1
|
Cell-Free Therapies: The Use of Cell Extracts to Mitigate Irradiation-Injured Salivary Glands. BIOLOGY 2023; 12:biology12020305. [PMID: 36829582 PMCID: PMC9953449 DOI: 10.3390/biology12020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Radiotherapy is a standard treatment for head and neck cancer patients worldwide. However, millions of patients who received radiotherapy consequently suffer from xerostomia because of irreversible damage to salivary glands (SGs) caused by irradiation (IR). Current treatments for IR-induced SG hypofunction only provide temporary symptom alleviation but do not repair the damaged SG, thus resulting in limited treatment efficacy. Therefore, there has recently been a growing interest in regenerative treatments, such as cell-free therapies. This review aims to summarize cell-free therapies for IR-induced SG, with a particular emphasis on utilizing diverse cell extract (CE) administrations. Cell extract is a group of heterogeneous mixtures containing multifunctional inter-cellular molecules. This review discusses the current knowledge of CE's components and efficacy. We propose optimal approaches to improve cell extract treatment from multiple perspectives (e.g., delivery routes, preparation methods, and other details regarding CE administration). In addition, the advantages and limitations of CE treatment are systematically discussed by comparing it to other cell-free (such as conditioned media and exosomes) and cell-based therapies. Although a comprehensive identification of the bioactive factors within CEs and their mechanisms of action have yet to be fully understood, we propose cell extract therapy as an effective, practical, user-friendly, and safe option to conventional therapies in IR-induced SG.
Collapse
|
2
|
Hasegawa K, Raudales JLM, I T, Yoshida T, Honma R, Iwatake M, Tran SD, Seki M, Asahina I, Sumita Y. Effective-mononuclear cell (E-MNC) therapy alleviates salivary gland damage by suppressing lymphocyte infiltration in Sjögren-like disease. Front Bioeng Biotechnol 2023; 11:1144624. [PMID: 37168614 PMCID: PMC10164970 DOI: 10.3389/fbioe.2023.1144624] [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: 01/14/2023] [Accepted: 04/14/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction: Sjögren syndrome (SS) is an autoimmune disease characterized by salivary gland (SG) destruction leading to loss of secretory function. A hallmark of the disease is the presence of focal lymphocyte infiltration in SGs, which is predominantly composed of T cells. Currently, there are no effective therapies for SS. Recently, we demonstrated that a newly developed therapy using effective-mononuclear cells (E-MNCs) improved the function of radiation-injured SGs due to anti-inflammatory and regenerative effects. In this study, we investigated whether E-MNCs could ameliorate disease development in non-obese diabetic (NOD) mice as a model for primary SS. Methods: E-MNCs were obtained from peripheral blood mononuclear cells (PBMNCs) cultured for 7 days in serum-free medium supplemented with five specific recombinant proteins (5G culture). The anti-inflammatory characteristics of E-MNCs were then analyzed using a co-culture system with CD3/CD28-stimulated PBMNCs. To evaluate the therapeutic efficacy of E-MNCs against SS onset, E-MNCs were transplanted into SGs of NOD mice. Subsequently, saliva secretion, histological, and gene expression analyses of harvested SG were performed to investigate if E-MNCs therapy delays disease development. Results: First, we characterized that both human and mouse E-MNCs exhibited induction of CD11b/CD206-positive cells (M2 macrophages) and that human E-MNCs could inhibit inflammatory gene expressions in CD3/CD28- stimulated PBMNCs. Further analyses revealed that Msr1-and galectin3-positive macrophages (immunomodulatory M2c phenotype) were specifically induced in E-MNCs of both NOD and MHC class I-matched mice. Transplanted E-MNCs induced M2 macrophages and reduced the expression of T cell-derived chemokine-related and inflammatory genes in SG tissue of NOD mice at SS-onset. Then, E-MNCs suppressed the infiltration of CD4-positive T cells and facilitated the maintenance of saliva secretion for up to 12 weeks after E-MNC administration. Discussion: Thus, the immunomodulatory actions of E-MNCs could be part of a therapeutic strategy targeting the early stage of primary SS.
Collapse
Affiliation(s)
- Kayo Hasegawa
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jorge Luis Montenegro Raudales
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takashi I
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takako Yoshida
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ryo Honma
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Unit of Translational Medicine, Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mayumi Iwatake
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Simon D. Tran
- Laboratory of Craniofacial Tissue Engineering and Stem Cells, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | | | - Izumi Asahina
- Unit of Translational Medicine, Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Depatment of Oral and Maxillofacial Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Yoshinori Sumita
- Department of Medical Research and Development for Oral Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- *Correspondence: Yoshinori Sumita,
| |
Collapse
|
3
|
Kagami H, Inoue M, Agata H, Asahina I, Nagamura-Inoue T, Taguri M, Tojo A. A Clinical Study of Alveolar Bone Tissue Engineering Using Autologous Bone Marrow Stromal Cells: Effect of Optimized Cell-Processing Protocol on Efficacy. J Clin Med 2022; 11:jcm11247328. [PMID: 36555944 PMCID: PMC9783548 DOI: 10.3390/jcm11247328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Objectives: The effect of cell-processing protocols on the clinical efficacy of bone tissue engineering is not well-known. To maximize efficacy, we optimized the cell-processing protocol for bone-marrow-derived mesenchymal stromal cells for bone tissue engineering. In this study, the efficacy of bone tissue engineering using this modified protocol was compared to that of the original protocol. (2) Materials and Methods: This single-arm clinical study included 15 patients. Cells were obtained from bone marrow aspirates and expanded in culture flasks containing basic fibroblast growth factor. The cells were seeded onto β-tricalcium phosphate granules and induced into osteogenic cells for two weeks. Then, the cell-scaffold composites were transplanted into patients with severe atrophic alveolar bone. Radiographic evaluations and bone biopsies were performed. The results were compared with those of a previous clinical study that used the original protocol. (3) Results: Panoramic X-ray and computed tomography showed bone regeneration at the transplantation site in all cases. The average bone area in the biopsy samples at 4 months was 44.0%, which was comparable to that in a previous clinical study at 6 months (41.9%) but with much less deviation. No side effects related to cell transplantation were observed. In regenerated bone, 100% of the implants were integrated. (4) Conclusions: Compared to the original protocol, the non-inferiority of this protocol was proven. The introduction of an optimized cell-processing protocol resulted in a comparable quality of regenerated bone, with less fluctuation. Optimized cell-processing protocols may contribute to stable bone regeneration.
Collapse
Affiliation(s)
- Hideaki Kagami
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri 399-0781, Japan
- Institute for Oral Science, Matsumoto Dental University, Shiojiri 399-0781, Japan
- Department of Dentistry and Oral Surgery, Aichi Medical University, Nagakute 480-1195, Japan
- Correspondence:
| | - Minoru Inoue
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri 399-0781, Japan
- Inoue Dental Clinic, Shizuoka 420-0866, Japan
| | - Hideki Agata
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Agata Dental Clinic, Hamamatsu 430-0929, Japan
| | - Izumi Asahina
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Unit of Translational Medicine, Department of Regenerative Oral Surgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Department of Oral and Maxillofacial Surgery, Juntendo University Hospital, Tokyo 113-8431, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, Research Hospital, The University of Tokyo, Tokyo 108-8639, Japan
| | - Masataka Taguri
- Division of Medical Data Science, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Arinobu Tojo
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Institute of Innovation Advancement, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
4
|
Nagamura-Inoue T, Kato S, Najima Y, Isobe M, Doki N, Yamamoto H, Uchida N, Takahashi A, Hori A, Nojima M, Ohashi K, Nagamura F, Tojo A. Immunological influence of serum-free manufactured umbilical cord-derived mesenchymal stromal cells for steroid-resistant acute graft-versus-host disease. Int J Hematol 2022; 116:754-769. [PMID: 35908021 DOI: 10.1007/s12185-022-03408-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/13/2022] [Indexed: 12/31/2022]
Abstract
This study investigated the safety, efficacy, and immunological influence of allogeneic umbilical cord-derived mesenchymal stromal cells (IMSUT-CORD) processed in serum-free medium and cryoprotectant, for treating steroid-resistant acute graft-versus-host disease (aGVHD). In a phase I dose-escalation trial, IMSUT-CORD were infused intravenously twice weekly over two cycles with up to two additional cycles. Four patients received a dose of 1 × 106 cells/kg, while three received 2 × 106/kg. Of 76 total adverse events, fourteen associated or possibly associated adverse events included 2 cases of a hot flash, headache, and peripheral neuropathy, 1 each of upper abdominal pain, hypoxia, increased γ-GTP, somnolence, peripheral vascular pain at the injection site, thrombocytopenia, hypertension, and decreased fibrinogen. At 16 weeks after the initial IMSUT-CORD infusion, three patients showed complete response (CR), two partial response (PR), one mixed response, and one no response. The overall response rate was 71.4%, and the continuous CR/PR rate was 100% for over 28 days after CR/PR. NK cell count significantly increased and correlated with treatment response, whereas IL-12, IL-17, and IL-33 levels decreased, but did not correlate with treatment response. CCL2 and CCL11 levels increased during IMSUT-CORD therapy. IMSUT-CORD are usable in patients with steroid-resistant aGVHD (UMIN000032819: https://www.umin.ac.jp/ctr ).
Collapse
Affiliation(s)
- Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan. .,Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan. .,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan. .,Department of Global Clinical Research, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Seiko Kato
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Masamichi Isobe
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | | | - Naoyuki Uchida
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akiko Hori
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.,IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kazuteru Ohashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Fumitaka Nagamura
- Department of Global Clinical Research, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Institute of Innovation Advancement, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
5
|
de Castro LF, Sworder BJ, Mui B, Futrega K, Berendsen A, Phillips MD, Burbach NJ, Cherman N, Kuznetsov S, Gabet Y, Holmbeck K, Robey PG. Secreted frizzled related-protein 2 (Sfrp2) deficiency decreases adult skeletal stem cell function in mice. Bone Res 2021; 9:49. [PMID: 34857734 PMCID: PMC8639730 DOI: 10.1038/s41413-021-00169-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/21/2021] [Accepted: 06/27/2021] [Indexed: 12/18/2022] Open
Abstract
In a previous transcriptomic study of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived "mesenchymal stem cells"), SFRP2 was highly over-represented in a subset of multipotent BMSCs (skeletal stem cells, SSCs), which recreate a bone/marrow organ in an in vivo ectopic bone formation assay. SFRPs modulate WNT signaling, which is essential to maintain skeletal homeostasis, but the specific role of SFRP2 in BMSCs/SSCs is unclear. Here, we evaluated Sfrp2 deficiency on BMSC/SSC function in models of skeletal organogenesis and regeneration. The skeleton of Sfrp2-deficient (KO) mice is overtly normal; but their BMSCs/SSCs exhibit reduced colony-forming efficiency, reflecting low SSC self-renewal/abundancy. Sfrp2 KO BMSCs/SSCs formed less trabecular bone than those from WT littermates in the ectopic bone formation assay. Moreover, regeneration of a cortical drilled hole defect was dramatically impaired in Sfrp2 KO mice. Sfrp2-deficient BMSCs/SSCs exhibited poor in vitro osteogenic differentiation as measured by Runx2 and Osterix expression and calcium accumulation. Interestingly, activation of the Wnt co-receptor, Lrp6, and expression of Wnt target genes, Axin2, C-myc and Cyclin D1, were reduced in Sfrp2-deficient BMSCs/SSCs. Addition of recombinant Sfrp2 restored most of these activities, suggesting that Sfrp2 acts as a Wnt agonist. We demonstrate that Sfrp2 plays a role in self-renewal of SSCs and in the recruitment and differentiation of adult SSCs during bone healing. SFRP2 is also a useful marker of BMSC/SSC multipotency, and a factor to potentially improve the quality of ex vivo expanded BMSC/SSC products.
Collapse
Affiliation(s)
- Luis Fernandez de Castro
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD, USA.
| | - Brian J. Sworder
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA ,grid.189504.10000 0004 1936 7558Department of Molecular Medicine, Boston University, Boston, MA USA
| | - Byron Mui
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Kathryn Futrega
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Agnes Berendsen
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Matthew D. Phillips
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Nathan J. Burbach
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA ,grid.17635.360000000419368657School of Dentistry, University of Minnesota—Twin Cities, Minneapolis, MN USA
| | - Natasha Cherman
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Sergei Kuznetsov
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Yankel Gabet
- grid.12136.370000 0004 1937 0546Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Kenn Holmbeck
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| | - Pamela G. Robey
- grid.94365.3d0000 0001 2297 5165Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
6
|
Asahina I, Kagami H, Agata H, Honda MJ, Sumita Y, Inoue M, Nagamura-Inoue T, Tojo A. Clinical Outcome and 8-Year Follow-Up of Alveolar Bone Tissue Engineering for Severely Atrophic Alveolar Bone Using Autologous Bone Marrow Stromal Cells with Platelet-Rich Plasma and β-Tricalcium Phosphate Granules. J Clin Med 2021; 10:jcm10225231. [PMID: 34830513 PMCID: PMC8623501 DOI: 10.3390/jcm10225231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/31/2021] [Accepted: 11/08/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Although bone tissue engineering for dentistry has been studied for many years, the clinical outcome for severe cases has not been established. Furthermore, there are limited numbers of studies that include long-term follow-up. In this study, the safety and efficacy of bone tissue engineering for patients with a severely atrophic alveolar bone were examined using autogenous bone marrow stromal cells (BMSCs), and the long-term stability was also evaluated. Methods: BMSCs from iliac bone marrow aspirate were cultured and expanded. Then, induced osteogenic cells were transplanted with autogenous platelet-rich plasma (PRP) and β-tricalcium phosphate granules (β-TCP) for maxillary sinus floor and alveolar ridge augmentation. Eight patients (two males and six females) with an average age of 54.2 years underwent cell transplantation. Safety was assessed by monitoring adverse events. Radiographic evaluation and bone biopsies were performed to evaluate the regenerated bone. Results: The major population of transplanted BMSCs belonged to the fraction of CD34−, CD45dim, and CD73+ cells, which was only 0.065% of the total bone marrow cells. Significant deviations were observed in cell growth and alkaline phosphatase activities among individuals. However, bone regeneration was observed in all patients and the average bone area in the biopsy samples was 41.9% 6 months following transplantation, although there were also significant deviations among each case. No adverse events related to the transplants were observed. In the regenerated bone, 27 out of 29 dental implants were integrated. Dental implants and regenerated bone were stable for an average follow-up period of 7 years and 10 months. Conclusions: Although individual variations were observed, the results showed that bone tissue engineering using BMSCs with PRP and β-TCP was feasible for patients with severe atrophic maxilla throughout a long-term follow-up period and was considered safe. However, further studies with a larger number of cases and controls to confirm the efficacy of BMSCs and the development of a protocol to establish a reproducible quality of stem cell-based graft material will be required.
Collapse
Affiliation(s)
- Izumi Asahina
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Hideaki Kagami
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri 399-0781, Japan
| | - Hideki Agata
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Masaki J Honda
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Oral Anatomy, Aichi-Gakuin University School of Dentistry, Nagoya 464-0821, Japan
| | - Yoshinori Sumita
- Division of Stem Cell Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| | - Minoru Inoue
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri 399-0781, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Arinobu Tojo
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
7
|
Thurairajah K, Briggs GD, Balogh ZJ. Stem cell therapy for fracture non-union: The current evidence from human studies. J Orthop Surg (Hong Kong) 2021; 29:23094990211036545. [PMID: 34396805 DOI: 10.1177/23094990211036545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Non-union is a taxing complication of fracture management for both the patient and their surgeon. Modern fracture fixation techniques have been developed to optimise the biomechanical environment for fracture healing but do not guarantee union. Patient biology has a critical role in achieving union and stem cell therapy has potential for improving fracture healing at a cellular level to treat or avoid non-union. This article reviews the current understanding of non-union, concepts in bone healing and the current literature on the application of stem cells in non-union.
Collapse
Affiliation(s)
- Kabilan Thurairajah
- Department of Traumatology, 37024John Hunter Hospital and University of Newcastle, Newcastle, Australia
| | - Gabrielle D Briggs
- School of Medicine and Public Health, 5982University of Newcastle, Newcastle, Australia
| | - Zsolt J Balogh
- Department of Traumatology, 37024John Hunter Hospital and University of Newcastle, Newcastle, Australia
| |
Collapse
|
8
|
Gonçalves AM, Moreira A, Weber A, Williams GR, Costa PF. Osteochondral Tissue Engineering: The Potential of Electrospinning and Additive Manufacturing. Pharmaceutics 2021; 13:983. [PMID: 34209671 PMCID: PMC8309012 DOI: 10.3390/pharmaceutics13070983] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
The socioeconomic impact of osteochondral (OC) damage has been increasing steadily over time in the global population, and the promise of tissue engineering in generating biomimetic tissues replicating the physiological OC environment and architecture has been falling short of its projected potential. The most recent advances in OC tissue engineering are summarised in this work, with a focus on electrospun and 3D printed biomaterials combined with stem cells and biochemical stimuli, to identify what is causing this pitfall between the bench and the patients' bedside. Even though significant progress has been achieved in electrospinning, 3D-(bio)printing, and induced pluripotent stem cell (iPSC) technologies, it is still challenging to artificially emulate the OC interface and achieve complete regeneration of bone and cartilage tissues. Their intricate architecture and the need for tight spatiotemporal control of cellular and biochemical cues hinder the attainment of long-term functional integration of tissue-engineered constructs. Moreover, this complexity and the high variability in experimental conditions used in different studies undermine the scalability and reproducibility of prospective regenerative medicine solutions. It is clear that further development of standardised, integrative, and economically viable methods regarding scaffold production, cell selection, and additional biochemical and biomechanical stimulation is likely to be the key to accelerate the clinical translation and fill the gap in OC treatment.
Collapse
Affiliation(s)
| | - Anabela Moreira
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| | - Achim Weber
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstrasse 12, 70569 Stuttgart, Germany;
| | - Gareth R. Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal; (A.M.G.); (A.M.)
| |
Collapse
|
9
|
Li G, Shen W, Tang X, Mo G, Yao L, Wang J. Combined use of calcium phosphate cement, mesenchymal stem cells and platelet-rich plasma for bone regeneration in critical-size defect of the femoral condyle in mini-pigs. Regen Med 2021; 16:451-464. [PMID: 34030462 DOI: 10.2217/rme-2020-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate the outcome of autologous bone marrow mesenchymal stem cells (BMMSCs) and platelet-rich plasma in combination with calcium phosphate cement (CPC) scaffold to reconstruct femoral critical bone defects in mini-pigs. Materials & methods: Scanning electron microscopy, micro-computed tomography evaluation and quantitative histological assessment were used. Results & conclusion: BMMSCs were attached to the CPC scaffold after 7 days of culture and decreased the residual CPC material in each group at 12 weeks compared with 6 weeks. The newly formed bone area was higher in the CPC+SC+P group than in the CPC group at each time point (all p < 0.05). The strategy of CPC combined with BMMSCs and platelet-rich plasma might be an effective method to repair bone defects.
Collapse
Affiliation(s)
- Guangjun Li
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Wen Shen
- Department of Radiology, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Xing Tang
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Guowei Mo
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Liqin Yao
- Department of Orthopedic, Deqing People's Hospital, Deqing, Zhejiang 313200, PR China
| | - Jixing Wang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| |
Collapse
|
10
|
Combination of optimized tissue engineering bone implantation with heel-strike like mechanical loading to repair segmental bone defect in New Zealand rabbits. Cell Tissue Res 2021; 385:639-658. [PMID: 33966092 DOI: 10.1007/s00441-021-03458-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
In this study, effects of combining optimized tissue engineering bone (TEB) implantation with heel-strike like mechanical loading to repair segmental bone defect in New Zealand rabbits were investigated. Physiological characteristics of bone marrow mesenchymal stem cells (BMMSCs), compact bone cells (CBCs), and bone marrow and compact bone coculture cells (BMMSC-CBCs) were compared to select the optimal seed cells for optimized TEB construction. Rabbits with segmental bone defects were treated in different ways (cancellous bone scaffold for group A, cancellous bone scaffold and mechanical loading for group B, optimized TEB for group C, optimized TEB and mechanical loading for group D, n = 4), and the bone repair were compared. BMMSC-CBCs showed better proliferation capacity than CBCs (p < 0.01) and stronger osteogenic differentiation ability than BMMSCs (p < 0.05). Heel-strike like mechanical loading improved proliferation and osteogenic differentiation ability and expression levels of TGFβ1 as well as BMP2 of seed cells in vitro (p < 0.05). At week 12 post-operation, group D showed the best bone repair, followed by groups B and C, while group A finished last (p < 0.05). During week 4 to 12 post-operation, group D peaked in terms of expression levels of TGFβ1, BMP2, and OCN, followed by groups B and C, while group A finished last (p < 0.05). Thus, BMMSC-CBCs showed good proliferation and osteogenic differentiation ability, and they were thought to be better as seed cells than BMMSCs and CBCs. The optimized TEB implantation combined with heel-strike like mechanical loading had a synergistic effect on bone defect healing, and enhanced expression of TGFβ1 and BMP2 played an important role in this process.
Collapse
|
11
|
Sumita Y, Iwamoto N, Seki M, Yoshida T, Honma R, Iwatake M, Ohba S, Takashi I, Hotokezaka Y, Harada H, Kuroshima S, Nagai K, Asahara T, Atsushi Kawakam I, Asahina I. Phase 1 clinical study of cell therapy with effective-mononuclear cells (E-MNC) for radiogenic xerostomia (first-in-human study) (FIH study on E-MNC therapy for radiogenic xerostomia). Medicine (Baltimore) 2020; 99:e20788. [PMID: 32590759 PMCID: PMC7328916 DOI: 10.1097/md.0000000000020788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Treatment for most patients with head and neck cancers includes ionizing radiation with or without chemotherapy. This treatment causes irreversible damage to salivary glands in the irradiation field accompanied by a loss of fluid-secreting acinar cells and a considerable decrease of saliva secretion. There is currently no adequate conventional treatment for this condition. In recent years, we developed an effective culture method to enhance the anti-inflammatory and vasculogenic phenotypes of peripheral blood mononuclear cells (PBMNCs), and such effectively conditioned PBMNC (E-MNC) therapy has shown promising improvements to the function of radiation-injured salivary glands in preclinical studies. However, the safety and effect of E-NMC therapy have yet assessed in human. The objective of this ongoing first-in-man study is to assess the safety, tolerability, and in part the efficacy of E-MNC therapy for treating radiation-induced xerostomia. METHODS/DESIGN This phase 1 first-in-man study is an open-label, single-center, two-step dose escalation study. A total of 6 patients, who had no recurrence of head and neck cancer over 5 years following radiation therapy and suffered from radiation-induced xerostomia, will receive a transplantation of E-NMCs derived from autologous PBMNCs to a submandibular gland. The duration of the intervention will be 1 year. To analyze the recovery of salivary secretion, a gum test will be performed. To analyze the recovery of atrophic salivary glands, computed tomography (CT), and magnetic resonance imaging (MRI) of salivary glands will be conducted. The primary endpoint is the safety of the protocol. The secondary endpoints are the changes from baseline in whole saliva secretion and salivary gland atrophy. DISCUSSION This will be the first clinical study of regenerative therapy using E-MNCs for patients with severe radiation-induced xerostomia. The results of this study are expected to contribute to developing the low-invasive cell-based therapy for radiation-induced xerostomia. TRIAL REGISTRATION This study was registered with the Japan Registry of Clinical Trials (http://jrct.niph.go.jp) as jRCTb070190057.
Collapse
Affiliation(s)
| | - Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | | | - Takako Yoshida
- Basic and Translational Research Center for Hard Tissue Disease
| | - Ryo Honma
- Basic and Translational Research Center for Hard Tissue Disease
- Department of Regenerative Oral Surgery, Unit of Translational Medicine
| | - Mayumi Iwatake
- Basic and Translational Research Center for Hard Tissue Disease
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Unit of Translational Medicine
| | - I. Takashi
- Department of Regenerative Oral Surgery, Unit of Translational Medicine
| | - Yuka Hotokezaka
- Department of Radiology and Cancer Biology, Nagasaki University Graduate School of Biomedical Sciences
| | | | - Shinichiro Kuroshima
- Department of Applied Prosthodontics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | | | - Takayuki Asahara
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | - I Atsushi Kawakam
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Unit of Translational Medicine
| |
Collapse
|
12
|
Miyaji T, Takami T, Fujisawa K, Matsumoto T, Yamamoto N, Sakaida I. Bone marrow-derived humoral factors suppress oxidative phosphorylation, upregulate TSG-6, and improve therapeutic effects on liver injury of mesenchymal stem cells. J Clin Biochem Nutr 2020; 66:213-223. [PMID: 32523248 DOI: 10.3164/jcbn.19-125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells, which have the potential to be used in regenerative medicine, require improvements in quality for patient use. To maintain stemness of cultured bone marrow-derived mesenchymal stem cells, we focused on the bone marrow microenvironment, generated a conditioned medium of whole bone marrow cells (BMC-CM), and assessed its effects on bone marrow-derived mesenchymal stem cells. BMC-CM suppressed morphological deterioration and proliferative decline in cultured bone marrow-derived mesenchymal stem cells, suppressed mitochondrial oxidative phosphorylation activity, a stemness indicator, and upregulated suppressors of oxidative phosphorylation such as hypoxia-inducible factor-1 alpha, Sirtuin 3, 4, and 5. Furthermore, BMC-CM upregulated TNF-stimulated gene 6 and ameliorated the therapeutic effects of cells on liver injury in carbon tetrachloride-administered rats. Since the elimination of 20-220-nm particles attenuated the effects of BMC-CM, we further analyzed exosomal microRNAs produced by whole bone marrow cells. Among the 49 microRNAs observed to be upregulated during the preparation of BMC-CM, several were identified that were associated with suppression of oxidative phosphorylation, upregulation of TNF-stimulated gene 6, and the pathogenesis of liver diseases. Thus, bone marrow-derived humoral factors including exosomal microRNAs may help to improve the therapeutic quality of bone marrow-derived mesenchymal stem cells for liver regenerative therapy.
Collapse
Affiliation(s)
- Takashi Miyaji
- Department of Gastroenterology & Hepatology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| | - Taro Takami
- Department of Liver Regenerative Medicine, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan.,Center for Regenerative and Cell Therapy, Yamaguchi University Organization for Research Initiatives, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| | - Koichi Fujisawa
- Department of Liver Regenerative Medicine, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| | - Toshihiko Matsumoto
- Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| | - Naoki Yamamoto
- Health Administration Center, Yamaguchi University, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| | - Isao Sakaida
- Department of Gastroenterology & Hepatology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan.,Center for Regenerative and Cell Therapy, Yamaguchi University Organization for Research Initiatives, Minamikogushi 1-1-1, Ube, Yamaguchi 755-0046, Japan
| |
Collapse
|
13
|
I T, Sumita Y, Yoshida T, Honma R, Iwatake M, Raudales JLM, Shizuno T, Kuroshima S, Masuda H, Seki M, Tran SD, Asahara T, Asahina I. Anti-inflammatory and vasculogenic conditioning of peripheral blood mononuclear cells reinforces their therapeutic potential for radiation-injured salivary glands. Stem Cell Res Ther 2019; 10:304. [PMID: 31623661 PMCID: PMC6798785 DOI: 10.1186/s13287-019-1414-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND There are currently no effective treatments available for patients with irreversible loss of salivary gland (SG) function caused by radiation therapy for head and neck cancer. In this study, we have developed an effective culture method to enhance the anti-inflammatory and vasculogenic phenotypes of peripheral blood mononuclear cells (PBMNCs) and investigated whether such effectively conditioned PBMNCs (E-MNCs) could regenerate radiation-injured SGs and ameliorate salivary secretory function in mice. METHODS Mouse PBMNCs were expanded in primary serum-free culture with five vasculogenic proteins for 5 days, and then the resulting cells (E-MNCs) were analyzed for their characteristics. Subsequently, 5 × 104 E-MNCs (labeled with EGFP in some experiments) were injected intra-glandularly into a mouse model of radiation-injured atrophic submandibular glands. After 2-3 weeks, the submandibular glands were harvested, and then the injected E-MNCs were tracked. Four, 8, and 12 weeks after irradiation (IR), salivary outputs were measured to evaluate the recovery of secretory function, and the gland tissues were harvested for histological and gene expression analyses to clarify the effects of cell transplantation. RESULTS The resulting E-MNCs contained an enriched population of definitive CD11b/CD206-positive (M2 macrophage-like) cells and showed anti-inflammatory and vasculogenic characteristics. Salivary secretory function in E-MNC-transplanted mice gradually recovered after 4 weeks post-irradiation (post-IR) and reached 3.8-fold higher than that of non-transplanted mice at 12 weeks. EGFP-expressing E-MNCs were detected in a portion of the vascular endothelium and perivascular gland tissues at 2 weeks post-IR, but mainly in some microvessels at 3 weeks. Between 4 and 12 weeks post-IR, mRNA expression and histological analyses revealed that E-MNC transplantation reduced the expression of inflammatory genes and increased the level of tissue-regenerative activities such as stem cell markers, cell proliferation, and blood vessel formation. At 12 weeks post-IR, the areas of acinar and ductal cells regenerated, and the glands had less fibrosis. CONCLUSIONS This effective conditioning of PBMNCs is a simple, rapid, and efficient method that provides a non-invasive source of therapeutic cells for regenerating radiation-injured atrophic SGs.
Collapse
Affiliation(s)
- Takashi I
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshinori Sumita
- Basic and Translational Research Center for Hard tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan.
| | - Takako Yoshida
- Basic and Translational Research Center for Hard tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Ryo Honma
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Basic and Translational Research Center for Hard tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Mayumi Iwatake
- Basic and Translational Research Center for Hard tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Jorge Luis Montenegro Raudales
- Basic and Translational Research Center for Hard tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | | | - Shinichiro Kuroshima
- Department of Applied Prosthodontics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Haruchika Masuda
- Department of Regenerative Medicine Science, Tokai University School of Medicine, Isehara, Japan
| | | | - Simon D Tran
- Laboratory of Craniofacial Tissue Engineering and Stem Cells, Faculty of Dentistry, McGill University, Montreal, Canada
| | - Takayuki Asahara
- Department of Applied Prosthodontics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
14
|
Spontaneously Formed Spheroids from Mouse Compact Bone-Derived Cells Retain Highly Potent Stem Cells with Enhanced Differentiation Capability. Stem Cells Int 2019; 2019:8469012. [PMID: 31191686 PMCID: PMC6525826 DOI: 10.1155/2019/8469012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 02/07/2023] Open
Abstract
The results from our recent study showed the presence of two distinct spheroid-forming mechanisms, i.e., spontaneous and mechanical. In this study, we focused on the spontaneously formed spheroids, and the character of spontaneously formed spheroids from mouse compact bone-derived cells (CBDCs) was explored. Cells from (C57BL/6J) mouse leg bones were isolated, and compact bone-derived cells were cultured after enzymatic digestion. Spontaneous spheroid formation was achieved on a culture plate with specific water contact angle as reported. The expression levels of embryonic stem cell markers were analyzed using immunofluorescence and quantitative reverse transcription polymerase chain reaction. Then, the cells from spheroids were induced into osteogenic and neurogenic lineages. The spontaneously formed spheroids from CBDCs were positive for ES cell markers such as SSEA1, Sox2, Oct4, and Nanog. Additionally, the expressions of fucosyltransferase 4/FUT4 (SSEA1), Sox2, and Nanog were significantly higher than those in monolayer cultured cells. The gene expression of mesenchymal stem cell markers was almost identical in both spheroids and monolayer-cultured cells, but the expression of Sca-1 was higher in spheroids. Spheroid-derived cells showed significantly higher osteogenic and neurogenic marker expression than monolayer-cultured cells after induction. Spontaneously formed spheroids expressed stem cell markers and showed enhanced osteogenic and neurogenic differentiation capabilities than cells from the conventional monolayer culture, which supports the superior stemness.
Collapse
|
15
|
Stem Cells in Dentistry: Types of Intra- and Extraoral Tissue-Derived Stem Cells and Clinical Applications. Stem Cells Int 2018; 2018:4313610. [PMID: 30057624 PMCID: PMC6051054 DOI: 10.1155/2018/4313610] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/05/2018] [Accepted: 06/07/2018] [Indexed: 12/13/2022] Open
Abstract
Stem cells are undifferentiated cells, capable of renewing themselves, with the capacity to produce different cell types to regenerate missing tissues and treat diseases. Oral facial tissues have been identified as a source and therapeutic target for stem cells with clinical interest in dentistry. This narrative review report targets on the several extraoral- and intraoral-derived stem cells that can be applied in dentistry. In addition, stem cell origins are suggested in what concerns their ability to differentiate as well as their particular distinguishing quality of convenience and immunomodulatory for regenerative dentistry. The development of bioengineered teeth to replace the patient's missing teeth was also possible because of stem cell technologies. This review will also focus our attention on the clinical application of stem cells in dentistry. In recent years, a variety of articles reported the advantages of stem cell-based procedures in regenerative treatments. The regeneration of lost oral tissue is the target of stem cell research. Owing to the fact that bone imperfections that ensue after tooth loss can result in further bone loss which limit the success of dental implants and prosthodontic therapies, the rehabilitation of alveolar ridge height is prosthodontists' principal interest. The development of bioengineered teeth to replace the patient's missing teeth was also possible because of stem cell technologies. In addition, a “dental stem cell banking” is available for regenerative treatments in the future. The main features of stem cells in the future of dentistry should be understood by clinicians.
Collapse
|
16
|
Stem Cells for Osteochondral Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1059:219-240. [DOI: 10.1007/978-3-319-76735-2_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
17
|
Farré-Guasch E, Bravenboer N, Helder MN, Schulten EAJM, ten Bruggenkate CM, Klein-Nulend J. Blood Vessel Formation and Bone Regeneration Potential of the Stromal Vascular Fraction Seeded on a Calcium Phosphate Scaffold in the Human Maxillary Sinus Floor Elevation Model. MATERIALS 2018; 11:ma11010161. [PMID: 29361686 PMCID: PMC5793659 DOI: 10.3390/ma11010161] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/12/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023]
Abstract
Bone substitutes are used as alternatives for autologous bone grafts in patients undergoing maxillary sinus floor elevation (MSFE) for dental implant placement. However, bone substitutes lack osteoinductive and angiogenic potential. Addition of adipose stem cells (ASCs) may stimulate osteogenesis and osteoinduction, as well as angiogenesis. We aimed to evaluate the vascularization in relation to bone formation potential of the ASC-containing stromal vascular fraction (SVF) of adipose tissue, seeded on two types of calcium phosphate carriers, within the human MSFE model, in a phase I study. Autologous SVF was obtained from ten patients and seeded on β-tricalcium phosphate (n = 5) or biphasic calcium phosphate carriers (n = 5), and used for MSFE in a one-step surgical procedure. After six months, biopsies were obtained during dental implant placement, and the quantification of the number of blood vessels was performed using histomorphometric analysis and immunohistochemical stainings for blood vessel markers, i.e., CD34 and alpha-smooth muscle actin. Bone percentages seemed to correlate with blood vessel formation and were higher in study versus control biopsies in the cranial area, in particular in β-tricalcium phosphate-treated patients. This study shows the safety, feasibility, and efficiency of the use of ASCs in the human MSFE, and indicates a pro-angiogenic effect of SVF.
Collapse
Affiliation(s)
- Elisabet Farré-Guasch
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands;
- Department of Oral and Maxillofacial Surgery, VU University Medical Center/Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, Amsterdam 1081 HV, The Netherlands; (M.N.H.); (E.A.J.M.S.); (C.M.t.B.)
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam 1007 MB, The Netherlands;
| | - Marco N. Helder
- Department of Oral and Maxillofacial Surgery, VU University Medical Center/Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, Amsterdam 1081 HV, The Netherlands; (M.N.H.); (E.A.J.M.S.); (C.M.t.B.)
| | - Engelbert A. J. M. Schulten
- Department of Oral and Maxillofacial Surgery, VU University Medical Center/Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, Amsterdam 1081 HV, The Netherlands; (M.N.H.); (E.A.J.M.S.); (C.M.t.B.)
| | - Christiaan M. ten Bruggenkate
- Department of Oral and Maxillofacial Surgery, VU University Medical Center/Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam Movement Sciences, Amsterdam 1081 HV, The Netherlands; (M.N.H.); (E.A.J.M.S.); (C.M.t.B.)
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands;
- Correspondence: ; Tel.: +31-(0)-205980-881; Fax: +31-(0)-205-980-333
| |
Collapse
|
18
|
Fujita K, Nozaki K, Horiuchi N, Yamashita K, Miura H, Nagai A. Regulation of periodontal ligament-derived cells by type III collagen-coated hydroxyapatite. Biomed Mater Eng 2017; 29:15-27. [DOI: 10.3233/bme-171709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kazuhisa Fujita
- Department of Fixed Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Kosuke Nozaki
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Naohiro Horiuchi
- Department of Inorganic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Kimihiro Yamashita
- Department of Inorganic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Hiroyuki Miura
- Department of Fixed Prosthodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Akiko Nagai
- Department of Biofunction Research, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| |
Collapse
|
19
|
Brennan MA, Renaud A, Guilloton F, Mebarki M, Trichet V, Sensebé L, Deschaseaux F, Chevallier N, Layrolle P. Inferior In Vivo Osteogenesis and Superior Angiogenesis of Human Adipose‐Derived Stem Cells Compared with Bone Marrow‐Derived Stem Cells Cultured in Xeno‐Free Conditions. Stem Cells Transl Med 2017; 6:2160-2172. [PMID: 29052365 PMCID: PMC5702520 DOI: 10.1002/sctm.17-0133] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
The possibility of using adipose tissue-derived stromal cells (ATSC) as alternatives to bone marrow-derived stromal cells (BMSC) for bone repair has garnered interest due to the accessibility, high cell yield, and rapid in vitro expansion of ATSC. For clinical relevance, their bone forming potential in comparison to BMSC must be proven. Distinct differences between ATSC and BMSC have been observed in vitro and comparison of osteogenic potential in vivo is not clear to date. The aim of the current study was to compare the osteogenesis of human xenofree-expanded ATSC and BMSC in vitro and in an ectopic nude mouse model of bone formation. Human MSC were implanted with biphasic calcium phosphate biomaterials in subcutis pockets for 8 weeks. Implant groups were: BMSC, ATSC, BMSC and ATSC mixed together in different ratios, as well as MSC primed with either osteogenic supplements (250 μM ascorbic acid, 10 mM β-glycerolphosphate, and 10 nM dexamethasone) or 50 ng/ml recombinant bone morphogenetic protein 4 prior to implantation. In vitro results show osteogenic gene expression and differentiation potentials of ATSC. Despite this, ATSC failed to form ectopic bone in vivo, in stark contrast to BMSC, although osteogenic priming did impart minor osteogenesis to ATSC. Neovascularization was enhanced by ATSC compared with BMSC; however, less ATSC engrafted into the implant compared with BMSC. Therefore, in the content of bone regeneration, the advantages of ATSC over BMSC including enhanced angiogenesis, may be negated by their lack of osteogenesis and prerequisite for osteogenic differentiation prior to transplantation. Stem Cells Translational Medicine 2017;6:2160-2172.
Collapse
Affiliation(s)
- Meadhbh A. Brennan
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Audrey Renaud
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Fabien Guilloton
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Miryam Mebarki
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Valerie Trichet
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| | - Luc Sensebé
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Frederic Deschaseaux
- STROMA Lab UMR UPS/CNRS 5273, U1031 INSERM, EFS‐Pyrénées‐MéditerranéeToulouseFrance
| | - Nathalie Chevallier
- INSERM, IMRB U955‐E10, Engineering and Cellular Therapy Unit, Etablissement Français du Sang, Faculty of Medicine, Paris Est UniversityCréteilFrance
| | - Pierre Layrolle
- INSERM, UMR 1238, PHYOS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of NantesNantesFrance
| |
Collapse
|
20
|
Noda S, Sumita Y, Ohba S, Yamamoto H, Asahina I. Soft tissue engineering with micronized-gingival connective tissues. J Cell Physiol 2017; 233:249-258. [PMID: 28233312 DOI: 10.1002/jcp.25871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 02/22/2017] [Indexed: 01/01/2023]
Abstract
The free gingival graft (FGG) and connective tissue graft (CTG) are currently considered to be the gold standards for keratinized gingival tissue reconstruction and augmentation. However, these procedures have some disadvantages in harvesting large grafts, such as donor-site morbidity as well as insufficient gingival width and thickness at the recipient site post-treatment. To solve these problems, we focused on an alternative strategy using micronized tissue transplantation (micro-graft). In this study, we first investigated whether transplantation of micronized gingival connective tissues (MGCTs) promotes skin wound healing. MGCTs (≤100 µm) were obtained by mincing a small piece (8 mm3 ) of porcine keratinized gingiva using the RIGENERA system. The MGCTs were then transplanted to a full skin defect (5 mm in diameter) on the dorsal surface of immunodeficient mice after seeding to an atelocollagen matrix. Transplantations of atelocollagen matrixes with and without micronized dermis were employed as experimental controls. The results indicated that MGCTs markedly promote the vascularization and epithelialization of the defect area 14 days after transplantation compared to the experimental controls. After 21 days, complete wound closure with low contraction was obtained only in the MGCT grafts. Tracking analysis of transplanted MGCTs revealed that some mesenchymal cells derived from MGCTs can survive during healing and may function to assist in wound healing. We propose here that micro-grafting with MGCTs represents an alternative strategy for keratinized tissue reconstruction that is characterized by low morbidity and ready availability.
Collapse
Affiliation(s)
- Sawako Noda
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hideyuki Yamamoto
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
21
|
Leegwater NC, Bakker AD, Hogervorst JMA, Nolte PA, Klein-Nulend J. Hypothermia reduces VEGF-165 expression, but not osteogenic differentiation of human adipose stem cells under hypoxia. PLoS One 2017; 12:e0171492. [PMID: 28166273 PMCID: PMC5293214 DOI: 10.1371/journal.pone.0171492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/20/2017] [Indexed: 01/01/2023] Open
Abstract
Cryotherapy is successfully used in the clinic to reduce pain and inflammation after musculoskeletal damage, and might prevent secondary tissue damage under the prevalent hypoxic conditions. Whether cryotherapy reduces mesenchymal stem cell (MSC) number and differentiation under hypoxic conditions, causing impaired callus formation is unknown. We aimed to determine whether hypothermia modulates proliferation, apoptosis, nitric oxide production, VEGF gene and protein expression, and osteogenic/chondrogenic differentiation of human MSCs under hypoxia. Human adipose MSCs were cultured under hypoxia (37°C, 1% O2), hypothermia and hypoxia (30°C, 1% O2), or control conditions (37°C, 20% O2). Total DNA, protein, nitric oxide production, alkaline phosphatase activity, gene expression, and VEGF protein concentration were measured up to day 8. Hypoxia enhanced KI67 expression at day 4. The combination of hypothermia and hypoxia further enhanced KI67 gene expression compared to hypoxia alone, but was unable to prevent the 1.2-fold reduction in DNA amount caused by hypoxia at day 4. Addition of hypothermia to hypoxic cells did not alter the effect of hypoxia alone on BAX-to-BCL-2 ratio, alkaline phosphatase activity, gene expression of SOX9, COL1, or osteocalcin, or nitric oxide production. Hypothermia decreased the stimulating effect of hypoxia on VEGF-165 gene expression by 6-fold at day 4 and by 2-fold at day 8. Hypothermia also decreased VEGF protein expression under hypoxia by 2.9-fold at day 8. In conclusion, hypothermia decreased VEGF-165 gene and protein expression, but did not affect differentiation, or apoptosis of MSCs cultured under hypoxia. These in vitro results implicate that hypothermia treatment in vivo, applied to alleviate pain and inflammation, is not likely to harm early stages of callus formation.
Collapse
Affiliation(s)
- Nick C. Leegwater
- Department of Orthopaedics, Spaarne Hospital, Hoofddorp, The Netherlands
| | - Astrid D. Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Jolanda M. A. Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Peter A. Nolte
- Department of Orthopaedics, Spaarne Hospital, Hoofddorp, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Mukai T, Nagamura-Inoue T, Shimazu T, Mori Y, Takahashi A, Tsunoda H, Yamaguchi S, Tojo A. Neurosphere formation enhances the neurogenic differentiation potential and migratory ability of umbilical cord-mesenchymal stromal cells. Cytotherapy 2016; 18:229-41. [PMID: 26794714 DOI: 10.1016/j.jcyt.2015.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/02/2015] [Accepted: 10/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS The human umbilical cord (UC) is a rich source of mesenchymal stromal cells (MSCs), which have been reported to have multi-lineage potential. The objectives of this study were to investigate the characteristics and capacity of UC-MSC neurosphere formation and whether this event enhances the propensity of UC-MSCs to undergo neural differentiation. METHODS UC-MSCs were collected by the improved explant method. UC-MSCs and neurosphere-forming UC-MSCs (UC-MSC-neurospheres) were induced to undergo neurogenic differentiation, the latter of which were induced by suspension culturing in the presence of epidermal growth factor and basic fibroblast growth factor. The differentiation and migratory capacities of the individual cultures were then compared on the basis of the expression of neural markers, as measured by immunocytochemistry, immunoblotting and quantitative real-time polymerase chain reaction and transwell assays, respectively. RESULTS Both UC-MSCs and UC-MSC-neurospheres were capable of differentiating into neurogenic cells when cultured in neurogenic differentiation medium. However, pre-conditioned UC-MSC-neurospheres exhibited significantly higher expression of neural markers--including microtubule-associated protein (MAP2), MUSASHI1, glial fibrillary acidic protein (GFAP), and NESTIN--compared with those derived from UC-MSCs directly. Moreover, UC-MSC-neurospheres expressed significantly higher levels of the stemness markers NANOG, KLF4 and OCT4 than did UC-MSCs. Migration assays also revealed that both UC-MSCs and UC-MSC-neurospheres actively migrate toward glucose-depleted cells. CONCLUSIONS Neurogenic differentiation potential probably is greater in UC-MSC-neurospheres than in UC-MSCs. Thus, UC-MSC-neurospheres may serve as a better source of cells for neurogenic regenerative medicine.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | | | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Prins HJ, Schulten EAJM, Ten Bruggenkate CM, Klein-Nulend J, Helder MN. Bone Regeneration Using the Freshly Isolated Autologous Stromal Vascular Fraction of Adipose Tissue in Combination With Calcium Phosphate Ceramics. Stem Cells Transl Med 2016; 5:1362-1374. [PMID: 27388241 DOI: 10.5966/sctm.2015-0369] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/18/2016] [Indexed: 02/07/2023] Open
Abstract
: In patients undergoing maxillary sinus floor elevation (MSFE) for dental implant placement, bone substitutes are currently evaluated as alternatives for autologous bone. However, bone substitutes have only osteoconductive properties and lack osteoinductive potential. Therefore, this phase I study evaluated the potential additive effect on bone regeneration by the addition of freshly isolated, autologous but heterologous stromal vascular fraction (SVF), which is highly enriched with adipose stromal/stem cells when compared with native adipose tissue. From 10 patients, SVF was procured using automatic processing, seeded on either β-tricalcium phosphate (n = 5) or biphasic calcium phosphate carriers (n = 5), and used for MSFE in a one-step surgical procedure. Primary objectives were feasibility and safety. The secondary objective was efficacy, evaluated by using biopsies of the augmented area taken 6 months postoperatively, concomitant with dental implant placement. Biopsies were assessed for bone, graft, and osteoid volumes. No adverse effects were reported during the procedure or follow-up (≥3 years). Bone and osteoid percentages were higher in study biopsies (SVF supplemented) than in control biopsies (ceramic only on contralateral side), in particular in β-tricalcium phosphate-treated patients. Paired analysis on the six bilaterally treated patients revealed markedly higher bone and osteoid volumes using microcomputed tomography or histomorphometric evaluations, demonstrating an additive effect of SVF supplementation, independent of the bone substitute. This study demonstrated for the first time the feasibility, safety, and potential efficacy of SVF seeded on bone substitutes for MSFE, providing the first step toward a novel treatment concept that might offer broad potential for SVF-based regenerative medicine applications. SIGNIFICANCE This is the first-in-human study using freshly isolated, autologous adipose stem cell preparations (the stromal vascular fraction [SVF] of adipose tissue) applied in a one-step surgical procedure with calcium phosphate ceramics (CaP) to increase maxillary bone height for dental implantations. All 10 patients received CaP plus SVF on one side, whereas bilaterally treated patients (6 of 10) received CaP only on the opposite side. This allowed intrapatient evaluation of the potential added value of SVF supplementation, assessed in biopsies obtained after 6 months. Feasibility, safety, and potential efficacy of SVF for bone regeneration were demonstrated, showing high potential for this novel concept.
Collapse
Affiliation(s)
- Henk-Jan Prins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute Amsterdam, VU University Amsterdam, Amsterdam, The Netherlands Department of Oral and Maxillofacial Surgery, MOVE Research Institute Amsterdam, VU University Medical Center/ACTA, Amsterdam, The Netherlands
| | - Engelbert A J M Schulten
- Department of Oral and Maxillofacial Surgery, MOVE Research Institute Amsterdam, VU University Medical Center/ACTA, Amsterdam, The Netherlands
| | - Christiaan M Ten Bruggenkate
- Department of Oral and Maxillofacial Surgery, MOVE Research Institute Amsterdam, VU University Medical Center/ACTA, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), MOVE Research Institute Amsterdam, VU University Amsterdam, Amsterdam, The Netherlands
| | - Marco N Helder
- Department of Orthopedic Surgery, MOVE Research Institute Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Akiyama H, Kobayashi A, Ichimura M, Tone H, Nakatani M, Inoue M, Tojo A, Kagami H. Comparison of manual and automated cultures of bone marrow stromal cells for bone tissue engineering. J Biosci Bioeng 2015; 120:570-6. [DOI: 10.1016/j.jbiosc.2015.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 02/24/2015] [Accepted: 03/16/2015] [Indexed: 01/05/2023]
|
25
|
Qi C, Yan X, Huang C, Melerzanov A, Du Y. Biomaterials as carrier, barrier and reactor for cell-based regenerative medicine. Protein Cell 2015; 6:638-53. [PMID: 26088192 PMCID: PMC4537472 DOI: 10.1007/s13238-015-0179-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/11/2015] [Indexed: 01/24/2023] Open
Abstract
Cell therapy has achieved tremendous success in regenerative medicine in the past several decades. However, challenges such as cell loss, death and immune-rejection after transplantation still persist. Biomaterials have been designed as carriers to deliver cells to desirable region for local tissue regeneration; as barriers to protect transplanted cells from host immune attack; or as reactors to stimulate host cell recruitment, homing and differentiation. With the assistance of biomaterials, improvement in treatment efficiency has been demonstrated in numerous animal models of degenerative diseases compared with routine free cell-based therapy. Emerging clinical applications of biomaterial assisted cell therapies further highlight their great promise in regenerative therapy and even cure for complex diseases, which have been failed to realize by conventional therapeutic approaches.
Collapse
Affiliation(s)
- Chunxiao Qi
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Xiaojun Yan
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
| | - Chenyu Huang
- />Department of Plastic and Reconstructive Surgery, Beijing Tsinghua Changgung Hospital; Medical Center, Tsinghua University, Beijing, 102218 China
| | - Alexander Melerzanov
- />Cellular and Molecular Technologies Laboratory, MIPT, Dolgoprudny, 141701 Russia
| | - Yanan Du
- />Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084 China
- />Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310003 China
| |
Collapse
|
26
|
He H, Nagamura-Inoue T, Takahashi A, Mori Y, Yamamoto Y, Shimazu T, Tsunoda H, Tojo A. Immunosuppressive properties of Wharton’s jelly-derived mesenchymal stromal cells in vitro. Int J Hematol 2015; 102:368-78. [DOI: 10.1007/s12185-015-1844-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 07/07/2015] [Accepted: 07/07/2015] [Indexed: 01/29/2023]
|
27
|
Wu H, Kang N, Wang Q, Dong P, Lv X, Cao Y, Xiao R. The Dose-Effect Relationship Between the Seeding Quantity of Human Marrow Mesenchymal Stem Cells and In Vivo Tissue-Engineered Bone Yield. Cell Transplant 2014; 24:1957-68. [PMID: 25398079 DOI: 10.3727/096368914x685393] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although the feasibility of human bone marrow mesenchymal stem cell (hBMMSC)-based tissue-engineered bone (TEB) has been proven in a number of studies, reaching a high positive fraction and bone yield of TEB still remains a challenge. Here we report a dose-effect relationship of the quantity of seeded cells with in vivo bone yield and the required quantity of hBMMSCs for the effective, stable bone formation of TEB. In our study, TEB was constructed using the static seeding technique with the gradient of seeding densities and volumes of passage 3 hBMMSCs. The in vitro characteristics of seeding efficiency, proliferation, viability, distribution, and osteogenic differentiation of hBMMSCs seeded on two commercial scaffolds of β-TCP and CHA were investigated using alamarBlue assay, live/dead staining, confocal laser scanning microscope, scanning electronic microscopy examination, and mRNA expression analysis of osteogenic differentiation markers. After 3 months of ectopic implantation, in vivo bone regeneration was examined by quantitative analysis of histology and micro-CT. The results showed that 10 × 10(6) cells/ml was the minimum cell seeding density for CHA and β-TCP to generate new bone in vivo. In addition, 20 × 10(6) cells/ml and 30 × 10(6) cells/ml were the saturating seeding densities for CHA and β-TCP to produce new bone effectively and stably, respectively. Thus, for different scaffolds, the saturating seeding density should be investigated first to ensure the effectiveness and stability of TEB construction with minimum donor injury, which is essential for the clinical application of TEB.
Collapse
Affiliation(s)
- Huanhuan Wu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | | | | | | | | | | | | |
Collapse
|
28
|
Li J, Baker BA, Mou X, Ren N, Qiu J, Boughton RI, Liu H. Biopolymer/Calcium phosphate scaffolds for bone tissue engineering. Adv Healthc Mater 2014; 3:469-84. [PMID: 24339420 DOI: 10.1002/adhm.201300562] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 11/15/2013] [Indexed: 11/08/2022]
Abstract
With nearly 30 years of progress, tissue engineering has shown promise in developing solutions for tissue repair and regeneration. Scaffolds, together with cells and growth factors, are key components of this development. Recently, an increasing number of studies have reported on the design and fabrication of scaffolding materials. In particular, inspired by the nature of bone, polymer/ceramic composite scaffolds have been studied extensively. The purpose of this paper is to review the recent progress of the naturally derived biopolymers and the methods applied to generate biomimetic biopolymer/calcium phosphate composites as well as their biomedical applications in bone tissue engineering.
Collapse
Affiliation(s)
- Jianhua Li
- State Key Lab of Crystal Materials, Shandong University; 27 Shandanan Road Jinan 250100 China
| | - Bryan. A. Baker
- Biosystems and Biomaterials Division, The National Institute of Standards and Technology; MD 20899-8300 USA
| | - Xiaoning Mou
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences; Beijing China
| | - Na Ren
- State Key Lab of Crystal Materials, Shandong University; 27 Shandanan Road Jinan 250100 China
| | - Jichuan Qiu
- State Key Lab of Crystal Materials, Shandong University; 27 Shandanan Road Jinan 250100 China
| | - Robert I. Boughton
- Department of Physics and Astronomy; Bowling Green State University; Bowling Green OH 43403 USA
| | - Hong Liu
- State Key Lab of Crystal Materials, Shandong University; 27 Shandanan Road Jinan 250100 China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences; Beijing China
| |
Collapse
|
29
|
Li J, Wang Y, Li Y, Sun J, Zhao G. The effect of combined regulation of the expression of peroxisome proliferator-activated receptor-γ and calcitonin gene-related peptide on alcohol-induced adipogenic differentiation of bone marrow mesenchymal stem cells. Mol Cell Biochem 2014; 392:39-48. [PMID: 24633961 DOI: 10.1007/s11010-014-2016-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/28/2014] [Indexed: 01/15/2023]
Abstract
Studies have shown that alcohol can upregulate the expression of peroxisome proliferator-activated receptor-γ (PPARγ) gene in bone marrow mesenchymal stem cells (BMSCs). High expression of PPARγ can promote adipogenic differentiation of BMSCs, and reduce their osteogenic differentiation. Abnormal proliferation of adipocytes and fatty accumulation in osteocytes can result in high intraosseous pressure and disturbance of blood circulation in the femoral head, which induces osteonecrosis of the femoral head (ONFH). Downregulation of PPARγ is efficient in inhibiting adipogenesis and maintaining osteogenesis of BMSCs, which might potentially reduce the incidence of ONFH. Calcitonin gene-related peptide (CGRP) is a neuropeptide gene which has been closely associated with bone regeneration. In this study, we aimed to observe the effect of combined regulation of the expression of PPARγ and CGRP genes on alcohol-induced adipogenic differentiation of BMSCs. Our results demonstrated that simultaneous downregulation of PPARγ and upregulation of CGRP was efficient in suppressing adipogenic differentiation of BMSCs and promoting their osteogenic differentiation. These findings might enlighten a novel approach for the prevention of ONFH.
Collapse
Affiliation(s)
- Jinfeng Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | | | | | | | | |
Collapse
|
30
|
He H, Nagamura-Inoue T, Tsunoda H, Yuzawa M, Yamamoto Y, Yorozu P, Agata H, Tojo A. Stage-specific embryonic antigen 4 in Wharton's jelly-derived mesenchymal stem cells is not a marker for proliferation and multipotency. Tissue Eng Part A 2014; 20:1314-24. [PMID: 24279891 DOI: 10.1089/ten.tea.2013.0333] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Umbilical cord Wharton's jelly (WJ) is a rich source of mesenchymal stem cells (MSCs) similar to bone marrow (BM) and adipose tissues. Stage-specific embryonic antigen (SSEA)4 has been reported as a stem cell marker in BM-derived MSCs, but whether SSEA4(+) cells have growth and differentiation advantages over SSEA4(-) cells remains controversial. To gain insight into the role of SSEA4, we studied SSEA4(+) cells in WJ-derived MSCs (WJ-MSCs). METHODS WJ-MSCs were collected by the explant (WJe-MSCs) or collagenase methods (WJc-MSCs) and analyzed by flow cytometry and reverse-transcription polymerase chain reaction (RT-PCR). To evaluate whether culture conditions influenced the SSEA4 expression, WJe-MSCs were cultured in the medium supplemented with different fetal bovine serum (FBS) concentrations. RESULTS SSEA4 was expressed for a long-term culture. In contrast, SSEA3(+) disappeared rapidly in early passages of the culture. The incidence of SSEA4(+) and SSEA3(+) cells was similar between WJe-MSCs and WJc-MSCs at passages P0-P9, except for transient depletion of SSEA4 expression in early passages of WJe-MSCs. These were CD73(+)CD105(+) cells that express embryonic stem cell markers detected by RT-PCR. No differences in growth and differentiation ability of osteocytes and adipocytes were observed between the sorted SSEA4(+) cells and SSEA4(-) cells. Further, SSEA4 expression in WJe-MSCs was significantly correlated with FBS concentration in the culture medium. DISCUSSION SSEA4, which may display altered expression profiles in response to culture conditions, may not be an essential marker of WJ-MSC multipotency.
Collapse
Affiliation(s)
- Haiping He
- 1 Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kagami H, Agata H, Inoue M, Asahina I, Tojo A, Yamashita N, Imai K. The use of bone marrow stromal cells (bone marrow-derived multipotent mesenchymal stromal cells) for alveolar bone tissue engineering: basic science to clinical translation. TISSUE ENGINEERING PART B-REVIEWS 2014; 20:229-32. [PMID: 24494719 DOI: 10.1089/ten.teb.2013.0578] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Bone tissue engineering is a promising field of regenerative medicine in which cultured cells, scaffolds, and osteogenic inductive signals are used to regenerate bone. Human bone marrow stromal cells (BMSCs) are the most commonly used cell source for bone tissue engineering. Although it is known that cell culture and induction protocols significantly affect the in vivo bone forming ability of BMSCs, the responsible factors of clinical outcome are poorly understood. The results from recent studies using human BMSCs have shown that factors such as passage number and length of osteogenic induction significantly affect ectopic bone formation, although such differences hardly affected the alkaline phosphatase activity or gene expression of osteogenic markers. Application of basic fibroblast growth factor helped to maintain the in vivo osteogenic ability of BMSCs. Importantly, responsiveness of those factors should be tested under clinical circumstances to improve the bone tissue engineering further. In this review, clinical application of bone tissue engineering was reviewed with putative underlying mechanisms.
Collapse
Affiliation(s)
- Hideaki Kagami
- 1 Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
In vitro induction of alkaline phosphatase levels predicts in vivo bone forming capacity of human bone marrow stromal cells. Stem Cell Res 2014; 12:428-40. [DOI: 10.1016/j.scr.2013.12.001] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 12/25/2022] Open
|
33
|
Ma J, Both SK, Yang F, Cui FZ, Pan J, Meijer GJ, Jansen JA, van den Beucken JJJP. Concise review: cell-based strategies in bone tissue engineering and regenerative medicine. Stem Cells Transl Med 2013; 3:98-107. [PMID: 24300556 DOI: 10.5966/sctm.2013-0126] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cellular strategies play an important role in bone tissue engineering and regenerative medicine (BTE/RM). Variability in cell culture procedures (e.g., cell types, cell isolation and expansion, cell seeding methods, and preculture conditions before in vivo implantation) may influence experimental outcome. Meanwhile, outcomes from initial clinical trials are far behind those of animal studies, which is suggested to be related to insufficient nutrient and oxygen supply inside the BTE/RM constructs as some complex clinical implementations require bone regeneration in too large a quantity. Coculture strategies, in which angiogenic cells are introduced into osteogenic cell cultures, might provide a solution for improving vascularization and hence increasing bone formation for cell-based constructs. So far, preclinical studies have demonstrated that cell-based tissue-engineered constructs generally induce more bone formation compared with acellular constructs. Further, cocultures have been shown to enhance vascularization and bone formation compared with monocultures. However, translational efficacy from animal studies to clinical use requires improvement, and the role implanted cells play in clinical bone regeneration needs to be further elucidated. In view of this, the present review provides an overview of the critical procedures during in vitro and in vivo phases for cell-based strategies (both monoculture and coculture) in BTE/RM to achieve more standardized culture conditions for future studies, and hence enhance bone formation.
Collapse
Affiliation(s)
- Jinling Ma
- Department of VIP Service and Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biomaterials and Department of Oral and Maxillofacial Surgery, Radboud University Medical Center, Nijmegen, The Netherlands; State Key Laboratory of New Ceramics and Fine Processing, Department of Materials Science & Engineering, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Inferior ectopic bone formation of mesenchymal stromal cells from adipose tissue compared to bone marrow: rescue by chondrogenic pre-induction. Stem Cell Res 2013; 11:1393-406. [PMID: 24140198 DOI: 10.1016/j.scr.2013.07.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 01/08/2023] Open
Abstract
Human mesenchymal stromal cells derived from bone marrow (BMSC) and adipose tissue (ATSC) represent a valuable source of progenitor cells for cell therapy and tissue engineering. While ectopic bone formation is a standard activity of human BMSC on calcium phosphate ceramics, the bone formation capacity of human ATSC has so far been unclear. The objectives of this study were to assess the therapeutic potency of ATSC for bone formation in an ectopic mouse model and determine molecular differences by standardized comparison with BMSC. Although ATSC contained less CD146(+) cells, exhibited better proliferation and displayed similar alkaline phosphatase activity upon osteogenic in vitro differentiation, cells did not develop into bone-depositing osteoblasts on β-TCP after 8weeks in vivo. Additionally, ATSC expressed less BMP-2, BMP-4, VEGF, angiopoietin and IL-6 and more adiponectin mRNA, altogether suggesting insufficient osteochondral commitment and reduced proangiogenic activity. Chondrogenic pre-induction of ATSC/β-TCP constructs with TGF-β and BMP-6 initiated ectopic bone formation in >75% of samples. Both chondrogenic pre-induction and the osteoconductive microenvironment of β-TCP were necessary for ectopic bone formation by ATSC pointing towards a need for inductive conditions/biomaterials to make this more easily accessible cell source attractive for future applications in bone regeneration.
Collapse
|
35
|
Naito H, Yoshimura M, Mizuno T, Takasawa S, Tojo T, Taniguchi S. The advantages of three-dimensional culture in a collagen hydrogel for stem cell differentiation. J Biomed Mater Res A 2013; 101:2838-45. [PMID: 23468218 DOI: 10.1002/jbm.a.34578] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/21/2012] [Accepted: 01/04/2013] [Indexed: 11/06/2022]
Abstract
We evaluated the advantages of three-dimensional (3D) culture in a collagen hydrogel for stem cell differentiation, including the morphology of differentiated cells, differentiation efficiency of stem cells from aged rat and cells after passaging and freeze/thawing. Rat mesenchymal stem cells (MSCs) from young and aged rats, and MSCs after passaging and freeze/thawing were induced to differentiate into osteoblasts in 3D and 2D cultures, and histological studies were performed. Differentiation efficiency was evaluated by markers of osteoblastic differentiation including Runx2 and osterix gene expressions, osteocalcin secretion and calcium deposition. MSCs were stained positive for alkaline phosphatase in 3D and 2D cultures. However, the morphology of differentiated cells in 3D culture, which was different from that in 2D culture, was similar to that of osteoblasts in vivo. Markers of osteoblastic differentiation in MSCs from aged rats in 3D culture were higher than those in MSCs from young rats in 2D culture. Markers of osteoblastic differentiation in MSCs after passaging and freeze/thawing in 3D culture were higher than those in nonpassaged MSCs in 2D culture. These results indicate that 3D culture in a collagen hydrogel has advantages for the differentiation of MSCs into osteoblasts with a similar phenotype to that of in vivo, when using even MSCs from aged donors or after passaging and freeze/thawing.
Collapse
Affiliation(s)
- Hiroshi Naito
- Department of Thoracic and Cardiovascular Surgery, Nara Medical University School of Medicine, Kashihara, Nara 634-8522, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Comparative sequential morphological analyses during in vitro chondrogenesis and osteogenesis of mesenchymal stem cells embedded in collagen gels. Med Mol Morphol 2013; 46:24-33. [DOI: 10.1007/s00795-012-0005-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 01/17/2012] [Indexed: 01/22/2023]
|
37
|
Alves H, Mentink A, Le B, van Blitterswijk CA, de Boer J. Effect of antioxidant supplementation on the total yield, oxidative stress levels, and multipotency of bone marrow-derived human mesenchymal stromal cells. Tissue Eng Part A 2013; 19:928-37. [PMID: 23173771 DOI: 10.1089/ten.tea.2011.0700] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone marrow-derived multipotent mesenchymal stromal cells (MSCs) are the most frequently investigated cell type for potential regenerative strategies because they are relatively easy to isolate and are able to differentiate into several mesenchymal lineages. Unfortunately, during ex vivo culture, MSCs present gradual loss of differentiation potential and reduced clinical efficacy. Reactive oxygen species (ROS) are associated with oxidative damage and accumulate during MSC expansion. Because ROS are believed to be involved in the loss of multipotency, we hypothesized that compounds with antioxidant activity have the capacity to scavenge ROS, prevent cellular damage, and rescue culture-induced loss of multipotency. In this manuscript, we show that antioxidant supplementation can partially rescue the loss of alkaline phosphatase expression induced by oxidizing agents and increases the yield of hMSCs, when supplemented to a fresh bone marrow aspirate. Concomitantly, oxidative DNA damage and ROS levels in hMSCs were reduced by antioxidants. We conclude that antioxidant supplementation during MSC expansion reduces the DNA damage load and increases the MSC yield.
Collapse
Affiliation(s)
- Hugo Alves
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
38
|
Prins HJ, Fernandes H, Rozemuller H, van Blitterswijk C, de Boer J, Martens ACM. Spatial distribution and survival of human and goat mesenchymal stromal cells on hydroxyapatite and β-tricalcium phosphate. J Tissue Eng Regen Med 2012; 10:233-44. [PMID: 23255230 DOI: 10.1002/term.1681] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 07/18/2012] [Accepted: 11/14/2012] [Indexed: 01/08/2023]
Abstract
The combination of scaffolds and mesenchymal stromal cells (MSCs) is a promising approach in bone tissue engineering (BTE). Knowledge on the survival, outgrowth and bone-forming capacity of MSCs in vivo is limited. Bioluminescence imaging (BLI), histomorphometry and immunohistochemistry were combined to study the fate of gene-marked goat and human MSCs (gMSCs, hMSCs) on scaffolds with different osteoinductive properties. Luciferase-GFP-labelled MSCs were seeded on hydroxyapatite (HA) or β-tricalcium phosphate (TCP), cultured for 7 days in vitro in osteogenic medium, implanted subcutaneously in immunodeficient mice and monitored with BLI for 6 weeks. The constructs were retrieved and processed for histomorphometry and detection of luciferase-positive cells (LPCs). For gMSCs, BLI revealed doubling of signal after 1 week, declining to 60% of input after 3 weeks and remaining constant until week 6. hMSCs showed a constant decrease of BLI signal to 25% of input, indicating no further expansion. Bone formation of gMSCs was two-fold higher on TCP than HA. hMSCs and gMSCs control samples produced equal amounts of bone on TCP. Upon transduction, there was a four-fold reduction in bone formation compared with untransduced hMSCs, and no bone was formed on HA. LPCs were detected at day 14, but were much less frequent at day 42. Striking differences were observed in spatial distribution. MSCs in TCP were found to be aligned and interconnected on the surface but were scattered in an unstructured fashion in HA. In conclusion, the spatial distribution of MSCs on the scaffold is critical for cell-scaffold-based BTE.
Collapse
Affiliation(s)
- Henk-Jan Prins
- Department of Immunology, University Medical Centre Utrecht, The Netherlands
| | - Hugo Fernandes
- Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | - Henk Rozemuller
- Department of Clinical Pharmacy, Cell Therapy Facility, University Medical Centre Utrecht, The Netherlands
| | | | - Jan de Boer
- Department of Tissue Regeneration, University of Twente, Enschede, The Netherlands
| | - Anton C M Martens
- Department of Immunology, University Medical Centre Utrecht, The Netherlands.,Department of Cell Biology, University Medical Centre Utrecht, The Netherlands
| |
Collapse
|
39
|
Stem cells in dentistry--Part II: Clinical applications. J Prosthodont Res 2012; 56:229-48. [PMID: 23137671 DOI: 10.1016/j.jpor.2012.10.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/03/2012] [Indexed: 12/29/2022]
Abstract
New technologies that facilitate solid alveolar ridge augmentation are receiving considerable attention in the field of prosthodontics because of the growing requirement for esthetic and functional reconstruction by dental implant treatments. Recently, several studies have demonstrated potential advantages for stem-cell-based therapies in regenerative treatments. Mesenchymal stem/stromal cells (MSCs) are now an excellent candidate for tissue replacement therapies, and tissue engineering approaches and chair-side cellular grafting approaches using autologous MSCs represent the clinical state of the art for stem-cell-based alveolar bone regeneration. Basic studies have revealed that crosstalk between implanted donor cells and recipient immune cells plays a key role in determining clinical success that may involve the recently observed immunomodulatory properties of MSCs. Part II of this review first overviews progress in regenerative dentistry to consider the implications of the stem cell technology in dentistry and then highlights cutting-edge stem-cell-based alveolar bone regenerative therapies. Factors that affect stem-cell-based bone regeneration as related to the local immune response are then discussed. Additionally, pre-clinical stem cell studies for the regeneration of teeth and other oral organs as well as possible applications of MSC-based immunotherapy in dentistry are outlined. Finally, the marketing of stem cell technology in dental stem cell banks with a view toward future regenerative therapies is introduced.
Collapse
|
40
|
Shiraishi T, Sumita Y, Wakamastu Y, Nagai K, Asahina I. Formation of Engineered Bone with Adipose Stromal Cells from Buccal Fat Pad. J Dent Res 2012; 91:592-7. [DOI: 10.1177/0022034512445633] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A robust method for inducing bone formation from adipose-derived stromal cells (ADSCs) has not been established. Moreover, the efficacy of strong osteogenic inducers including BMP-2 for ADSC-mediated bone engineering remains controversial. Meanwhile, the buccal fat pad (BFP), which is found in the oral cavity as an adipose-encapsulated mass, has been shown to have potential as a new accessible source of ADSCs for oral surgeons. However, to date, there have been no reports that define the practical usefulness of ADSCs from BFP (B-ADSCs) for bone engineering. Here, we report an efficient method of generating bone from B-ADSCs using rhBMP-2. The analyses show that B-ADSCs can differentiate in vitro toward the osteoblastic lineage by the addition of rhBMP-2 to culture medium, regardless of the presence of osteoinductive reagents (OSR), as demonstrated by measurements of ALP activity, in vitro calcification, and osteogenic gene expression. Interestingly, adipogenic genes were clearly detectable only in cultures with rhBMP-2 and OSR. However, in vivo bone formation was most substantial when B-ADSCs cultured in this condition were transplanted. Thus, B-ADSCs reliably formed engineered bone when pre-treated with rhBMP-2 for inducing mature osteoblastic differentiation. This study supports the potential translation for B-ADSC use in the clinical treatment of bone defects.
Collapse
Affiliation(s)
- T. Shiraishi
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Y. Sumita
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Y. Wakamastu
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - K. Nagai
- Blood Transfusion Service, Nagasaki University Hospital, Nagasaki, Japan
| | - I. Asahina
- Department of Regenerative Oral Surgery, Unit of Translational Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
41
|
Agata H, Yamazaki M, Uehara M, Hori A, Sumita Y, Tojo A, Kagami H. Characteristic differences among osteogenic cell populations of rat bone marrow stromal cells isolated from untreated, hemolyzed or Ficoll-treated marrow. Cytotherapy 2012; 14:791-801. [PMID: 22494074 DOI: 10.3109/14653249.2012.674639] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Although bone marrow (BM) stromal cells (SC; BMSC) isolated from adherent cultures of untreated BM are known to contain both committed and uncommitted osteogenic cells, it remains unknown whether BMSC isolated either by hemolysis or Ficoll centrifugation also contain both of these populations. METHODS Differences in the osteogenic cell populations of rat BMSC isolated from untreated, hemolyzed or Ficoll-treated BM were analyzed by in vivo transplantation, flow cytometry, alkaline phosphatase (ALP) assay, real-time polymerase chain reaction (PCR) and alizarin red staining. RESULTS Transplantation of non-cultured samples indicated that the Ficolled BMSC contained the lowest number of committed osteogenic cells. Flow cytometric analysis of cultured, non-induced samples showed that the percentage of ALP-positive cells was significantly lower in Ficolled BMSC. Quantitative ALP assays confirmed that the lowest ALP activity was in the Ficolled BMSC. Hemolyzed BMSC also contained lower numbers of committed osteogenic cells than untreated BMSC, but still more than Ficolled BMSC. Interestingly, the Ficolled BMSC showed the greatest levels of osteogenic ability when cultured in osteogenic induction medium. CONCLUSIONS These findings suggest that, although Ficolled BMSC rarely contain committed osteogenic cells, they are able to show comparable or even greater levels of osteogenic ability after induction, possibly because they contain a greater proportion of uncommitted stem cells. In contrast, induction is optional but recommended for both untreated and hemolyzed BMSC before use, because both these groups contain both committed and uncommitted osteogenic cells. These findings are of significant importance when isolating BMSC for use in bone tissue engineering.
Collapse
Affiliation(s)
- Hideki Agata
- Tissue Engineering Research Group, Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
42
|
Effects of osteogenic differentiation inducers on in vitro expanded adult mesenchymal stromal cells. Int J Artif Organs 2012; 34:998-1011. [PMID: 22161283 DOI: 10.5301/ijao.5000001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2011] [Indexed: 02/07/2023]
Abstract
PURPOSE For bone regeneration therapy using stem cells, well-defined ex vivo protocols to expand mesenchymal stromal cells (MSC), as well as assays to show their potential differentiation into the osteogenic lineage, are needed. Aim of this study was to analyze the role of the biochemical osteogenic inducers, i.e. ascorbic acid, dexamethasone, and ß-glycerophosphate, employed in the current protocols for osteogenic differentiation of MSC in vitro, to address the requirements for reliable differentiation systems. METHODS MSC were isolated from the bone marrow of donors (46-73 years of age) undergoing total hip replacement, and expanded in vitro. At confluence, MSC were cultured under four different conditions: α-MEM plus serum (basal medium or C1), basal medium plus ascorbate (C2), basal medium plus ascorbate and dexamethasone (C3), or basal medium plus ascorbate, dexamethasone and ß-glycerophosphate (C4). Morphology, proliferation, mineralization, alkaline phosphatase, collagen and expression of bone-related genes of MSC under the different media were analyzed at fixed time points. RESULTS MSC proliferation and the number of colony forming units were increased by ascorbic acid, whereas dexamethasone enhanced the proportion of ALP-positive CFU and was critical for mineral deposition. Runx-2 and type I collagen gene expression decreased along with additive-induced MSC differentiation, i.e. from C1 to C4, while ALP and osteocalcin were differently regulated. CONCLUSION Our findings support the role of different inducers on the sequential stages of MSC expansion and osteogenic differentiation in vitro, suggesting the addition of DEX following proliferation to ensure mineralization, as an index of in vivo osteogenic potency of human mesenchymal cells.
Collapse
|
43
|
Nguyen LTH, Liao S, Ramakrishna S, Chan CK. The role of nanofibrous structure in osteogenic differentiation of human mesenchymal stem cells with serial passage. Nanomedicine (Lond) 2011; 6:961-74. [DOI: 10.2217/nnm.11.26] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Using scaffolds with autologous stem cells is a golden strategy for the treatment of bone defects. In this strategy, human mesenchymal stem cells (hMSCs) have often been isolated and expanded in vitro on a plastic surface to obtain a sufficient cell number before seeding on a suitable scaffold. Materials & Methods: Investigating the influence of serial passages (from passage two to passage eight) on the abilities of proliferation and osteogenic differentiation of hMSCs on 24-well tissue culture polystyrene plates and poly L-lactic acid electrospun nanofibrous scaffolds was performed to determine how prolonged culture affected these cellular abilities and how the nanofibrous scaffolds supported the osteogenic differentiation potential of hMSCs. Results & Conclusion: Serial passage caused adverse changes in hMSCs characteristics, which were indicated by the decline in both proliferation and osteogenic differentiation abilities. Interestingly, the poly L-lactic acid nanofibrous scaffolds showed a significant support in recovering the osteogenic abilities of hMSCs, which had been severely affected by prolonged culture.
Collapse
Affiliation(s)
| | - Susan Liao
- Nanyang Technological University, 637551 Singapore
| | - Seeram Ramakrishna
- National University of Singapore, 117576 Singapore
- Institute of Materials Research & Engineering, 117602 Singapore
| | - Casey K Chan
- National University of Singapore, 117576 Singapore
| |
Collapse
|
44
|
Kagami H, Agata H, Tojo A. Bone marrow stromal cells (bone marrow-derived multipotent mesenchymal stromal cells) for bone tissue engineering: basic science to clinical translation. Int J Biochem Cell Biol 2010; 43:286-9. [PMID: 21147252 DOI: 10.1016/j.biocel.2010.12.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/29/2010] [Accepted: 12/06/2010] [Indexed: 12/13/2022]
Abstract
Bone tissue engineering is a promising field of regenerative medicine in which cultured cells, scaffolds, and osteogenic inductive signals are used to regenerate bone. This technology has already been used in several clinical studies and its efficacy has been reported. In this review, we focus on bone marrow stromal cells, which are the most commonly used cell source for bone tissue engineering. The nature of the cells, suitable culture conditions for bone tissue engineering, and their potential therapeutic applications are reviewed with possible caveats. Furthermore, recent advances in bone marrow stromal cell biology are discussed with reference to clinical translation.
Collapse
Affiliation(s)
- Hideaki Kagami
- Tissue Engineering Research Group, Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.
| | | | | |
Collapse
|
45
|
Kretlow JD, Spicer PP, Jansen JA, Vacanti CA, Kasper FK, Mikos AG. Uncultured marrow mononuclear cells delivered within fibrin glue hydrogels to porous scaffolds enhance bone regeneration within critical-sized rat cranial defects. Tissue Eng Part A 2010; 16:3555-68. [PMID: 20715884 DOI: 10.1089/ten.tea.2010.0471] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For bone tissue engineering, the benefits of incorporating mesenchymal stem cells (MSCs) into porous scaffolds are well established. There is, however, little consensus on the effects of or need for MSC handling ex vivo. Culture and expansion of MSCs adds length and cost, and likely increases risk associated with treatment. We evaluated the effect of using uncultured bone marrow mononuclear cells (bmMNCs) encapsulated within fibrin glue hydrogels and seeded into porous scaffolds to regenerate bone over 12 weeks in an 8-mm-diameter, critical-sized rat cranial defect. A full factorial experimental design was used to evaluate bone formation within model poly(L-lactic acid) and corraline hydroxyapatite scaffolds with or without platelet-rich plasma (PRP) and bmMNCs. Mechanical push-out testing, microcomputed tomographical analyses, and histology were performed. PRP showed no benefit for bone formation. Cell-laden poly(L-lactic acid) scaffolds without PRP required significantly greater force to displace from surrounding tissues than control (cell-free) scaffolds, but no differences were observed during push-out testing of coral scaffolds. For bone volume formation as analyzed by microcomputed tomography, significant positive overall effects were observed with bmMNC incorporation. These data suggest that bmMNCs may provide therapeutic advantages in bone tissue engineering applications without the need for culture, expansion, and purification.
Collapse
Affiliation(s)
- James D Kretlow
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
46
|
Rozemuller H, Prins HJ, Naaijkens B, Staal J, Bühring HJ, Martens AC. Prospective isolation of mesenchymal stem cells from multiple mammalian species using cross-reacting anti-human monoclonal antibodies. Stem Cells Dev 2010; 19:1911-21. [PMID: 20367498 DOI: 10.1089/scd.2009.0510] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) of human and nonhuman mammalian species are often studied for various applications in regenerative medicine research. These MSCs can be derived from human bone marrow (BM) and identified by their ability to form fibroblast-like colony forming units that develop into stromal like cells when expanded in culture. These cells are characterized by their spindle-shaped morphology, their characteristic phenotype (CD73(+), CD90(+), CD105(+), CD45⁻, and CD34⁻), and their ability to differentiate into cells of the osteogenic, adipogenic, and chondrogenic lineages. However, the identification and purification of MSCs from nonhuman mammalian species is hampered by the lack of suitable monoclonal antibodies (mAb). In this report, primary BM and cultured BM-derived MSCs of human and monkey, goat, sheep, dog, and pig were screened for cross-reactivity using a panel of 43 mAb, of which 22 react with either human BM mononuclear cells or cultured human MSCs. We found 7 mAb with specificity for CD271, MSCA-1 (W8B2 antigen), W4A5, CD56, W3C4 (CD349), W5C4, and 58B1, which showed interspecies cross-reactivity. These mAb proved to be useful for prospective sorting of MSCs from the BM of the 6 mammalian species studied as well as for the characterization of their cultured offspring. Flow sorting with the cross-reacting mAb resulted in up to 2400-fold enrichment of the clonogenic cell fraction (fibroblast-like colony forming units). This study provides an important contribution for the comparative prospective isolation of primary BM-MSCs and the characterization of cultured MSCs from multiple mammalian species for preclinical research.
Collapse
Affiliation(s)
- Henk Rozemuller
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
47
|
|