201
|
Lam J, Bellayr IH, Marklein RA, Bauer SR, Puri RK, Sung KE. Functional Profiling of Chondrogenically Induced Multipotent Stromal Cell Aggregates Reveals Transcriptomic and Emergent Morphological Phenotypes Predictive of Differentiation Capacity. Stem Cells Transl Med 2018; 7:664-675. [PMID: 30084545 PMCID: PMC6127231 DOI: 10.1002/sctm.18-0065] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/07/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Multipotent stromal cells (MSCs) are an attractive cell source for bone and cartilage tissue repair strategies. However, the functional heterogeneity of MSCs derived from different donors and manufacturing conditions has limited clinical translation, emphasizing the need for improved methods to assess MSC chondrogenic capacity. We used functionally relevant morphological profiling to dynamically monitor emergent morphological phenotypes of chondrogenically induced MSC aggregates to identify morphological features indicative of MSC chondrogenesis. Toward this goal, we characterized the morphology of chondrogenically stimulated MSC aggregates from eight different human cell-lines at multiple passages and demonstrated that MSC aggregates exhibited unique morphological dynamics that were both cell line- and passage-dependent. This variation in 3D morphology was shown to be informative of long-term MSC chondrogenesis based on multiple quantitative functional assays. We found that the specific morphological features of spheroid area, radius, minimum feret diameter, and minor axis length to be strongly correlated with MSC chondrogenic synthetic activity but not gene expression as early as day 4 in 3D culture. Our high-throughput, nondestructive approach could potentially serve as a tool to identify MSC lines with desired chondrogenic capacity toward improving manufacturing strategies for MSC-based cellular products for cartilage tissue repair. Stem Cells Translational Medicine 2018;1-12.
Collapse
Affiliation(s)
- Johnny Lam
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ian H Bellayr
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Raj K Puri
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kyung E Sung
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
202
|
Kim M, Yun HW, Park DY, Choi BH, Min BH. Three-Dimensional Spheroid Culture Increases Exosome Secretion from Mesenchymal Stem Cells. Tissue Eng Regen Med 2018; 15:427-436. [PMID: 30603566 PMCID: PMC6171656 DOI: 10.1007/s13770-018-0139-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/22/2018] [Accepted: 06/24/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Mass production of exosomes is a prerequisite for their commercial utilization. This study investigated whether three-dimensional (3D) spheroid culture of mesenchymal stem cells (MSCs) could improve the production efficiency of exosomes and if so, what was the mechanism involved. METHODS We adopted two models of 3D spheroid culture using the hanging-drop (3D-HD) and poly(2-hydroxyethyl methacrylate) (poly-HEMA) coating methods (3D-PH). The efficiency of exosome production from MSCs in the 3D spheroids was compared with that of monolayer culture in various conditions. We then investigated the mechanism of the 3D spheroid culture-induced increase in exosome production. RESULTS The 3D-HD formed a single larger spheroid, while the 3D-PH formed multiple smaller ones. However, MSCs cultured on both types of spheroids produced significantly more exosomes than those cultured in conventional monolayer culture (2D). We then investigated the cause of the increased exosome production in terms of hypoxia within the 3D spheroids, high cell density, and non-adherent cell morphology. With increasing spheroid size, the efficiency of exosome production was the largest with the least amount of cells in both 3D-HD and 3D-PH. An increase in cell density in 2D culture (2D-H) was less efficient in exosome production than the conventional, lower cell density, 2D culture. Finally, when cells were plated at normal density on the poly-HEMA coated spheroids (3D-N-PH); they formed small aggregates of less than 10 cells and still produced more exosomes than those in the 2D culture when plated at the same density. We also found that the expression of F-actin was markedly reduced in the 3D-N-PH culture. CONCLUSION These results suggested that 3D spheroid culture produces more exosomes than 2D culture and the non-adherent round cell morphology itself might be a causative factor. The result of the present study could provide useful information to develop an optimal process for the mass production of exosomes.
Collapse
Affiliation(s)
- Mijin Kim
- Department of Molecular Sciences and Technology, Ajou University, 206, World cup-ro, Yeongtong-gu, Woncheon-dong, Suwon, 16499 Republic of Korea
- Cell Therapy Center, Ajou University Medical Center, 206 World Cup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| | - Hee-Woong Yun
- Department of Molecular Sciences and Technology, Ajou University, 206, World cup-ro, Yeongtong-gu, Woncheon-dong, Suwon, 16499 Republic of Korea
- Cell Therapy Center, Ajou University Medical Center, 206 World Cup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| | - Do Young Park
- Department of Orthopedic Surgery, School of Medicine, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100 Inha-ro, Nam-gu, Incheon, 22212 Republic of Korea
| | - Byoung-Hyun Min
- Department of Molecular Sciences and Technology, Ajou University, 206, World cup-ro, Yeongtong-gu, Woncheon-dong, Suwon, 16499 Republic of Korea
- Cell Therapy Center, Ajou University Medical Center, 206 World Cup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
- Department of Orthopedic Surgery, School of Medicine, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| |
Collapse
|
203
|
Kim E, Kim JC, Min K, Goh M, Tae G. Rapid and Versatile Cell Aggregate Formation Using Lipid-Conjugated Heparin. ACS APPLIED MATERIALS & INTERFACES 2018; 10:24431-24439. [PMID: 29953811 DOI: 10.1021/acsami.8b07731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cell aggregates hold significant therapeutic promise for in vitro cell analysis, ex vivo tissue models, and in vivo cell therapy and tissue engineering. Traditional methods of making cell aggregates require long incubation times and can only produce three-dimensional-spheroid-shaped aggregates. We propose a novel method of making cell aggregates of diverse sizes and shapes using lipid-conjugated heparin. Shaking the cell suspension containing a small amount of lipid-conjugated heparin for approximately 30 min produced cell aggregates. This approach can be applied to any cell type, including stem cells, fibroblast cells, and T lymphocytes. The shape of biocompatible templates could modulate the shape of cell aggregates. In addition to layered, multicompartmental cell aggregates on template, template-free, tube-shaped cell aggregates could also be made. The cell aggregates formed were alive and maintained biological activities.
Collapse
Affiliation(s)
- Eunsol Kim
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , 123 Cheomdan-gwagiro , Buk-gu, Gwangju 61005 , Republic of Korea
| | - Jong Chul Kim
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , 123 Cheomdan-gwagiro , Buk-gu, Gwangju 61005 , Republic of Korea
| | - Kiyoon Min
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , 123 Cheomdan-gwagiro , Buk-gu, Gwangju 61005 , Republic of Korea
| | - MeeiChyn Goh
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , 123 Cheomdan-gwagiro , Buk-gu, Gwangju 61005 , Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering , Gwangju Institute of Science and Technology , 123 Cheomdan-gwagiro , Buk-gu, Gwangju 61005 , Republic of Korea
| |
Collapse
|
204
|
Yan L, Jiang B, Li E, Wang X, Ling Q, Zheng D, Park JW, Chen X, Cheung E, Du X, Li Y, Cheng G, He E, Xu RH. Scalable Generation of Mesenchymal Stem Cells from Human Embryonic Stem Cells in 3D. Int J Biol Sci 2018; 14:1196-1210. [PMID: 30123069 PMCID: PMC6097489 DOI: 10.7150/ijbs.25023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
Human embryonic stem cell (hESC) derived mesenchymal stem cells (EMSC) are efficacious in treating a series of autoimmune, inflammatory, and degenerative diseases in animal models. However, all the EMSC derivation methods reported so far rely on two-dimensional (2D) culture systems, which are inefficient, costive and difficult for large-scale production. HESC, as an unlimited source, can be successively propagated in spheroids. Here, we demonstrate that hESC spheroids can directly differentiate into MSC spheroids (EMSCSp) within 20 days in one vessel without passaging and the system is scalable to any desired size. EMSCSp can further differentiate into osteocytes and chondrocytes in spheres or demineralized bone matrix. EMSCSp also retains immune-modulatory effects in vitro and therapeutic effects on two mouse models of colitis after dissociation. Compared to EMSC differentiated in monolayer, EMSCSp-derived cells have faster proliferation and higher yield and develop less apoptosis and slower senescence. Thus, the 3D differentiation system allows simple, cost-effective, and scalable production of high-quality EMSC and subsequently bone and cartilage tissues for therapeutic application.
Collapse
Affiliation(s)
- Li Yan
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Bin Jiang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Enqin Li
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xiaoyan Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Qinjie Ling
- Department of Orthopedics, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejin Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jung Woo Park
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xin Chen
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Edwin Cheung
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xin Du
- Department of Hematology, Guangdong General Hospital, Guangzhou, Guangdong, China
| | - Yingcui Li
- Department of Biology, University of Hartford, West Hartford, Connecticut, USA
| | - Gregory Cheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Erxing He
- Department of Orthopedics, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ren-He Xu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
205
|
Egger D, Tripisciano C, Weber V, Dominici M, Kasper C. Dynamic Cultivation of Mesenchymal Stem Cell Aggregates. Bioengineering (Basel) 2018; 5:E48. [PMID: 29921755 PMCID: PMC6026937 DOI: 10.3390/bioengineering5020048] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/24/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered as primary candidates for cell-based therapies due to their multiple effects in regenerative medicine. Pre-conditioning of MSCs under physiological conditions—such as hypoxia, three-dimensional environments, and dynamic cultivation—prior to transplantation proved to optimize their therapeutic efficiency. When cultivated as three-dimensional aggregates or spheroids, MSCs display increased angiogenic, anti-inflammatory, and immunomodulatory effects as well as improved stemness and survival rates after transplantation, and cultivation under dynamic conditions can increase their viability, proliferation, and paracrine effects, alike. Only few studies reported to date, however, have utilized dynamic conditions for three-dimensional aggregate cultivation of MSCs. Still, the integration of dynamic bioreactor systems, such as spinner flasks or stirred tank reactors might pave the way for a robust, scalable bulk expansion of MSC aggregates or MSC-derived extracellular vesicles. This review summarizes recent insights into the therapeutic potential of MSC aggregate cultivation and focuses on dynamic generation and cultivation techniques of MSC aggregates.
Collapse
Affiliation(s)
- Dominik Egger
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| | - Carla Tripisciano
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria.
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, 3500 Krems, Austria.
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Via Università 4, 41121 Modena, Italy.
- Technopole of Mirandola TPM, 41037 Mirandola, Modena, Italy.
| | - Cornelia Kasper
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
206
|
Aijaz A, Li M, Smith D, Khong D, LeBlon C, Fenton OS, Olabisi RM, Libutti S, Tischfield J, Maus MV, Deans R, Barcia RN, Anderson DG, Ritz J, Preti R, Parekkadan B. Biomanufacturing for clinically advanced cell therapies. Nat Biomed Eng 2018; 2:362-376. [PMID: 31011198 PMCID: PMC6594100 DOI: 10.1038/s41551-018-0246-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
The achievements of cell-based therapeutics have galvanized efforts to bring cell therapies to the market. To address the demands of the clinical and eventual commercial-scale production of cells, and with the increasing generation of large clinical datasets from chimeric antigen receptor T-cell immunotherapy, from transplants of engineered haematopoietic stem cells and from other promising cell therapies, an emphasis on biomanufacturing requirements becomes necessary. Robust infrastructure should address current limitations in cell harvesting, expansion, manipulation, purification, preservation and formulation, ultimately leading to successful therapy administration to patients at an acceptable cost. In this Review, we highlight case examples of cutting-edge bioprocessing technologies that improve biomanufacturing efficiency for cell therapies approaching clinical use.
Collapse
Affiliation(s)
- Ayesha Aijaz
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Matthew Li
- Department of Surgery, Center for Surgery, Innovation, and Bioengineering, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, USA
| | - David Smith
- Hitachi Chemical Advanced Therapeutics Solutions, Allendale, NJ, USA
| | - Danika Khong
- Department of Surgery, Center for Surgery, Innovation, and Bioengineering, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, USA
| | - Courtney LeBlon
- Hitachi Chemical Advanced Therapeutics Solutions, Allendale, NJ, USA
| | - Owen S Fenton
- Department of Chemical Engineering, Institute for Medical Engineering and Science, Division of Health Science and Technology, and the David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ronke M Olabisi
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | | | - Jay Tischfield
- Human Genetics Institute of New Jersey, RUCDR, Piscataway, NJ, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | | | | | - Daniel G Anderson
- Department of Chemical Engineering, Institute for Medical Engineering and Science, Division of Health Science and Technology, and the David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jerome Ritz
- Cell Manipulation Core Facility, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Robert Preti
- Hitachi Chemical Advanced Therapeutics Solutions, Allendale, NJ, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
- Department of Surgery, Center for Surgery, Innovation, and Bioengineering, Massachusetts General Hospital, Harvard Medical School and Shriners Hospitals for Children, Boston, MA, USA.
- Sentien Biotechnologies, Inc, Lexington, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
207
|
Candiello J, Grandhi TSP, Goh SK, Vaidya V, Lemmon-Kishi M, Eliato KR, Ros R, Kumta PN, Rege K, Banerjee I. 3D heterogeneous islet organoid generation from human embryonic stem cells using a novel engineered hydrogel platform. Biomaterials 2018; 177:27-39. [PMID: 29883914 DOI: 10.1016/j.biomaterials.2018.05.031] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 01/05/2023]
Abstract
Organoids, which exhibit spontaneous organ specific organization, function, and multi-cellular complexity, are in essence the in vitro reproduction of specific in vivo organ systems. Recent work has demonstrated human pluripotent stem cells (hPSCs) as a viable regenerative cell source for tissue-specific organoid engineering. This is especially relevant for engineering islet organoids, due to the recent advances in generating functional beta-like cells from human pluripotent stem cells. In this study, we report specific engineering of regenerative islet organoids of precise size and cellular heterogeneity, using a novel hydrogel system, Amikagel. Amikagel facilitated controlled and spontaneous aggregation of human embryonic stem cell derived pancreatic progenitor cells (hESC-PP) into robust homogeneous spheroids. This platform further allowed fine control over the integration of multiple cell populations to produce heterogeneous spheroids, which is a necessity for complex organoid engineering. Amikagel induced hESC-PP spheroid formation enhanced pancreatic islet-specific Pdx-1 and NKX6.1 gene and protein expression, while also increasing the percentage of committed population. hESC-PP spheroids were further induced towards mature beta-like cells which demonstrated increased Beta-cell specific INS1 gene and C-peptide protein expression along with functional insulin production in response to in vitro glucose challenge. Further integration of hESC-PP with biologically relevant supporting endothelial cells resulted in multicellular organoids which demonstrated spontaneous maturation towards islet-specific INS1 gene and C-peptide protein expression along with a significantly developed extracellular matrix support system. These findings establish Amikagel -facilitated platform ideal for islet organoid engineering.
Collapse
Affiliation(s)
- Joseph Candiello
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | | | - Saik Kia Goh
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | - Vimal Vaidya
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States
| | - Maya Lemmon-Kishi
- Department of Bioengineering, University of Pittsburgh, PA, United States
| | - Kiarash Rahmani Eliato
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Robert Ros
- Department of Physics, Center for Biological Physics, and Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Prashant N Kumta
- Department of Bioengineering, University of Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States; Department of Mechanical Engineering and Material Science, University of Pittsburgh, PA, United States; Center for Complex Engineered Multifunctional Materials, University of Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, United States
| | - Kaushal Rege
- Chemical Engineering, Arizona State University, Tempe, AZ, United States
| | - Ipsita Banerjee
- Department of Bioengineering, University of Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA, United States.
| |
Collapse
|
208
|
Lafuente MT, Alférez F, González-Candelas L. Light-emitting Diode Blue Light Alters the Ability ofPenicillium digitatumto Infect Citrus Fruits. Photochem Photobiol 2018; 94:1003-1009. [DOI: 10.1111/php.12929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022]
Affiliation(s)
- María T. Lafuente
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC); Consejo Superior de Investigaciones Científicas; Paterna-Valencia Spain
| | - Fernando Alférez
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC); Consejo Superior de Investigaciones Científicas; Paterna-Valencia Spain
| | - Luis González-Candelas
- Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC); Consejo Superior de Investigaciones Científicas; Paterna-Valencia Spain
| |
Collapse
|
209
|
Zhou Q, Li SL, Ma YJ, de Tal V, Li W, Zhao YH. Detection of Repair of the Zone of Calcified Cartilage with Osteoarthritis through Mesenchymal Stem Cells by Ultrashort Echo Time Magnetic Resonance Imaging. Chin Med J (Engl) 2018; 131:1092-1098. [PMID: 29451138 PMCID: PMC5937319 DOI: 10.4103/0366-6999.224725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Objective: Currently, magnetic resonance imaging (MRI) is the most commonly used imaging modality for observing the growth and development of mesenchymal stem cells (MSCs) after in vivo transplantation to treat osteoarthritis (OA). However, it is a challenge to accurately monitor the treatment effects of MSCs in the zone of calcified cartilage (ZCC) with OA. This is especially true in the physiological and biochemical views that are not accurately detected by MRI contrast agents. In contrast, ultrashort time echo (UTE) MRI has been shown to be sensitive to the presence of the ZCC, creating the potential for more effectively observing the repair of the ZCC in OA by MSCs. A special focus is given to the outlook of the use of UTE MRI to detect repair of the ZCC with OA through MSCs. The limitations of the current techniques for clinical applications and future directions are also discussed. Data Sources: Using the combined keywords: “osteoarthritis”, “mesenchymal stem cells”, “calcified cartilage”, and “magnetic resonance imaging”, the PubMed/MEDLINE literature search was conducted up to June 1, 2017. Study Selection: A total of 132 published articles were initially identified citations. Of the 132 articles, 48 articles were selected after further detailed review. This study referred to all the important English literature in full. Results: In contrast, UTE MRI has been shown to be sensitive to the presence of the ZCC, creating the potential for more effectively observing the repair of the ZCC in OA by MSCs. Conclusions: The current studies showed that the ZCC could be described in terms of its histomorphology and biochemistry by UTE MRI. We prospected that UTE MRI has been shown the potential for more effectively observing the repair of the ZCC in OA by MSCs in vivo.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, Guangdong 510630, China
| | - Shao-Lin Li
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, Guangdong 510630; Department of Medical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Ya-Jun Ma
- Department of Radiology, University of California, San Diego, CA 92103-8226, USA
| | - Vicki de Tal
- Department of Radiology, University of California, San Diego, CA 92103-8226, USA
| | - Wei Li
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, Guangdong 510630, China
| | - Ying-Hua Zhao
- Department of Radiology, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Guangzhou, Guangdong 510630, China
| |
Collapse
|
210
|
Abbasi F, Ghanian MH, Baharvand H, Vahidi B, Eslaminejad MB. Engineering mesenchymal stem cell spheroids by incorporation of mechanoregulator microparticles. J Mech Behav Biomed Mater 2018; 84:74-87. [PMID: 29751274 DOI: 10.1016/j.jmbbm.2018.04.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 11/19/2022]
Abstract
Mechanical forces throughout human mesenchymal stem cell (hMSC) spheroids (mesenspheres) play a predominant role in determining cellular functions of cell growth, proliferation, and differentiation through mechanotransductional mechanisms. Here, we introduce microparticle (MP) incorporation as a mechanical intervention method to alter tensional homeostasis of the mesensphere and explore MSC differentiation in response to MP stiffness. The microparticulate mechanoregulators with different elastic modulus (34 kPa, 0.6 MPa, and 2.2 MPa) were prepared by controlled crosslinking cell-sized microdroplets of polydimethylsiloxane (PDMS). Preparation of MP-MSC composite spheroids enabled us to study the possible effects of MPs through experimental and computational assays. Our results showed that MP incorporation selectively primed MSCs toward osteogenesis, yet hindered adipogenesis. Interestingly, this behavior depended on MP mechanics, as the spheroids that contained MPs with intermediate stiffness behaved similar to control MP-free mesenspheres with more tendencies toward chondrogenesis. However, by using the soft or stiff MPs, the MP-mesenspheres significantly showed signs of osteogenesis. This could be explained by the complex of forces which acted in the cell spheroid and, totally, provided a homeostasis situation. Incorporation of cell-sized polymer MPs as mechanoregulators of cell spheroids could be utilized as a new engineering toolkit for multicellular organoids in disease modeling and tissue engineering applications.
Collapse
Affiliation(s)
- Fatemeh Abbasi
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran; Department of Cell Engineering, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Bahman Vahidi
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
211
|
Park MJ, Lee J, Byeon JS, Jeong DU, Gu NY, Cho IS, Cha SH. Effects of three-dimensional spheroid culture on equine mesenchymal stem cell plasticity. Vet Res Commun 2018; 42:171-181. [PMID: 29721754 DOI: 10.1007/s11259-018-9720-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 04/02/2018] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) are useful candidates for tissue engineering and cell therapy fields. We optimize culture conditions of equine adipose tissue-derived MSCs (eAD-MSCs) for treatment of horse fractures. To investigate enhancing properties of three-dimensional (3D) culture system in eAD-MSCs, we performed various sized spheroid formation and determined changes in gene expression levels to obtain different sized spheroid for cell therapy. eAD-MSCs were successfully isolated from horse tailhead. Using hanging drop method, spheroid formation was generated for three days. Quantitative real-time PCR was performed to analyze gene expression. As results, expression levels of pluripotent markers were increased depending on spheroid size and the production of PGE2 was increased in spheroid formation compared to that in monolayer. Ki-67 showed a remarkable increase in the spheroid formed with 2.0 × 105 cells/drop as compared to that in the monolayer. Expression levels of angiogenesis-inducing factors such as VEGF, IL-6, IL-8, and IL-18 were significantly increased in spheroid formation compared to those in the monolayer. Expression levels of bone morphogenesis-inducing factors such as Cox-2 and TGF-β1 were also significantly increased in spheroid formation compared to those in the monolayer. Expression levels of osteocyte-specific markers such as RUNX2, osteocalcin, and differentiation potential were also significantly increased in spheroid formation compared to those in the monolayer. Therefore, spheroid formation of eAD-MSCs through the hanging drop method can increases the expression of angiogenesis-inducing and bone morphogenesis-inducing factors under optimal culture conditions.
Collapse
Affiliation(s)
- Mi Jeong Park
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jeong Su Byeon
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Da-Un Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - In-Soo Cho
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Sang-Ho Cha
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea.
| |
Collapse
|
212
|
Joshi J, Mahajan G, Kothapalli CR. Three-dimensional collagenous niche and azacytidine selectively promote time-dependent cardiomyogenesis from human bone marrow-derived MSC spheroids. Biotechnol Bioeng 2018; 115:2013-2026. [PMID: 29665002 DOI: 10.1002/bit.26714] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/16/2018] [Accepted: 04/09/2018] [Indexed: 12/22/2022]
Abstract
Endogenous adult cardiac regenerative machinery is not capable of replacing the lost cells following myocardial infarction, often leading to permanent alterations in structure-function-mechanical properties. Regenerative therapies based on delivering autologous stem cells within an appropriate 3D milieu could meet such demand, by enabling homing and directed differentiation of the transplanted cells into lost specialized cell populations. Since type I collagen is the predominant cardiac tissue matrix protein, we here optimized the 3D niche which could promote time-dependent evolution of cardiomyogenesis from human bone marrow-derived mesenchymal stem cells (BM-MSC). 3D collagen gel physical and mechanical characteristics were assessed using SEM and AFM, respectively, while the standalone and combined effects of collagen concentration, culture duration, and 5-azacytidine (aza) dose on the phenotype and genotype of MSC spheroids were quantified using immunofluorescence labeling and RT-PCR analysis. Increasing collagen concentration led to a significant increase in Young's modulus (p < 0.01) but simultaneous decrease in the mean pore size, resulting in stiffer gels. Spheroid formation significantly modulated MSC differentiation and genotype, mostly due to better cell-cell interactions. Among the aza dosages tested, 10 μM appears to be optimal, while 3 mg/ml gels resulted in significantly lower cell viability compared to 1 or 2 mg/ml gels. Stiffer gels (2 and 3 mg/ml) and exposure to 10 μM aza upregulated early and late cardiac marker expressions in a time-dependent fashion. On the other hand, cell-cell signaling within the MSC spheroids seem to have a strong role in influencing mature cardiac markers expression, since neither aza nor gel stiffness seem to significantly improve their expression. Western blot analysis suggested that canonical Wnt/β-catenin signaling pathway might be primarily mediating the observed benefits of aza on cardiac differentiation of MSC spheroids. In conclusion, 2 mg/ml collagen and 10 μM aza appears to offer optimal 3D microenvironment in terms of cell viability and time-dependent evolution of cardiomyogenesis from human BM-MSCs, with significant applications in cardiac tissue engineering and stem cell transplantation for regenerating lost cardiac tissue.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | - Gautam Mahajan
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, Ohio
| | | |
Collapse
|
213
|
Ino K, Onodera T, Kanno Y, Suda A, Kunikata R, Matsue T, Shiku H. Electrochemicolor imaging of endogenous alkaline phosphatase and respiratory activities of mesenchymal stem cell aggregates in early-stage osteodifferentiation. Electrochim Acta 2018. [DOI: 10.1016/j.electacta.2018.02.094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
214
|
Shin HS, Lee S, Hong HJ, Lim YC, Koh WG, Lim JY. Stem cell properties of human clonal salivary gland stem cells are enhanced by three-dimensional priming culture in nanofibrous microwells. Stem Cell Res Ther 2018; 9:74. [PMID: 29566770 PMCID: PMC5863805 DOI: 10.1186/s13287-018-0829-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cultures recapitulate the microenvironment of tissue-resident stem cells and enable them to modulate their properties. We determined whether salivary gland-resident stem cells (SGSCs) are primed by a 3D spheroid culture prior to treating irradiation-induced salivary hypofunction using in-vitro coculture and in-vivo transplant models. METHODS 3D spheroid-derived SGSCs (SGSCs3D) were obtained from 3D culture in microwells consisting of a nanofiber bottom and cell-repellent hydrogel walls, and were examined for salivary stem or epithelial gene/protein expression, differentiation potential, and paracrine secretory function compared with monolayer-cultured SGSCs (SGSCs2D) in vitro and in vivo. RESULTS SGSCs3D expressed increased salivary stem cell markers (LGR5 and THY1) and pluripotency markers (POU5F1 and NANOG) compared with SGSCs2D. Also, SGSCs3D exhibited enhanced potential to differentiate into salivary epithelial cells upon differentiation induction and increased paracrine secretion as compared to SGSCs2D. Wnt signaling was activated by 3D spheroid formation in the microwells and suppression of the Wnt/β-catenin pathway led to reduced stemness of SGSCs3D. Enhanced radioprotective properties of SGSCs3D against radiation-induced salivary hypofunction was confirmed by an organotypic 3D coculture and in-vivo transplantation experiments. CONCLUSION The 3D spheroid culture of SGSCs in nanofibrous microwells promotes stem cell properties via activation of Wnt signaling. This may contribute to SGSC priming prior to regenerative therapy to restore salivary hypofunction after radiotherapy.
Collapse
Affiliation(s)
- Hyun-Soo Shin
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Songyi Lee
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Young Chang Lim
- Department of Otorhinolaryngology, Head and Neck Surgery, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Republic of Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
215
|
Motoike S, Kajiya M, Komatsu N, Takewaki M, Horikoshi S, Matsuda S, Ouhara K, Iwata T, Takeda K, Fujita T, Kurihara H. Cryopreserved clumps of mesenchymal stem cell/extracellular matrix complexes retain osteogenic capacity and induce bone regeneration. Stem Cell Res Ther 2018; 9:73. [PMID: 29562931 PMCID: PMC5863484 DOI: 10.1186/s13287-018-0826-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/16/2018] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Three-dimensional (3D) cultured clumps of mesenchymal stem cell (MSC)/extracellular matrix (ECM) complexes (C-MSCs) consist of cells and self-produced ECM. C-MSCs can regulate cellular functions in vitro and can be grafted into a defect site without an artificial scaffold to induce bone regeneration. Long-term cryopreservation of C-MSCs, which can enable them to serve as a ready-to-use cell preparation, may be helpful in developing beneficial cell therapy for bone regeneration. Therefore, the aim of this study was to investigate the effect of cryopreservation on C-MSCs. METHODS MSCs isolated from rat femurs were cultured in growth medium supplemented with ascorbic acid. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and were then torn off. The sheet was rolled to make a round clumps of cells. The C-MSCs were cryopreserved in cryomedium including 10% dimethyl sulfoxide. RESULTS Cryopreserved C-MSCs retained their 3D structure and did not exhibit a decrease in cell viability. In addition, stem cell marker expression levels and the osteogenic differentiation properties of C-MSCs were not reduced by cryopreservation. However, C-MSCs pretreated with collagenase before cryopreservation showed a lower level of type I collagen and could not retain their 3D structure, and their rates of cell death increased during cryopreservation. Both C-MSC and cryopreserved C-MSC transplantation into rat calvarial defects induced successful bone regeneration. CONCLUSION These data indicate that cryopreservation does not reduce the biological properties of C-MSCs because of its abundant type I collagen. More specifically, cryopreserved C-MSCs could be applicable for novel bone regenerative therapies.
Collapse
Affiliation(s)
- Souta Motoike
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan.
| | - Nao Komatsu
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Manabu Takewaki
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Susumu Horikoshi
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Shinji Matsuda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Tomoyuki Iwata
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Katsuhiro Takeda
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Tsuyoshi Fujita
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| | - Hidemi Kurihara
- Department of Periodontal Medicine, Applied Life Sciences, Institute of Biomedical & Health Sciences, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, Japan
| |
Collapse
|
216
|
Petry F, Weidner T, Czermak P, Salzig D. Three-Dimensional Bioreactor Technologies for the Cocultivation of Human Mesenchymal Stem/Stromal Cells and Beta Cells. Stem Cells Int 2018; 2018:2547098. [PMID: 29731775 PMCID: PMC5872596 DOI: 10.1155/2018/2547098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023] Open
Abstract
Diabetes is a prominent health problem caused by the failure of pancreatic beta cells. One therapeutic approach is the transplantation of functional beta cells, but it is difficult to generate sufficient beta cells in vitro and to ensure these cells remain viable at the transplantation site. Beta cells suffer from hypoxia, undergo apoptosis, or are attacked by the host immune system. Human mesenchymal stem/stromal cells (hMSCs) can improve the functionality and survival of beta cells in vivo and in vitro due to direct cell contact or the secretion of trophic factors. Current cocultivation concepts with beta cells are simple and cannot exploit the favorable properties of hMSCs. Beta cells need a three-dimensional (3D) environment to function correctly, and the cocultivation setup is therefore more complex. This review discusses 3D cultivation forms (aggregates, capsules, and carriers) for hMSCs and beta cells and strategies for large-scale cultivation. We have determined process parameters that must be balanced and considered for the cocultivation of hMSCs and beta cells, and we present several bioreactor setups that are suitable for such an innovative cocultivation approach. Bioprocess engineering of the cocultivation processes is necessary to achieve successful beta cell therapy.
Collapse
Affiliation(s)
- Florian Petry
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Tobias Weidner
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
- Department of Chemical Engineering, Kansas State University, Manhattan, KS, USA
- Project Group Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Winchesterstr. 3, 35394 Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstraße 14, 35390 Giessen, Germany
| |
Collapse
|
217
|
Zhang J, Yun S, Bi J, Dai S, Du Y, Zannettino ACW, Zhang H. Enhanced multi-lineage differentiation of human mesenchymal stem/stromal cells within poly(N-isopropylacrylamide-acrylic acid) microgel-formed three-dimensional constructs. J Mater Chem B 2018; 6:1799-1814. [PMID: 32254252 DOI: 10.1039/c8tb00376a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human mesenchymal stem/stromal cells (hMSCs) are a potential cell source of stem cell therapy for many serious diseases and hMSC spheroids have emerged to replace single cell suspensions for cell therapy. Three-dimensional (3D) scaffolds or hydrogels which can mimic properties of the extracellular matrix (ECM) have been widely explored for their application in tissue regeneration. However, there are considerably less studies on inducing differentiation of hMSC spheroids using 3D scaffolds or hydrogels. This study is the first to explore multi-lineage differentiation of a stem cell line and primary stem cells within poly(N-isopropylacrylamide) (p(NIPAAm))-based thermosensitive microgel-formed constructs. We first demonstrated that poly(N-isopropylacrylamide-co-acrylic acid) (p(NIPAAm-AA)) was not toxic to hMSCs and the microgel-formed constructs facilitated formation of uniform stem cell spheroids. Due to functional enhancement of cell spheroids, hMSCs within the 3D microgel-formed constructs were induced for multi-lineage differentiation as evidenced by significant up-regulation of messenger RNA (mRNA) expression of chondrogenic and osteogenic genes even in the absence of induction media on day 9. When induction media were in situ supplied on day 9, mRNA expression of chondrogenic, osteogenic and adipogenic genes within the microgel-formed constructs were significantly higher than that in the pellet and 2D cultures, respectively, on day 37. In addition, histological and immunofluorescent images also confirmed successful multi-lineage differentiation of hMSCs within the 3D microgel-formed constructs. Hence, the thermosensitive p(NIPAAm-AA) microgel can be potentially used in an in vitro model for cell differentiation or in vivo transplantation of pre-differentiated human mesenchymal stromal cells into patients for specific lineage differentiation.
Collapse
Affiliation(s)
- Jiabin Zhang
- School of Chemical Engineering, The University of Adelaide, Adelaide, SA 5005, Australia.
| | | | | | | | | | | | | |
Collapse
|
218
|
Messina V, Valtieri M, Rubio M, Falchi M, Mancini F, Mayor A, Alano P, Silvestrini F. Gametocytes of the Malaria Parasite Plasmodium falciparum Interact With and Stimulate Bone Marrow Mesenchymal Cells to Secrete Angiogenetic Factors. Front Cell Infect Microbiol 2018; 8:50. [PMID: 29546035 PMCID: PMC5838020 DOI: 10.3389/fcimb.2018.00050] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/09/2018] [Indexed: 12/31/2022] Open
Abstract
The gametocytes of Plasmodium falciparum, responsible for the transmission of this malaria parasite from humans to mosquitoes, accumulate and mature preferentially in the human bone marrow. In the 10 day long sexual development of P. falciparum, the immature gametocytes reach and localize in the extravascular compartment of this organ, in contact with several bone marrow stroma cell types, prior to traversing the endothelial lining and re-entering in circulation at maturity. To investigate the host parasite interplay underlying this still obscure process, we developed an in vitro tridimensional co-culture system in a Matrigel scaffold with P. falciparum gametocytes and self-assembling spheroids of human bone marrow mesenchymal cells (hBM-MSCs). Here we show that this co-culture system sustains the full maturation of the gametocytes and that the immature, but not the mature, gametocytes adhere to hBM-MSCs via trypsin-sensitive parasite ligands exposed on the erythrocyte surface. Analysis of a time course of gametocytogenesis in the co-culture system revealed that gametocyte maturation is accompanied by the parasite induced stimulation of hBM-MSCs to secrete a panel of 14 cytokines and growth factors, 13 of which have been described to play a role in angiogenesis. Functional in vitro assays on human bone marrow endothelial cells showed that supernatants from the gametocyte mesenchymal cell co-culture system enhance ability of endothelial cells to form vascular tubes. These results altogether suggest that the interplay between immature gametocytes and hBM-MSCs may induce functional and structural alterations in the endothelial lining of the human bone marrow hosting the P. falciparum transmission stages.
Collapse
Affiliation(s)
- Valeria Messina
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Valtieri
- Dipartimento di Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Mercedes Rubio
- ISGlobal, Barcelona Ctr. Int. Health Res, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Mario Falchi
- AIDS National Center, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Mancini
- Dipartimento di Biotecnologie Cellulari ed Ematologia, Umberto I - Policlinico di Roma, Rome, Italy
| | - Alfredo Mayor
- ISGlobal, Barcelona Ctr. Int. Health Res, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça, Maputo, Mozambique
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
219
|
Zhang S, Buttler-Buecher P, Denecke B, Arana-Chavez VE, Apel C. A comprehensive analysis of human dental pulp cell spheroids in a three-dimensional pellet culture system. Arch Oral Biol 2018; 91:1-8. [PMID: 29621667 DOI: 10.1016/j.archoralbio.2018.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/20/2018] [Accepted: 02/12/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Three-dimensional (3D) cell culture methods are of high importance to studies of biological processes. This is particularly the case with spheroid cultures, which create 3D cell aggregates without the use of exogenous materials. Compared to conventional monolayer cultures, cellular spheroid cultures have been demonstrated to improve multilineage potential and extracellular matrix production. To address this issue in depth, we present a more comprehensive analysis of 3D human dental pulp cell (hDPC) spheroids. DESIGN hDPC spheroids were fabricated by the pellet culture method and were cultured without adding any reagent to induce differentiation. The gene-expression profiles of the 3D and two-dimensional (2D) cultured hDPCs were compared by complementary DNA microarray analysis. Odontoblastic and osteoblastic differentiation marker gene expression was evaluated by quantitative real-time PCR (RT-qPCR). Hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM) were applied to examine the morphology of hDPC spheroids and extracellular matrix components. RESULTS Compared with 2D monolayer culture, microarray analysis identified 405 genes and 279 genes with twofold or greater differential expression after 3 days and 28 days of 3D culture, respectively. In 3D hDPC spheroids, gene ontology analysis revealed upregulation of extracellular matrix-related genes and downregulation of cell growth-related genes. RT-qPCR analysis showed higher expression levels of osteocalcin, dentin sialophosphoprotein, and alkaline phosphatase. TEM revealed the morphological characteristics of the fibrillar collagen-rich matrix and cell-cell interactions. CONCLUSIONS The present findings provide clues to understanding the mechanisms of pellet-cultured hDPCs and contribute to future research in the comparative studies of different 3D culture methods.
Collapse
Affiliation(s)
- Siyuan Zhang
- Department of Biohybrid & Medical Textiles, Institute of Applied Medical Engineering, Helmholtz-Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Patricia Buttler-Buecher
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research (IZKF) Aachen, RWTH Aachen University, Germany
| | - Victor E Arana-Chavez
- Department of Biomaterials and Oral Biology, School of Dentistry, University of São Paulo, Brazil
| | - Christian Apel
- Department of Biohybrid & Medical Textiles, Institute of Applied Medical Engineering, Helmholtz-Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
220
|
Suarez Muñoz M, Confalonieri D, Walles H, van Dongen EMWM, Dandekar G. Recombinant Collagen I Peptide Microcarriers for Cell Expansion and Their Potential Use As Cell Delivery System in a Bioreactor Model. J Vis Exp 2018:57363. [PMID: 29443081 PMCID: PMC5912385 DOI: 10.3791/57363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Tissue engineering is a promising field, focused on developing solutions for the increasing demand on tissues and organs regarding transplantation purposes. The process to generate such tissues is complex, and includes an appropriate combination of specific cell types, scaffolds, and physical or biochemical stimuli to guide cell growth and differentiation. Microcarriers represent an appealing tool to expand cells in a three-dimensional (3D) microenvironment, since they provide higher surface-to volume ratios and mimic more closely the in vivo situation compared to traditional two-dimensional methods. The vascular system, supplying oxygen and nutrients to the cells and ensuring waste removal, constitutes an important building block when generating engineered tissues. In fact, most constructs fail after being implanted due to lacking vascular support. In this study, we present a protocol for endothelial cell expansion on recombinant collagen-based microcarriers under dynamic conditions in spinner flask and bioreactors, and we explain how to determine in this setting cell viability and functionality. In addition, we propose a method for cell delivery for vascularization purposes without additional detachment steps necessary. Furthermore, we provide a strategy to evaluate the cell vascularization potential in a perfusion bioreactor on a decellularized biological matrix. We believe that the use of the presented methods could lead to the development of new cell-based therapies for a large range of tissue engineering applications in the clinical practice.
Collapse
Affiliation(s)
- Melva Suarez Muñoz
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg;
| | - Davide Confalonieri
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg
| | - Heike Walles
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC
| | | | - Gudrun Dandekar
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg; Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC
| |
Collapse
|
221
|
Al Madhoun A, Alkandari S, Ali H, Carrio N, Atari M, Bitar MS, Al-Mulla F. Chemically Defined Conditions Mediate an Efficient Induction of Mesodermal Lineage from Human Umbilical Cord- and Bone Marrow- Mesenchymal Stem Cells and Dental Pulp Pluripotent-Like Stem Cells. Cell Reprogram 2018; 20:9-16. [PMID: 29412734 DOI: 10.1089/cell.2017.0028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The human umbilical cord Wharton's Jelly- and the bone marrow- mesenchymal stem cells (WJ-MSCs and BM-MSCs, respectively) and the newly identified dental pulp pluripotent-like stem cells (DPPSCs) are new sources for stem cells with prospective use in cell regeneration and therapy. These cells are self-renewable, can be differentiated into several lineages, and can potentiate the immune responses. We hypothesized that three-dimensional (3D) culture conditions and directed differentiation using specific signaling regulators will enhance an efficient generation of mesoderm (MD) lineage independent from the origin or source of the stem cells. For a period of 3-days, cell aggregates were generated in a serum-free media containing ascorbic acid, retinoic acid, and keratinocyte growth factor; sonic hedgehog and bone morphogenic protein-4 signaling were inhibited using small molecules. In all cell types used, the biochemical and molecular analysis revealed a time course-dependent induction of the mesodermal, but not endodermal or ectodermal makers. In this study, we utilized a novel and efficient serum-free protocol to differentiate WJ-MSCs, BM-MSCs, and DPPSCs into MD-cells. Successful development of an efficient differentiation protocol can further be utilized and expanded on to obtain MD- derivative cell lineages.
Collapse
Affiliation(s)
- Ashraf Al Madhoun
- 1 Functional Genomic Unit, Research Division, Dasman Diabetes Institute , Dasman, Kuwait
| | - Sarah Alkandari
- 1 Functional Genomic Unit, Research Division, Dasman Diabetes Institute , Dasman, Kuwait
| | - Hamad Ali
- 1 Functional Genomic Unit, Research Division, Dasman Diabetes Institute , Dasman, Kuwait
- 2 Department of Medical Laboratory Sciences (MLS), Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University , Kuwait
| | - Neus Carrio
- 3 Regenerative Medicine Research Institute , UIC Barcelona, Barcelona, Spain
| | - Maher Atari
- 3 Regenerative Medicine Research Institute , UIC Barcelona, Barcelona, Spain
| | - Milad S Bitar
- 4 Department of Pharmacology and Toxicology, Health Sciences Center, Kuwait University , Kuwait
| | - Fahd Al-Mulla
- 1 Functional Genomic Unit, Research Division, Dasman Diabetes Institute , Dasman, Kuwait
| |
Collapse
|
222
|
Ko DY, Patel M, Lee HJ, Jeong B. Coordinating Thermogel for Stem Cell Spheroids and Their Cyto-Effectiveness. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1706286. [DOI: 10.1002/adfm.201706286] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Du Young Ko
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Madhumita Patel
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience; Ewha Womans University; 52 Ewhayeodae-gil Seodaemun-gu Seoul 03760 Korea
| |
Collapse
|
223
|
Efficient scalable production of therapeutic microvesicles derived from human mesenchymal stem cells. Sci Rep 2018; 8:1171. [PMID: 29352188 PMCID: PMC5775399 DOI: 10.1038/s41598-018-19211-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022] Open
Abstract
Microvesicles (MVs) released by cells are involved in a multitude of physiological events as important mediators of intercellular communication. MVs derived from mesenchymal stem cells (MSCs) contain various paracrine factors from the cells that primarily contribute to their therapeutic efficacy observed in numerous clinical trials. As nano-sized and bi-lipid layered vesicles retaining therapeutic potency equivalent to that of MSCs, MSC-derived MVs have been in focus as ideal medicinal candidates for regenerative medicine, and are preferred over MSC infusion therapy with their improved safety profiles. However, technical challenges in obtaining sufficient amounts of MVs have limited further progress in studies and clinical application. Of the multiple efforts to reinforce the therapeutic capacity of MSCs, few studies have reportedly examined the scale-up of MSC-derived MV production. In this study, we successfully amplified MV secretion from MSCs compared to the conventional culture method using a simple and efficient 3D-bioprocessing method. The MSC-derived MVs produced in our dynamic 3D-culture contained numerous therapeutic factors such as cytokines and micro-RNAs, and showed their therapeutic potency in in vitro efficacy evaluation. Our results may facilitate diverse applications of MSC-derived MVs from the bench to the bedside, which requires the large-scale production of MVs.
Collapse
|
224
|
Redondo-Castro E, Cunningham CJ, Miller J, Brown H, Allan SM, Pinteaux E. Changes in the secretome of tri-dimensional spheroid-cultured human mesenchymal stem cells in vitro by interleukin-1 priming. Stem Cell Res Ther 2018; 9:11. [PMID: 29343288 PMCID: PMC5773162 DOI: 10.1186/s13287-017-0753-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 12/30/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are one of the most promising candidates for the treatment of major neurological disorders. Desirable therapeutic properties of MSCs include reparative and regenerative potential but, despite their proven safety, the efficacy of MSCs remains controversial. Therefore, it is essential to optimise culture protocols to enhance the therapeutic potential of the MSC secretome. Here we aimed to: assess the increase in secretion of cytokines that may induce repair, regeneration, or immunomodulation when cultured in three dimensions; study the effect of interleukin (IL)-1 priming on two- (2D) and three-dimensional (3D) cultures of MSC; and evaluate the potential use of the modified secretome using microglial-MSC co-cultures. Methods We established a 3D spheroid culture of human MSCs, and compared the secretome in 2D and 3D cultures under primed (IL-1) and unprimed conditions. BV2 microglial cells were stimulated with lipopolysaccharide (LPS) and treated with spheroid conditioned media (CM) or were co-cultured with whole spheroids. Concentrations of secreted cytokines were determined by enzyme-linked immunosorbent assay (ELISA). Protein arrays were used to further evaluate the effect of IL-1 priming in 2D and 3D cultures. Results 3D culture of MSCs significantly increased secretion of the IL-1 receptor antagonist (IL-1Ra), vascular endothelial growth factor (VEGF), and granulocyte-colony stimulating factor (G-CSF) compared with 2D culture, despite priming treatments with IL-1 being more effective in 2D than in 3D. The addition of CM of 3D-MSCs reduced LPS-induced tumour necrosis factor (TNF)-α secretion from BV2 cells, while the 3D spheroid co-cultured with the BV2 cells induced an increase in IL-6, but had no effect on TNF-α release. Protein arrays indicated that priming treatments trigger a more potent immune profile which is necessary to orchestrate an effective tissue repair. This effect was lost in 3D, partly because of the overexpression of IL-6. Conclusions Increased secretion of anti-inflammatory markers occurs when MSCs are cultured in 3D, but this specific secretome did not translate into anti-inflammatory effects on LPS-treated BV2 cells in co-culture. These data highlight the importance of optimising priming treatments and culture conditions to maximise the therapeutic potential of MSC spheroids.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona J Cunningham
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Helena Brown
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
225
|
Large-Scale Expansion and Differentiation of Mesenchymal Stem Cells in Microcarrier-Based Stirred Bioreactors. Methods Mol Biol 2018; 1502:87-102. [PMID: 26892015 DOI: 10.1007/7651_2015_314] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cells (MSCs) have emerged as an important tool for tissue engineering, thanks to their differentiation potential and their broad trophic activities. However, for clinical purposes or for relevant in vitro applications, large quantities of MSCs are required, which could hardly be reached using conventional cultivation in plastic dishes. Microcarriers have high surface to volume ratio, which enables the easy scale-up of the expansion and differentiation of MSCs. In addition, the agitation in stirred tank bioreactors limits the diffusion gradient of nutrients or morphogens, thus providing a physiologically relevant environment to favor MSC production at large scale. This work describes a simple method for the mass expansion and differentiation of MSCs, including the procedures to monitor the proliferation, metabolic status and phenotype of MSCs during suspension culture. Moreover, this work proposes suitable materials for cGMP compliant culture conditions enabling the clinical grade production of MSCs.
Collapse
|
226
|
Bauman E, Feijão T, Carvalho DTO, Granja PL, Barrias CC. Xeno-free pre-vascularized spheroids for therapeutic applications. Sci Rep 2018; 8:230. [PMID: 29321569 PMCID: PMC5762877 DOI: 10.1038/s41598-017-18431-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/12/2017] [Indexed: 12/16/2022] Open
Abstract
Spheroid culture has gained increasing popularity, arising as a promising tool for regenerative medicine applications. Importantly, spheroids may present advantages over single-cell suspensions in cell-based therapies (CT). Unfortunately, most growth media used for spheroid culture contain animal origin-components, such as fetal bovine serum (FBS). The presence of FBS compromises the safety of CT and presents economic and ethical constraints. SCC (supplement for cell culture) is a novel xeno-free (XF) industrial cell culture supplement, derived from well-controlled pooled human plasma and processed under good manufacturing practice rules. Here, we developed a XF SCC-based formulation for 2D-culture of outgrowth endothelial cells (OEC), and then used it for generating co-culture spheroids of OEC and mesenchymal stem cells (MSC). XF MSC-OEC spheroids were characterized in detail and compared to spheroids cultured in FBS-supplemented medium. XF spheroids presented comparable integrity, size and morphology as the reference culture. The use of both media resulted in spheroids with similar structure, abundant extracellular matrix deposition and specific patterns of OEC distribution and organization. Notably, XF spheroids presented significantly enhanced angiogenic potential, both in vitro (fibrin sprouting assay) and in vivo (CAM assay). These findings are particularly promising in the context of potential therapeutic applications.
Collapse
Affiliation(s)
- E Bauman
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal
| | - T Feijão
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - D T O Carvalho
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - P L Granja
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - C C Barrias
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal. .,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal. .,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
227
|
Song L, Tsai AC, Yuan X, Bejoy J, Sart S, Ma T, Li Y. Neural Differentiation of Spheroids Derived from Human Induced Pluripotent Stem Cells-Mesenchymal Stem Cells Coculture. Tissue Eng Part A 2018; 24:915-929. [PMID: 29160172 DOI: 10.1089/ten.tea.2017.0403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Organoids, the condensed three-dimensional (3D) tissues emerged at the early stage of organogenesis, are a promising approach to regenerate functional and vascularized organ mimics. While incorporation of heterotypic cell types, such as human mesenchymal stem cells (hMSCs) and human induced pluripotent stem cells (hiPSCs)-derived neural progenitors aid neural organ development, the interactions of secreted factors during neurogenesis have not been well understood. The objective of this study is to investigate the impact of the composition and structure of 3D hybrid spheroids of hiPSCs and hMSCs on dorsal cortical differentiation and the secretion of extracellular matrices and trophic factors in vitro. The hybrid spheroids were formed at different hiPSC:hMSC ratios (100:0, 75:25, 50:50, 25:75, 0:100) using direct mixing or pre-hiPSC aggregation method, which generated dynamic spheroid structure. The cellular organization, proliferation, neural marker expression, and the secretion of extracellular matrix proteins and the cytokines were characterized. The incorporation of MSCs upregulated Nestin and β-tubulin III expression (the dorsal cortical identity was shown by Pax6 and TBR1 expression), matrix remodeling proteins, and the secretion of transforming growth factor-β1 and prostaglandin E2. This study indicates that the appropriate composition and structure of hiPSC-MSC spheroids promote neural differentiation and trophic factor and matrix secretion due to the heterotypic cell-cell interactions.
Collapse
Affiliation(s)
- Liqing Song
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Ang-Chen Tsai
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Xuegang Yuan
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Julie Bejoy
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Sébastien Sart
- 2 Hydrodynamics Laboratory (LadHyX) , Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, Palaiseau, France
| | - Teng Ma
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| | - Yan Li
- 1 Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University; Tallahassee , Florida
| |
Collapse
|
228
|
Vorwald CE, Ho SS, Whitehead J, Leach JK. High-Throughput Formation of Mesenchymal Stem Cell Spheroids and Entrapment in Alginate Hydrogels. Methods Mol Biol 2018; 1758:139-149. [PMID: 29679328 DOI: 10.1007/978-1-4939-7741-3_11] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are a promising cell source for tissue repair and regeneration due to their multilineage capacity, potential for autologous use, and secretion of potent bioactive factors to catalyze the endogenous repair program. However, a major limitation to current cell-based tissue engineering approaches is the drastic loss of cells upon transplantation. The causation of this loss, whether due to apoptosis following a dramatic change in the microenvironment or migration away from the defect site, has yet to be determined. MSCs formed into aggregates, known as spheroids, possess a strong therapeutic advantage compared to the more commonly used dissociated cells due to their improved resistance to apoptosis and increased secretion of endogenous trophic factors. Furthermore, the use of biomaterials such as alginate hydrogels to transplant cells in situ improves cell survival, localizes payloads at the defect site, and facilitates continued instruction of cells by manipulating the biophysical properties of the biomaterial. Transplantation of MSC spheroids without a vehicle into tissue defects comprises the majority of studies to date, ceding control of spheroid function due to the cell's interaction with the native tissue extracellular matrix and abrogating the established benefits of spheroid formation. Thus, there is a significant need to consider the role of biomaterials in transplanting MSC spheroids using an appropriate carrier. In this chapter, we describe high-throughput formation of spheroids, steps for further characterization, and encapsulation in alginate hydrogels with an eye toward localizing MSC spheroids at the target site.
Collapse
Affiliation(s)
- Charlotte E Vorwald
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA
| | - Steve S Ho
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA
| | - Jacklyn Whitehead
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California-Davis, Davis, CA, USA. .,Department of Orthopaedic Surgery, School of Medicine, University of California-Davis, Sacramento, CA, USA.
| |
Collapse
|
229
|
Successful Low-Cost Scaffold-Free Cartilage Tissue Engineering Using Human Cartilage Progenitor Cell Spheroids Formed by Micromolded Nonadhesive Hydrogel. Stem Cells Int 2017. [PMID: 29527227 PMCID: PMC5750468 DOI: 10.1155/2017/7053465] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The scaffold-free tissue engineering using spheroids is pointed out as an approach for optimizing the delivery system of cartilage construct. In this study, we aimed to evaluate the micromolded nonadhesive hydrogel (MicroTissues®) for spheroid compaction (2-day culture) and spontaneous chondrogenesis (21-day culture) using cartilage progenitors cells (CPCs) from human nasal septum without chondrogenic stimulus. CPC spheroids showed diameter stability (486 μm ± 65), high percentage of viable cells (88.1 ± 2.1), and low percentage of apoptotic cells (2.3%). After spheroid compaction, the synthesis of TGF-β1, TGF-β2, and TGF-β3 was significantly higher compared to monolayer (p < 0.005). Biomechanical assay revealed that the maximum forces applied to spheroids after chondrogenesis were 2.6 times higher than for those cultured for 2 days. After spontaneous chondrogenesis, CPC spheroids were entirely positive for N-cadherin, collagen type II and type VI, and aggrecan and chondroitin sulfate. Comparing to monolayer, the expression of SOX5 and SOX6 genes analyzed by qPCR was significantly upregulated (p < 0.01). Finally, we observed the capacity of CPC spheroids starting to fuse. To the best of our knowledge, this is the first time in the scientific literature that human CPC spheroids were formed by micromolded nonadhesive hydrogel, achieving a successful scaffold-free cartilage engineering without chondrogenic stimulus (low cost).
Collapse
|
230
|
Aggregation of Culture Expanded Human Mesenchymal Stem Cells in Microcarrier-based Bioreactor. Biochem Eng J 2017; 131:39-46. [PMID: 29736144 DOI: 10.1016/j.bej.2017.12.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Three-dimensional aggregation of human mesenchymal stem cells (hMSCs) has been used to enhance their therapeutic properties but current fabrication protocols depend on laboratory methods and are not scalable. In this study, we developed thermal responsive poly(N-isopropylacrylamide) grafted microcarriers (PNIPAM-MCs), which supported expansion and thermal detachment of hMSCs at reduced temperature (23.0 °C). hMSCs were cultured on the PNIPAM-MCs in both spinner flask (SF) and PBS Vertical-Wheel (PBS-VW) bioreactors for expansion. At room temperature, hMSCs were detached as small cell sheets, which subsequently self-assembled into 3D hMSC aggregates in PBS-VW bioreactor and remain as single cells in SF bioreactor owing to different hydrodynamic conditions. hMSC aggregates generated from the bioreactor maintained comparable immunomodulation and cytokine secretion properties compared to the ones made from the AggreWell®. The results of the current study demonstrate the feasibility of scale-up production of hMSC aggregates in the suspension bioreactor using thermal responsive microcarriers for integrated cell expansion and 3D aggregation in a close bioreactor system and highlight the critical role of hydrodynamics in self-assembly of detached hMSC in suspension.
Collapse
|
231
|
Lo H, Huang A, Lee P, Chung T, Wang S. Morphological transformation of h
BMSC
from 2
D
monolayer to 3
D
microtissue on low‐crystallinity
SF‐PCL
patch with promotion of cardiomyogenesis. J Tissue Eng Regen Med 2017; 12:e1852-e1864. [DOI: 10.1002/term.2616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/19/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Hsin‐Yu Lo
- Department of Biomedical EngineeringNational Yang‐Ming University Taipei Taiwan
| | - An‐Li Huang
- Department of Biomedical EngineeringNational Yang‐Ming University Taipei Taiwan
| | - Pei‐Chi Lee
- Department of Biomedical EngineeringNational Yang‐Ming University Taipei Taiwan
| | - Tze‐Wen Chung
- Department of Biomedical EngineeringNational Yang‐Ming University Taipei Taiwan
| | - Shoei‐Shen Wang
- Department of SurgeryFu Jen Catholic University Hospital, and Fu Jen Catholic University College of Medicine New Taipei City Taiwan
| |
Collapse
|
232
|
|
233
|
Mohammadi Ghahhari N, Maghsood F, Jahandideh S, Lotfinia M, Lak S, Johari B, Azarnezhad A, Kadivar M. Secretome of Aggregated Embryonic Stem Cell-Derived Mesenchymal Stem Cell Modulates the Release of
Inflammatory Factors in Lipopolysaccharide-Induced
Peripheral Blood Mononuclear Cells. IRANIAN BIOMEDICAL JOURNAL 2017; 22:237-45. [PMID: 29132205 PMCID: PMC5949126 DOI: 10.22034/ibj.22.4.237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Bone marrow mesenchymal stem cells (BM-MSCs) have emerged as a potential therapy for various inflammatory diseases. Because of some limitations, several recent studies have suggested the use of embryonic stem cell-derived MSCs (ESC-MSCs) as an alternative for BM-MSCs. Some of the therapeutic effects of the ESC-MSCs are related to the secretion of a broad array of cytokines and growth factors, known as secretome. Harnessing this secretome for therapeutic applications requires the optimization of production of secretary molecules. It has been shown that aggregation of MSCs into 3D spheroids, as a preconditioning strategy, can enhance immunomodulatory potential of such cells. In this study, we investigated the effect of secretome derived from human ESC-MSCs (hESC-MSCs) spheroids on secretion of IL-1β, IL-10, and tumor necrosis factor α (TNF-α) from lipopolysaccharide (LPS)-induced peripheral blood mononuclear cells (PBMCs). Methods: In the present study, after immunophenotyping and considering mesodermal differentiation of hESC-MSCs, the cells were non-adherently grown to prepare 3D aggregates, and then conditioned medium or secretome was extracted from the cultures. Afterwards, the anti-inflammatory effects of the secretome were assessed in an in vitro model of inflammation. Results: Results from this study showed that aggregate-prepared secretome from hESC-MSCs was able to significantly decrease the secretion of TNF-α (301.7 ± 5.906, p < 0.0001) and IL-1β (485.2 ± 48.38, p < 0.001) from LPS-induced PBMCs as the indicators of inflammation, in comparison with adherent culture-prepared secretome (TNF-α: 166.6 ± 8.04, IL-1β: 125.2 ± 2.73). Conclusion: Our study indicated that cell aggregation can be an appropriate strategy to increase immunomodulatory characteristics of hESC-MSCs.
Collapse
Affiliation(s)
| | - Faezeh Maghsood
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Jahandideh
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Majid Lotfinia
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Shirin Lak
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Behrooz Johari
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Asaad Azarnezhad
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Kadivar
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
234
|
Murphy KC, Hung BP, Browne-Bourne S, Zhou D, Yeung J, Genetos DC, Leach JK. Measurement of oxygen tension within mesenchymal stem cell spheroids. J R Soc Interface 2017; 14:rsif.2016.0851. [PMID: 28179546 DOI: 10.1098/rsif.2016.0851] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/17/2017] [Indexed: 02/07/2023] Open
Abstract
Spheroids formed of mesenchymal stem cells (MSCs) exhibit increased cell survival and trophic factor secretion compared with dissociated MSCs, making them therapeutically advantageous for cell therapy. Presently, there is no consensus for the mechanism of action. Many hypothesize that spheroid formation potentiates cell function by generating a hypoxic core within spheroids of sufficiently large diameters. The purpose of this study was to experimentally determine whether a hypoxic core is generated in MSC spheroids by measuring oxygen tension in aggregates of increasing diameter and correlating oxygen tension values with cell function. MSC spheroids were formed with 15 000, 30 000 or 60 000 cells per spheroid, resulting in radii of 176 ± 8 µm, 251 ± 12 µm and 353 ± 18 µm, respectively. Oxygen tension values coupled with mathematical modelling revealed a gradient that varied less than 10% from the outer diameter within the largest spheroids. Despite the modest radial variance in oxygen tension, cellular metabolism from spheroids significantly decreased as the number of cells and resultant spheroid size increased. This may be due to adaptive reductions in matrix deposition and packing density with increases in spheroid diameter, enabling spheroids to avoid the formation of a hypoxic core. Overall, these data provide evidence that the enhanced function of MSC spheroids is not oxygen mediated.
Collapse
Affiliation(s)
- Kaitlin C Murphy
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Ben P Hung
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Stephen Browne-Bourne
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Dejie Zhou
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Jessica Yeung
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Damian C Genetos
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - J Kent Leach
- Department of Biomedical Engineering, School of Veterinary Medicine, University of California, Davis, CA 95616, USA .,Department of Orthopaedic Surgery, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
235
|
Xie AW, Binder BYK, Khalil AS, Schmitt SK, Johnson HJ, Zacharias NA, Murphy WL. Controlled Self-assembly of Stem Cell Aggregates Instructs Pluripotency and Lineage Bias. Sci Rep 2017; 7:14070. [PMID: 29070799 PMCID: PMC5656593 DOI: 10.1038/s41598-017-14325-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022] Open
Abstract
Stem cell-derived organoids and other 3D microtissues offer enormous potential as models for drug screening, disease modeling, and regenerative medicine. Formation of stem/progenitor cell aggregates is common in biomanufacturing processes and critical to many organoid approaches. However, reproducibility of current protocols is limited by reliance on poorly controlled processes (e.g., spontaneous aggregation). Little is known about the effects of aggregation parameters on cell behavior, which may have implications for the production of cell aggregates and organoids. Here we introduce a bioengineered platform of labile substrate arrays that enable simple, scalable generation of cell aggregates via a controllable 2D-to-3D "self-assembly". As a proof-of-concept, we show that labile substrates generate size- and shape-controlled embryoid bodies (EBs) and can be easily modified to control EB self-assembly kinetics. We show that aggregation method instructs EB lineage bias, with faster aggregation promoting pluripotency loss and ectoderm, and slower aggregation favoring mesoderm and endoderm. We also find that aggregation kinetics of EBs markedly influence EB structure, with slower kinetics resulting in increased EB porosity and growth factor signaling. Our findings suggest that controlling internal structure of cell aggregates by modifying aggregation kinetics is a potential strategy for improving 3D microtissue models for research and translational applications.
Collapse
Affiliation(s)
- Angela W Xie
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Bernard Y K Binder
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Andrew S Khalil
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Samantha K Schmitt
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Hunter J Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Nicholas A Zacharias
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53705, United States.
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, 53705, United States.
| |
Collapse
|
236
|
Huang X, Shan L, Cheng K, Weng W. Cytocompatibility of Titanium Microsphere-Based Surfaces. ACS Biomater Sci Eng 2017; 3:3254-3260. [DOI: 10.1021/acsbiomaterials.7b00551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaoxiao Huang
- School
of Materials Science and Engineering, State Key Laboratory of Silicon
Materials, Zhejiang University, Hangzhou 310027, China
| | - Lijun Shan
- Department
of Chemical and Process Engineering, Faculty of Engineering and Built
Environment, University Kebangsaan Malaysia, Bangi, Malaysia
| | - Kui Cheng
- School
of Materials Science and Engineering, State Key Laboratory of Silicon
Materials, Zhejiang University, Hangzhou 310027, China
| | - Wenjian Weng
- School
of Materials Science and Engineering, State Key Laboratory of Silicon
Materials, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
237
|
Ahrens CC, Dong Z, Li W. Engineering cell aggregates through incorporated polymeric microparticles. Acta Biomater 2017; 62:64-81. [PMID: 28782721 DOI: 10.1016/j.actbio.2017.08.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/16/2022]
Abstract
Ex vivo cell aggregates must overcome significant limitations in the transport of nutrients, drugs, and signaling proteins compared to vascularized native tissue. Further, engineered extracellular environments often fail to sufficiently replicate tethered signaling cues and the complex architecture of native tissue. Co-cultures of cells with microparticles (MPs) is a growing field directed towards overcoming many of these challenges by providing local and controlled presentation of both soluble and tethered proteins and small molecules. Further, co-cultured MPs offer a mechanism to better control aggregate architecture and even to report key characteristics of the local microenvironment such as pH or oxygen levels. Herein, we provide a brief introduction to established and developing strategies for MP production including the choice of MP materials, fabrication techniques, and techniques for incorporating additional functionality. In all cases, we emphasize the specific utility of each approach to form MPs useful for applications in cell aggregate co-culture. We review established techniques to integrate cells and MPs. We highlight those strategies that promote targeted heterogeneity or homogeneity, and we describe approaches to engineer cell-particle and particle-particle interactions that enhance aggregate stability and biological response. Finally, we review advances in key application areas of MP aggregates and future areas of development. STATEMENT OF SIGNIFICANT Cell-scaled polymer microparticles (MPs) integrated into cellular aggregates have been shown to be a powerful tool to direct cell response. MPs have supported the development of healthy cartilage, islets, nerves, and vasculature by the maintenance of soluble gradients as well as by the local presentation of tethered cues and diffusing proteins and small molecules. MPs integrated with pluripotent stem cells have directed in vivo expansion and differentiation. Looking forward, MPs are expected to support both the characterization and development of in vitro tissue systems for applications such as drug testing platforms. However, useful co-cultures must be designed keeping in mind the limitations and attributes of each material strategy within the context of the overall tissue biology. The present review integrates prospectives from materials development, drug delivery, and tissue engineering to provide a toolbox for the development and application of MPs useful for long-term co-culture within cell aggregates.
Collapse
Affiliation(s)
- Caroline C Ahrens
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States
| | - Ziye Dong
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States
| | - Wei Li
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States.
| |
Collapse
|
238
|
Tseng TC, Wong CW, Hsieh FY, Hsu SH. Biomaterial Substrate-Mediated Multicellular Spheroid Formation and Their Applications in Tissue Engineering. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201700064] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/01/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ting-Chen Tseng
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Chui-Wei Wong
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Fu-Yu Hsieh
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
| | - Shan-hui Hsu
- Institute of Polymer Science and Engineering; National Taiwan University; Taipei Taiwan
- Institute of Cellular and System Medicine; National Health Research Institutes; Miaoli Taiwan
| |
Collapse
|
239
|
Abbasalizadeh S, Pakzad M, Cabral JMS, Baharvand H. Allogeneic cell therapy manufacturing: process development technologies and facility design options. Expert Opin Biol Ther 2017; 17:1201-1219. [PMID: 28699788 DOI: 10.1080/14712598.2017.1354982] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Currently, promising outcomes from clinical trials of allogeneic cells, especially allogeneic mesenchymal stromal cells, fibroblasts, keratinocytes, and human cardiac stem cells, have encouraged research institutions, small and medium enterprises (SMEs), and big pharmaceutical companies to invest and focus on developing allogeneic cell therapy products. Commercial and large-scale production of allogeneic cell therapy products requires unique capabilities to develop technologies that generate safe and effective allogeneic cells/cell lines and their fully characterized master/working banks. In addition, it is necessary to design robust upstream and downstream manufacturing processes, and establish integrated, well-designed manufacturing facilities to produce high quality affordable products in accordance with current GMP regulations for the production of cell therapy products. Areas covered: The authors highlight: the recent advances in the development of allogeneic products, the available options to develop robust manufacturing processes, and facility design considerations. Expert opinion: Currently, there are multiple challenges in development of allogeneic cell therapy products. Indeed, the field is still in its infancy; with technologies and regulations still under development, as is our understanding of the mechanisms of action in the body and their interaction with the host immune system. Their characterization and testing is also an emerging and very complex area.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Mohammad Pakzad
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
| | - Joaquim M S Cabral
- b Department of Bioengineering and Institute for Bioengineering and Biosciences , Instituto Superior Técnico, Universidade de Lisboa , Lisboa , Portugal
| | - Hossein Baharvand
- a Department of Stem Cells and Developmental Biology, Cell Science Research Center , Royan Institute for Stem Cell Biology and Technology, ACECR , Tehran , Iran
- c Department of Developmental Biology , University of Science and Culture , Tehran , Iran
| |
Collapse
|
240
|
Yan Y, Song L, Madinya J, Ma T, Li Y. Derivation of Cortical Spheroids from Human Induced Pluripotent Stem Cells in a Suspension Bioreactor. Tissue Eng Part A 2017; 24:418-431. [PMID: 28825364 DOI: 10.1089/ten.tea.2016.0400] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) emerge as a promising source to construct human brain-like tissues, spheroids, or organoids in vitro for disease modeling and drug screening. A suspension bioreactor can be used to generate large size of brain organoids from hiPSCs through enhanced diffusion, but the influence of a dynamic bioreactor culture environment on neural tissue patterning from hiPSCs has not been well understood. The objective of this study is to assess the influence of a suspension bioreactor culture on cortical spheroid (i.e., forebrain-like aggregates) formation from hiPSCs. Single undifferentiated hiPSK3 cells or preformed embryoid bodies were inoculated into the bioreactor. Aggregate size distribution, neural marker expression (e.g., Nestin, PAX6, β-tubulin III, and MAP-2), and cortical tissue patterning markers (e.g., TBR1, BRN2, SATB2, and vGlut1) were evaluated with static control. Bioreactor culture was found to promote the expression of TBR1, a deep cortical layer VI marker, and temporally affect SATB2, a superficial cortical layer II-IV marker that appears later according to inside-out cortical tissue development. Prolonged culture after 70 days showed layer-specific cortical structure in the spheroids. Differential expression of matrix metalloproteinase-2 and -3 was also observed for bioreactor and static culture. The altered expression of cortical markers by a suspension bioreactor indicates the importance of culture environment on cortical tissue development from hiPSCs.
Collapse
Affiliation(s)
- Yuanwei Yan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Jason Madinya
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
241
|
Sart S, Tomasi RFX, Amselem G, Baroud CN. Multiscale cytometry and regulation of 3D cell cultures on a chip. Nat Commun 2017; 8:469. [PMID: 28883466 PMCID: PMC5589863 DOI: 10.1038/s41467-017-00475-x] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/30/2017] [Indexed: 12/23/2022] Open
Abstract
Three-dimensional cell culture is emerging as a more relevant alternative to the traditional two-dimensional format. Yet the ability to perform cytometry at the single cell level on intact three-dimensional spheroids or together with temporal regulation of the cell microenvironment remains limited. Here we describe a microfluidic platform to perform high-density three-dimensional culture, controlled stimulation, and observation in a single chip. The method extends the capabilities of droplet microfluidics for performing long-term culture of adherent cells. Using arrays of 500 spheroids per chip, in situ immunocytochemistry and image analysis provide multiscale cytometry that we demonstrate at the population scale, on 104 single spheroids, and over 105 single cells, correlating functionality with cellular location within the spheroids. Also, an individual spheroid can be extracted for further analysis or culturing. This will enable a shift towards quantitative studies on three-dimensional cultures, under dynamic conditions, with implications for stem cells, organs-on-chips, or cancer research.3D cell culture is more relevant than the two-dimensional format, but methods for parallel analysis and temporal regulation of the microenvironment are limited. Here the authors develop a droplet microfluidics system to perform long-term culture of 3D spheroids, enabling multiscale cytometry of individual cells within the spheroid.
Collapse
Affiliation(s)
- Sébastien Sart
- Laboratory of Hydrodynamics (LadHyX)-Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, 91128, Palaiseau, France
| | - Raphaël F-X Tomasi
- Laboratory of Hydrodynamics (LadHyX)-Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, 91128, Palaiseau, France
| | - Gabriel Amselem
- Laboratory of Hydrodynamics (LadHyX)-Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, 91128, Palaiseau, France
| | - Charles N Baroud
- Laboratory of Hydrodynamics (LadHyX)-Department of Mechanics, Ecole Polytechnique, CNRS-UMR7646, 91128, Palaiseau, France.
| |
Collapse
|
242
|
Confalonieri D, La Marca M, van Dongen EMWM, Walles H, Ehlicke F. An Injectable Recombinant Collagen I Peptide–Based Macroporous Microcarrier Allows Superior Expansion of C2C12 and Human Bone Marrow-Derived Mesenchymal Stromal Cells and Supports Deposition of Mineralized Matrix. Tissue Eng Part A 2017; 23:946-957. [DOI: 10.1089/ten.tea.2016.0436] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Davide Confalonieri
- Translational Center Wuerzburg “Regenerative Therapies in Oncology and Musculoskeletal Disease,” Wuerzburg, Germany
| | | | | | - Heike Walles
- Translational Center Wuerzburg “Regenerative Therapies in Oncology and Musculoskeletal Disease,” Wuerzburg, Germany
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Franziska Ehlicke
- Department Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
243
|
Cha JM, Park H, Shin EK, Sung JH, Kim O, Jung W, Bang OY, Kim J. A novel cylindrical microwell featuring inverted-pyramidal opening for efficient cell spheroid formation without cell loss. Biofabrication 2017; 9:035006. [DOI: 10.1088/1758-5090/aa8111] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
244
|
Sart S, Bejoy J, Li Y. Characterization of 3D pluripotent stem cell aggregates and the impact of their properties on bioprocessing. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
245
|
|
246
|
Shift of EMT gradient in 3D spheroid MSCs for activation of mesenchymal niche function. Sci Rep 2017; 7:6859. [PMID: 28761088 PMCID: PMC5537359 DOI: 10.1038/s41598-017-07049-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/23/2017] [Indexed: 12/19/2022] Open
Abstract
Despite the wide use of mesenchymal stromal cells (MSCs) for paracrine support in clinical trials, their variable and heterogeneous supporting activity pose major challenges. While three-dimensional (3D) MSC cultures are emerging as alternative approaches, key changes in cellular characteristics during 3D-spheroid formation remain unclear. Here, we show that MSCs in 3D spheroids undergo further progression towards the epithelial-mesenchymal transition (EMT), driven by upregulation of EMT-promoting microRNAs and suppression of EMT-inhibitory miRNAs. The shift of EMT in MSCs is associated with widespread histone modifications mimicking the epigenetic reprogramming towards enhanced chromatin dynamics and stem cell-like properties, but without changes in their surface phenotype. Notably, these molecular shifts towards EMT in 3D MSCs caused enhanced stem cell niche activity, resulting in higher stimulation of hematopoietic progenitor self-renewal and cancer stem cell metastasis. Moreover, miRNA-mediated induction of EMT in 2D MSCs were sufficient to mimic the enhanced niche activity of 3D spheroid MSCs. Thus, the molecular hierarchy in the EMT gradient among phenotypically indistinguishable MSCs revealed the previously unrecognized functional parameters in MSCs, and the EMT-enhanced “naïve” mesenchymal state represents an ‘activated mesenchymal niche’ in 3D spheroid MSCs.
Collapse
|
247
|
Balikov DA, Crowder SW, Boire TC, Lee JB, Gupta MK, Fenix AM, Lewis HN, Ambrose CM, Short PA, Kim CS, Burnette DT, Reilly MA, Murthy NS, Kang ML, Kim WS, Sung HJ. Tunable Surface Repellency Maintains Stemness and Redox Capacity of Human Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22994-23006. [PMID: 28621931 PMCID: PMC5687519 DOI: 10.1021/acsami.7b06103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Human bone marrow derived mesenchymal stem cells (hMSCs) hold great promise for regenerative medicine due to their multipotent differentiation capacity and immunomodulatory capabilities. Substantial research has elucidated mechanisms by which extracellular cues regulate hMSC fate decisions, but considerably less work has addressed how material properties can be leveraged to maintain undifferentiated stem cells. Here, we show that synthetic culture substrates designed to exhibit moderate cell-repellency promote high stemness and low oxidative stress-two indicators of naïve, healthy stem cells-in commercial and patient-derived hMSCs. Furthermore, the material-mediated effect on cell behavior can be tuned by altering the molar percentage (mol %) and/or chain length of poly(ethylene glycol) (PEG), the repellant block linked to hydrophobic poly(ε-caprolactone) (PCL) in the copolymer backbone. Nano- and angstrom-scale characterization of the cell-material interface reveals that PEG interrupts the adhesive PCL domains in a chain-length-dependent manner; this prevents hMSCs from forming mature focal adhesions and subsequently promotes cell-cell adhesions that require connexin-43. This study is the first to demonstrate that intrinsic properties of synthetic materials can be tuned to regulate the stemness and redox capacity of hMSCs and provides new insight for designing highly scalable, programmable culture platforms for clinical translation.
Collapse
Affiliation(s)
- Daniel A. Balikov
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Spencer W. Crowder
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Timothy C. Boire
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jung Bok Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Aidan M. Fenix
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Holley N. Lewis
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Caitlyn M. Ambrose
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Philip A. Short
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Chang Soo Kim
- Department of Materials Science and Engineering, University of Wisconsin, Milwaukee, Wisconsin 53211, United States
| | - Dylan T. Burnette
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Matthew A. Reilly
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - N. Sanjeeva Murthy
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Mi-Lan Kang
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Won Shik Kim
- Department of Otorhinolaryngology, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center for Stem Cell Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul 120-752, Republic of Korea
| |
Collapse
|
248
|
Hanga MP, Murasiewicz H, Pacek AW, Nienow AW, Coopman K, Hewitt CJ. Expansion of bone marrow-derived human mesenchymal stem/stromal cells (hMSCs) using a two-phase liquid/liquid system. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017; 92:1577-1589. [PMID: 28706339 PMCID: PMC5485050 DOI: 10.1002/jctb.5279] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 05/05/2023]
Abstract
BACKGROUND Human mesenchymal stem/stromal cells (hMSCs) are at the forefront of regenerative medicine applications due to their relatively easy isolation and availability in adults, potential to differentiate and to secrete a range of trophic factors that could determine specialised tissue regeneration. To date, hMSCs have been successfully cultured in vitro on substrates such as polystyrene dishes (TCPS) or microcarriers. However, hMSC sub-cultivation and harvest typically employs proteolytic enzymes that act by cleaving important cell membrane proteins resulting in long-term cell damage. In a process where the cells themselves are the product, a non-enzymatic and non-damaging harvesting approach is desirable. RESULTS An alternative system for hMSC expansion and subsequent non-enzymatic harvest was investigated here. A liquid/liquid two-phase system was proposed, comprising a selected perfluorocarbon (FC40) and growth medium (DMEM). The cells exhibited similar cell morphologies compared with TCPS. Moreover, they retained their identity and differentiation potential post-expansion and post-harvest. Further, no significant difference was found when culturing hMSCs in the culture systems prepared with either fresh or recycled FC40 perfluorocarbon. CONCLUSIONS These findings make the FC40/DMEM system an attractive alternative for traditional cell culture substrates due to their ease of cell recovery and recyclability, the latter impacting on overall process costs. © 2017 The Authors. Journal of Chemical Technology & Biotechnology published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Mariana P Hanga
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Halina Murasiewicz
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- West Pomeranian University of Technology SzczecinFaculty of Chemical Technology and EngineeringSzczecinPoland
| | - Andrzej W Pacek
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| | - Alvin W Nienow
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Karen Coopman
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
| | - Christopher J Hewitt
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| |
Collapse
|
249
|
Hanga MP, Murasiewicz H, Pacek AW, Nienow AW, Coopman K, Hewitt CJ. Expansion of bone marrow-derived human mesenchymal stem/stromal cells (hMSCs) using a two-phase liquid/liquid system. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017. [PMID: 28706339 DOI: 10.1002/jctb.5279m] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Human mesenchymal stem/stromal cells (hMSCs) are at the forefront of regenerative medicine applications due to their relatively easy isolation and availability in adults, potential to differentiate and to secrete a range of trophic factors that could determine specialised tissue regeneration. To date, hMSCs have been successfully cultured in vitro on substrates such as polystyrene dishes (TCPS) or microcarriers. However, hMSC sub-cultivation and harvest typically employs proteolytic enzymes that act by cleaving important cell membrane proteins resulting in long-term cell damage. In a process where the cells themselves are the product, a non-enzymatic and non-damaging harvesting approach is desirable. RESULTS An alternative system for hMSC expansion and subsequent non-enzymatic harvest was investigated here. A liquid/liquid two-phase system was proposed, comprising a selected perfluorocarbon (FC40) and growth medium (DMEM). The cells exhibited similar cell morphologies compared with TCPS. Moreover, they retained their identity and differentiation potential post-expansion and post-harvest. Further, no significant difference was found when culturing hMSCs in the culture systems prepared with either fresh or recycled FC40 perfluorocarbon. CONCLUSIONS These findings make the FC40/DMEM system an attractive alternative for traditional cell culture substrates due to their ease of cell recovery and recyclability, the latter impacting on overall process costs. © 2017 The Authors. Journal of Chemical Technology & Biotechnology published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Mariana P Hanga
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Halina Murasiewicz
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- West Pomeranian University of Technology SzczecinFaculty of Chemical Technology and EngineeringSzczecinPoland
| | - Andrzej W Pacek
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| | - Alvin W Nienow
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Karen Coopman
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
| | - Christopher J Hewitt
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| |
Collapse
|
250
|
Hanga MP, Murasiewicz H, Pacek AW, Nienow AW, Coopman K, Hewitt CJ. Expansion of bone marrow-derived human mesenchymal stem/stromal cells (hMSCs) using a two-phase liquid/liquid system. JOURNAL OF CHEMICAL TECHNOLOGY AND BIOTECHNOLOGY (OXFORD, OXFORDSHIRE : 1986) 2017. [PMID: 28706339 DOI: 10.1002/jctb.5166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Human mesenchymal stem/stromal cells (hMSCs) are at the forefront of regenerative medicine applications due to their relatively easy isolation and availability in adults, potential to differentiate and to secrete a range of trophic factors that could determine specialised tissue regeneration. To date, hMSCs have been successfully cultured in vitro on substrates such as polystyrene dishes (TCPS) or microcarriers. However, hMSC sub-cultivation and harvest typically employs proteolytic enzymes that act by cleaving important cell membrane proteins resulting in long-term cell damage. In a process where the cells themselves are the product, a non-enzymatic and non-damaging harvesting approach is desirable. RESULTS An alternative system for hMSC expansion and subsequent non-enzymatic harvest was investigated here. A liquid/liquid two-phase system was proposed, comprising a selected perfluorocarbon (FC40) and growth medium (DMEM). The cells exhibited similar cell morphologies compared with TCPS. Moreover, they retained their identity and differentiation potential post-expansion and post-harvest. Further, no significant difference was found when culturing hMSCs in the culture systems prepared with either fresh or recycled FC40 perfluorocarbon. CONCLUSIONS These findings make the FC40/DMEM system an attractive alternative for traditional cell culture substrates due to their ease of cell recovery and recyclability, the latter impacting on overall process costs. © 2017 The Authors. Journal of Chemical Technology & Biotechnology published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
- Mariana P Hanga
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Halina Murasiewicz
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- West Pomeranian University of Technology SzczecinFaculty of Chemical Technology and EngineeringSzczecinPoland
| | - Andrzej W Pacek
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| | - Alvin W Nienow
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| | - Karen Coopman
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
| | - Christopher J Hewitt
- Centre for Biological EngineeringLoughborough UniversityLoughboroughUK
- Aston Medical Research InstituteAston UniversityBirminghamUK
| |
Collapse
|