1
|
Huang H, Ye K, Jin S. Cell Seeding Strategy Influences Metabolism and Differentiation Potency of Human Induced Pluripotent Stem Cells Into Pancreatic Progenitors. Biotechnol J 2025; 20:e70022. [PMID: 40285386 PMCID: PMC12032514 DOI: 10.1002/biot.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Human induced pluripotent stem cells (iPSCs) are an invaluable endless cell source for generating various therapeutic cells and tissues. However, their differentiation into specific cell lineages, such as definitive endoderm (DE) and pancreatic progenitor (PP), often suffers from poor reproducibility, due partially to their pluripotency. In this work, we investigated the impact of iPSC confluency during cell self-renewal and seeding density on cell metabolic activity, glycolysis to oxidative phosphorylation shift, and differentiation potential toward DE and PP lineages. Our findings demonstrated that cell seeding strategy influences cellular metabolic activity and the robustness of iPSC differentiation. iPSCs maintained at higher seeding density exhibited lower initial oxygen consumption rate (OCR) and metabolic activity. There is an optimal seeding density to ensure sufficient oxygen consumption during differentiation and to yield high expression of SOX17 in the DE lineage and high PDX1/NKX6.1 dual-positive cells in PPs. Interestingly, we found that cell confluency at the time of harvest has less impact on the efficacy of pancreatic lineage formation or metabolic activity. This study sheds light on the interplay between metabolic activity and iPSC lineage specification, offering new insights into the robustness of iPSC self-renewal and differentiation for creating human tissues.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| | - Kaiming Ye
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
- Center of Biomanufacturing for Regenerative MedicineBinghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| | - Sha Jin
- Department of Biomedical Engineering Thomas J. Watson College of Engineering and Applied Science Binghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
- Center of Biomanufacturing for Regenerative MedicineBinghamton UniversityState University of New York (SUNY)BinghamtonNew YorkUSA
| |
Collapse
|
2
|
Khan PA, Raheem A, Kalirajan C, Prashanth KG, Manivasagam G. In Vivo Assessment of a Triple Periodic Minimal Surface Based Biomimmetic Gyroid as an Implant Material in a Rabbit Tibia Model. ACS MATERIALS AU 2024; 4:479-488. [PMID: 39280806 PMCID: PMC11393938 DOI: 10.1021/acsmaterialsau.4c00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 09/18/2024]
Abstract
Biomimetic approaches to implant construction are a rising frontier in implantology. Triple Periodic Minimal Surface (TPMS)-based additively manufactured gyroid structures offer a mean curvature of zero, rendering this structure an ideal porous architecture. Previous studies have demonstrated the ability of these structures to effectively mimic the mechanical cues required for optimal implant construction. The porous nature of gyroid materials enhances bone ingrowth, thereby improving implant stability within the body. This enhancement is attributed to the increased surface area of the gyroid structure, which is approximately 185% higher than that of a dense material of the same form factor. This larger surface area allows for enhanced cellular attachment and nutrient circulation facilitated by the porous channels. This study aims to evaluate the biological performance of a gyroid-based Ti6Al-4V implant material compared to a dense alloy counterpart. Cellular viability was assessed using the lactate dehydrogenase (LDH) assay, which demonstrated that the gyroid surface allowed marginally higher viability than dense material. The in vivo integration was studied over 6 weeks using a rabbit tibia model and characterized using X-ray, micro-CT, and histopathological examination. With a metal volume of 8.1%, the gyroid exhibited a bone volume/total volume (BV/TV) ratio of 9.6%, which is 11-fold higher than that of dense metal (0.8%). Histological assessments revealed neovascularization, in-bone growth, and the presence of a Haversian system in the gyroid structure, hinting at superior osteointegration.
Collapse
Affiliation(s)
- Pearlin Amaan Khan
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore 632014, India
| | - Ansheed Raheem
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore 632014, India
| | - Cheirmadurai Kalirajan
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore 632014, India
| | - Konda Gokuldoss Prashanth
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore 632014, India
- Tallinn University of Technology, Ehitajate tee 5, 19086 Tallinn, Estonia
| | - Geetha Manivasagam
- Centre for Biomaterials, Cellular, and Molecular Theranostics, Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
3
|
Joyce CM, Gordon EB, McGivney A, Li X, Lim T, Cohen MA, Kaplan DL. Methods to Screen the Adhesion of Fish Cells on Plant-, Algal- and Fungal-Derived Biomaterials. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39969-39980. [PMID: 39024341 DOI: 10.1021/acsami.4c06543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cellular agriculture, an alternative and innovative approach to sustainable food production, has gained momentum in recent years. However, there is limited research into the production of cultivated seafood. Here, we investigated the ability of fish mackerel cells (Scomber scombrus) to adhere to plant, algal and fungal-based biomaterial scaffolds, aiming to optimize the cultivation of fish cells for use in cellular agriculture. A mackerel cell line was utilized, and metabolic assays and confocal imaging were utilized to track cell adhesion, growth, and differentiation on the different biomaterials. The mackerel cells adhered and grew on gelatin (positive control), zein, and soy proteins, as well as on alginate, chitosan, and cellulose polysaccharides. The highest adhesion and growth were on the zein and chitosan substrates, apart from the gelatin control. These findings provide a blueprint to enhance scaffold selection and design, contributing to the broader field of cellular agriculture through the development of scalable and eco-conscious solutions for meeting the growing global demand for seafood.
Collapse
Affiliation(s)
- Connor M Joyce
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Edward B Gordon
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Aelish McGivney
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Xinxin Li
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Taehwan Lim
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Malkiel A Cohen
- Wanda Fish Technologies LTD, 7 Pinhas Sapir St., Ness Ziona 7403630, Israel
| | - David L Kaplan
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
4
|
Faqeer A, Liu J, Zhang L, Wang C, Zhou G, Zhang Y. Establishment and validation of an efficient method for the 3D culture of osteoclasts in vitro. J Dent 2024; 144:104957. [PMID: 38527517 DOI: 10.1016/j.jdent.2024.104957] [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: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION Osteoclasts (OCs) play a crucial role in maintaining bone health. Changes in OC activity are linked to different bone diseases, making them an intriguing focus for research. However, most studies on OCs have relied on 2D cultures, limiting our understanding of their behavior. Yet, there's a lack of knowledge regarding platforms that effectively support osteoclast formation in 3D cultures. METHODS In our investigation, we explored the capacity of collagen and GelMA hydrogels to facilitate osteoclast development in 3D culture settings. We assessed the osteoclast development by using different hydrogels and cell seeding strategies and optimizing cell seeding density and cytokine concentration. The osteoclast development in 3D cultures was further validated by biochemical assays and immunochemical staining. RESULTS Our findings revealed that 0.3 % (w/v) collagen was conducive to osteoclast formation in both 2D and 3D cultures, demonstrated by increased multinucleation and higher TRAP activity compared to 0.6 % collagen and 5 % to 10 % (w/v) GelMA hydrogels. Additionally, we devised a "sandwich" technique using collagen substrates and augmented the initial macrophage seeding density and doubling cytokine concentrations, significantly enhancing the efficiency of OC culture in 3D conditions. Notably, we validated osteoclasts derived from macrophages in our 3D cultures express key osteoclast markers like cathepsin K and TRAP. CONCLUSIONS To conclude, our study contributes to establishing an effective method for cultivating osteoclasts in 3D environments in vitro. This innovative approach not only promises a more physiologically relevant platform to study osteoclast behavior during bone remodeling but also holds potential for applications in bone tissue engineering. CLINICAL SIGNIFICANCE This study introduces an efficient method for cultivating osteoclasts in 3D environments in vitro. It offers a more physiologically relevant platform to investigate osteoclast behavior and holds promise to advance research in bone biology and regenerative dentistry.
Collapse
Affiliation(s)
- Abdullah Faqeer
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China; School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
| | - Jie Liu
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Geriatric Orthopeadics, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518118, China
| | - Li Zhang
- Department of Stomatology, Shenzhen Children's Hospital, Shenzhen 518026, China
| | - Changde Wang
- Department of Geriatric Orthopeadics, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518118, China
| | - Guangqian Zhou
- School of Basic Medicine, Shenzhen University Medical School, Shenzhen 518015, China.
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China; School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China.
| |
Collapse
|
5
|
de Leeuw AM, Graf R, Lim PJ, Zhang J, Schädli GN, Peterhans S, Rohrbach M, Giunta C, Rüger M, Rubert M, Müller R. Physiological cell bioprinting density in human bone-derived cell-laden scaffolds enhances matrix mineralization rate and stiffness under dynamic loading. Front Bioeng Biotechnol 2024; 12:1310289. [PMID: 38419730 PMCID: PMC10900528 DOI: 10.3389/fbioe.2024.1310289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Human organotypic bone models are an emerging technology that replicate bone physiology and mechanobiology for comprehensive in vitro experimentation over prolonged periods of time. Recently, we introduced a mineralized bone model based on 3D bioprinted cell-laden alginate-gelatin-graphene oxide hydrogels cultured under dynamic loading using commercially available human mesenchymal stem cells. In the present study, we created cell-laden scaffolds from primary human osteoblasts isolated from surgical waste material and investigated the effects of a previously reported optimal cell printing density (5 × 106 cells/mL bioink) vs. a higher physiological cell density (10 × 106 cells/mL bioink). We studied mineral formation, scaffold stiffness, and cell morphology over a 10-week period to determine culture conditions for primary human bone cells in this microenvironment. For analysis, the human bone-derived cell-laden scaffolds underwent multiscale assessment at specific timepoints. High cell viability was observed in both groups after bioprinting (>90%) and after 2 weeks of daily mechanical loading (>85%). Bioprinting at a higher cell density resulted in faster mineral formation rates, higher mineral densities and remarkably a 10-fold increase in stiffness compared to a modest 2-fold increase in the lower printing density group. In addition, physiological cell bioprinting densities positively impacted cell spreading and formation of dendritic interconnections. We conclude that our methodology of processing patient-specific human bone cells, subsequent biofabrication and dynamic culturing reliably affords mineralized cell-laden scaffolds. In the future, in vitro systems based on patient-derived cells could be applied to study the individual phenotype of bone disorders such as osteogenesis imperfecta and aid clinical decision making.
Collapse
Affiliation(s)
| | - Reto Graf
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Pei Jin Lim
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jianhua Zhang
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | | - Marianne Rohrbach
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cecilia Giunta
- Connective Tissue Unit, Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Rüger
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Department of Pediatric Orthopaedics and Traumatology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Marina Rubert
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Turner J, Nandakumar A, Anilbhai N, Boccaccini AR, Jones JR, Jell G. The effect of Si species released from bioactive glasses on cell behaviour: A quantitative review. Acta Biomater 2023; 170:39-52. [PMID: 37714247 DOI: 10.1016/j.actbio.2023.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/25/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
Despite over 50 years of silicate bioactive glass (SBG) research, commercial success, and 6000+ published articles, there remains a lack of understanding of how soluble silicate (Si) species released from SBGs influences cellular responses. Using a systematic approach, this article quantitatively compares the in vitro responses of cells to SBG dissolution products reported in the literature and determines if there is a Si concentration ([Si]) dependent effect on cell behaviour. Cell behavioural responses to SBGs [Si] in dissolution products included metabolic activity (reported in 52 % of articles), cell number (24 %), protein production (22 %), gene expression (22 %) and biomineralization (24 %). There was a difference in the [Si] reported to cause increased (desirable) cellular responses (median = 30.2 ppm) compared to the [Si] reported to cause decreased (undesirable) cellular responses (median = 52.0 ppm) (P ≤ 0.001). The frequency of undesirable outcomes increased with increasing [Si], with ∼3 times more negative outcomes reported above 52 ppm. We also investigated the effect of [Si] on specific cellular outcomes (e.g., metabolic activity, angiogenesis, osteogenesis), if cell type/species influenced these responses and the impact of other ions (Ca, P, Na) within the SBG dissolution media on cell behaviour. This review has, for the first time, quantitatively compared the cellular responses to SBGs from the literature, providing a quantitative overview of SBG in vitro practices and presents evidence of a range of [Si] where desirable cellular responses may be more likely (30-52 ppm). This review also demonstrates the need for greater standardisation of in vitro methodological approaches and recommends some minimum reporting standards. STATEMENT OF SIGNIFICANCE: This systematic review investigates the relationship between the concentration of Si released from Si-bioactive glasses (SBG) and in vitro cellular responses. Si releasing materials continue to be of considerable scientific, commercial, and medical interest (with 1500+ articles published in the last 3 years) but there is considerable variation in the reported biologically effective Si concentrations and on the importance of Si on cell behaviour. Despite the variation in methodological approaches, this article demonstrated statistical commonalities in the Si concentrations that cause desirable and undesirable cellular behaviours, suggesting a window where positive cellular outcomes are more likely. This review also provides a quantitative analysis of in vitro practices within the bioactive glass field and highlights the need for greater standardisation.
Collapse
Affiliation(s)
- Joel Turner
- Division of Surgery and Interventional Sciences/ UCL, Royal Free Hospital, Pond St, London, UK; Department of Materials, Imperial College London, South Kensington Campus, London, SW7 2AZ UK
| | - Arkhash Nandakumar
- Division of Surgery and Interventional Sciences/ UCL, Royal Free Hospital, Pond St, London, UK
| | - Nikhit Anilbhai
- Division of Surgery and Interventional Sciences/ UCL, Royal Free Hospital, Pond St, London, UK
| | - Aldo R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Julian R Jones
- Department of Materials, Imperial College London, South Kensington Campus, London, SW7 2AZ UK
| | - Gavin Jell
- Division of Surgery and Interventional Sciences/ UCL, Royal Free Hospital, Pond St, London, UK.
| |
Collapse
|
7
|
Wang J, Qiao Q, Sun Y, Yu W, Wang J, Zhu M, Yang K, Huang X, Bai Y. Osteogenic Differentiation Effect of Human Periodontal Ligament Stem-Cell Initial Cell Density on Autologous Cells and Human Bone Marrow Stromal Cells. Int J Mol Sci 2023; 24:ijms24087133. [PMID: 37108296 PMCID: PMC10138982 DOI: 10.3390/ijms24087133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Stem cells have differentiation and regulation functions. Here, we discussed the impact of cell culture density on stem cell proliferation, osteoblastogenesis, and regulation. To discuss the effect of the initial culture density of human periodontal ligament stem cells (hPDLSCs) on the osteogenic differentiation of autologous cells, we found that the hPDLSC proliferation rate decreased with an increase in the initial plating density (0.5-8 × 104 cells/cm2) for the 48 h culture cycle. After hPDLSCs induced osteogenic differentiation for 14 days with different initial cell culture densities, the expression of osteoprotegerin (OPG) and runt-related transcription factor 2(RUNX2) and the OPG/ Receptor Activator of Nuclear Factor-κ B Ligand (RANKL) ratio were the highest in the hPDLSCs initially plated at a density of 2 × 104 cells/cm2, and the average cell calcium concentration was also the highest. To study hPDLSCs regulating the osteoblastic differentiation of other cells, we used 50 μg/mL of secreted exosomes derived from hPDLSCs cultured using different initial cell densities to induce human bone marrow stromal cell (hBMSC) osteogenesis. After 14 days, the results indicated that the gene expression of OPG, Osteocalcin(OCN,)RUNX2, and osterix and the OPG/RANKL ratio were the highest in the 2 × 104 cells/cm2 initial cell density group, and the average calcium concentration was also the highest. This provides a new idea for the clinical application of stem cell osteogenesis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Wenting Yu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Jiran Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Minjia Zhu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Xiaofeng Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| |
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
|
Moncal KK, Yeo M, Celik N, Acri TM, Rizk E, Wee H, Lewis GS, Salem AK, Ozbolat IT. Comparison of in-situversus ex-situdelivery of polyethylenimine-BMP-2 polyplexes for rat calvarial defect repair via intraoperative bioprinting. Biofabrication 2022; 15:10.1088/1758-5090/ac9f70. [PMID: 36322966 PMCID: PMC10012389 DOI: 10.1088/1758-5090/ac9f70] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/02/2022] [Indexed: 11/07/2022]
Abstract
Gene therapeutic applications combined with bio- and nano-materials have been used to address current shortcomings in bone tissue engineering due to their feasibility, safety and potential capability for clinical translation. Delivery of non-viral vectors can be altered using gene-activated matrices to improve their efficacy to repair bone defects.Ex-situandin-situdelivery strategies are the most used methods for bone therapy, which have never been directly compared for their potency to repair critical-sized bone defects. In this regard, we first time explore the delivery of polyethylenimine (PEI) complexed plasmid DNA encoding bone morphogenetic protein-2 (PEI-pBMP-2) using the two delivery strategies,ex-situandin-situdelivery. To realize these gene delivery strategies, we employed intraoperative bioprinting (IOB), enabling us to 3D bioprint bone tissue constructs directly into defect sites in a surgical setting. Here, we demonstrated IOB of an osteogenic bioink loaded with PEI-pBMP-2 for thein-situdelivery approach, and PEI-pBMP-2 transfected rat bone marrow mesenchymal stem cells laden bioink for theex-situdelivery approach as alternative delivery strategies. We found thatin-situdelivery of PEI-pBMP-2 significantly improved bone tissue formation compared toex-situdelivery. Despite debates amongst individual advantages and disadvantages ofex-situandin-situdelivery strategies, our results ruled in favor of thein-situdelivery strategy, which could be desirable to use for future clinical applications.
Collapse
Affiliation(s)
- Kazim K Moncal
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States of America
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA, United States of America
| | - Miji Yeo
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States of America
| | - Nazmiye Celik
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States of America
| | - Timothy M Acri
- Division of Pharmaceutics and Translational Therapeutics, Collage of Pharmacy, University of Iowa, Iowa City, IA, United States of America
| | - Elias Rizk
- Department of Neurosurgery, Penn State University, College of Medicine, Hershey, PA, United States of America
| | - Hwabok Wee
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Department of Orthopedics and Rehabilitation, Penn State University, College of Medicine, Hershey, PA, United States of America
| | - Gregory S Lewis
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Department of Orthopedics and Rehabilitation, Penn State University, College of Medicine, Hershey, PA, United States of America
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, Collage of Pharmacy, University of Iowa, Iowa City, IA, United States of America
- Department of Chemical and Biochemical Engineering, College of Engineering, University of Iowa, Iowa City, IA, United States of America
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics, Pennsylvania State University, University Park, PA, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States of America
- Department of Neurosurgery, Penn State University, College of Medicine, Hershey, PA, United States of America
- Biomedical Engineering, Pennsylvania State University, University Park, PA, United States of America
- Materials Research Institute, Pennsylvania State University, University Park, PA, United States of America
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| |
Collapse
|
10
|
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: 26] [Impact Index Per Article: 8.7] [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
|
11
|
Kageyama T, Akieda H, Sonoyama Y, Sato K, Yoshikawa H, Isono H, Hirota M, Kitajima H, Chun YS, Maruo S, Fukuda J. Bone Beads Enveloped with Vascular Endothelial Cells for Bone Regenerative Medicine. Acta Biomater 2022:S1742-7061(22)00520-7. [PMID: 36030051 DOI: 10.1016/j.actbio.2022.08.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/27/2022]
Abstract
The transplantation of pre-vascularized bone grafts is a promising strategy to improve the efficacy of engraftment and bone regeneration. We propose a hydrogel microbead-based approach for preparing vascularized and high-density tissue grafts. Mesenchymal stem cell-encapsulated collagen microgels (2 µL), termed bone beads, were prepared through spontaneous constriction, which improved the density of the mesenchymal stem cells and collagen molecules by more than 15-fold from the initial day of culture. Constriction was attributed to cell-attractive forces and involved better osteogenic differentiation of mesenchymal stem cells than that of spheroids. This approach was scalable, and ∼2,000 bone beads were prepared semi-automatically using a liquid dispenser and spinner flask. The mechanical stimuli in the spinner flask further improved the osteogenic differentiation of the mesenchymal stem cells in the bone beads compared with that in static culture. Vascular endothelial cells readily attach to and cover the surface of bone beads. The in vitro assembly of the endothelial cell-enveloped bone beads resulted in microchannel formation in the interspaces between the bone beads. Significant effects of endothelialization on in vivo bone regeneration were shown in rats with cranial bone defects. The use of endothelialized bone beads may be a scalable and robust approach for treating large bone defects. STATEMENT OF SIGNIFICANCE: A unique aspect of this study is that the hMSC-encapsulated collagen microgels were prepared through spontaneous constriction, leading to the enrichment of collagen and cell density. This constriction resulted in favorable microenvironments for the osteogenic differentiation of hMSCs, which is superior to conventional spheroid culture. The microgel beads were then enveloped with vascular endothelial cells and assembled to fabricate a tissue graft with vasculature in the interspaces among the beads. The significant effects of endothelialization on in vivo bone regeneration were clearly demonstrated in rats with cranial bone defects. We believe that microgel beads covered with vascular endothelial cells provide a promising approach for engineering better tissue grafts for bone-regenerative medicine.
Collapse
Affiliation(s)
- Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN
| | - Hikaru Akieda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Yukie Sonoyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Ken Sato
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hiroshi Yoshikawa
- Department of Chemistry, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Sakura-ku, Saitama City, Saitama 338-8570, JAPAN
| | - Hitoshi Isono
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku Yokohama, Kanagawa 236-0004, JAPAN
| | - Makoto Hirota
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Hiroaki Kitajima
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Ura-fune, Minami-ku Yokohama, Kanagawa 232-0024, JAPAN
| | - Yang-Sook Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 110-799, KOREA
| | - Shoji Maruo
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, JAPAN; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa, 213-0012, JAPAN.
| |
Collapse
|
12
|
Extensive cell seeding densities adaptable SF/PGA electrospinning scaffolds for bone tissue engineering. BIOMATERIALS ADVANCES 2022; 137:212834. [PMID: 35929266 DOI: 10.1016/j.bioadv.2022.212834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/31/2022]
|
13
|
In vitro 3D cocultured tumor-vascular barrier model based on alginate hydrogel and Transwell system for anti-cancer drug evaluation. Tissue Cell 2022; 76:101796. [DOI: 10.1016/j.tice.2022.101796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 11/23/2022]
|
14
|
Park JH, Ahn M, Park SH, Kim H, Bae M, Park W, Hollister SJ, Kim SW, Cho DW. 3D bioprinting of a trachea-mimetic cellular construct of a clinically relevant size. Biomaterials 2021; 279:121246. [PMID: 34775331 PMCID: PMC8663475 DOI: 10.1016/j.biomaterials.2021.121246] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/16/2022]
Abstract
Despite notable advances in extrusion-based 3D bioprinting, it remains a challenge to create a clinically-sized cellular construct using extrusion-based 3D printing due to long printing times adversely affecting cell viability and functionality. Here, we present an advanced extrusion-based 3D bioprinting strategy composed of a two-step printing process to facilitate creation of a trachea-mimetic cellular construct of clinically relevant size. A porous bellows framework is first printed using typical extrusion-based 3D printing. Selective printing of cellular components, such as cartilage rings and epithelium lining, is then performed on the outer grooves and inner surface of the bellows framework by a rotational printing process. With this strategy, 3D bioprinting of a trachea-mimetic cellular construct of clinically relevant size is achieved in significantly less total printing time compared to a typical extrusion-based 3D bioprinting strategy which requires printing of an additional sacrificial material. Tracheal cartilage formation was successfully demonstrated in a nude mouse model through a subcutaneous implantation study of trachea-mimetic cellular constructs wrapped with a sinusoidal-patterned tubular mesh preventing rapid resorption of cartilage rings in vivo. This two-step 3D bioprinting for a trachea-mimetic cellular construct of clinically relevant size can provide a fundamental step towards clinical translation of 3D bioprinting based tracheal reconstruction.
Collapse
Affiliation(s)
- Jeong Hun Park
- Wallace H. Coulter Department of Biomedical Engineering and Center for 3D Medical Fabrication, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA
| | - Minjun Ahn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Sun Hwa Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, 137-710, Republic of Korea
| | - Hyeonji Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Scott J Hollister
- Wallace H. Coulter Department of Biomedical Engineering and Center for 3D Medical Fabrication, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, 137-710, Republic of Korea.
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Kyungbuk, 37673, Republic of Korea.
| |
Collapse
|
15
|
Ghaffari S, Torabi-Rahvar M, Aghayan S, Jabbarpour Z, Moradzadeh K, Omidkhoda A, Ahmadbeigi N. Optimizing interleukin-2 concentration, seeding density and bead-to-cell ratio of T-cell expansion for adoptive immunotherapy. BMC Immunol 2021; 22:43. [PMID: 34217218 PMCID: PMC8254233 DOI: 10.1186/s12865-021-00435-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The successful ex vivo expansion of T-cells in great numbers is the cornerstone of adoptive cell therapy. We aimed to achieve the most optimal T-cell expansion condition by comparing the expansion of T-cells at various seeding densities, IL-2 concentrations, and bead-to-cell ratios. we first expanded the peripheral blood mononuclear cells (PBMCs) of a healthy donor at a range of 20 to 500 IU/mL IL-2 concentrations, 125 × 103 to 1.5 × 106 cell/mL, and 1:10 to 10:1 B:C (Bead-to-cell) ratios and compared the results. We then expanded the PBMC of three healthy donors using the optimized conditions and examined the growth kinetics. On day 28, CD3, CD4, and CD8 expression of the cell populations were analyzed by flow cytometry. RESULTS T-cells of the first donor showed greater expansion results in IL-2 concentrations higher than 50 IU/mL compared to 20 IU/mL (P = 0.02). A seeding density of 250 × 103 cell/mL was superior to higher or lower densities in expanding T-cells (P = 0.025). Also, we witnessed a direct correlation between the B:C ratio and T-cell expansion, in which, in 5:1 and 10:1 B:C ratios T-cell significantly expanded more than lower B:C ratios. The results of PBMC expansions of three healthy donors were similar in growth kinetics. In the optimized condition, 96-98% of the lymphocyte population expressed CD3. While the majority of these cells expressed CD8, the mean expression of CD4 in the donors was 19.3, 16.5, and 20.4%. CONCLUSIONS Our methodology demonstrates an optimized culture condition for the production of large quantities of polyclonal T-cells, which could be useful for future clinical and research studies.
Collapse
Affiliation(s)
- Sasan Ghaffari
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Monireh Torabi-Rahvar
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sajjad Aghayan
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Jabbarpour
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kobra Moradzadeh
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Omidkhoda
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
The mTORC2 Regulator Homer1 Modulates Protein Levels and Sub-Cellular Localization of the CaSR in Osteoblast-Lineage Cells. Int J Mol Sci 2021; 22:ijms22126509. [PMID: 34204449 PMCID: PMC8234890 DOI: 10.3390/ijms22126509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
We recently found that, in human osteoblasts, Homer1 complexes to Calcium-sensing receptor (CaSR) and mediates AKT initiation via mechanistic target of rapamycin complex (mTOR) complex 2 (mTORC2) leading to beneficial effects in osteoblasts including β-catenin stabilization and mTOR complex 1 (mTORC1) activation. Herein we further investigated the relationship between Homer1 and CaSR and demonstrate a link between the protein levels of CaSR and Homer1 in human osteoblasts in primary culture. Thus, when siRNA was used to suppress the CaSR, we observed upregulated Homer1 levels, and when siRNA was used to suppress Homer1 we observed downregulated CaSR protein levels using immunofluorescence staining of cultured osteoblasts as well as Western blot analyses of cell protein extracts. This finding was confirmed in vivo as the bone cells from osteoblast specific CaSR-/- mice showed increased Homer1 expression compared to wild-type (wt). CaSR and Homer1 protein were both expressed in osteocytes embedded in the long bones of wt mice, and immunofluorescent studies of these cells revealed that Homer1 protein sub-cellular localization was markedly altered in the osteocytes of CaSR-/- mice compared to wt. The study identifies additional roles for Homer1 in the control of the protein level and subcellular localization of CaSR in cells of the osteoblast lineage, in addition to its established role of mTORC2 activation downstream of the receptor.
Collapse
|
17
|
Schott NG, Friend NE, Stegemann JP. Coupling Osteogenesis and Vasculogenesis in Engineered Orthopedic Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:199-214. [PMID: 32854589 PMCID: PMC8349721 DOI: 10.1089/ten.teb.2020.0132] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Inadequate vascularization of engineered tissue constructs is a main challenge in developing a clinically impactful therapy for large, complex, and recalcitrant bone defects. It is well established that bone and blood vessels form concomitantly during development, as well as during repair after injury. Endothelial cells (ECs) and mesenchymal stromal cells (MSCs) are known to be key players in orthopedic tissue regeneration and vascularization, and these cell types have been used widely in tissue engineering strategies to create vascularized bone. Coculture studies have demonstrated that there is crosstalk between ECs and MSCs that can lead to synergistic effects on tissue regeneration. At the same time, the complexity in fabricating, culturing, and characterizing engineered tissue constructs containing multiple cell types presents a challenge in creating multifunctional tissues. In particular, the timing, spatial distribution, and cell phenotypes that are most conducive to promoting concurrent bone and vessel formation are not well understood. This review describes the processes of bone and vascular development, and how these have been harnessed in tissue engineering strategies to create vascularized bone. There is an emphasis on interactions between ECs and MSCs, and the culture systems that can be used to understand and control these interactions within a single engineered construct. Developmental engineering strategies to mimic endochondral ossification are discussed as a means of generating vascularized orthopedic tissues. The field of tissue engineering has made impressive progress in creating tissue replacements. However, the development of larger, more complex, and multifunctional engineered orthopedic tissues will require a better understanding of how osteogenesis and vasculogenesis are coupled in tissue regeneration. Impact statement Vascularization of large engineered tissue volumes remains a challenge in developing new and more biologically functional bone grafts. A better understanding of how blood vessels develop during bone formation and regeneration is needed. This knowledge can then be applied to develop new strategies for promoting both osteogenesis and vasculogenesis during the creation of engineered orthopedic tissues. This article summarizes the processes of bone and blood vessel development, with a focus on how endothelial cells and mesenchymal stromal cells interact to form vascularized bone both during development and growth, as well as tissue healing. It is meant as a resource for tissue engineers who are interested in creating vascularized tissue, and in particular to those developing cell-based therapies for large, complex, and recalcitrant bone defects.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
18
|
Chen P, Xie J, Zhu J, Hu Y. Design of two natural deproteinized bovine bone scaffolds and evaluation of the effect of initial cell seeding density on repairing bone defects. Microsc Res Tech 2021; 84:1612-1620. [PMID: 33768719 DOI: 10.1002/jemt.23721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 11/07/2022]
Abstract
Engineering functional bone using combinations of cells, scaffolds, and bioactive factors is a promising strategy for the bone-tissue regeneration, while challenge remains. Chemical methods deproteinizing natural bovine cancellous bone to remove immunogenic are poorly understood, and the cell seeding density to promote bone formation still needs to be clarified. In this study, 8.0 × 8.0 × 2.0 mm bovine cancellous bones were either treated with H2 O2 for 8 hr or pepsin for 24 hr and then inoculated with MC3T3-E1 osteoblasts with two cell densities (1 × 106 cells/ml or 4 × 106 cells/ml)separately. We compared the appearance of the bones treated by the two chemical deproteinizing methods, as well as the proliferation ability of the inoculating cell density at 1 × 106 cells/ml. Moreover, scanning electron microscopy was done to analyze the growth of cells on the surface of the material, and an alkaline phosphatase assay was performed to assess osteogenic differentiation. We showed that both treated bones treatments are biocompatible, but bones treated with H2 O2 were more conducive to osteoblast differentiation and ALP secretion, especially when seeded at the higher cell density at 4 × 106 cells/ml. We concluded that chemical deproteinized bovine cancellous bones met the basic bone graft material requirements. Cell seeding density is an important factor to promote the material's osteogenic ability, with H2 O2 -deproteinized bones exhibiting enhanced osteoblast differentiation.
Collapse
Affiliation(s)
- Peng Chen
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jie Xie
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jianxi Zhu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Akcay G, Luttge R. Stiff-to-Soft Transition from Glass to 3D Hydrogel Substrates in Neuronal Cell Culture. MICROMACHINES 2021; 12:mi12020165. [PMID: 33567528 PMCID: PMC7915240 DOI: 10.3390/mi12020165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/26/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023]
Abstract
Over the past decade, hydrogels have shown great potential for mimicking three- dimensional (3D) brain architectures in vitro due to their biocompatibility, biodegradability, and wide range of tunable mechanical properties. To better comprehend in vitro human brain models and the mechanotransduction processes, we generated a 3D hydrogel model by casting photo-polymerized gelatin methacryloyl (GelMA) in comparison to poly (ethylene glycol) diacrylate (PEGDA) atop of SH-SY5Y neuroblastoma cells seeded with 150,000 cells/cm2 according to our previous experience in a microliter-sized polydimethylsiloxane (PDMS) ring serving for confinement. 3D SH-SY5Y neuroblastoma cells in GelMA demonstrated an elongated, branched, and spreading morphology resembling neurons, while the cell survival in cast PEGDA was not supported. Confocal z-stack microscopy confirmed our hypothesis that stiff-to-soft material transitions promoted neuronal migration into the third dimension. Unfortunately, large cell aggregates were also observed. A subsequent cell seeding density study revealed a seeding cell density above 10,000 cells/cm2 started the formation of cell aggregates, and below 1500 cells/cm2 cells still appeared as single cells on day 6. These results allowed us to conclude that the optimum cell seeding density might be between 1500 and 5000 cells/cm2. This type of hydrogel construct is suitable to design a more advanced layered mechanotransduction model toward 3D microfluidic brain-on-a-chip applications.
Collapse
|
20
|
Wu S, Lei L, Bao C, Liu J, Weir MD, Ren K, Schneider A, Oates TW, Liu J, Xu HHK. An injectable and antibacterial calcium phosphate scaffold inhibiting Staphylococcus aureus and supporting stem cells for bone regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111688. [PMID: 33545850 DOI: 10.1016/j.msec.2020.111688] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023]
Abstract
Staphylococcus aureus (S. aureus) is the major pathogen for osteomyelitis, which can lead to bone necrosis and destruction. There has been no report on antibacterial calcium phosphate cement (CPC) against S. aureus. The aims of this study were to: (1) develop novel antibacterial CPC-chitosan-alginate microbead scaffold; (2) investigate mechanical and antibacterial properties of CPC-chitosan-penicillin-alginate scaffold; (3) evaluate the encapsulation and delivery of human umbilical cord mesenchymal stem cells (hUCMSCs). Flexural strength, elastic modulus and work-of-fracture of the CPC-chitosan-penicillin-alginate microbeads scaffold and CPC-chitosan scaffold were evaluated. Penicillin release profile and antibacterial effects on S. aureus were determined. The hUCMSC delivery and release from penicillin-alginate microbeads were investigated. Injectable CPC-chitosan-penicillin-alginate microbeads scaffold was developed for the first time. CPC-chitosan-penicillin-alginate microbeads scaffold had a flexural strength of 3.16 ± 0.55 MPa, matching that of cancellous bone. With sustained penicillin release, the new scaffold had strong antibacterial effects on S. aureus, with an inhibition zone diameter of 32.2 ± 2.5 mm, greater than that of penicillin disk control (15.1 ± 2.0 mm) (p < 0.05). Furthermore, this injectable and antibacterial scaffold had no toxic effects, yielding excellent hUCMSC viability, which was similar to that of CPC control without antibacterial activity (p > 0.05). CPC-chitosan-penicillin-microbeads scaffold had injectability, good strength, strong antibacterial effects, and good biocompatibility to support stem cell viability for osteogenesis. CPC-chitosan-penicillin-microbeads scaffold is promising for dental, craniofacial and orthopedic applications to combat infections and promote bone regeneration.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jin Liu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Key Laboratory of Shannxi for Craniofacial Precision Medicine Research, Clinical Research Center of Shannxi for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Shannxi 710004, China
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Ke Ren
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, University of Maryland, Baltimore, MD 21201, USA
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas W Oates
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
21
|
Juan CH, Chen MH, Lin FH, Wong CS, Chien CC, Chen MH. In Vitro Differentiation of Human Placenta-Derived Multipotent Cells into Schwann-Like Cells. Biomolecules 2020; 10:biom10121657. [PMID: 33322066 PMCID: PMC7763858 DOI: 10.3390/biom10121657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/31/2022] Open
Abstract
Human placenta-derived multipotent stem cells (PDMCs) resembling embryonic stem cells can differentiate into three germ layer cells, including ectodermal lineage cells, such as neurons, astrocytes, and oligodendrocytes. The favorable characteristics of noninvasive cell harvesting include fewer ethical, religious, and legal considerations as well as accessible and limitless supply. Thus, PDMCs are attractive for cell-based therapy. The Schwann cell (SC) is the most common cell type used for tissue engineering such as nerve regeneration. However, the differentiation potential of human PDMCs into SCs has not been demonstrated until now. In this study, we evaluated the potential of PDMCs to differentiate into SC-like cells in a differentiation medium. After induction, PDMCs not only exhibited typical SC spindle-shaped morphology but also expressed SC markers, including S100, GFAP, p75, MBP, and Sox 10, as revealed by immunocytochemistry. Moreover, a reverse transcription-quantitative polymerase chain reaction analysis revealed the elevated gene expression of S100, GFAP, p75, MBP, Sox-10, and Krox-20 after SC induction. A neuroblastoma cell line, SH-SY5Y, was cultured in the conditioned medium (CM) collected from PDMC-differentiated SCs. The growth rate of the SH-SY5Y increased in the CM, indicating the function of PDMC-induced SCs. In conclusion, human PDMCs can be differentiated into SC-like cells and thus are an attractive alternative to SCs for cell-based therapy in the future.
Collapse
Affiliation(s)
- Chung-Hau Juan
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
- Department of Biomedical Sciences, National Central University, Taoyuan 32001, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 220216, Taiwan;
- Department of Biomedical Engineering, Ming Chuan University, Taoyuan 333321, Taiwan
| | - Feng-Hui Lin
- Department of Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan;
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
| | - Chih-Cheng Chien
- Department of Anesthesiology, Cathay General Hospital, Taipei 106438, Taiwan; (C.-H.J.); (C.-S.W.); (C.-C.C.)
| | - Ming-Hong Chen
- Department of Neurosurgery, Taipei Municipal Wangfang Hospital, Taipei 116081, Taiwan
- Department of Biomedical Sciences, Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence:
| |
Collapse
|
22
|
Vurat MT, Şeker Ş, Lalegül-Ülker Ö, Parmaksiz M, Elçin AE, Elçin YM. Development of a multicellular 3D-bioprinted microtissue model of human periodontal ligament-alveolar bone biointerface: Towards a pre-clinical model of periodontal diseases and personalized periodontal tissue engineering. Genes Dis 2020; 9:1008-1023. [PMID: 35685479 PMCID: PMC9170773 DOI: 10.1016/j.gendis.2020.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/20/2022] Open
Abstract
While periodontal (PD) disease is among principal causes of tooth loss worldwide, regulation of concomitant soft and mineralized PD tissues, and PD pathogenesis have not been completely clarified yet. Besides, relevant pre-clinical models and in vitro platforms have limitations in simulating human physiology. Here, we have harnessed three-dimensional bioprinting (3DBP) technology for developing a multi-cellular microtissue model resembling PD ligament-alveolar bone (PDL-AB) biointerface for the first time. 3DBP parameters were optimized; the physical, chemical, rheological, mechanical, and thermal properties of the constructs were assessed. Constructs containing gelatin methacryloyl (Gel-MA) and hydroxyapatite-magnetic iron oxide nanoparticles showed higher level of compressive strength when compared with that of Gel-MA constructs. Bioprinted self-supporting microtissue was cultured under flow in a microfluidic platform for >10 days without significant loss of shape fidelity. Confocal microscopy analysis indicated that encapsulated cells were homogenously distributed inside the matrix and preserved their viability for >7 days under microfluidic conditions. Immunofluorescence analysis showed the cohesion of stromal cell surface marker-1+ human PDL fibroblasts containing PDL layer with the osteocalcin+ human osteoblasts containing mineralized layer in time, demonstrating some permeability of the printed constructs to cell migration. Preliminary tetracycline interaction study indicated the uptake of model drug by the cells inside the 3D-microtissue. Also, the non-toxic levels of tetracycline were determined for the encapsulated cells. Thus, the effects of tetracyclines on PDL-AB have clinical significance for treating PD diseases. This 3D-bioprinted multi-cellular periodontal/osteoblastic microtissue model has potential as an in vitro platform for studying processes of the human PDL.
Collapse
|
23
|
Elnaggar MA, El-Fawal HAN, Allam NK. Biocompatible PCL-nanofibers scaffold with immobilized fibronectin and laminin for neuronal tissue regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111550. [PMID: 33321614 DOI: 10.1016/j.msec.2020.111550] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/08/2020] [Accepted: 09/22/2020] [Indexed: 10/23/2022]
Abstract
Recent advances in regenerative medicine have given hope in overcoming and rehabilitating complex medical conditions. In this regard, the biopolymer poly-ε-caprolactone (PCL) may be a promising candidate for tissue regeneration, despite lacking the essential bioactivity. The present study used PCL nanofibers (NFs) scaffold decorated with the extracellular matrix proteins fibronectin and laminin combined for neuronal regeneration. The potential for the dual proteins to support neuronal cells and promote axonal growth was investigated. Two NFs scaffolds were produced with PLC concentrations of 12% or 15%. Under scanning electron microscopy, both scaffolds evidenced uniform diameter distribution in the range of 358 nm and 887 nm, respectively, with >80% porosity. The Brunauer-Emmett-Teller (BET) test confirmed that the fabricated NFs mats had a high surface area, especially for the 12% NFs with 652 m2/g compared to 254 m2/g for the 15% NFs. The proteins of interest were successfully conjugated to the 12% PCL scaffold through chemical carbodiimide reaction as confirmed by Fourier-transform infrared spectroscopy. The addition of fibronectin and laminin together was shown to be the most favorable for cellular attachment and elongation of neuroblastoma SH-SY5Y cells compared to other formulations. Light microscopy revealed longer neurite outgrowth, higher cellular projected area, and lower shape index for the cells cultured on the combined proteins conjugated fibers, indicating enhanced cellular spread on the scaffold. This preliminary study suggests that PCL nanoscaffolding conjugated with matrix proteins can support neuronal cell viability and neurite growth.
Collapse
Affiliation(s)
- Manar A Elnaggar
- Nanotechnology Program, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Hassan A N El-Fawal
- Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
| | - Nageh K Allam
- Nanotechnology Program, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt; Energy Materials Laboratory, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt.
| |
Collapse
|
24
|
Berent ZT, Wagoner Johnson AJ. Cell seeding simulation on micropatterned islands shows cell density depends on area to perimeter ratio, not on island size or shape. Acta Biomater 2020; 107:152-163. [PMID: 32112979 DOI: 10.1016/j.actbio.2020.02.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/21/2020] [Accepted: 02/21/2020] [Indexed: 01/24/2023]
Abstract
Protein micropatterned substrates have been used to control cell size, shape, and cell-cell contacts, characteristics that influence a range of cell behaviors from early cell differentiation to late stages of maturation. Knowing the initial island cell seeding density is important to interpreting results and understanding downstream cell behavior. While studies routinely report the intended or target cell seeding density, they do not report the actual cell seeding density on the islands. As cells proliferate, differences in initial cell seeding density could compound and may lead to misinterpretation of results. In this work, we present a cell seeding simulation and apply it to 100s of islands with a range of geometries (sizes and shapes) to explore how island cell seeding density relates to the target or unpatterned cell seeding density. We first experimentally validate the simulation and then show that normalized island cell seeding density depends on island size, shape, and spacing, but can be predicted solely from island area to perimeter ratio, A2P, via a power law relationship for a wide range of island geometries. Interestingly, normalized island cell seeding density is the same as the normalized unpatterned cell seeding density for A2P ≥ 17 µm. This simulation will help to design micropatterned substrates and to have more accurate representation of the island cell seeding density at the start of experiments. By knowing the island cell seeding density, we can more easily reproduce results across research groups to understand the roles of cell-cell contact and cell size and shape on cell behavior. STATEMENT OF SIGNIFICANCE: We present a cell seeding simulation on protein-micropatterned substrates and use it to simulate seeding across 100s of island geometries (size, shape, and spacing) covering two orders of magnitude in size. The simulation shows that island cell density varies significantly with island geometry compared to the target seeding density. However, island cell density can be predicted from one geometric parameter - the island's area to perimeter ratio. Results will help direct researchers on how to achieve uniform cell density across all island geometries. Since cell density and island shape both influence cell behaviors, such as differentiation, this simulation may help to isolate these factors, facilitate micropatterned substrate design, and provide a mechanism for more reproduceable results across studies.
Collapse
|
25
|
The Influence of Cell Culture Density on the Cytotoxicity of Adipose-Derived Stem Cells Induced by L-Ascorbic Acid-2-Phosphate. Sci Rep 2020; 10:104. [PMID: 31919399 PMCID: PMC6952413 DOI: 10.1038/s41598-019-56875-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Ascorbic acid-2-phosphate (A2-P) is an oxidation-resistant derivative of ascorbic acid that has been widely employed in culturing adipose-derived stem cells (ASCs) for faster expansion and cell sheet formation. While high dose ascorbic acid is known to induce cellular apoptosis via metabolic stress and genotoxic effects, potential cytotoxic effects of A2-P at high concentrations has not been explored. In this study, the relationship between ASC seeding density and A2-P-induced cytotoxicity was investigated. Spheroid-derived ASCs with smaller cellular dimensions were generated to investigate the effect of cell-cell contact on the resistance to A2-P-induced cytotoxicity. Decreased viability of ASC, fibroblast, and spheroid-derived ASC was noted at higher A2-P concentration, and it could be reverted with high seeding density. Compared to control ASCs, spheroid-derived ASCs seeded at the same density exhibited decreased viability in the A2-P-supplemented medium. The expression of antioxidant enzymes (catalase, SOD1, and SOD2) was enhanced in ASCs at higher seeding densities. However, their enhanced expression in spheroid-derived ASCs was less evident. Furthermore, we found that co-administration of catalase or N-acetylcysteine nullified the observed cytotoxicity. Collectively, A2-P can induce ASC cytotoxicity at higher concentrations, which can be prevented by seeding ASCs at high density or co-administration of another antioxidant.
Collapse
|
26
|
Rubí-Sans G, Recha-Sancho L, Pérez-Amodio S, Mateos-Timoneda MÁ, Semino CE, Engel E. Development of a Three-Dimensional Bioengineered Platform for Articular Cartilage Regeneration. Biomolecules 2019; 10:E52. [PMID: 31905668 PMCID: PMC7023234 DOI: 10.3390/biom10010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022] Open
Abstract
Degenerative cartilage pathologies are nowadays a major problem for the world population. Factors such as age, genetics or obesity can predispose people to suffer from articular cartilage degeneration, which involves severe pain, loss of mobility and consequently, a loss of quality of life. Current strategies in medicine are focused on the partial or total replacement of affected joints, physiotherapy and analgesics that do not address the underlying pathology. In an attempt to find an alternative therapy to restore or repair articular cartilage functions, the use of bioengineered tissues is proposed. In this study we present a three-dimensional (3D) bioengineered platform combining a 3D printed polycaprolactone (PCL) macrostructure with RAD16-I, a soft nanofibrous self-assembling peptide, as a suitable microenvironment for human mesenchymal stem cells' (hMSC) proliferation and differentiation into chondrocytes. This 3D bioengineered platform allows for long-term hMSC culture resulting in chondrogenic differentiation and has mechanical properties resembling native articular cartilage. These promising results suggest that this approach could be potentially used in articular cartilage repair and regeneration.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Biomaterials for Regenerative Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (G.R.-S.); (S.P.-A.); (M.Á.M.-T.)
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Tissue Engineering Laboratory, IQS School of Engineering, Ramon Llull University, 08017 Barcelona, Spain;
| | - Lourdes Recha-Sancho
- Tissue Engineering Laboratory, IQS School of Engineering, Ramon Llull University, 08017 Barcelona, Spain;
| | - Soledad Pérez-Amodio
- Biomaterials for Regenerative Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (G.R.-S.); (S.P.-A.); (M.Á.M.-T.)
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Department of Materials Science and Metallurgical Engineering, EEBE campus, Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| | - Miguel Ángel Mateos-Timoneda
- Biomaterials for Regenerative Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (G.R.-S.); (S.P.-A.); (M.Á.M.-T.)
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Department of Materials Science and Metallurgical Engineering, EEBE campus, Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| | - Carlos Eduardo Semino
- Tissue Engineering Laboratory, IQS School of Engineering, Ramon Llull University, 08017 Barcelona, Spain;
- Hebe Biolab S.L., C/Can Castellvi 27, 08017 Barcelona, Spain
| | - Elisabeth Engel
- Biomaterials for Regenerative Therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; (G.R.-S.); (S.P.-A.); (M.Á.M.-T.)
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
- Department of Materials Science and Metallurgical Engineering, EEBE campus, Technical University of Catalonia (UPC), 08019 Barcelona, Spain
| |
Collapse
|
27
|
Ghaffari S, Torabi‐Rahvar M, Omidkhoda A, Ahmadbeigi N. Impact of various culture conditions on
ex vivo
expansion of polyclonal T cells for adoptive immunotherapy. APMIS 2019; 127:737-745. [DOI: 10.1111/apm.12981] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Sasan Ghaffari
- Cell‐Based Therapies Research Center, Digestive Disease Research InstituteTehran University of Medical Sciences TehranIran
- Student Scientific Research Center Tehran University of Medical Sciences TehranIran
| | - Monireh Torabi‐Rahvar
- Cell‐Based Therapies Research Center, Digestive Disease Research InstituteTehran University of Medical Sciences TehranIran
- SABZ Biomedicals Science‐Based Company TehranIran
| | - Azadeh Omidkhoda
- Department of Hematology, School of Allied Medical Sciences Tehran University of Medical Sciences Tehran Iran
| | - Naser Ahmadbeigi
- Cell‐Based Therapies Research Center, Digestive Disease Research InstituteTehran University of Medical Sciences TehranIran
| |
Collapse
|
28
|
Malakpour Permlid A, Roci P, Fredlund E, Fält F, Önell E, Johansson F, Oredsson S. Unique animal friendly 3D culturing of human cancer and normal cells. Toxicol In Vitro 2019; 60:51-60. [DOI: 10.1016/j.tiv.2019.04.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 10/26/2022]
|
29
|
Zou J, Wang W, Nie Y, Xu X, Ma N, Lendlein A. Microscale roughness regulates laminin-5 secretion of bone marrow mesenchymal stem cells. Clin Hemorheol Microcirc 2019; 73:237-247. [PMID: 31561334 DOI: 10.3233/ch-199205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Laminin-5 (Ln-5), an important extracellular matrix (ECM) protein, plays a critical role in regulating the growth and differentiation of mesodermal tissues, including bone. Ln-5 can be secreted by the mesenchymal stem cells (MSCs), and Ln-5 promotes MSCs osteogenic differentiation. It has been demonstrated that a substrate's surface topography could regulate MSC secretion and differentiation. A better understanding of the mechanism of how Ln-5 and surface roughness regulate MSC osteogenic differentiation would guide the design of surface topography and coatings of orthopedic implants and cell culture substrates. However, few studies have investigated the relationship between surface roughness and the secretion of Ln-5 in MSC osteogenic differentiation. Whether substrate surface topography regulates MSC differentiation via regulating Ln-5 secretion and how surface topography contributes to the secretion of Ln-5 are still not known. In this study, the influence of microscale roughness at different levels (R0, R1 and R2) on the secretion of Ln-5 of human bone marrow MSCs (hBMSCs) and subsequent osteogenic differentiation were examined. hBMSCs spreading, distribution and morphology were greatly affected by different roughness levels. A significantly higher level of Ln-5 secretion was detected on R2, which correlated to the local cell density regulated by the rough surface. Ln-5 binding integrins (α2 and α3) were strongly activated on R2. In addition, the results from hBMSCs on R0 inserts with different cell densities further confirmed that local cell density regulated Ln-5 secretion and cell surface integrin activation. In addition, the mineralization level of MSCs on R2 was remarkably higher than that on R0 and R1. These results suggest that hBMSC osteogenic differentiation level on R2 roughness was enhanced via increased Ln-5 secretion that was attributed to rough surface regulated local cell density. Thus, the microroughness could serve as effective topographical stimulus in cell culture devices and bone implant materials.
Collapse
Affiliation(s)
- Jie Zou
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Yan Nie
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, Berlin, Germany
- Institute of Chemistry, University of Potsdam, Potsdam, Germany
| |
Collapse
|
30
|
Evaluation of Proliferation and Osteogenic Differentiation of Human Umbilical Cord-Derived Mesenchymal Stem Cells in Porous Scaffolds. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1084:207-220. [PMID: 31214911 DOI: 10.1007/5584_2019_343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Human umbilical cord-derived mesenchymal stem cells (UCMSCs) are multiple potential stem cells that can differentiate into various kinds of functional cells, including adipocytes, osteoblasts, and chondroblasts. Thus, UCMSCs have recently been used in both stem cell therapy and tissue engineering applications to produce various functional tissues. This study aimed to evaluate the proliferation and differentiation of UCMSCs on porous scaffolds. METHODS UCMSCs were established in a previous study and kept in liquid nitrogen. They were thawed and expanded in vitro to yield enough cells for further experiments. The cells were characterized as having MSC phenotype. They were seeded onto culture medium-treated porous scaffolds or on non-treated porous scaffolds at different densities of UCMSCs (105, 2.1 × 105, and 5 × 105 cells/0.005 g scaffold). The existence of UCMSCs on the scaffold was evaluated by nucleic staining using Hoechst 33342 dye, while cell proliferation on the scaffold was determined by MTT assay. Osteogenic differentiation was evaluated by changes in cellular morphology, accumulation of extracellular calcium, and expression of osteoblast-specific genes (including runx2, osteopontin (OPN), and osteocalcin (OCN)). RESULTS The data showed that UCMSCs could attach, proliferate, and differentiate on both treated and non-treated scaffolds but were better on the treated scaffold. At a cell density of 105 cells/0.005 g scaffold, the adherent and proliferative abilities of UCMSCs were higher than that of the other densities after 14 days of culture (p < 0.05). Adherent UCMSCs on the scaffold could be induced into osteoblasts in the osteogenic medium after 21 days of induction. These cells accumulated calcium in the extracellular matrix that was positive with Alizarin Red staining. They also expressed some genes related to osteoblasts, including runx2, OPN, and OCN. CONCLUSION UCMSCs could adhere, proliferate, and differentiate into osteoblasts on porous scaffolds. Therefore, porous scaffolds (such as Variotis) may be suitable scaffolds for producing bone tissue in combination with UCMSCs.
Collapse
|
31
|
Meng D, Dong L, Yuan Y, Jiang Q. In vitro and in vivo analysis of the biocompatibility of two novel and injectable calcium phosphate cements. Regen Biomater 2018; 6:13-19. [PMID: 30740238 PMCID: PMC6362821 DOI: 10.1093/rb/rby027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/05/2018] [Accepted: 11/20/2018] [Indexed: 11/29/2022] Open
Abstract
Calcium phosphate cements (CPCs) have been widely used as bone graft substitutes for many years. The aim of this study was to evaluate the biocompatibility of two novel injectable, bioactive cements: β-tricalcium phosphate (β-TCP)/CPC and chitosan microsphere/CPC in vitro and in vivo. This was accomplished by culturing mouse pre-osteoblastic cells (MC3T3-E1) on discs and pastes of CPCs. Cell growth, adhesion, proliferation and differentiation were assessed by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide and alkaline phosphatase assays as well as by scanning electron microscopy and fluorescence. The effect of CPC paste curing was also evaluated. Implantation of two materials into the muscle tissue of rabbits was also studied and evaluated by histological analysis. Cell analysis indicated good biocompatibility in vitro. The fluorescence assay suggested that the cured material discs had no obvious effect on cell growth, while the curing process did. Histological examination showed no inflammatory cell infiltration into soft tissue. These data suggest that β-TCP/CPC and chitosan microsphere/CPC composites may be promising injectable material for bone tissue engineering.
Collapse
Affiliation(s)
- Dan Meng
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Limin Dong
- Beijing Key Laboratory of Fine Ceramics, Institute of Nuclear and New Energy Technology, Tsinghua University, Energy Science Building, Beijing, China
| | - Yafei Yuan
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Qingsong Jiang
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Talò G, Turrisi C, Arrigoni C, Recordati C, Gerges I, Tamplenizza M, Cappelluti A, Riboldi S, Moretti M. Industrialization of a perfusion bioreactor: Prime example of a non‐straightforward process. J Tissue Eng Regen Med 2017; 12:405-415. [DOI: 10.1002/term.2480] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 01/17/2023]
Affiliation(s)
- G. Talò
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
| | - C. Turrisi
- Dipartimento di Elettronica, Informazione e BioingegneriaPolitecnico di Milano Milan Italy
| | - C. Arrigoni
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
| | | | | | | | - A. Cappelluti
- Fondazione Filarete Milan Italy
- SEMM European School of Molecular Medicine Milano Italy
| | | | - M. Moretti
- Cell and Tissue Engineering LaboratoryIRCCS Istituto Ortopedico Galeazzi Milan Italy
- Regenerative Medicine Technologies LaboratoryEnte Ospedaliero Cantonale (EOC) Lugano Switzerland
- Swiss Institute of Regenerative Medicine (SIRM) Torricella‐Taverne Switzerland
| |
Collapse
|
33
|
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: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Tang J, Saito T. Human plasma fibronectin promotes proliferation and differentiation of odontoblast. J Appl Oral Sci 2017; 25:299-309. [PMID: 28678949 PMCID: PMC5482253 DOI: 10.1590/1678-7757-2016-0442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/18/2016] [Indexed: 11/21/2022] Open
Abstract
Objective To assess the effect of fibronectin (Fn) and porcine type I collagen (PCOL) on odontoblast-like cells in vitro. Material and Methods Rat odontoblast-like cells (MDPC-23 cells) were inoculated and cultured on Fn-coated or type I collagen-coated substrates. Proliferation assay, alkaline phosphatase activity (ALP activity), mRNA expression of hard tissue-forming markers, and Alizarin red staining were investigated over a period of 10 days. Results Cells maintained a high proliferation activity on Fn and PCOL even at a low seeding concentration (0.5×104/mL) as demonstrated by CCK-8 assay. The proliferation activity of cells on Fn increases in a concentration-dependent manner while it reached a plateau after 10 µg/mL. Cells adopted long, thin and spindle shape on Fn(10-50) and PCOL. Parallel actin filaments were observed in MDPC-23 cells cultured on Fn and PCOL. ALP activity was markedly up-regulated on Fn and PCOL-coated surfaces. Importantly, gene expression of BSP (Fn10: 2.44±0.32; Fn20: 3.05±0.01; Fn30: 2.90±0.21; Fn40: 2.74±0.30; Fn50: 2.64±0.12; PCOL: 2.20±0.03) and OCN (Fn10: 2.52±0.23; Fn20: 2.28±0.24; Fn30: 2.34±0.21; Fn40: 2.34±0.25; Fn50: 2.20±0.22; PCOL: 1.56±0.16) was significantly enhanced on Fn and PCOL substrates as compared with control; moreover, expression of integrin beta 1 (ITGB1), an ubiquitous cell surface receptor was augmented in Fn(10-50) and PCOL groups simultaneously. In accordance with the ALP activity and gene expression data, calcific deposition in cells grown on Fn(10-50) and PCOL was observed as well. Conclusion Despite the limitation of this study, the findings indicate that a surface coating of Fn enhances the proliferation, differentiation and mineralization of odontoblast-like cells by activation of integrin beta 1 (ITG B1). The promoting effects of Fn on MDPC-23 cells were achieved at a comparatively lower coating concentration than type I collagen (300 µg/mL). Specifically, it is suggested that the optimum coating concentration of Fn to be 10 µg/mL.
Collapse
Affiliation(s)
- Jia Tang
- Health Sciences University of Hokkaido, School of Dentistry, Department of Oral Rehabilitation, Division of Clinical Cariology and Endodontology, Hokkaido, Japan
| | - Takashi Saito
- Health Sciences University of Hokkaido, School of Dentistry, Department of Oral Rehabilitation, Division of Clinical Cariology and Endodontology, Hokkaido, Japan
| |
Collapse
|
35
|
Kim DK, Kim JI, Hwang TI, Sim BR, Khang G. Bioengineered Osteoinductive Broussonetia kazinoki/Silk Fibroin Composite Scaffolds for Bone Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2017; 9:1384-1394. [PMID: 28001353 DOI: 10.1021/acsami.6b14351] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In this article, Broussonetia kazinoki (BK) powdery extract is utilized to modify the silk fibroin (SF) scaffold and applied to the bone defect area. The BK/SF scaffold is an efficient cell carrier which promotes cell proliferation and osteogenic differentiation of rBMSCs (bone marrow derived mesenchymal stem cells). We confirmed biocompatibility and osteogenic differentiation capacity of BK/SF scaffolds compared to pristine SF scaffold in both in vitro and in vivo evaluation. Gene expression related to osteogenic differentiation and bone regeneration significantly upregulated in the BK/SF scaffold group. The implanted scaffolds were attached well to the surface of the bone defect region and integrated with surrounding tissues without significant inflammatory reaction. Furthermore, almost 45% of bone volume has been recovered at 8 weeks postsurgery, while the SF and control group showed 20% recovery. These results suggest that BK powdery extract incorporated with an SF scaffold might be a suitable substitute for an alternative bone graft for bone regeneration.
Collapse
Affiliation(s)
- Do Kyung Kim
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology and Polymer BIN Research Center, Chonbuk National University , Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Jeong In Kim
- Department of Bionanosystem Engineering, Graduate School, Chonbuk National University , Jeonju 561-756, Republic of Korea
| | - Tae In Hwang
- Department of Bionanosystem Engineering, Graduate School, Chonbuk National University , Jeonju 561-756, Republic of Korea
| | - Bo Ra Sim
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology and Polymer BIN Research Center, Chonbuk National University , Deokjin-gu, Jeonju 561-756, Republic of Korea
| | - Gilson Khang
- Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology and Polymer BIN Research Center, Chonbuk National University , Deokjin-gu, Jeonju 561-756, Republic of Korea
| |
Collapse
|
36
|
Selli C, Erac Y, Tosun M. Effects of cell seeding density on real-time monitoring of anti-proliferative effects of transient gene silencing. ACTA ACUST UNITED AC 2016; 23:20. [PMID: 27981039 PMCID: PMC5133759 DOI: 10.1186/s40709-016-0057-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 11/25/2016] [Indexed: 01/15/2023]
Abstract
Background Real-time cellular analysis systems enable impedance-based label-free and dynamic monitoring of various cellular events such as proliferation. In this study, we describe the effects of initial cell seeding density on the anti-proliferative effects of transient gene silencing monitored via real-time cellular analysis. We monitored the real-time changes in proliferation of Huh7 hepatocellular carcinoma and A7r5 vascular smooth muscle cells with different initial seeding densities following transient receptor potential canonical 1 (TRPC1) silencing using xCELLigence system. Huh7 and A7r5 cells were seeded on E-plate 96 at 10,000, 5000, 1250 and 5000, 2500 cells well−1, respectively, following silencing vector transfection. The inhibitory effects of transient silencing on cell proliferation monitored every 30 min for 72 h. Results TRPC1 silencing did not inhibit the proliferation rates of Huh7 cells at 10,000 cells well−1 seeding density. However, a significant anti-proliferative effect was observed at 1250 cells well−1 density at each time point throughout 72 h. Furthermore, significant inhibitory effects on A7r5 proliferation were observed at both 5000 and 2500 cells well−1 for 72 h. Conclusions Data suggest that the effects of transient silencing on cell proliferation differ depending on the initial cell seeding density. While high seeding densities mask the significant changes in proliferation, the inhibitory effects of silencing become apparent at lower seeding densities as the entry into log phase is delayed. Using the optimal initial seeding density is crucial when studying the effects of transient gene silencing. In addition, the results suggest that TRPC1 may contribute to proliferation and phenotypic switching of vascular smooth muscle cells.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR UK ; Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey
| | - Yasemin Erac
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey
| | - Metiner Tosun
- Department of Pharmacology, Faculty of Pharmacy, Ege University, 35040 Izmir, Turkey ; Faculty of Medicine, Izmir University of Economics, 35330 Izmir, Turkey
| |
Collapse
|
37
|
Najafabadi MM, Bayati V, Orazizadeh M, Hashemitabar M, Absalan F. Impact of Cell Density on Differentiation Efficiency of Rat Adipose-derived Stem Cells into Schwann-like Cells. Int J Stem Cells 2016; 9:213-220. [PMID: 27788569 PMCID: PMC5155717 DOI: 10.15283/ijsc16031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2016] [Indexed: 01/20/2023] Open
Abstract
Background and Objectives Schwann-like (SC-like) cells induced from adipose-derived stem cells (ASCs) may be one of the ideal alternative cell sources for obtaining Schwann cells (SCs). They can be used for treating peripheral nerve injuries. Co-culture with SCs or exposure to glial growth factors are commonly used for differentiation of ASCs to SC-like cells. However, the effect of initial cell density as an inductive factor on the differentiation potential of ASCs into the SC-like cells has not been yet investigated. Methods and Results ASCs were harvested from rat and characterized. The cells were seeded into the culture flasks at three different initial cell densities i.e. 2×103, 4×103 and 8×103 cells/cm2 an overnight and differentiated toward SC-like cells using glial growth factors. After two weeks, the differentiation rate of ASCs to SC-like cells at different densities was assessed by immunofluorescence, fluorescence-activated cell sorting analysis and real time RT-PCR. Expression of the typical SCs markers, S-100 proteins and glial fibrillary acidic protein (GFAP) protein, was observed in all cell densities groups although the number of S100-positive and GFAP-positive cells, and the expression of p75NTR mRNA, another SC marker, were significantly higher at the density of 8×103 cells/cm2 when compared with the other cell densities groups (p<0.001). Conclusions The results suggest that the higher differentiation rate of ASCs to SC-like cells can be obtained at initial cell density of 8×103 cells/cm2, possibly via increased cell-cell interaction and cell density-dependent influence of glial growth factors.
Collapse
Affiliation(s)
- Mahtab Maghzi Najafabadi
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Hashemitabar
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Forouzan Absalan
- Cellular and Molecular Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
38
|
Zhang L, Xu Y, Xu J, Wei Y, Xu X. Protein kinase A inhibitor, H89, significantly enhances survival rate of dissociated human embryonic stem cells following cryopreservation. Cell Prolif 2016; 49:589-98. [PMID: 27484641 DOI: 10.1111/cpr.12278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 07/01/2016] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Human embryonic stem cells (hESCs) have huge potential for establishment of disease models and for treating degenerative diseases. However, the extremely low survival level of dissociated hESCs following cryopreservation is been a tremendous problem to allow for their rapid expansion, genetic manipulation and future medical applications. In this study, we have aimed to develop an efficient strategy to improve survival of dissociated hESCs after cryopreservation. MATERIALS AND METHODS Human embryonic stem cells (H9 line), dissociated into single cells, were cryopreserved using the slow-freezing method. Viable cells and their colony numbers in culture after cryopreservation were evaluated when treated with protein kinase A inhibitor H89. Western blotting was carried out to investigate mechanisms of low survival levels of dissociated hESCs following cryopreservation. Immunofluorescence, reverse transcription-polymerase chain reaction (RT-PCR), in vitro and in vivo differentiation were performed to testify to pluripotency and differentiation ability of hte cryopreserved cells treated with H89. RESULTS H89 significantly improved survival level of dissociated hESCs after cryopreservation through ROCK inhibition. H89-treated cells still maintained their pluripotency and differentiation capacity. CONCLUSIONS This new approach for cryopreservation of single hESCs, using H89, can promote potential use of hESCs in regenerative medicine in the future.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanqing Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jiandong Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuping Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xia Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
39
|
Current View on Osteogenic Differentiation Potential of Mesenchymal Stromal Cells Derived from Placental Tissues. Stem Cell Rev Rep 2016; 11:570-85. [PMID: 25381565 PMCID: PMC4493719 DOI: 10.1007/s12015-014-9569-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells (MSC) isolated from human term placental tissues possess unique characteristics, including their peculiar immunomodulatory properties and their multilineage differentiation potential. The osteogenic differentiation capacity of placental MSC has been widely disputed, and continues to be an issue of debate. This review will briefly discuss the different MSC populations which can be obtained from different regions of human term placenta, along with their unique properties, focusing specifically on their osteogenic differentiation potential. We will present the strategies used to enhance osteogenic differentiation potential in vitro, such as through the selection of subpopulations more prone to differentiate, the modification of the components of osteo-inductive medium, and even mechanical stimulation. Accordingly, the applications of three-dimensional environments in vitro and in vivo, such as non-synthetic, polymer-based, and ceramic scaffolds, will also be discussed, along with results obtained from pre-clinical studies of placental MSC for the regeneration of bone defects and treatment of bone-related diseases.
Collapse
|
40
|
Marklein RA, Lo Surdo JL, Bellayr IH, Godil SA, Puri RK, Bauer SR. High Content Imaging of Early Morphological Signatures Predicts Long Term Mineralization Capacity of Human Mesenchymal Stem Cells upon Osteogenic Induction. Stem Cells 2016; 34:935-47. [PMID: 26865267 DOI: 10.1002/stem.2322] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/30/2015] [Indexed: 01/05/2023]
Abstract
Human bone marrow-derived multipotent mesenchymal stromal cells, often referred to as mesenchymal stem cells (MSCs), represent an attractive cell source for many regenerative medicine applications due to their potential for multi-lineage differentiation, immunomodulation, and paracrine factor secretion. A major complication for current MSC-based therapies is the lack of well-defined characterization methods that can robustly predict how they will perform in a particular in vitro or in vivo setting. Significant advances have been made with identifying molecular markers of MSC quality and potency using multivariate genomic and proteomic approaches, and more recently with advanced techniques incorporating high content imaging to assess high-dimensional single cell morphological data. We sought to expand upon current methods of high dimensional morphological analysis by investigating whether short term cell and nuclear morphological profiles of MSCs from multiple donors (at multiple passages) correlated with long term mineralization upon osteogenic induction. Using the combined power of automated high content imaging followed by automated image analysis, we demonstrated that MSC morphology after 3 days was highly correlated with 35 day mineralization and comparable to other methods of MSC osteogenesis assessment (such as alkaline phosphatase activity). We then expanded on this initial morphological characterization and identified morphological features that were highly predictive of mineralization capacities (>90% accuracy) of MSCs from additional donors and different manufacturing techniques using linear discriminant analysis. Together, this work thoroughly demonstrates the predictive power of MSC morphology for mineralization capacity and motivates further studies into MSC morphology as a predictive marker for additional in vitro and in vivo responses.
Collapse
Affiliation(s)
- Ross A Marklein
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jessica L Lo Surdo
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ian H Bellayr
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Saniya A Godil
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Raj K Puri
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Cellular and Tissue Therapies Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
41
|
Zhang L, Xu Y, Xu J, Wei Y, Xu X. Protein kinase A inhibitor, H89, enhances survival and clonogenicity of dissociated human embryonic stem cells through Rho-associated coiled-coil containing protein kinase (ROCK) inhibition. Hum Reprod 2016; 31:832-43. [PMID: 26848187 DOI: 10.1093/humrep/dew011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/12/2016] [Indexed: 12/27/2022] Open
Abstract
STUDY QUESTION Can cell survival of dissociated human embryonic stem cells (hESCs) be increased during culture? SUMMARY ANSWER A protein kinase A (PKA) inhibitor, H89, can significantly enhance survival and clonogenicity of dissociated hESCs without affecting their pluripotency. WHAT IS KNOWN ALREADY hESCs are vulnerable to massive cell death upon cellular detachment and dissociation. STUDY DESIGN, SIZE, DURATION hESCs were dissociated into single cells and then cultured in feeder-dependent and -independent manners. H89 was added to the culture medium at different concentrations for 1 day. The statistical results were obtained from at least three independent experiments (n ≥ 4). The group without treatment was used as the negative control. PARTICIPANTS/MATERIALS, SETTING, METHODS 4 µM H89 was added in the culture medium to promote cell survival and colony formation of dissociated hESCs. MTT method and propidium iodide (PI) staining were used to determine cell proliferation, cell death and cell cycle, respectively. To count colony formation, alkaline phosphatase (AP) staining was carried out. Western blot was performed to determine protein expression. Except AP staining, immunofluorescence, RT-PCR and karyotype analysis were used to confirm the pluripotent state of H89 treated hESCs. MAIN RESULTS AND THE ROLE OF CHANCE H89 inhibits the dissociation-induced phosphorylation of PKA and two substrates of Rho-associated coiled-coil containing protein kinase (ROCK), myosin light chain (MLC2) and myosin phosphatase target subunit 1 (MYPT1), significantly increases cell survival and colony formation, and strongly depresses dissociation-induced cell death and cell blebbing without affecting the pluripotency of hESCs and their differentiation in vitro. LIMITATIONS, REASONS FOR CAUTION Appropriate H89 concentration should be used and 1 day of H89 treatment is sufficient for promoting survival and colony formation of dissociated hESCs. WIDER IMPLICATIONS OF THE FINDINGS These results provide an alternative for human pluripotent stem cell (hPSC) culture, broaden the scope of participants in the cell death of single hES cells after dissociation and further enlighten clues to understand the mechanism of dissociation-induced cell death. STUDY FUNDING/COMPETING INTERESTS This research was supported by the National Natural Science Foundation of China (21176238, 21576266), and Chinese Academy of Sciences. There is no conflict of interest to declare. TRIAL REGISTRATION NUMBER Nil.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yanqing Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Jiandong Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Yuping Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Xia Xu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| |
Collapse
|
42
|
Sivolella S, Brunello G, Ferroni L, Berengo M, Meneghello R, Savio G, Piattelli A, Gardin C, Zavan B. A Novel In Vitro Technique for Assessing Dental Implant Osseointegration. Tissue Eng Part C Methods 2016; 22:132-141. [DOI: 10.1089/ten.tec.2015.0158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Stefano Sivolella
- Section of Dentistry, Department of Neurosciences, University of Padova, Padova, Italy
| | - Giulia Brunello
- Section of Dentistry, Department of Neurosciences, University of Padova, Padova, Italy
| | - Letizia Ferroni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Mario Berengo
- Section of Dentistry, Department of Neurosciences, University of Padova, Padova, Italy
| | - Roberto Meneghello
- Department of Management and Engineering, University of Padova, Vicenza, Italy
| | - Gianpaolo Savio
- Department of Civil, Environmental and Architectural Engineering, University of Padova, Padova, Italy
| | - Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Chiara Gardin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Barbara Zavan
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
43
|
Yang B, Zuo Y, Zou Q, Li L, Li J, Man Y, Li Y. Effect of ultrafine poly(ε-caprolactone) fibers on calcium phosphate cement: in vitro degradation and in vivo regeneration. Int J Nanomedicine 2016; 11:163-77. [PMID: 26792992 PMCID: PMC4708242 DOI: 10.2147/ijn.s91596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We incorporated ultrafine polymer fibers into calcium phosphate cement (CPC) to improve the resorption rate of CPC with fiber degradation. Different weight percentages of electrospun poly(ε-caprolactone) fibers (0%, 3%, and 7%, named as ultrafine fiber-incorporated CPC0 [UFICPC0], UFICPC3, and UFICPC7) were included into preset CPC specimens for in vitro immersion in lipase phosphate-buffered solution and long-term in vivo implantation in the femoral condyle of rabbits. The effect of the ultrafine poly(ε-caprolactone) fibers with a diameter ranging from nanometer to micrometer on CPC degradation was evaluated by measuring the pH of the medium, mass loss, porosity, and physiochemical properties. For the in vivo evaluation, histomorphometrical analysis as well as three-dimensional (3D) reconstruction was applied to assess the osteogenic properties of the CPC composite. After in vitro immersion and in vivo implantation, the total porosity and macroporosity as well as the bone formation and ingrowth increased significantly during time in the fiber-incorporated CPC specimens. After 24 weeks of implantation, the degraded space was occupied by newly formed bone, and the UFICPC3 and UFICPC7 composites showed ~3.5 times higher fraction of bone volume than that of the pristine CPC (UFICPC0). In vitro and in vivo results proved that the introduction of ultrafine degradable fibers within a CPC matrix can be used to improve macroporosity efficiently and enhance CPC degradation and bone ingrowth largely.
Collapse
Affiliation(s)
- Boyuan Yang
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| | - Yi Zuo
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| | - Qin Zou
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| | - Limei Li
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| | - Jidong Li
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| | - Yi Man
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, People’s Republic of China
| | - Yubao Li
- Research Center for Nano Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
44
|
Wang P, Song Y, Weir MD, Sun J, Zhao L, Simon CG, Xu HHK. A self-setting iPSMSC-alginate-calcium phosphate paste for bone tissue engineering. Dent Mater 2015; 32:252-63. [PMID: 26743965 DOI: 10.1016/j.dental.2015.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/07/2015] [Accepted: 11/30/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Calcium phosphate cements (CPCs) are promising for dental and craniofacial repairs. The objectives of this study were to: (1) develop an injectable cell delivery system based on encapsulation of induced pluripotent stem cell-derived mesenchymal stem cells (iPSMSCs) in microbeads; (2) develop a novel tissue engineered construct by dispersing iPSMSC-microbeads in CPC to investigate bone regeneration in an animal model for the first time. METHODS iPSMSCs were pre-osteoinduced for 2 weeks (OS-iPSMSCs), or transduced with bone morphogenetic protein-2 (BMP2-iPSMSCs). Cells were encapsulated in fast-degradable alginate microbeads. Microbeads were mixed with CPC paste and filled into cranial defects in nude rats. Four groups were tested: (1) CPC-microbeads without cells (CPC control); (2) CPC-microbeads-iPSMSCs (CPC-iPSMSCs); (3) CPC-microbeads-OS-iPSMSCs (CPC-OS-iPSMSCs); (4) CPC-microbeads-BMP2-iPSMSCs (CPC-BMP2-iPSMSCs). RESULTS Cells maintained good viability inside microbeads after injection. The microbeads were able to release the cells which had more than 10-fold increase in live cell density from 1 to 14 days. The cells exhibited up-regulation of osteogenic markers and deposition of minerals. In vivo, new bone area fraction (mean±SD; n=5) for CPC-iPSMSCs group was (22.5±7.6)%. New bone area fractions were (38.9±18.4)% and (44.7±22.8)% for CPC-OS-iPSMSCs group and CPC-BMP2-iPSMSCs group, respectively, 2-3 times the (15.6±11.2)% in CPC control at 12 weeks (p<0.05). Cell-CPC constructs accelerated scaffold resorption, with CPC-BMP2-iPSMSCs having remaining scaffold material that was 7-fold less than CPC control. SIGNIFICANCE Novel injectable CPC-microbead-cell constructs promoted bone regeneration, with OS-iPSMSCs and BMP2-iPSMSCs having 2-3 fold the new bone of CPC control. Cell delivery accelerated scaffold resorption, with CPC-BMP2-iPSMSC having remaining scaffold material that was 7-fold less than CPC control. Therefore, CPC-microbead-iPSMSC is a promising injectable material for orthopedic, dental and craniofacial bone regenerations.
Collapse
Affiliation(s)
- Ping Wang
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Song
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jinyu Sun
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Liang Zhao
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Carl G Simon
- Biosystems and Biomaterials Division, National Institute of Standards & Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Endodontics, Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Mechanical Engineering Department, University of Maryland Baltimore County, Baltimore County, MD 21250, USA.
| |
Collapse
|
45
|
Léotot J, Lebouvier A, Hernigou P, Bierling P, Rouard H, Chevallier N. Bone-Forming Capacity and Biodistribution of Bone Marrow-Derived Stromal Cells Directly Loaded into Scaffolds: A Novel and Easy Approach for Clinical Application of Bone Regeneration. Cell Transplant 2015; 24:1945-55. [DOI: 10.3727/096368914x685276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the context of clinical applications of bone regeneration, cell seeding into scaffolds needs to be safe and easy. Moreover, cell density also plays a crucial role in the development of efficient bone tissue engineering constructs. The aim of this study was to develop and evaluate a simple and rapid cell seeding procedure on hydroxyapatite/ β-tricalcium phosphate (HA/βTCP), as well as define optimal cell density and control the biodistribution of grafted cells. To this end, human bone marrow-derived stromal cells (hBMSCs) were seeded on HA/βTCP scaffolds, and we have compared bone formation using an ectopic model. Our results demonstrated a significantly higher bone-forming capacity of hBMSCs directly loaded on HA/βTCP during surgery compared to hBMSCs preseeded for 7 days in vitro on HA/βTCP before ectopic implantation. The extent of new bone formation increases with increasing hBMSC densities quantitatively, qualitatively, and in frequency. Also, this study showed that grafted hBMSCs remained confined to the implantation site and did not spread toward other tissues, such as liver, spleen, lungs, heart, and kidneys. In conclusion, direct cell loading into a scaffold during surgery is more efficient for bone regeneration, as well as quick and safe. Therefore direct cell loading is suitable for clinical requirements and cell production control, making it a promising approach for orthopedic applications. Moreover, our results have provided evidence that the formation of a mature bone organ containing hematopoietic islets needs a sufficiently high local density of grafted hBMSCs, which should guide the optimal dose of cells for clinical use.
Collapse
Affiliation(s)
- Julie Léotot
- Université Paris-Est Créteil, Faculté de médecine, Laboratoire de “Bioingénierie Cellulaire, Tissulaire et Sanguine,” Créteil, France
- Etablissement Français du Sang d'Ile-de-France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
| | - Angélique Lebouvier
- Université Paris-Est Créteil, Faculté de médecine, Laboratoire de “Bioingénierie Cellulaire, Tissulaire et Sanguine,” Créteil, France
- Etablissement Français du Sang d'Ile-de-France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
| | - Philippe Hernigou
- Université Paris-Est Créteil, Faculté de médecine, Laboratoire de “Bioingénierie Cellulaire, Tissulaire et Sanguine,” Créteil, France
- Service de Chirurgie Orthopédique et Traumatologique, AP-HP Hôpital Henri-Mondor, Créteil, France
| | - Philippe Bierling
- Etablissement Français du Sang d'Ile-de-France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
- INSERM UMR955, Paris-Est University, Créteil, France
| | - Hélène Rouard
- Université Paris-Est Créteil, Faculté de médecine, Laboratoire de “Bioingénierie Cellulaire, Tissulaire et Sanguine,” Créteil, France
- Etablissement Français du Sang d'Ile-de-France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
- AP-HP Hôpital Henri-Mondor – A. Chenevier, Service Hospitalier, Créteil, France
| | - Nathalie Chevallier
- Université Paris-Est Créteil, Faculté de médecine, Laboratoire de “Bioingénierie Cellulaire, Tissulaire et Sanguine,” Créteil, France
- Etablissement Français du Sang d'Ile-de-France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
| |
Collapse
|
46
|
Yassin MA, Leknes KN, Pedersen TO, Xing Z, Sun Y, Lie SA, Finne-Wistrand A, Mustafa K. Cell seeding density is a critical determinant for copolymer scaffolds-induced bone regeneration. J Biomed Mater Res A 2015; 103:3649-58. [PMID: 26013960 PMCID: PMC4744655 DOI: 10.1002/jbm.a.35505] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/15/2022]
Abstract
Constructs intended for bone tissue engineering (TE) are influenced by the initial cell seeding density. Therefore, the objective of this study was to determine the effect of bone marrow stromal stem cells (BMSCs) density loaded onto copolymer scaffolds on bone regeneration. BMSCs were harvested from rat's bone marrow and cultured in media with or without osteogenic supplements. Cells were seeded onto poly(l‐lactide‐co‐ε‐caprolactone) [poly(LLA‐co‐CL)] scaffolds at two different densities: low density (1 × 106 cells/scaffold) or high density (2 × 106 cells/scaffold) using spinner modified flasks and examined after 1 and 3 weeks. Initial attachment and spread of BMSC onto the scaffolds was recorded by scanning electron microscopy. Cell proliferation was assessed by DNA quantification and cell differentiation by quantitative real‐time reverse transcriptase‐polymerized chain reaction analysis (qRT‐PCR). Five‐millimeter rat calvarial defects (24 defects in 12 rats) were implanted with scaffolds seeded with either low or high density expanded with or without osteogenic supplements. Osteogenic supplements significantly increased cell proliferation (p < 0.001). Scaffolds seeded at high cell density exhibited higher mRNA expressions of Runx2 p = 0.001, Col1 p = 0.001, BMP2 p < 0.001, BSP p < 0.001, and OC p = 0.013. More bone was formed in response to high cell seeding density (p = 0.023) and high seeding density with osteogenic medium (p = 0.038). Poly (LLA‐co‐CL) scaffolds could be appropriate candidates for bone TE. The optimal number of cells to be loaded onto scaffolds is critical for promoting Extracellular matrix synthesis and bone formation. Cell seeding density and osteogenic supplements may have a synergistic effect on the induction of new bone. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 103A: 3649–3658, 2015.
Collapse
Affiliation(s)
- Mohammed A Yassin
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Knut N Leknes
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Torbjorn O Pedersen
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Zhe Xing
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Yang Sun
- Department of Fibre and Polymer Technology, School of Chemical Science and Engineering, KTH Royal Institute of Technology, Teknikringen 42, SE-100 44, Stockholm, Sweden
| | - Stein A Lie
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, School of Chemical Science and Engineering, KTH Royal Institute of Technology, Teknikringen 42, SE-100 44, Stockholm, Sweden
| | - Kamal Mustafa
- Faculty of Medicine and Dentistry, Department of Clinical Dentistry, Center for Clinical Dental Research, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| |
Collapse
|
47
|
Wang P, Liu X, Zhao L, Weir MD, Sun J, Chen W, Man Y, Xu HHK. Bone tissue engineering via human induced pluripotent, umbilical cord and bone marrow mesenchymal stem cells in rat cranium. Acta Biomater 2015; 18:236-48. [PMID: 25712391 DOI: 10.1016/j.actbio.2015.02.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 02/03/2015] [Accepted: 02/13/2015] [Indexed: 02/05/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are an exciting cell source with great potential for tissue engineering. Human bone marrow mesenchymal stem cells (hBMSCs) have been used in clinics but are limited by several disadvantages, hence alternative sources of MSCs such as umbilical cord MSCs (hUCMSCs) are being investigated. However, there has been no report comparing hiPSCs, hUCMSCs and hBMSCs for bone regeneration. The objectives of this pilot study were to investigate hiPSCs, hUCMSCs and hBMSCs for bone tissue engineering, and compare their bone regeneration via seeding on biofunctionalized macroporous calcium phosphate cement (CPC) in rat cranial defects. For all three types of cells, approximately 90% of the cells remained alive on CPC scaffolds. Osteogenic genes were up-regulated, and mineral synthesis by cells increased with time in vitro for all three types of cells. The new bone area fractions at 12weeks (mean±sd; n=6) were (30.4±5.8)%, (27.4±9.7)% and (22.6±4.7)% in hiPSC-MSC-CPC, hUCMSC-CPC and hBMSC-CPC respectively, compared to (11.0±6.3)% for control (p<0.05). No significant differences were detected among the three types of stem cells (p>0.1). New blood vessel density was higher in cell-seeded groups than control (p<0.05). De novo bone formation and participation by implanted cells was confirmed via immunohistochemical staining. In conclusion, (1) hiPSCs, hUCMSCs and hBMSCs greatly enhanced bone regeneration, more than doubling the new bone amount of cell-free CPC control; (2) hiPSC-MSCs and hUCMSCs represented viable alternatives to hBMSCs; (3) biofunctionalized macroporous CPC-stem cell constructs had a robust capacity for bone regeneration.
Collapse
Affiliation(s)
- Ping Wang
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xian Liu
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Zhao
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Michael D Weir
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Jirun Sun
- Dr. Anthony Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| | - Wenchuan Chen
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Man
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hockin H K Xu
- Biomaterials & Tissue Engineering Division, Department of Endodontics Prosthodontics and Operative Dentistry, University of Maryland Dental School, Baltimore, MD 21201, USA; Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Mechanical Engineering Department, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
48
|
Perez RA, Shin SH, Han CM, Kim HW. Bioactive injectables based on calcium phosphates for hard tissues: A recent update. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-015-0096-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
49
|
He Z, Zhai Q, Hu M, Cao C, Wang J, Yang H, Li B. Bone cements for percutaneous vertebroplasty and balloon kyphoplasty: Current status and future developments. J Orthop Translat 2015; 3:1-11. [PMID: 30035034 PMCID: PMC5982384 DOI: 10.1016/j.jot.2014.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/16/2014] [Accepted: 11/25/2014] [Indexed: 11/26/2022] Open
Abstract
Osteoporotic vertebral compression fractures (OVCFs) have gradually evolved into a serious health care problem globally. In order to reduce the morbidity of OVCF patients and improve their life quality, two minimally invasive surgery procedures, vertebroplasty (VP) and balloon kyphoplasty (BKP), have been developed. Both VP and BKP require the injection of bone cement into the vertebrae of patients to stabilize fractured vertebra. As such, bone cement as the filling material plays an essential role in the effectiveness of these treatments. In this review article, we summarize the bone cements that are currently available in the market and those still under development. Two major categories of bone cements, nondegradable acrylic bone cements (ABCs) and degradable calcium phosphate cements (CPCs), are introduced in detail. We also provide our perspectives on the future development of bone cements for VP and BKP.
Collapse
Affiliation(s)
- Zhiwei He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qingpan Zhai
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Muli Hu
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Chengbin Cao
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Jihui Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Orthopedic Institute, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
50
|
Liu J, Chen W, Zhao Z, Xu HH. Effect of NELL1 gene overexpression in iPSC-MSCs seeded on calcium phosphate cement. Acta Biomater 2014; 10:5128-5138. [PMID: 25220281 DOI: 10.1016/j.actbio.2014.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/05/2014] [Accepted: 08/15/2014] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) are a promising source of patient-specific stem cells with great regenerative potential. There has been no report on NEL-like protein 1 (NELL1) gene modification of iPSC-MSCs. The objectives of this study were to genetically modify iPSC-MSCs with NELL1 overexpression for bone tissue engineering, and investigate the osteogenic differentiation of NELL1 gene-modified iPSC-MSCs seeded on Arg-Gly-Asp (RGD)-grafted calcium phosphate cement (CPC) scaffold. Cells were transduced with red fluorescence protein (RFP-iPSC-MSCs) or NELL1 (NELL1-iPSC-MSCs) by a lentiviral vector. Cell proliferation on RGD-grafted CPC scaffold, osteogenic differentiation and bone mineral synthesis were evaluated. RFP-iPSC-MSCs stably expressed high levels of RFP. Both the NELL1 gene and NELL1 protein levels were confirmed higher in NELL1-iPSC-MSCs than in RFP-iPSC-MSCs using RT-PCR and Western blot (P<0.05). Alkaline phosphatase activity was increased by 130% by NELL1 overexpression at 14days (P<0.05), indicating that NELL1 promoted iPSC-MSC osteogenic differentiation. When seeded on RGD-grafted CPC, NELL1-iPSC-MSCs attached and expanded similarly well to RFP-iPSC-MSCs. At 14days, the runt-related transcription factor 2 (RUNX2) gene level of NELL1-iPSC-MSCs was 2.0-fold that of RFP-iPSC-MSCs. The osteocalcin (OC) level of NELL1-iPSC-MSCs was 3.1-fold that of RFP-iPSC-MSCs (P<0.05). The collagen type I alpha 1 (COL1A1) gene level of NELL1-iPSC-MSCs was 1.7-fold that of RFP-iPSC-MSCs at 7days (P<0.05). Mineral synthesis was increased by 81% in NELL1-iPSC-MSCs at 21days. In conclusion, NELL1 overexpression greatly enhanced the osteogenic differentiation and mineral synthesis of iPSC-MSCs on RGD-grafted CPC scaffold for the first time. The novel NELL1-iPSC-MSC seeded RGD-CPC construct is promising for enhancing bone engineering.
Collapse
|