1
|
Kessler F, Arnke K, Eggerschwiler B, Neldner Y, Märsmann S, Gröninger O, Casanova EA, Weber FA, König MA, Stark WJ, Pape HC, Cinelli P, Tiziani S. Murine iPSC-Loaded Scaffold Grafts Improve Bone Regeneration in Critical-Size Bone Defects. Int J Mol Sci 2024; 25:5555. [PMID: 38791592 PMCID: PMC11121928 DOI: 10.3390/ijms25105555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
In certain situations, bones do not heal completely after fracturing. One of these situations is a critical-size bone defect where the bone cannot heal spontaneously. In such a case, complex fracture treatment over a long period of time is required, which carries a relevant risk of complications. The common methods used, such as autologous and allogeneic grafts, do not always lead to successful treatment results. Current approaches to increasing bone formation to bridge the gap include the application of stem cells on the fracture side. While most studies investigated the use of mesenchymal stromal cells, less evidence exists about induced pluripotent stem cells (iPSC). In this study, we investigated the potential of mouse iPSC-loaded scaffolds and decellularized scaffolds containing extracellular matrix from iPSCs for treating critical-size bone defects in a mouse model. In vitro differentiation followed by Alizarin Red staining and quantitative reverse transcription polymerase chain reaction confirmed the osteogenic differentiation potential of the iPSCs lines. Subsequently, an in vivo trial using a mouse model (n = 12) for critical-size bone defect was conducted, in which a PLGA/aCaP osteoconductive scaffold was transplanted into the bone defect for 9 weeks. Three groups (each n = 4) were defined as (1) osteoconductive scaffold only (control), (2) iPSC-derived extracellular matrix seeded on a scaffold and (3) iPSC seeded on a scaffold. Micro-CT and histological analysis show that iPSCs grafted onto an osteoconductive scaffold followed by induction of osteogenic differentiation resulted in significantly higher bone volume 9 weeks after implantation than an osteoconductive scaffold alone. Transplantation of iPSC-seeded PLGA/aCaP scaffolds may improve bone regeneration in critical-size bone defects in mice.
Collapse
Affiliation(s)
- Franziska Kessler
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Kevin Arnke
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Benjamin Eggerschwiler
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Yvonne Neldner
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Sonja Märsmann
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Olivier Gröninger
- Institute for Chemical and Bioengineering, ETH Zurich, 8093 Zurich, Switzerland
| | - Elisa A. Casanova
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Fabienne A. Weber
- Institute of Laboratory Animal Science, University of Zurich, 8091 Zurich, Switzerland
| | | | - Wendelin J. Stark
- Institute for Chemical and Bioengineering, ETH Zurich, 8093 Zurich, Switzerland
| | - Hans-Christoph Pape
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| | - Paolo Cinelli
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
- Center for Applied Biotechnology and Molecular Medicine (CABMM), University of Zurich, 8057 Zurich, Switzerland
| | - Simon Tiziani
- Department of Trauma Surgery, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091 Zurich, Switzerland (E.A.C.); (P.C.)
| |
Collapse
|
2
|
Jiang N, Tian X, Wang Q, Hao J, Jiang J, Wang H. Regulation Mechanisms and Maintenance Strategies of Stemness in Mesenchymal Stem Cells. Stem Cell Rev Rep 2024; 20:455-483. [PMID: 38010581 DOI: 10.1007/s12015-023-10658-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Stemness pertains to the intrinsic ability of mesenchymal stem cells (MSCs) to undergo self-renewal and differentiate into multiple lineages, while simultaneously impeding their differentiation and preserving crucial differentiating genes in a state of quiescence and equilibrium. Owing to their favorable attributes, including uncomplicated isolation protocols, ethical compliance, and ease of procurement, MSCs have become a focal point of inquiry in the domains of regenerative medicine and tissue engineering. As age increases or ex vivo cultivation is prolonged, the functionality of MSCs decreases and their stemness gradually diminishes, thereby limiting their potential therapeutic applications. Despite the existence of several uncertainties surrounding the comprehension of MSC stemness, considerable advancements have been achieved in the clarification of the potential mechanisms that lead to stemness loss, as well as the associated strategies for stemness maintenance. This comprehensive review provides a systematic overview of the factors influencing the preservation of MSC stemness, the molecular mechanisms governing it, the strategies for its maintenance, and the therapeutic potential associated with stemness. Finally, we underscore the obstacles and prospective avenues in present investigations, providing innovative perspectives and opportunities for the preservation and therapeutic utilization of MSC stemness.
Collapse
Affiliation(s)
- Nizhou Jiang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiliang Tian
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Quanxiang Wang
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China
| | - Jian Jiang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China.
| | - Hong Wang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China.
| |
Collapse
|
3
|
Arakawa M, Sakamoto Y, Miyagawa Y, Nito C, Takahashi S, Nitahara-Kasahara Y, Suda S, Yamazaki Y, Sakai M, Kimura K, Okada T. iPSC-derived mesenchymal stem cells attenuate cerebral ischemia-reperfusion injury by inhibiting inflammatory signaling and oxidative stress. Mol Ther Methods Clin Dev 2023; 30:333-349. [PMID: 37637385 PMCID: PMC10448333 DOI: 10.1016/j.omtm.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/11/2023] [Indexed: 08/29/2023]
Abstract
Induced pluripotent stem cell-derived mesenchymal stem cells (iMSCs) hold great promise as a cell source for transplantation into injured tissues to alleviate inflammation. However, the therapeutic efficacy of iMSC transplantation for ischemic stroke remains unknown. In this study, we evaluated the therapeutic effects of iMSC transplantation on brain injury after ischemia-reperfusion using a rat transient middle cerebral artery occlusion model and compared its therapeutic efficacy with that of bone marrow mesenchymal stem cells (BMMSCs). We showed that iMSCs and BMMSCs reduced infarct volumes after reperfusion and significantly improved motor function on days 3, 7, 14, 28, and 56 and cognitive function on days 28 and 56 after reperfusion compared with the vehicle group. Furthermore, immunological analyses revealed that transplantation of iMSCs and BMMSCs inhibited microglial activation and expression of proinflammatory cytokines and suppressed oxidative stress and neuronal cell death in the cerebral cortex at the ischemic border zone. No difference in therapeutic effect was observed between the iMSC and BMMSC groups. Taken together, our results demonstrate that iMSC therapy can be a practical alternative as a cell source for attenuation of brain injury and improvement of neurological function because of the unlimited supply of uniform therapeutic cells.
Collapse
Affiliation(s)
- Masafumi Arakawa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuki Sakamoto
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshitaka Miyagawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Laboratory for Clinical Research, Collaborative Research Center, Nippon Medical School, Tokyo, Japan
| | - Shiro Takahashi
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yoshiyuki Yamazaki
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Mashito Sakai
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Dias IX, Cordeiro A, Guimarães JAM, Silva KR. Potential and Limitations of Induced Pluripotent Stem Cells-Derived Mesenchymal Stem Cells in Musculoskeletal Disorders Treatment. Biomolecules 2023; 13:1342. [PMID: 37759742 PMCID: PMC10526864 DOI: 10.3390/biom13091342] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 09/29/2023] Open
Abstract
The burden of musculoskeletal disorders (MSK) is increasing worldwide. It affects millions of people worldwide, decreases their quality of life, and can cause mortality. The treatment of such conditions is challenging and often requires surgery. Thus, it is necessary to discuss new strategies. The therapeutic potential of mesenchymal stem cells (MSC) in several diseases has been investigated with relative success. However, this potential is hindered by their limited stemness and expansion ability in vitro and their high donor variability. MSC derived from induced pluripotent stem cells (iPSC) have emerged as an alternative treatment for MSK diseases. These cells present distinct features, such as a juvenile phenotype, in addition to higher stemness, proliferation, and differentiation potential than those of MSC. Here, we review the opportunities, challenges, and applications of iPSC as relevant clinical therapeutic cell sources for MSK disorders. We discuss iPSC sources from which to derive iMSC and the advantages and disadvantages of iMSC over MSC as a therapeutic approach. We further summarize the main preclinical and clinical studies exploring the therapeutic potential of iMSC in MSK disorders.
Collapse
Affiliation(s)
- Isabelle Xavier Dias
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - Aline Cordeiro
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - João Antonio Matheus Guimarães
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
| | - Karina Ribeiro Silva
- Teaching and Research Division, National Institute of Traumatology and Orthopaedics, Rio de Janeiro 20940-070, Brazil; (A.C.); (J.A.M.G.)
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550-170, Brazil
| |
Collapse
|
5
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
6
|
Dewey MJ, Collins AJ, Tiffany A, Barnhouse VR, Lu C, Kolliopoulos V, Mutreja I, Hickok NJ, Harley BAC. Evaluation of bacterial attachment on mineralized collagen scaffolds and addition of manuka honey to increase mesenchymal stem cell osteogenesis. Biomaterials 2023; 294:122015. [PMID: 36701999 PMCID: PMC9928779 DOI: 10.1016/j.biomaterials.2023.122015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023]
Abstract
The design of biomaterials to regenerate bone is likely to increasingly require modifications that reduce bacterial attachment and biofilm formation as infection during wound regeneration can significantly impede tissue repair and typically requires surgical intervention to restart the healing process. Further, much research on infection prevention in bone biomaterials has focused on modeling of non-resorbable metal alloy materials, whereas an expanding direction of bone regeneration has focused on development of bioresorbable materials. This represents a need for the prevention and understanding of infection in resorbable biomaterials. Here, we investigate the ability of a mineralized collagen biomaterial to natively resist infection and examine how the addition of manuka honey, previously identified as an antimicrobial agent, affects gram positive and negative bacterial colonization and mesenchymal stem cell osteogenesis and vasculature formation. We incorporate manuka honey into these scaffolds via either direct fabrication into the scaffold microarchitecture or via soaking the scaffold in a solution of manuka honey after fabrication. Direct incorporation results in a change in the surface characteristics and porosity of mineralized collagen scaffolds. Soaking scaffolds in honey concentrations higher than 10% had significant negative effects on mesenchymal stem cell metabolic activity. Soaking or incorporating 5% honey had no impact on endothelial cell tube formation. Although solutions of 5% honey reduced metabolic activity of mesenchymal stem cells, MSC-seeded scaffolds displayed increased calcium and phosphorous mineral formation, osteoprotegerin release, and alkaline phosphatase activity. Bacteria cultured on mineralized collagen scaffolds demonstrated surfaces covered in bacteria and no method of preventing infection, and using 10 times the minimal inhibitory concentration of antibiotics did not completely kill bacteria within the mineralized collagen scaffolds, indicating bioresorbable scaffold materials may act to shield bacteria from antibiotics. The addition of 5% manuka honey to scaffolds was not sufficient to prevent P. aeruginosa attachment or consistently reduce the activity of methicillin resistant staphylococcus aureus, and concentrations above 7% manuka honey are likely necessary to impact MRSA. Together, our results suggest bioresorbable scaffolds may create an environment conducive to bacterial growth, and potential trade-offs exist for the incorporation of low levels of honey in scaffolds to increase osteogenic potential of osteoprogenitors while high-levels of honey may be sufficient to reduce gram positive or negative bacteria activity but at the cost of reduced osteogenesis.
Collapse
Affiliation(s)
- Marley J Dewey
- Dept. of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Alan J Collins
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Aleczandria Tiffany
- Dept. of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Victoria R Barnhouse
- Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Crislyn Lu
- School of Chemical Sciences, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Vasiliki Kolliopoulos
- Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Isha Mutreja
- Department of Restorative Science, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Brendan A C Harley
- Dept. of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Dept. of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Dept. of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
7
|
Osteogenesis of Human iPSC-Derived MSCs by PLLA/SF Nanofiber Scaffolds Loaded with Extracellular Matrix. J Tissue Eng Regen Med 2023. [DOI: 10.1155/2023/5280613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bone defects that arise from trauma, skeletal diseases, or tumor resections have become the commonest and most thorny problems in orthopedic clinics. Recently, biocomposite materials used as artificial bone repair materials have provided a promising approach for bone regeneration. In this study, poly (l-lactide acid) (PLLA) and silk fibroin (SF) were used to fabricate nanofiber scaffolds by electrospinning technology. In order to simulate a biomimetic osteoblast microenvironment, decellularized extracellular matrix from osteoblasts was loaded into the biocomposite scaffolds (O-ECM/PLLA/SF). It was found that the O-ECM/PLLA/SF scaffolds were nontoxic for L929 cells and had good cytocompatibility. Their effects on mesenchymal stem cells derived from human-induced pluripotent stem cell (iPSC-MSC) behavior were investigated. As a result, the scaffolds with the addition of O-ECM showed enhanced alizarin red S (ARS) activity. In addition, higher expression of osteogenic gene markers such as runt-related transcription factor 2 (Runx2), collagen type I (Col-1), and osteocalcin (OCN) as well as upregulated expression of osteogenic marker protein osteopontin (OPN) and Col-1 further substantiated the applicability of O-ECM/PLLA/SF scaffolds for osteogenesis. Furthermore, the in vivo study also indicated maximal new bone formation in the skull defect model of Sprague Dawley (SD) rats treated with the O-ECM/PLLA/SF carried by human iPSC-MSCs. Hence, this study suggests that O-ECM/PLLA/SF scaffolds have a potential application in bone tissue engineering.
Collapse
|
8
|
Li G, Shen W, Chu M, Mo G, Yao L, Xu W. Effect of inoculation density of bone marrow mesenchymal stem cells cultured on calcium phosphate cement scaffold on osteogenic differentiation. Biomed Mater Eng 2023; 34:111-121. [PMID: 35871314 DOI: 10.3233/bme-221394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Calcium phosphate cements (CPCs) are biocompatible materials that have been evaluated as scaffolds in bone tissue engineering. At present, the stem cell density of inoculation on CPC scaffold varies. OBJECTIVE The aim of this study is to analyze the effect of seeding densities on cell growth and osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs) on a calcium phosphate cements (CPCs) scaffold. METHODS BMMSCs derived from minipigs were seeded onto a CPC scaffold at three densities [1 million/mL (1M), 5 million/mL (5M) and 25 million/mL 25M)], and cultured for osteogenic induction for 1, 4 and 8 days. RESULTS Well adhered and extended BMMSCs on the CPC scaffold showed significantly different proliferation rates within each seeding density group at different time points (P < 0.05). The number of live cells per unit area in 1M, 5M and 25M increased by 3.5, 3.9 and 2.5 folds respectively. The expression of ALP peaked at 4 days post inoculation with the fold-change being 2.6 and 2.8 times higher in 5M and 25M respectively as compared to 1M. The expression levels of OC, Coll-1 and Runx-2 peaked at 8 days post inoculation. CONCLUSIONS An optimal seeding density may be more conducive for cell proliferation, differentiation, and extracellular matrix synthesis on scaffolds. We suggest the optimal seeding density should be 5 million/mL.
Collapse
Affiliation(s)
- Guangjun Li
- Department of Orthopedics, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Wen Shen
- Department of Radiology, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Minghui Chu
- Department of Orthopedics, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Guowei Mo
- Department of Orthopedics, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Liqin Yao
- Department of Orthopedics, Deqing People's Hospital, Deqing, Zhejiang, China
| | - Weidong Xu
- Department of Orthopedics, Deqing People's Hospital, Deqing, Zhejiang, China
| |
Collapse
|
9
|
Regeneration of periodontal bone defects with mesenchymal stem cells in animal models. Systematic review and meta-analysis. Odontology 2023; 111:105-122. [PMID: 35788845 PMCID: PMC9810679 DOI: 10.1007/s10266-022-00725-5] [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: 03/12/2022] [Accepted: 06/08/2022] [Indexed: 01/07/2023]
Abstract
The aim of this study was to evaluate the efficacy of mesenchymal stem cells (MSCs) in the regeneration of periodontal bone defects in animal models. A systematic review and meta-analysis were conducted following the PRISMA guidelines, and the study was recorded in PROSPERO under reference number CDR42021247462. The PICO question was: is periodontal regeneration (cementum, periodontal ligament and alveolar bone) with MSCs more effective than other techniques? Three groups were considered: Group 1: MSCs alone or mixed with regenerative materials. Group 2: only regenerative materials. Group 3: no regenerative material nor MSCs. The search was conducted using MeSH with a total of 18 articles for qualitative analysis and 5 for quantitative analysis. For the meta-analysis, a modification of the effect size algorithm was developed, which considered a comparison of means between treatments using the Student's t sample distribution. When comparing the effect size between Group 1 and Group 2, the effect size for the new cementum was 2.83 mm with an estimated confidence interval of 95% (CI 95%) between 0.48 and 5.17 mm. When considering the fit to a random-effects model, the combined variance (τ2) was 6.1573 mm, with a standard deviation (SD) of 5.6008 mm and a percentage of total heterogeneity I2 of 92.33% (p < 0.0001). For new bone, the effect size was 0.88 mm, CI 95% - 0.25 to 2.01 mm, τ2 = 1.3108 mm (SD = 1.2021 mm) and I2 = 80.46%, p = 0.0004). With regard to the new periodontal ligament, it was not possible for the meta-analysis to be performed. MSCs have a greater capacity for tissue regeneration in root cementum than in alveolar bone compared to other regenerative materials.
Collapse
|
10
|
Current Application of iPS Cells in the Dental Tissue Regeneration. Biomedicines 2022; 10:biomedicines10123269. [PMID: 36552025 PMCID: PMC9775967 DOI: 10.3390/biomedicines10123269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
When teeth and periodontal tissues are severely damaged by severe caries, trauma, and periodontal disease, such cases may be subject to tooth extraction. As tooth loss leads to the deterioration of quality of life, the development of regenerative medicine for tooth and periodontal tissue is desired. Induced pluripotent stem cells (iPS cells) are promising cell resources for dental tissue regeneration because they offer high self-renewal and pluripotency, along with fewer ethical issues than embryonic stem cells. As iPS cells retain the epigenetic memory of donor cells, they have been established from various dental tissues for dental tissue regeneration. This review describes the regeneration of dental tissue using iPS cells. It is important to mimic the process of tooth development in dental tissue regeneration using iPS cells. Although iPS cells had safety issues in clinical applications, they have been overcome in recent years. Dental tissue regeneration using iPS cells has not yet been established, but it is expected in the future.
Collapse
|
11
|
Yazdanian M, Alam M, Abbasi K, Rahbar M, Farjood A, Tahmasebi E, Tebyaniyan H, Ranjbar R, Hesam Arefi A. Synthetic materials in craniofacial regenerative medicine: A comprehensive overview. Front Bioeng Biotechnol 2022; 10:987195. [PMID: 36440445 PMCID: PMC9681815 DOI: 10.3389/fbioe.2022.987195] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
The state-of-the-art approach to regenerating different tissues and organs is tissue engineering which includes the three parts of stem cells (SCs), scaffolds, and growth factors. Cellular behaviors such as propagation, differentiation, and assembling the extracellular matrix (ECM) are influenced by the cell's microenvironment. Imitating the cell's natural environment, such as scaffolds, is vital to create appropriate tissue. Craniofacial tissue engineering refers to regenerating tissues found in the brain and the face parts such as bone, muscle, and artery. More biocompatible and biodegradable scaffolds are more commensurate with tissue remodeling and more appropriate for cell culture, signaling, and adhesion. Synthetic materials play significant roles and have become more prevalent in medical applications. They have also been used in different forms for producing a microenvironment as ECM for cells. Synthetic scaffolds may be comprised of polymers, bioceramics, or hybrids of natural/synthetic materials. Synthetic scaffolds have produced ECM-like materials that can properly mimic and regulate the tissue microenvironment's physical, mechanical, chemical, and biological properties, manage adherence of biomolecules and adjust the material's degradability. The present review article is focused on synthetic materials used in craniofacial tissue engineering in recent decades.
Collapse
Affiliation(s)
- Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Rahbar
- Department of Restorative Dentistry, School of Dentistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amin Farjood
- Orthodontic Department, Dental School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Arian Hesam Arefi
- Dental Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
12
|
Hua Z, Li S, Liu Q, Yu M, Liao M, Zhang H, Xiang X, Wu Q. Low-Intensity Pulsed Ultrasound Promotes Osteogenic Potential of iPSC-Derived MSCs but Fails to Simplify the iPSC-EB-MSC Differentiation Process. Front Bioeng Biotechnol 2022; 10:841778. [PMID: 35656194 PMCID: PMC9152674 DOI: 10.3389/fbioe.2022.841778] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) are a promising cell source for bone tissue engineering. However, iMSCs have less osteogenic potential than BMSCs, and the classical iPSC-EB-iMSC process to derive iMSCs from iPSCs is too laborious as it involves multiple in vitro steps. Low-intensity pulsed ultrasound (LIPUS) is a safe therapeutic modality used to promote osteogenic differentiation of stem cells. Whether LIPUS can facilitate osteogenic differentiation of iMSCs and simplify the iPSC-EB-iMSC process is unknown. We stimulated iMSCs with LIPUS at different output intensities (20, 40, and 60 mW/cm2) and duty cycles (20, 50, and 80%). Results of ALP activity assay, osteogenic gene expression, and mineralization quantification demonstrated that LIPUS was able to promote osteogenic differentiation of iMSCs, and it worked best at the intensity of 40 mW/cm2 and the duty cycle of 50% (LIPUS40/50). The Wnt/β-catenin signaling pathway was involved in LIPUS40/50-mediated osteogenesis. When cranial bone defects were implanted with iMSCs, LIPUS40/50 stimulation resulted in a significant higher new bone filling rate (72.63 ± 17.04)% than the non-stimulated ones (34.85 ± 4.53)%. Daily exposure to LIPUS40/50 may accelerate embryoid body (EB)-MSC transition, but it failed to drive iPSCs or EB cells to an osteogenic lineage directly. This study is the first to demonstrate the pro-osteogenic effect of LIPUS on iMSCs. Although LIPUS40/50 failed to simplify the classical iPSC-EB-MSC differentiation process, our preliminary results suggest that LIPUS with a more suitable parameter set may achieve the goal. LIPUS is a promising method to establish an efficient model for iPSC application.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingqing Wu
- *Correspondence: Qingqing Wu, ; Xuerong Xiang,
| |
Collapse
|
13
|
Liu TM. Application of mesenchymal stem cells derived from human pluripotent stem cells in regenerative medicine. World J Stem Cells 2021; 13:1826-1844. [PMID: 35069985 PMCID: PMC8727229 DOI: 10.4252/wjsc.v13.i12.1826] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/29/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent the most clinically used stem cells in regenerative medicine. However, due to the disadvantages with primary MSCs, such as limited cell proliferative capacity and rarity in the tissues leading to limited MSCs, gradual loss of differentiation during in vitro expansion reducing the efficacy of MSC application, and variation among donors increasing the uncertainty of MSC efficacy, the clinical application of MSCs has been greatly hampered. MSCs derived from human pluripotent stem cells (hPSC-MSCs) can circumvent these problems associated with primary MSCs. Due to the infinite self-renewal of hPSCs and their differentiation potential towards MSCs, hPSC-MSCs are emerging as an attractive alternative for regenerative medicine. This review summarizes the progress on derivation of MSCs from human pluripotent stem cells, disease modelling and drug screening using hPSC-MSCs, and various applications of hPSC-MSCs in regenerative medicine. In the end, the challenges and concerns with hPSC-MSC applications are also discussed.
Collapse
Affiliation(s)
- Tong-Ming Liu
- Agency for Science, Technology and Research, Institute of Molecular and Cell Biology, Singapore 138648, Singapore
| |
Collapse
|
14
|
Bahraminasab M, Janmohammadi M, Arab S, Talebi A, Nooshabadi VT, Koohsarian P, Nourbakhsh MS. Bone Scaffolds: An Incorporation of Biomaterials, Cells, and Biofactors. ACS Biomater Sci Eng 2021; 7:5397-5431. [PMID: 34797061 DOI: 10.1021/acsbiomaterials.1c00920] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Large injuries to bones are still one of the most challenging musculoskeletal problems. Tissue engineering can combine stem cells, scaffold biomaterials, and biofactors to aid in resolving this complication. Therefore, this review aims to provide information on the recent advances made to utilize the potential of biomaterials for making bone scaffolds and the assisted stem cell therapy and use of biofactors for bone tissue engineering. The requirements and different types of biomaterials used for making scaffolds are reviewed. Furthermore, the importance of stem cells and biofactors (growth factors and extracellular vesicles) in bone regeneration and their use in bone scaffolds and the key findings are discussed. Lastly, some of the main obstacles in bone tissue engineering and future trends are highlighted.
Collapse
Affiliation(s)
- Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Mahsa Janmohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, Semnan University, Semnan 3513119111, Iran
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Athar Talebi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Parisa Koohsarian
- Department of Biochemistry and Hematology, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | | |
Collapse
|
15
|
Dewey MJ, Milner DJ, Weisgerber D, Flanagan CL, Rubessa M, Lotti S, Polkoff KM, Crotts S, Hollister SJ, Wheeler MB, Harley BAC. Repair of critical-size porcine craniofacial bone defects using a collagen-polycaprolactone composite biomaterial. Biofabrication 2021; 14. [PMID: 34663761 DOI: 10.1088/1758-5090/ac30d5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 10/18/2021] [Indexed: 12/16/2022]
Abstract
Regenerative medicine approaches for massive craniomaxillofacial (CMF) bone defects face challenges associated with the scale of missing bone, the need for rapid graft-defect integration, and challenges related to inflammation and infection. Mineralized collagen scaffolds have been shown to promote mesenchymal stem cell osteogenesis due to their porous nature and material properties, but are mechanically weak, limiting surgical practicality. Previously, these scaffolds were combined with 3D-printed polycaprolactone (PCL) mesh to form a scaffold-mesh composite to increase strength and promote bone formation in sub-critical sized porcine ramus defects. Here, we compare the performance of mineralized collagen-PCL composites to the PCL mesh in a critical-sized porcine ramus defect model. While there were no differences in overall healing response between groups, our data demonstrated broadly variable metrics of healing regarding new bone infiltration and fibrous tissue formation. Abscesses were present surrounding some implants and PCL polymer was still present after 9-10 months of implantation. Overall, while there was limited successful healing, with 2 of 22 implants showed substantial levels of bone regeneration, and others demonstrating some form of new bone formation, the results suggest targeted improvements to improve repair of large animal models to more accurately represent CMF bone healing. Notably, strategies to increase osteogenesis throughout the implant, modulate the immune system to support repair, and employ shape-fitting tactics to avoid implant micromotion and resultant fibrosis. Improvements to the mineralized collagen scaffolds involve changes in pore size and shape to increase cell migration and osteogenesis and inclusion or delivery of factors to aid vascular ingrowth and bone regeneration.
Collapse
Affiliation(s)
- Marley J Dewey
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Derek J Milner
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Daniel Weisgerber
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Colleen L Flanagan
- Department of Bioengineering, University of Michigan, Ann Arbor, MI, 30332, United States of America
| | - Marcello Rubessa
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Sammi Lotti
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Kathryn M Polkoff
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Sarah Crotts
- Center for 3D Medical Fabrication, Wallace A. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, United States of America
| | - Scott J Hollister
- Center for 3D Medical Fabrication, Wallace A. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, United States of America
| | - Matthew B Wheeler
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| | - Brendan A C Harley
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America.,Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, United States of America
| |
Collapse
|
16
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2021; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
17
|
Alsahafi RA, Mitwalli HA, Balhaddad AA, Weir MD, Xu HHK, Melo MAS. Regenerating Craniofacial Dental Defects With Calcium Phosphate Cement Scaffolds: Current Status and Innovative Scope Review. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.743065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The management and treatment of dental and craniofacial injuries have continued to evolve throughout the last several decades. Limitations with autograft, allograft, and synthetics created the need for more advanced approaches in tissue engineering. Calcium phosphate cements (CPC) are frequently used to repair bone defects. Since their discovery in the 1980s, extensive research has been conducted to improve their properties, and emerging evidence supports their increased application in bone tissue engineering. This review focuses on the up-to-date performance of calcium phosphate cement (CPC) scaffolds and upcoming promising dental and craniofacial bone regeneration strategies. First, we summarized the barriers encountered in CPC scaffold development. Second, we compiled the most up to date in vitro and in vivo literature. Then, we conducted a systematic search of scientific articles in MEDLINE and EMBASE to screen the related studies. Lastly, we revealed the current developments to effectively design CPC scaffolds and track the enhanced viability and therapeutic efficacy to overcome the current limitations and upcoming perspectives. Finally, we presented a timely and opportune review article focusing on the significant potential of CPC scaffolds for dental and craniofacial bone regeneration, which will be discussed thoroughly. CPC offers multiple capabilities that may be considered toward the oral defects, expecting a future outlook in nanotechnology design and performance.
Collapse
|
18
|
Dupuis V, Oltra E. Methods to produce induced pluripotent stem cell-derived mesenchymal stem cells: Mesenchymal stem cells from induced pluripotent stem cells. World J Stem Cells 2021; 13:1094-1111. [PMID: 34567428 PMCID: PMC8422924 DOI: 10.4252/wjsc.v13.i8.1094] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/03/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have received significant attention in recent years due to their large potential for cell therapy. Indeed, they secrete a wide variety of immunomodulatory factors of interest for the treatment of immune-related disorders and inflammatory diseases. MSCs can be extracted from multiple tissues of the human body. However, several factors may restrict their use for clinical applications: the requirement of invasive procedures for their isolation, their limited numbers, and their heterogeneity according to the tissue of origin or donor. In addition, MSCs often present early signs of replicative senescence limiting their expansion in vitro, and their therapeutic capacity in vivo. Due to the clinical potential of MSCs, a considerable number of methods to differentiate induced pluripotent stem cells (iPSCs) into MSCs have emerged. iPSCs represent a new reliable, unlimited source to generate MSCs (MSCs derived from iPSC, iMSCs) from homogeneous and well-characterized cell lines, which would relieve many of the above mentioned technical and biological limitations. Additionally, the use of iPSCs prevents some of the ethical concerns surrounding the use of human embryonic stem cells. In this review, we analyze the main current protocols used to differentiate human iPSCs into MSCs, which we classify into five different categories: MSC Switch, Embryoid Body Formation, Specific Differentiation, Pathway Inhibitor, and Platelet Lysate. We also evaluate common and method-specific culture components and provide a list of positive and negative markers for MSC characterization. Further guidance on material requirements to produce iMSCs with these methods and on the phenotypic features of the iMSCs obtained is added. The information may help researchers identify protocol options to design and/or refine standardized procedures for large-scale production of iMSCs fitting clinical demands.
Collapse
Affiliation(s)
- Victoria Dupuis
- Faculté des Sciences et d’Ingénierie, Sorbonne Université, Paris 75252, France
| | - Elisa Oltra
- Department of Pathology, Universidad Católica de Valencia San Vicente Mártir, Valencia 46001, Spain
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, Valencia 46001, Spain
| |
Collapse
|
19
|
Kaboodkhani R, Mehrabani D, Karimi-Busheri F. Achievements and Challenges in Transplantation of Mesenchymal Stem Cells in Otorhinolaryngology. J Clin Med 2021; 10:2940. [PMID: 34209041 PMCID: PMC8267672 DOI: 10.3390/jcm10132940] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022] Open
Abstract
Otorhinolaryngology enrolls head and neck surgery in various tissues such as ear, nose, and throat (ENT) that govern different activities such as hearing, breathing, smelling, production of vocal sounds, the balance, deglutition, facial animation, air filtration and humidification, and articulation during speech, while absence of these functions can lead to high morbidity and even mortality. Conventional therapies for head and neck damaged tissues include grafts, transplants, and artificial materials, but grafts have limited availability and cause morbidity in the donor site. To improve these limitations, regenerative medicine, as a novel and rapidly growing field, has opened a new therapeutic window in otorhinolaryngology by using cell transplantation to target the healing and replacement of injured tissues. There is a high risk of rejection and tumor formation for transplantation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs); mesenchymal stem cells (MSCs) lack these drawbacks. They have easy expansion and antiapoptotic properties with a wide range of healing and aesthetic functions that make them a novel candidate in otorhinolaryngology for craniofacial defects and diseases and hold immense promise for bone tissue healing; even the tissue sources and types of MSCs, the method of cell introduction and their preparation quality can influence the final outcome in the injured tissue. In this review, we demonstrated the anti-inflammatory and immunomodulatory properties of MSCs, from different sources, to be safely used for cell-based therapies in otorhinolaryngology, while their achievements and challenges have been described too.
Collapse
Affiliation(s)
- Reza Kaboodkhani
- Otorhinolaryngology Research Center, Department of Otorhinolaryngology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71936-36981, Iran;
| | - Davood Mehrabani
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz 71987-74731, Iran
- Comparative and Experimental Medicine Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Li Ka Shing Center for Health Research and Innovation, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Feridoun Karimi-Busheri
- Department of Oncology, Faculty of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
20
|
Li C, Mills Z, Zheng Z. Novel cell sources for bone regeneration. MedComm (Beijing) 2021; 2:145-174. [PMID: 34766140 PMCID: PMC8491221 DOI: 10.1002/mco2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
A plethora of both acute and chronic conditions, including traumatic, degenerative, malignant, or congenital disorders, commonly induce bone disorders often associated with severe persisting pain and limited mobility. Over 1 million surgical procedures involving bone excision, bone grafting, and fracture repair are performed each year in the U.S. alone, resulting in immense levels of public health challenges and corresponding financial burdens. Unfortunately, the innate self-healing capacity of bone is often inadequate for larger defects over a critical size. Moreover, as direct transplantation of committed osteoblasts is hindered by deficient cell availability, limited cell spreading, and poor survivability, an urgent need for novel cell sources for bone regeneration is concurrent. Thanks to the development in stem cell biology and cell reprogramming technology, many multipotent and pluripotent cells that manifest promising osteogenic potential are considered the regenerative remedy for bone defects. Considering these cells' investigation is still in its relative infancy, each of them offers their own particular challenges that must be conquered before the large-scale clinical application.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zane Mills
- College of DentistryUniversity of OklahomaOklahoma CityOklahomaUSA
| | - Zhong Zheng
- Division of Growth and Development, School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
21
|
Jung SH, You JE, Choi SW, Kang KS, Cho JY, Lyu J, Kim PH. Polycystin-1 Enhances Stemmness Potential of Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22094868. [PMID: 34064452 PMCID: PMC8125233 DOI: 10.3390/ijms22094868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 01/01/2023] Open
Abstract
Polycystic Kidney Disease (PKD) is a disorder that affects the kidneys and other organs, and its major forms are encoded by polycystin-1 (PC1) and polycystin-2 (PC2), as PKD1 and PKD2. It is located sandwiched inside and outside cell membranes and interacts with other cells. This protein is most active in kidney cells before birth, and PC1 and PC2 work together to help regulate cell proliferation, cell migration, and interactions with other cells. The molecular relationship and the function between PKD1 and cancer is well known, such as increased or decreased cell proliferation and promoting or suppressing cell migration depending on the cancer cell type specifically. However, its function in stem cells has not been revealed. Therefore, in this study, we investigated the biological function of PC1 and umbilical cord blood-derived mesenchymal stem cell (UCB-MSC). Furthermore, we assessed how it affects cell migration, proliferation, and the viability of cells when expressed in the PKD1 gene. In addition, we confirmed in an ex vivo artificial tooth model generated by the three-dimension printing technique that the ability to differentiate into osteocytes improved according to the expression level of the stemness markers when PKD1 was expressed. This study is the first report to examine the biological function of PKD1 in UCB-MSC. This gene may be capable of enhancing differentiation ability and maintaining long-term stemness for the therapeutic use of stem cells.
Collapse
Affiliation(s)
- Se-Hwa Jung
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
| | - Ji-Eun You
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
| | - Soon-Won Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.-W.C.); (K.-S.K.)
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.-W.C.); (K.-S.K.)
| | - Je-Yeol Cho
- Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea;
| | - Jungmook Lyu
- Myung-Gok Eye Research Institute, Department of Medical Science, Konyang University, Daejeon 320-832, Korea;
| | - Pyung-Hwan Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
- Correspondence: ; Tel.: +82-42-600-8436; Fax: +82-42-600-8408
| |
Collapse
|
22
|
Oliveira CS, Carreira M, Correia CR, Mano JF. The Therapeutic Potential of Hematopoietic Stem Cells in Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:379-392. [PMID: 33683146 DOI: 10.1089/ten.teb.2021.0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The repair process of bone fractures is a complex biological mechanism requiring the recruitment and in situ functionality of stem/stromal cells from the bone marrow (BM). BM mesenchymal stem/stromal cells have been widely explored in multiple bone tissue engineering applications, whereas the use of hematopoietic stem cells (HSCs) has been poorly investigated in this context. A reasonable explanation is the fact that the role of HSCs and their combined effect with other elements of the hematopoietic niches in the bone-healing process is still elusive. Therefore, in this review we intend to highlight the influence of HSCs in the bone repair process, mainly through the promotion of osteogenesis and angiogenesis at the bone injury site. For that, we briefly describe the main biological characteristics of HSCs, as well as their hematopoietic niches, while reviewing the biomimetic engineered BM niche models. Moreover, we also highlighted the role of HSCs in translational in vivo transplantation or implantation as promoters of bone tissue repair.
Collapse
Affiliation(s)
- Cláudia S Oliveira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Mariana Carreira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Clara R Correia
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
23
|
Song B, Fu H, Liu J, Ren K, Weir MD, Schneider A, Wang P, Song Y, Zhao L, Xu H. Bioactive small molecules in calcium phosphate scaffold enhanced osteogenic differentiation of human induced pluripotent stem cells. Dent Mater J 2021; 40:615-624. [PMID: 33814531 DOI: 10.4012/dmj.2019-263] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) are exciting for regenerative medicine due to their multi-potent differentiation. SB431542 bioactive molecule can activate bone morphogenetic protein-signalling in osteoblasts. The objectives were to: (1) develop a novel injectable calcium phosphate cement (CPC)-SB431542 scaffold for dental/craniofacial bone engineering; and (2) investigate cell proliferation and osteo-differentiation of hiPSC-derived mesenchymal stem cells (hiPSC-MSCs) on CPC-SB431542 scaffold. Three groups were tested: CPC control; CPC with SB431542 inside CPC (CPCSM); CPC with SB431542 in osteogenic medium (CPC+SMM). SB431542 in CPC promoted stem cell proliferation and viability. hiPSC-MSCs differentiated into osteogenic lineage and synthesized bone minerals. CPC with SB431542 showed much greater osteo-expressions and more bone minerals than those without SB431542. In conclusion, hiPSC-MSCs on CPC scaffold containing SB431542 showed excellent osteo-differentiation and bone mineral synthesis for the first time. CPC was a suitable scaffold for delivering stem cells and SB431542 to promote bone regeneration in dental/craniofacial applications.
Collapse
Affiliation(s)
- Bing Song
- Department of Orthopedic Surgery, Shunde Hospital of Southern Medical University.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Haijun Fu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology
| | - Jianwei Liu
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Stomatology
| | - Ke Ren
- Department of Neural and Pain Sciences, University of Maryland School of Dentistry
| | - Michael D Weir
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry
| | - Ping Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Yang Song
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry.,Department of Prosthodontics, Guanghua School of Stomatology, Sun Yat-sen University
| | - Liang Zhao
- Department of Orthopedic Surgery, Shunde Hospital of Southern Medical University.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry
| | - Huakun Xu
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine.,University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine
| |
Collapse
|
24
|
Tynecka M, Moniuszko M, Eljaszewicz A. Old Friends with Unexploited Perspectives: Current Advances in Mesenchymal Stem Cell-Based Therapies in Asthma. Stem Cell Rev Rep 2021; 17:1323-1342. [PMID: 33649900 PMCID: PMC7919631 DOI: 10.1007/s12015-021-10137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have a great regenerative and immunomodulatory potential that was successfully tested in numerous pre-clinical and clinical studies of various degenerative, hematological and inflammatory disorders. Over the last few decades, substantial immunoregulatory effects of MSC treatment were widely observed in different experimental models of asthma. Therefore, it is tempting to speculate that stem cell-based treatment could become an attractive means to better suppress asthmatic airway inflammation, especially in subjects resistant to currently available anti-inflammatory therapies. In this review, we discuss mechanisms accounting for potent immunosuppressive properties of MSCs and the rationale for their use in asthma. We describe in detail an intriguing interplay between MSCs and other crucial players in the immune system as well as lung microenvironment. Finally, we reveal the potential of MSCs in maintaining airway epithelial integrity and alleviating lung remodeling.
Collapse
Affiliation(s)
- Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
- Department of Allergology and Internal Medicine, Medical University of Bialystok, ul. M. Skłodowskiej-Curie 24A, Białystok, 15-276, Poland.
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, ul. Waszyngtona 13, 15-269, Białystok, Poland.
| |
Collapse
|
25
|
Sfougataki I, Varela I, Stefanaki K, Karagiannidou A, Roubelakis MG, Kalodimou V, Papathanasiou I, Traeger-Synodinos J, Kitsiou-Tzeli S, Kanavakis E, Kitra V, Tsezou A, Tzetis M, Goussetis E. Proliferative and chondrogenic potential of mesenchymal stromal cells from pluripotent and bone marrow cells. Histol Histopathol 2020; 35:1415-1426. [PMID: 32959885 DOI: 10.14670/hh-18-259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) can be derived from a wide range of fetal and adult sources including pluripotent stem cells (PSCs). The properties of PSC-derived MSCs need to be fully characterized, in order to evaluate the feasibility of their use in clinical applications. PSC-MSC proliferation and differentiation potential in comparison with bone marrow (BM)-MSCs is still under investigation. The objective of this study was to determine the proliferative and chondrogenic capabilities of both human induced pluripotent stem cell (hiPSC-) and embryonic stem cell (hESC-) derived MSCs, by comparing them with BM-MSCs. METHODS MSCs were derived from two hiPSC lines (hiPSC-MSCs), the well characterized Hues9 hESC line (hESC-MSCs) and BM from two healthy donors (BM-MSCs). Proliferation potential was investigated using appropriate culture conditions, with serial passaging, until cells entered into senescence. Differentiation potential to cartilage was examined after in vitro chondrogenic culture conditions. RESULTS BM-MSCs revealed a fold expansion of 1.18x10⁵ and 2.3x10⁵ while the two hiPSC-MSC lines and hESC-MSC showed 5.88x10¹⁰, 3.49x10⁸ and 2.88x10⁸, respectively. Under chondrogenic conditions, all MSC lines showed a degree of chondrogenesis. However, when we examined the formed chondrocyte micromasses by histological analysis of the cartilage morphology and immunohistochemistry for the chondrocyte specific markers Sox9 and Collagen II, we observed that PSC-derived MSC lines had formed pink rather than hyaline cartilage, in contrast to BM-MSCs. CONCLUSION In conclusion, MSCs derived from both hESCs and hiPSCs had superior proliferative capacity compared to BM-MSCs, but they were inefficient in their ability to form hyaline cartilage.
Collapse
Affiliation(s)
- Irene Sfougataki
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece.,Research Institute for the Study of Genetic and Malignant Disorders in Childhood, Aghia Sophia Children's Hospital, Athens, Greece.
| | - Ioanna Varela
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| | - Kalliope Stefanaki
- Department of Histopathology, Aghia Sophia Children's Hospital, Athens, Greece
| | | | - Maria G Roubelakis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Kalodimou
- Flow Cytometry-Research and Regenerative Medicine Department, IASO Hospital, Athens, Greece
| | - Ioanna Papathanasiou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessally, Thessally, Greece
| | - Joanne Traeger-Synodinos
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Kitsiou-Tzeli
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanuel Kanavakis
- Genesis Genoma Lab, Genetic diagnosis, Clinical Genetics and Research, Chalandri, Greece
| | - Vasiliki Kitra
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessally, Thessally, Greece
| | - Maria Tzetis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evgenios Goussetis
- Stem Cell Transplant Unit, Aghia Sophia Children's Hospital, Athens, Greece
| |
Collapse
|
26
|
Lopes JH, Souza LP, Domingues JA, Ferreira FV, Alencar Hausen M, Camilli JA, Martin RA, Rezende Duek EA, Mazali IO, Bertran CA. In vitro and in vivo osteogenic potential of niobium‐doped 45S5 bioactive glass: A comparative study. J Biomed Mater Res B Appl Biomater 2020; 108:1372-1387. [DOI: 10.1002/jbm.b.34486] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/22/2019] [Accepted: 08/29/2019] [Indexed: 01/03/2023]
Affiliation(s)
- João H. Lopes
- Department of Chemistry, Division of Fundamental Sciences (IEF)Aeronautics Institute of Technology (ITA) Sao Jose dos Campos Brazil
| | - Lucas P. Souza
- Department of Structural and Functional BiologyInstitute of Biology, University of Campinas – UNICAMP Campinas Brazil
| | - Juliana A. Domingues
- Department of Structural and Functional BiologyInstitute of Biology, University of Campinas – UNICAMP Campinas Brazil
| | - Filipe V. Ferreira
- School of Chemical EngineeringUniversity of Campinas – UNICAMP Campinas Brazil
| | - Moema Alencar Hausen
- Department of Physiological Sciences, Biomaterials LaboratoryPontifical Catholic University of São Paulo Sorocaba Brazil
| | - José A. Camilli
- Department of Structural and Functional BiologyInstitute of Biology, University of Campinas – UNICAMP Campinas Brazil
| | - Richard A. Martin
- School of Engineering & Aston Research Centre for Healthy AgeingAston University Birmingham UK
| | - Eliana A. Rezende Duek
- Department of Physiological Sciences, Biomaterials LaboratoryPontifical Catholic University of São Paulo Sorocaba Brazil
| | - Italo O. Mazali
- Department of Inorganic ChemistryInstitute of Chemistry, University of Campinas – UNICAMP Campinas Brazil
| | - Celso A. Bertran
- Department of Physical ChemistryInstitute of Chemistry, University of Campinas – UNICAMP Campinas Brazil
| |
Collapse
|
27
|
Induced Pluripotent Stem Cells in Dental and Nondental Tissue Regeneration: A Review of an Unexploited Potential. Stem Cells Int 2020; 2020:1941629. [PMID: 32300365 PMCID: PMC7146092 DOI: 10.1155/2020/1941629] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/06/2020] [Indexed: 12/16/2022] Open
Abstract
Cell-based therapies currently represent the state of art for tissue regenerative treatment approaches for various diseases and disorders. Induced pluripotent stem cells (iPSCs), reprogrammed from adult somatic cells, using vectors carrying definite transcription factors, have manifested a breakthrough in regenerative medicine, relying on their pluripotent nature and ease of generation in large amounts from various dental and nondental tissues. In addition to their potential applications in regenerative medicine and dentistry, iPSCs can also be used in disease modeling and drug testing for personalized medicine. The current review discusses various techniques for the production of iPSC-derived osteogenic and odontogenic progenitors, the therapeutic applications of iPSCs, and their regenerative potential in vivo and in vitro. Through the present review, we aim to explore the potential applications of iPSCs in dental and nondental tissue regeneration and to highlight different protocols used for the generation of different tissues and cell lines from iPSCs.
Collapse
|
28
|
Guo NN, Liu LP, Zheng YW, Li YM. Inducing human induced pluripotent stem cell differentiation through embryoid bodies: A practical and stable approach. World J Stem Cells 2020; 12:25-34. [PMID: 32110273 PMCID: PMC7031760 DOI: 10.4252/wjsc.v12.i1.25] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 09/30/2019] [Accepted: 12/15/2019] [Indexed: 02/06/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are invaluable resources for producing high-quality differentiated cells in unlimited quantities for both basic research and clinical use. They are particularly useful for studying human disease mechanisms in vitro by making it possible to circumvent the ethical issues of human embryonic stem cell research. However, significant limitations exist when using conventional flat culturing methods especially concerning cell expansion, differentiation efficiency, stability maintenance and multicellular 3D structure establishment, differentiation prediction. Embryoid bodies (EBs), the multicellular aggregates spontaneously generated from iPSCs in the suspension system, might help to address these issues. Due to the unique microenvironment and cell communication in EB structure that a 2D culture system cannot achieve, EBs have been widely applied in hiPSC-derived differentiation and show significant advantages especially in scaling up culturing, differentiation efficiency enhancement, ex vivo simulation, and organoid establishment. EBs can potentially also be used in early prediction of iPSC differentiation capability. To improve the stability and feasibility of EB-mediated differentiation and generate high quality EBs, critical factors including iPSC pluripotency maintenance, generation of uniform morphology using micro-pattern 3D culture systems, proper cellular density inoculation, and EB size control are discussed on the basis of both published data and our own laboratory experiences. Collectively, the production of a large quantity of homogeneous EBs with high quality is important for the stability and feasibility of many PSCs related studies.
Collapse
Affiliation(s)
- Ning-Ning Guo
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Li-Ping Liu
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, University of Tsukuba Faculty of Medicine, Tsukuba, Ibaraki 305-8575, Japan
- Yokohama City University School of Medicine, Yokohama, Kanagawa 234-0006, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, the University of Tokyo, Tokyo 108-8639, Japan
| | - Yu-Mei Li
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu Province, China
| |
Collapse
|
29
|
Abstract
The generation of tissue engineered organs from autologous cells will allow replacement of diseased or absent organs without the need for immunosuppression. Common steps of tissue engineering include isolation of pluripotent or multipotent stem cells, preparation of synthetic or biologic scaffold, and implantation into a host to support the proliferation of engineered tissue. Some organs have been successfully transplanted in human patients; gastrointestinal tract tissues are nearing clinical introduction. The state of the science has progressed rapidly and providers and researchers alike must take appropriate steps to ensure strict adherence to ethical standards before introduction to human therapy.
Collapse
Affiliation(s)
- Daniel Levin
- Division of Pediatric Surgery, Department of Surgery, University of Virginia, 1300 Jefferson Park Avenue, PO BOX 800709, Charlottesville, VA 22908-0709, USA.
| |
Collapse
|
30
|
Wang AYL, Loh CYY. Episomal Induced Pluripotent Stem Cells: Functional and Potential Therapeutic Applications. Cell Transplant 2019; 28:112S-131S. [PMID: 31722555 PMCID: PMC7016470 DOI: 10.1177/0963689719886534] [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] [Indexed: 12/19/2022] Open
Abstract
The term episomal induced pluripotent stem cells (EiPSCs) refers to somatic cells that are reprogrammed into induced pluripotent stem cells (iPSCs) using non-integrative episomal vector methods. This reprogramming process has a better safety profile compared with integrative methods using viruses. There is a current trend toward using episomal plasmid reprogramming to generate iPSCs because of the improved safety profile. Clinical reports of potential human cell sources that have been successfully reprogrammed into EiPSCs are increasing, but no review or summary has been published. The functional applications of EiPSCs and their potential uses in various conditions have been described, and these may be applicable to clinical scenarios. This review summarizes the current direction of EiPSC research and the properties of these cells with the aim of explaining their potential role in clinical applications and functional restoration.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,*Both the authors contributed equally to this article
| | - Charles Yuen Yung Loh
- St Andrew's Center for Burns and Plastic Surgery, Chelmsford, United Kingdom.,*Both the authors contributed equally to this article
| |
Collapse
|
31
|
Rana D, Kumar S, Webster TJ, Ramalingam M. Impact of Induced Pluripotent Stem Cells in Bone Repair and Regeneration. Curr Osteoporos Rep 2019; 17:226-234. [PMID: 31256323 DOI: 10.1007/s11914-019-00519-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The main objective of this article is to investigate the current trends in the use of induced pluripotent stem cells (iPSCs) for bone tissue repair and regeneration. RECENT FINDINGS Pluripotent stem cell-based tissue engineering has extended innovative therapeutic approaches for regenerative medicine. iPSCs have shown osteogenic differentiation capabilities and would be an innovative resource of stem cells for bone tissue regenerative applications. This review recapitulates the current knowledge and recent progress regarding utilization of iPSCs for bone therapy. A review of current findings suggests that a combination of a three-dimensional scaffolding system with iPSC technology to mimic the physiological complexity of the native stem cell niche is highly favorable for bone tissue repair and regeneration.
Collapse
Affiliation(s)
- Deepti Rana
- Department of Biomechanical Engineering, Technical Medical Centre, University of Twente, 7522 NB, Enschede, The Netherlands
| | - Sanjay Kumar
- Centre for Stem Cell Research, Christian Medical College Campus, Vellore, 632002, India
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA.
| | - Murugan Ramalingam
- Biomaterials and Stem Cell Engineering Lab, Centre for Biomaterials, Cellular and Molecular Theranostics, School of Mechanical Engineering, Vellore Institute of Technology (Deemed to be University), Vellore, 632014, India.
| |
Collapse
|
32
|
Abdal Dayem A, Lee SB, Kim K, Lim KM, Jeon TI, Seok J, Cho ASG. Production of Mesenchymal Stem Cells Through Stem Cell Reprogramming. Int J Mol Sci 2019; 20:ijms20081922. [PMID: 31003536 PMCID: PMC6514654 DOI: 10.3390/ijms20081922] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/10/2019] [Accepted: 04/15/2019] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess a broad spectrum of therapeutic applications and have been used in clinical trials. MSCs are mainly retrieved from adult or fetal tissues. However, there are many obstacles with the use of tissue-derived MSCs, such as shortages of tissue sources, difficult and invasive retrieval methods, cell population heterogeneity, low purity, cell senescence, and loss of pluripotency and proliferative capacities over continuous passages. Therefore, other methods to obtain high-quality MSCs need to be developed to overcome the limitations of tissue-derived MSCs. Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are considered potent sources for the derivation of MSCs. PSC-derived MSCs (PSC-MSCs) may surpass tissue-derived MSCs in proliferation capacity, immunomodulatory activity, and in vivo therapeutic applications. In this review, we will discuss basic as well as recent protocols for the production of PSC-MSCs and their in vitro and in vivo therapeutic efficacies. A better understanding of the current advances in the production of PSC-MSCs will inspire scientists to devise more efficient differentiation methods that will be a breakthrough in the clinical application of PSC-MSCs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology, Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, Gwangjin-gu, Seoul 05029, Korea.
| | | | | | | | | | | | | |
Collapse
|
33
|
Xu M, Shaw G, Murphy M, Barry F. Induced Pluripotent Stem Cell-Derived Mesenchymal Stromal Cells Are Functionally and Genetically Different From Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells 2019; 37:754-765. [PMID: 30779868 PMCID: PMC6591688 DOI: 10.1002/stem.2993] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/21/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022]
Abstract
There has been considerable interest in the generation of functional mesenchymal stromal cell (MSC) preparations from induced pluripotent stem cells (iPSCs) and this is now regarded as a potential source of unlimited, standardized, high‐quality cells for therapeutic applications in regenerative medicine. Although iMSCs meet minimal criteria for defining MSCs in terms of marker expression, there are substantial differences in terms of trilineage potential, specifically a marked reduction in chondrogenic and adipogenic propensity in iMSCs compared with bone marrow‐derived (BM) MSCs. To reveal the cellular basis underlying these differences, we conducted phenotypic, functional, and genetic comparisons between iMSCs and BM‐MSCs. We found that iMSCs express very high levels of both KDR and MSX2 compared with BM‐MSCs. In addition, BM‐MSCs had significantly higher levels of PDGFRα. These distinct gene expression profiles were maintained during culture expansion, suggesting that prepared iMSCs are more closely related to vascular progenitor cells (VPCs). Although VPCs can differentiate along the chondrogenic, osteogenic, and adipogenic pathways, they require different inductive conditions compared with BM‐MSCs. These observations suggest to us that iMSCs, based on current widely used preparation protocols, do not represent a true alternative to primary MSCs isolated from BM. Furthermore, this study highlights the fact that high levels of expression of typical MSC markers such as CD73, CD90, and CD105 are insufficient to distinguish MSCs from other mesodermal progenitors in differentiated induced pluripotent stem cell cultures. stem cells2019;37:754–765
Collapse
Affiliation(s)
- Maojia Xu
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Georgina Shaw
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Mary Murphy
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| | - Frank Barry
- The Regenerative Medicine Institute, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
34
|
Agrawal M, Alexander A, Khan J, Giri TK, Siddique S, Dubey SK, Ajazuddin, Patel RJ, Gupta U, Saraf S, Saraf S. Recent Biomedical Applications on Stem Cell Therapy: A Brief Overview. Curr Stem Cell Res Ther 2019; 14:127-136. [DOI: 10.2174/1574888x13666181002161700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/29/2018] [Accepted: 09/02/2018] [Indexed: 12/16/2022]
Abstract
Stem cells are the specialized cell population with unique self-renewal ability and act as the
precursor of all the body cells. Broadly, stem cells are of two types one is embryonic stem cells while
the other is adult or somatic stem cells. Embryonic stem cells are the cells of zygote of the blastocyst
which give rise to all kind of body cells including embryonic cells, and it can reconstruct a complete
organism. While the adult stem cells have limited differentiation ability in comparison with embryonic
stem cells and it proliferates into some specific kind of cells. This unique ability of the stem cell makes
it a compelling biomedical and therapeutic tool. Stem cells primarily serve as regenerative medicine for
particular tissue regeneration or the whole organ regeneration in any physical injury or disease condition
(like diabetes, cancer, periodontal disorder, etc.), tissue grafting and plastic surgery, etc. Along
with this, it is also used in various preclinical and clinical investigations, biomedical engineering and as
a potential diagnostic tool (such as the development of biomarkers) for non-invasive diagnosis of severe
disorders. In this review article, we have summarized the application of stem cell as regenerative
medicine and in the treatment of various chronic diseases.
Collapse
Affiliation(s)
- Mukta Agrawal
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490 024, India
| | - Amit Alexander
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490 024, India
| | - Junaid Khan
- University Teaching Department (Pharmacy), Sarguja University, Ambikapur, Chhattisgarh 497001, India
| | - Tapan K. Giri
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490 024, India
| | - Sabahuddin Siddique
- Patel College of Pharmacy, Madhyanchal Professional University, Bhopal, Madhya Pradesh, India
| | - Sunil K. Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490 024, India
| | - Ravish J. Patel
- Ramanbhai Patel College of Pharmacy (RPCP), Charotar University of Science and Technology (CHARUSAT), Gujarat 388 421, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer - 305817, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492 010, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492 010, India
| |
Collapse
|
35
|
Kamaldinov T, Erndt-Marino J, Diaz-Rodriguez P, Chen H, Gharat T, Munoz-Pinto D, Arduini B, Hahn MS. Tuning Forkhead Box D3 overexpression to promote specific osteogenic differentiation of human embryonic stem cells while reducing pluripotency in a three-dimensional culture system. J Tissue Eng Regen Med 2018; 12:2256-2265. [PMID: 30350469 DOI: 10.1002/term.2757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/15/2018] [Accepted: 10/18/2018] [Indexed: 12/15/2022]
Abstract
Clinical use of human embryonic stem cells (hESCs) in bone regeneration applications requires that their osteogenic differentiation be highly controllable as well as time- and cost-effective. The main goals of the current work were thus (a) to assess whether overexpression of pluripotency regulator Forkhead Box D3 (FOXD3) can enhance the osteogenic commitment of hESCs seeded in three-dimensional (3D) scaffolds and (b) to evaluate if the degree of FOXD3 overexpression regulates the strength and specificity of hESC osteogenic commitment. In conducting these studies, an interpenetrating hydrogel network consisting of poly(ethylene glycol) diacrylate and collagen I was utilized as a 3D culture platform. Expression of osteogenic, chondrogenic, pluripotency, and germ layer markers by encapsulated hESCs was measured after 2 weeks of culture in osteogenic medium in the presence or absence doxycycline-induced FOXD3 transgene expression. Towards the first goal, FOXD3 overexpression initiated 24 hr prior to hESC encapsulation, relative to unstimulated controls, resulted in upregulation of osteogenic markers and enhanced calcium deposition, without promoting off-target effects. However, when initiation of FOXD3 overexpression was increased from 24 to 48 hr prior to encapsulation, hESC osteogenic commitment was not further enhanced and off-target effects were noted. Specifically, relative to 24-hr prestimulation, initiation of FOXD3 overexpression 48 hr prior to encapsulation yielded increased expression of pluripotency markers while reducing mesodermal but increasing endodermal germ layer marker expression. Combined, the current results indicate that the controlled overexpression of FOXD3 warrants further investigation as a mechanism to guide enhanced hESC osteogenic commitment.
Collapse
Affiliation(s)
- Timothy Kamaldinov
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Josh Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | | | - Hongyu Chen
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Tanmay Gharat
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Dany Munoz-Pinto
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Brigitte Arduini
- Rensselaer Center for Stem Cell Research, Rensselaer Polytechnic Institute, Troy, New York
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
36
|
Sparks NRL, Martinez IKC, Soto CH, Zur Nieden NI. Low Osteogenic Yield in Human Pluripotent Stem Cells Associates with Differential Neural Crest Promoter Methylation. Stem Cells 2018; 36:349-362. [PMID: 29193426 DOI: 10.1002/stem.2746] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/20/2017] [Accepted: 10/23/2017] [Indexed: 01/06/2023]
Abstract
Human pluripotent stem cell-derived osteoblasts possess great potential for use in bone disorder elucidation and repair; however, while the general ability of human pluripotent stem cells to differentiate into osteoblasts and lay down bone-specific matrix has been shown, previous studies lack the complete characterization of the process whereby such osteoblasts are derived as well as a comparison between the osteogenic efficiency of multiple cell lines. Here, we compared the osteogenic potential of two human induced pluripotent stem cell lines (RIV9 and RIV4) to human H9 embryonic stem cells. Generally capable of osteogenic differentiation, the overall osteogenic yield was lower in the RIV9 and RIV4 lines and correlated with differential expression of osteocalcin (OCN) in mature cultures and PAX7 and TWIST1 during early differentiation. In the undifferentiated cells, the promoters of the latter two genes were differentially methylated potentially explaining the variation in differentiation efficiency. Furthermore, the expression signatures of selected neural crest and mesodermal genes and proteins suggested that H9 cells preferentially gave rise to neural crest-derived osteoblasts, whereas the osteoblasts in the RIV9 cultures were generated both through a mesodermal and a neural crest route although each at a lower rate. These data suggest that epigenetic dissimilarities between multiple PSC lines may lead to differences in lineage derivation and mineralization. Since osteoblast progenitors from one origin inadequately repair a defect in the other, these data underscore the importance of screening human pluripotent stem cells lines for the identity of the osteoprogenitors they lay down. Stem Cells 2018;36:349-362.
Collapse
Affiliation(s)
- Nicole Renee Lee Sparks
- Department of Molecular, Cell and Systems Biology and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, California, 92521, USA
| | - Ivann Kenneth Carvajal Martinez
- Department of Molecular, Cell and Systems Biology and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, California, 92521, USA
| | - Cristina Helen Soto
- Department of Molecular, Cell and Systems Biology and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, California, 92521, USA
| | - Nicole Isolde Zur Nieden
- Department of Molecular, Cell and Systems Biology and Stem Cell Center, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, California, 92521, USA
| |
Collapse
|
37
|
Jin YZ, Lee JH. Mesenchymal Stem Cell Therapy for Bone Regeneration. Clin Orthop Surg 2018; 10:271-278. [PMID: 30174801 PMCID: PMC6107811 DOI: 10.4055/cios.2018.10.3.271] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/27/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinic for approximately 20 years. During this period, various new populations of MSCs have been found or manipulated. However, their characters and relative strength for bone regeneration have not been well known. For a comprehensive understanding of MSCs, we reviewed the literature on the multipotent cells ranging from the definition to the current research progress for bone regeneration. Based on our literature review, bone marrow MSCs have been most widely studied and utilized in clinical settings. Among other populations of MSCs, adipose-derived MSCs and perivascular MSCs might be potential candidates for bone regeneration, whose efficacy and safety still require further investigation.
Collapse
Affiliation(s)
- Yuan-Zhe Jin
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul, Korea.,Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
38
|
Zhu S, Ehnert S, Rouß M, Häussling V, Aspera-Werz RH, Chen T, Nussler AK. From the Clinical Problem to the Basic Research-Co-Culture Models of Osteoblasts and Osteoclasts. Int J Mol Sci 2018; 19:ijms19082284. [PMID: 30081523 PMCID: PMC6121694 DOI: 10.3390/ijms19082284] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Bone tissue undergoes constant remodeling and healing when fracture happens, in order to ensure its structural integrity. In order to better understand open biological and clinical questions linked to various bone diseases, bone cell co-culture technology is believed to shed some light into the dark. Osteoblasts/osteocytes and osteoclasts dominate the metabolism of bone by a multitude of connections. Therefore, it is widely accepted that a constant improvement of co-culture models with both cell types cultured on a 3D scaffold, is aimed to mimic an in vivo environment as closely as possible. Although in recent years a considerable knowledge of bone co-culture models has been accumulated, there are still many open questions. We here try to summarize the actual knowledge and address open questions.
Collapse
Affiliation(s)
- Sheng Zhu
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Marc Rouß
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Victor Häussling
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Romina H Aspera-Werz
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Tao Chen
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Andreas K Nussler
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Institute for Trauma Research, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| |
Collapse
|
39
|
Wu Q, Yang B, Cao C, Hu K, Wang P, Man Y. Therapeutic antibody directed osteogenic differentiation of induced pluripotent stem cell derived MSCs. Acta Biomater 2018; 74:222-235. [PMID: 29778895 DOI: 10.1016/j.actbio.2018.05.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/31/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are regarded as a new cell source for regenerative medicine. Recent advances in tissue engineering have brought to light the therapeutic application of induced pluripotent stem cells (iPSCs) in bone defect repair. However, a safe and efficient way to differentiate iPSCs into osteogenic lineage remains to be a major challenge. Here we describe an approach using anti-BMP2 antibodies (Abs) to mediate osteogenic differentiation of iPSC-derived mesenchymal stromal cells (iMSCs). We first proved that 3G7 (an anti-BMP2 Ab) not only bound to BMP2, but also allowed the bound BMP2 to engage the BMP2 receptors on iMSCs. Subcutaneous implantation sites loaded with iMSCs + 3G7 group showed significant bone formation and vascularization in mice while those sites with exogenous BMP2 exhibited dystrophic calcification and significantly lower vascularization. Our in vitro study demonstrated that the anti-BMP2 Ab/BMP2 immune complex were capable of dictating the acquisition of osteogenic phenotype of iMSCs and subsequent mineralization. The study provided the first evidence of antibody-mediated differentiation of iMSCs and osseous regeneration in vivo. This novel strategy takes full advantage of the endogenous bioactive molecules for osseous regeneration and its potential therapeutic application is promising. STATEMENT OF SIGNIFICANCE Induced pluripotent stem cells (iPSCs) and its derived cells hold significant promise for the treatment of bone defects. In present study, we carried out the concept of antibody-mediated bone regeneration into the iPSC research for the first time. We demonstrated that anti-BMP2 Ab/BMP2 immune complex was capable of promoting osteogenic differentiation of iPSC-derived MSCs (iMSCs), likely through the classical BMP2/Smad1/Runx2 pathway. Subcutaneous co-delivery of iMSCs and anti-BMP2 Abs resulted in significant bone formation and vascularization. These findings suggested antibody mediated osteogenic differentiation may be a favorable approach for iPSC-based bone tissue engineering.
Collapse
|
40
|
Fliefel R, Ehrenfeld M, Otto S. Induced pluripotent stem cells (iPSCs) as a new source of bone in reconstructive surgery: A systematic review and meta-analysis of preclinical studies. J Tissue Eng Regen Med 2018; 12:1780-1797. [DOI: 10.1002/term.2697] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 04/16/2018] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Riham Fliefel
- Experimental Surgery and Regenerative Medicine (ExperiMed), Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry; Alexandria University; Alexandria Egypt
| | - Michael Ehrenfeld
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine; Ludwig Maximilian University of Munich; Munich Germany
| |
Collapse
|
41
|
Nassiri Asl M, Aali E. Review on the mesenchymal stem cells and their potential application in regenerative medicine. THE JOURNAL OF QAZVIN UNIVERSITY OF MEDICAL SCIENCES 2018. [DOI: 10.29252/qums.21.6.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
42
|
Zhang C, Hu K, Liu X, Reynolds MA, Bao C, Wang P, Zhao L, Xu HH. Novel hiPSC-based tri-culture for pre-vascularization of calcium phosphate scaffold to enhance bone and vessel formation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Wang P, Ma T, Guo D, Hu K, Shu Y, Xu HHK, Schneider A. Metformin induces osteoblastic differentiation of human induced pluripotent stem cell-derived mesenchymal stem cells. J Tissue Eng Regen Med 2017; 12:437-446. [PMID: 28494141 DOI: 10.1002/term.2470] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/12/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022]
Abstract
Metformin, a first-line antidiabetic drug used by millions of patients, has been shown to have potential osteogenic properties. The present study was performed to test the hypothesis that clinically relevant doses of metformin promote the osteogenic differentiation and mineralization of induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs). iPSC-MSCs were treated with metformin (10 μm) to assess cell viability, osteogenic differentiation, mineralization and activation of the LKB1/AMP-activated protein kinase (AMPK) signalling pathway, a surrogate marker of metformin action. To determine its potential application in MSC-based bone and periodontal tissue engineering, iPSC-MSCs were also treated with metformin when seeded on to calcium phosphate cement (CPC) scaffolds. Immunoblotting and cellular uptake assays showed that iPSC-MSCs express functional organic cation transporter-1 (OCT-1), a transmembrane protein that mediates the intracellular uptake of metformin. Although metformin treatment did not impair iPSC-MSC viability, it significantly stimulated alkaline phosphatase activity, enhanced mineralized nodule formation and increased expression of osteogenic markers, including Runt-related transcription factor 2 (RUNX2) and osterix. Inhibition of LKB1 activity, a common upstream AMPK kinase, markedly reversed metformin-induced AMPK activation, RUNX2 expression and nuclear localization. Moreover, metformin substantially increased mineralized nodule formation of iPSC-MSC seeded on CPC scaffolds. Collectively, functional OCT-expressing iPSC-MSCs responded to metformin by inducing an osteogenic effect in part mediated by the LKB1/AMPK pathway. Considering the widespread use of metformin in diabetics, this work may lead to novel tissue-engineering platforms where autogenous OCT-expressing iPSC-MSCs might be used to enhance bone and periodontal regeneration in diabetic patients prescribed with daily doses of metformin.
Collapse
Affiliation(s)
- Ping Wang
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Dong Guo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Kevin Hu
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Hockin H K Xu
- Department of Endodontics, Periodontics and Prosthodontics, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
44
|
Tu C, Xu R, Koleti M, Zoldan J. Glycogen synthase kinase-3 inhibition sensitizes human induced pluripotent stem cells to thiol-containing antioxidants induced apoptosis. Stem Cell Res 2017; 23:182-187. [DOI: 10.1016/j.scr.2017.07.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/20/2017] [Accepted: 07/19/2017] [Indexed: 01/22/2023] Open
|
45
|
Li Y, Li L, Chen ZN, Gao G, Yao R, Sun W. Engineering-derived approaches for iPSC preparation, expansion, differentiation and applications. Biofabrication 2017; 9:032001. [DOI: 10.1088/1758-5090/aa7e9a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
Caddeo S, Boffito M, Sartori S. Tissue Engineering Approaches in the Design of Healthy and Pathological In Vitro Tissue Models. Front Bioeng Biotechnol 2017; 5:40. [PMID: 28798911 PMCID: PMC5526851 DOI: 10.3389/fbioe.2017.00040] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022] Open
Abstract
In the tissue engineering (TE) paradigm, engineering and life sciences tools are combined to develop bioartificial substitutes for organs and tissues, which can in turn be applied in regenerative medicine, pharmaceutical, diagnostic, and basic research to elucidate fundamental aspects of cell functions in vivo or to identify mechanisms involved in aging processes and disease onset and progression. The complex three-dimensional (3D) microenvironment in which cells are organized in vivo allows the interaction between different cell types and between cells and the extracellular matrix, the composition of which varies as a function of the tissue, the degree of maturation, and health conditions. In this context, 3D in vitro models can more realistically reproduce a tissue or organ than two-dimensional (2D) models. Moreover, they can overcome the limitations of animal models and reduce the need for in vivo tests, according to the "3Rs" guiding principles for a more ethical research. The design of 3D engineered tissue models is currently in its development stage, showing high potential in overcoming the limitations of already available models. However, many issues are still opened, concerning the identification of the optimal scaffold-forming materials, cell source and biofabrication technology, and the best cell culture conditions (biochemical and physical cues) to finely replicate the native tissue and the surrounding environment. In the near future, 3D tissue-engineered models are expected to become useful tools in the preliminary testing and screening of drugs and therapies and in the investigation of the molecular mechanisms underpinning disease onset and progression. In this review, the application of TE principles to the design of in vitro 3D models will be surveyed, with a focus on the strengths and weaknesses of this emerging approach. In addition, a brief overview on the development of in vitro models of healthy and pathological bone, heart, pancreas, and liver will be presented.
Collapse
Affiliation(s)
- Silvia Caddeo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, Amsterdam, Netherlands
| | - Monica Boffito
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Susanna Sartori
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
47
|
Wu Z, Yang T, Bao C, Li M, Xu HHK, Liao X, Li L, Huang R, Pan J, Liu X. Effect of Electrospun Fibrous Scaffolds with Different Fiber Orientations on the Alignment of Microvessel-Like Structures. J Med Biol Eng 2017. [DOI: 10.1007/s40846-017-0284-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Kim YM, Kang YG, Park SH, Han MK, Kim JH, Shin JW, Shin JW. Effects of mechanical stimulation on the reprogramming of somatic cells into human-induced pluripotent stem cells. Stem Cell Res Ther 2017; 8:139. [PMID: 28595633 PMCID: PMC5465448 DOI: 10.1186/s13287-017-0594-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/24/2017] [Accepted: 05/22/2017] [Indexed: 12/26/2022] Open
Abstract
Background Mechanical stimuli play important roles in the proliferation and differentiation of adult stem cells. However, few studies on their effects on induced pluripotent stem cells (iPSCs) have been published. Methods Human dermal fibroblasts were seeded onto flexible membrane-bottom plates, and infected with retrovirus expressing the four reprogramming factors OCT4, SOX2, KLF, and c-MYC (OSKM). The cells were subjected to equiaxial stretching (3% or 8% for 2, 4, or 7 days) and seeded on feeder cells (STO). The reprogramming into iPSCs was evaluated by the expression of pluripotent markers, in vitro differentiation into three germ layers, and teratoma formation. Results Equiaxial stretching enhanced reprogramming efficiency without affecting the viral transduction rate. iPSCs induced by transduction of four reprogramming factors and application of equiaxial stretching had characteristics typical of iPSCs in terms of pluripotency and differentiation potentials. Conclusions This is the first study to show that mechanical stimuli can increase reprogramming efficiency. However, it did not enhance the infection rate, indicating that mechanical stimuli, defined as stretching in this study, have positive effects on reprogramming rather than on infection. Additional studies should evaluate the mechanism underlying the modulation of reprogramming of somatic cells into iPSCs.
Collapse
Affiliation(s)
- Young Mi Kim
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongsangnam-do, Republic of Korea
| | - Yun Gyeong Kang
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongsangnam-do, Republic of Korea
| | - So Hee Park
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongsangnam-do, Republic of Korea
| | - Myung-Kwan Han
- Department of Microbiology, Chonbuk National University Medical School, Jeonju, Jeollabuk-do, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
| | - Ji Won Shin
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongsangnam-do, Republic of Korea
| | - Jung-Woog Shin
- Department of Biomedical Engineering, Inje University, Gimhae, Gyeongsangnam-do, Republic of Korea. .,Department of Health Science and Technology/Cardiovascular and Metabolic Disease Center/Institute of Aged Life Redesign/UHARC, Gimhae, Gyeongsangnam-do, Republic of Korea.
| |
Collapse
|
49
|
He W, Zhu W, Cao Q, Shen Y, Zhou Q, Yu P, Liu X, Ma J, Li Y, Hong K. Generation of Mesenchymal-Like Stem Cells From Urine in Pediatric Patients. Transplant Proc 2017; 48:2181-5. [PMID: 27569968 DOI: 10.1016/j.transproceed.2016.02.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 01/05/2016] [Accepted: 02/24/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Mesenchymal stem cells (MSCs) have been widely used for regenerative medicine. Traditionally, the procedures of MSC isolation are usually invasive and time-consuming. Urine is merely a body waste, and recent studies have suggested that urine represents an alternative source of stem cells. We, therefore, determined whether the possibility of isolating mesenchymal-like stem cells was practical from human urine. METHODS A total of 16 urine samples were collected from pediatric patients. Urine-derived cells were isolated, expanded, and identified for specific cell surface markers using flow cytometry. Cell morphology was observed by microscopy. Osteogenic and adipogenic differentiation potential were determinded by culturing cells in specific induction medium, and assessed by alkaline phosphatase and oil red O stainings, respectively. RESULTS Clones were established and passaged successfully from primary cultures of urine cells. Cultured urine-derived cells at passage 3 were fusiform and arranged with certain directionality. Urine-derived cells at passage 5 displayed expressions of cell surface markers (CD29, CD105, CD166, CD90, and CD13). There was no expression of the general hematopoietic cell markers (CD45, CD34, and HLA-DR). Under in vitro induction conditions, urine-derived cells at passage 5 were able to differentiate into osteoblasts, but not adipocytes. CONCLUSIONS Urine may be a noninvasive source for mesenchymal-like stem cells. These cells could potentially provide a new source of autologous stem cells for regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- W He
- Jiangxi Key Laboratory of Molecular Medicine, Jiangxi, China; Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - W Zhu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Q Cao
- Jiangxi Key Laboratory of Molecular Medicine, Jiangxi, China; Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Y Shen
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Q Zhou
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - P Yu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - X Liu
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - J Ma
- Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Y Li
- Department of Cardiovascular Surgery and Institute of Cardiovascular Science, the First Affiliated Hospital of Soochow University, Jiangsu, China
| | - K Hong
- Jiangxi Key Laboratory of Molecular Medicine, Jiangxi, China; Department of Cardiovascular Medicine, the Second Affiliated Hospital of Nanchang University, Jiangxi, China.
| |
Collapse
|
50
|
Bastami F, Nazeman P, Moslemi H, Rezai Rad M, Sharifi K, Khojasteh A. Induced pluripotent stem cells as a new getaway for bone tissue engineering: A systematic review. Cell Prolif 2017; 50:e12321. [PMID: 27905670 PMCID: PMC6529104 DOI: 10.1111/cpr.12321] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 10/31/2016] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Mesenchymal stem cells (MSCs) are frequently used for bone regeneration, however, they are limited in quantity. Moreover, their proliferation and differentiation capabilities reduce during cell culture expansion. Potential application of induced pluripotent stem cells (iPSCs) has been reported as a promising alternative source for bone regeneration. This study aimed to systematically review the available literature on osteogenic potential of iPSCs and to discuss methods applied to enhance their osteogenic potential. METHODS AND MATERIALS A thorough search of MEDLINE database was performed from January 2006 to September 2016, limited to English-language articles. All in vitro and in vivo studies on application of iPSCs in bone regeneration were included. RESULTS The current review is organized according to the PRISMA statement. Studies were categorized according to three different approaches used for osteo-induction of iPSCs. Data are summarized and reported according to the following variables: types of study, cell sources used for iPSC generation, applied reprogramming methods, applied osteo-induction methods and treatment groups. CONCLUSION According to the articles reviewed, osteo-induced iPSCs revealed osteogenic capability equal to or superior than MSCs; cell sources do not significantly affect osteogenic potential of iPSCs; addition of resveratrol to the osteogenic medium (OM) and irradiatiation after osteogenic induction reduce teratoma formation in animal models; transfection with lentiviral bone morphogenetic protein 2 results in higher mineralization compared to osteo-induction in OM; addition of TGF-β, IGF-1 and FGF-β to OM increases osteogenic capability of iPSCs.
Collapse
Affiliation(s)
- Farshid Bastami
- Medical Nano‐Technology & Tissue Engineering Research CenterSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Pantea Nazeman
- Medical Nano‐Technology & Tissue Engineering Research CenterSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Hamidreza Moslemi
- School of DentistryShahid Beheshti University of Medical SciencesTehranIran
| | - Maryam Rezai Rad
- Medical Nano‐Technology & Tissue Engineering Research CenterSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Kazem Sharifi
- Department of BiotechnologySchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Arash Khojasteh
- Department of Tissue EngineeringSchool of Advanced Technologies in MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineUniversity of AntwerpAntwerpBelgium
| |
Collapse
|