of Biological and Environmental
Science, University of Stirling, Stirling, UK
| | - Matthew J Dalby
- Centre for Cell Engineering, Institute
of Molecular, Cell and Systems Biology, CMVLS, University of Glasgow, Glasgow,
UK
| | - Richard OC Oreffo
- Centre for Human Development, Stem Cells
and Regeneration, Institute of Developmental Sciences, Faculty of Medicine,
University of Southampton, Southampton, UK
| |
Collapse
|
45
|
Tang D, Yang LY, Ou KL, Oreffo ROC. Repositioning Titanium: An In Vitro Evaluation of Laser-Generated Microporous, Microrough Titanium Templates As a Potential Bridging Interface for Enhanced Osseointegration and Durability of Implants. Front Bioeng Biotechnol 2017; 5:77. [PMID: 29322044 PMCID: PMC5732141 DOI: 10.3389/fbioe.2017.00077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/23/2017] [Indexed: 11/21/2022] Open
Abstract
Although titanium alloys remain the preferred biomaterials for the manufacture of biomedical implants today, such devices can fail within 15 years of implantation due to inadequate osseointegration. Furthermore, wear debris toxicity due to alloy metal ion release has been found to cause side-effects including neurotoxicity and chronic inflammation. Titanium, with its known biocompatibility, corrosion resistance, and high elastic modulus, could if harnessed in the form of a superficial scaffold or bridging device, resolve such issues. A novel three-dimensional culture approach was used to investigate the potential osteoinductive and osseointegrative capabilities of a laser-generated microporous, microrough medical grade IV titanium template on human skeletal stem cells (SSCs). Human SSCs seeded on a rough 90-µm pore surface of ethylene oxide-sterilized templates were observed to be strongly adherent, and to display early osteogenic differentiation, despite their inverted culture in basal conditions over 21 days. Limited cellular migration across the template surface highlighted the importance of high surface wettability in maximizing cell adhesion, spreading and cell-biomaterial interaction, while restricted cell ingrowth within the conical-shaped pores underlined the crucial role of pore geometry and size in determining the extent of osseointegration of an implant device. The overall findings indicate that titanium only devices, with appropriate optimizations to porosity and surface wettability, could yet play a major role in improving the long-term efficacy, durability, and safety of future implant technology.
Collapse
Affiliation(s)
- Daniel Tang
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Research Center for Biotechnology, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Keng-Liang Ou
- Department of Dentistry, Cathay General Hospital, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan.,3D Global Biotech Inc., New Taipei City, Taiwan
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
46
|
Moura CC, Tare RS, Oreffo ROC, Mahajan S. Raman spectroscopy and coherent anti-Stokes Raman scattering imaging: prospective tools for monitoring skeletal cells and skeletal regeneration. J R Soc Interface 2017; 13:rsif.2016.0182. [PMID: 27170652 DOI: 10.1098/rsif.2016.0182] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/13/2016] [Indexed: 12/20/2022] Open
Abstract
The use of skeletal stem cells (SSCs) for cell-based therapies is currently one of the most promising areas for skeletal disease treatment and skeletal tissue repair. The ability for controlled modification of SSCs could provide significant therapeutic potential in regenerative medicine, with the prospect to permanently repopulate a host with stem cells and their progeny. Currently, SSC differentiation into the stromal lineages of bone, fat and cartilage is assessed using different approaches that typically require cell fixation or lysis, which are invasive or even destructive. Raman spectroscopy and coherent anti-Stokes Raman scattering (CARS) microscopy present an exciting alternative for studying biological systems in their natural state, without any perturbation. Here we review the applications of Raman spectroscopy and CARS imaging in stem-cell research, and discuss the potential of these two techniques for evaluating SSCs, skeletal tissues and skeletal regeneration as an exemplar.
Collapse
Affiliation(s)
- Catarina Costa Moura
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Rahul S Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Sumeet Mahajan
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK
| |
Collapse
|
47
|
Wu YX, Jing XZ, Sun Y, Ye YP, Guo JC, Huang JM, Xiang W, Zhang JM, Guo FJ. CD146+ skeletal stem cells from growth plate exhibit specific chondrogenic differentiation capacity in vitro. Mol Med Rep 2017; 16:8019-8028. [PMID: 28983600 PMCID: PMC5779886 DOI: 10.3892/mmr.2017.7616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023] Open
Abstract
Skeletal stem cells (SSCs) are a population of progenitor cells which give rise to postnatal skeletal tissues including bone, cartilage and bone marrow stroma, however not to adipose, haematopoietic or muscle tissue. Growth plate chondrocytes exhibit the ability of continuous proliferation and differentiation, which contributes to the continuous physiological growth. The growth plate has been hypothesized to contain SSCs which exhibit a desirable differentiation capacity to generate bone and cartilage. Due to the heterogeneity of the growth plate chondrocytes, SSCs in the growth plate are not well studied. The present study used cluster of differentiation (CD)146 and CD105 as markers to isolate purified SSCs. CD105+ SSCs and CD146+ SSCs were isolated using a magnetic activated cell sorting method. To quantitatively investigate the proliferation and differentiation ability, the colony-forming efficiency (CFE) and multi‑lineage differentiation capacity of CD105+ SSCs and CD146+ SSCs were compared with unsorted cells and adipose-derived stem cells (ASCs). It was revealed that CD105+ and CD146+ subpopulations represented subsets of SSCs which generated chondrocytes and osteocytes, however not adipocytes. Compared with CD105+ subpopulations and ASCs, the CD146+ subpopulation exhibited a greater CFE and continuous high chondrogenic differentiation capacity in vitro. Therefore, the present study suggested that the CD146+ subpopulation represented a chondrolineage‑restricted subpopulation of SSCs and may therefore act as a valuable cell source for cartilage regeneration.
Collapse
Affiliation(s)
- Ying-Xing Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xing-Zhi Jing
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yue Sun
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ya-Ping Ye
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jia-Chao Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jun-Ming Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Wei Xiang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jia-Ming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng-Jing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
48
|
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: 9.0] [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
|
49
|
Predicting the Remaining Lifespan and Cultivation-Related Loss of Osteogenic Capacity of Bone Marrow Multipotential Stromal Cells Applicable across a Broad Donor Age Range. Stem Cells Int 2017; 2017:6129596. [PMID: 28298930 PMCID: PMC5337353 DOI: 10.1155/2017/6129596] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/16/2017] [Indexed: 12/28/2022] Open
Abstract
Background and Objectives. Culture expanded multipotential stromal cells (MSCs) have considerable potential for bone regeneration therapy but their wider use is constrained by the lack of simple and predictive assays of functional potency. Extended passaging leads to loss of multipotency but speed of decline depends on MSC donor age. The aim of this study was to develop an assay predictive of MSC culture longevity applicable to a broad donor age range. Materials and Methods. Bone marrow (BM, n = 7) was obtained from a diverse range (2–72 years) of healthy donors. MSCs were culture expanded to senescence and their osteoprogenitor content, gene expression profiles, epigenetic signature, and telomere behaviour were measured throughout. Output data was combined for modelling purposes. Results. Regardless of donor age, cultures' osteoprogenitor content correlated better with remaining lifespan (population doublings before senescence, PD-BS) than proliferative history (accrued PDs). Individual gene's expression or telomere length did not predict PD-BS but methylation of individual CpG islands did, PRAMEF2 in particular (r = 0.775). Coupling the steep relationship of relative SPARC expression with PD-BS (r = −0.753) the formula SPARC × 1/PREMEF2 gave an improved correlation (r = −0.893). Conclusion. A formula based on SPARC mRNA and PRAMEF2 methylation may be used to predict remaining BM-MSC longevity and related loss of multipotentiality independent of donor age.
Collapse
|
50
|
Guerado E, Caso E. Challenges of bone tissue engineering in orthopaedic patients. World J Orthop 2017; 8:87-98. [PMID: 28251059 PMCID: PMC5314152 DOI: 10.5312/wjo.v8.i2.87] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/23/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Bone defects may impede normal biomechanics and the structural stability of bone as an organ. In many cases, the correction of bone defects requires extensive surgical intervention involving the use of bone-grafting techniques and other procedures in which healing is slow, there is a high risk of infection and considerable pain is provoked - with no guarantee of complete correction of the defect. Therefore, the search for surgical alternatives continues to present a major challenge in orthopaedic traumatology. The reamer-irrigator-aspirator (RIA) system, which was devised to avoid the problems that can arise with autograft harvesting from the iliac crest, consists of collecting the product of the femoral canal after reaming. The RIA technique improves osteogenic differentiation of mesenchymal stem cells, compared to bone marrow aspiration or cancellous bone harvesting from the iliac crest using a spoon. Another approach, the Masquelet technique, consists of reconstructing a long bone defect by means of an induced membrane grown onto an acrylic cement rod inserted to fill the defect; in a second surgical step, once the membrane is constituted, the cement rod is removed and cancellous autograft is used to fill the defect. Both in RIA and in the Masquelet technique, osteosynthesis is usually needed. Bone transportation by compression-distraction lengthening principles is commonly implemented for the treatment of large bone loss. However, complications are frequently encountered with these techniques. Among new techniques that have been proposed to address the problem of large bone loss, the application of stem cells in conjunction with tissue engineering techniques is very promising, as is the creation of personalised medicine (or precision medicine), in which molecular profiling technologies are used to tailor the therapeutic strategy, to ensure the right method is applied for the right person at the right time, after determining the predisposition to disease among the general population. All of the above techniques for addressing bone defects are discussed in this paper.
Collapse
|