1
|
Scala P, Lovecchio J, Lamparelli EP, Vitolo R, Giudice V, Giordano E, Selleri C, Rehak L, Maffulli N, Della Porta G. Myogenic commitment of human stem cells by myoblasts Co-culture: a static vs. a dynamic approach. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:49-58. [PMID: 35188030 DOI: 10.1080/21691401.2022.2039684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An in-vitro model of human bone marrow mesenchymal stem cells (hBM-MSCs) myogenic commitment by synergic effect of a differentiation media coupled with human primary skeletal myoblasts (hSkMs) co-culture was developed adopting both conventional static co-seeding and perfused culture systems. Static co-seeding provided a notable outcome in terms of gene expression with a significant increase of Desmin (141-fold) and Myosin heavy chain II (MYH2, 32-fold) at day 21, clearly detected also by semi-quantitative immunofluorescence. Under perfusion conditions, myogenic induction ability of hSkMs on hBM-MSCs was exerted by paracrine effect with an excellent gene overexpression and immunofluorescence detection of MYH2 protein; furthermore, due to the dynamic cell culture in separate wells, western blot data were acquired confirming a successful cell commitment at day 14. A significant increase of anti-inflammatory cytokine gene expression, including IL-10 and IL-4 (15-fold and 11-fold, respectively) at day 14, with respect to the pro-inflammatory cytokines IL-12A (7-fold at day 21) and IL-1β (1.4-fold at day 7) was also detected during dynamic culture, confirming the immunomodulatory activity of hBM-MSCs along with commitment events. The present study opens interesting perspectives on the use of dynamic culture based on perfusion as a versatile tool to study myogenic events and paracrine cross-talk compared to the simple co-seeding static culture.
Collapse
Affiliation(s)
- Pasqualina Scala
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - J Lovecchio
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy
| | - E P Lamparelli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - R Vitolo
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - V Giudice
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - E Giordano
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy.,Advanced Research Center on Electronic Systems (ARCES), University of Bologna, Via Vincenzo Toffano 2/2, 40125 Bologna (BO), Italy
| | - C Selleri
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - L Rehak
- Athena Biomedical innovations, Viale Europa 139, Florence (FI), 50126, Italy
| | - N Maffulli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - G Della Porta
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy.,Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, Salerno (SA), Italy
| |
Collapse
|
2
|
Lowen GB, Vanderburgh JP, Florian D, Scott T, Sterling JAR, Guelcher SA. A Perfusion Bioreactor Model of Tumor-Induced Bone Disease Using Human Cells. Curr Protoc 2022; 2:e333. [PMID: 34985830 DOI: 10.1002/cpz1.333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Advanced solid tumors often metastasize to bone. Once established in bone, these tumors can induce bone destruction resulting in decreased quality of life and increased mortality. Neither 2D in vitro models nor 3D animal models sufficiently recapitulate the human bone-tumor microenvironment needed to fully understand the complexities of bone metastasis, highlighting the need for new models. A 3D in vitro humanized model of tumor-induced bone disease was developed by dynamically culturing human osteoblast, osteoclast, and metastatic cancer cells together within tissue-engineered bone constructs. Cell-mediated resorption can be observed by micro-computed tomography and can be quantified by change in mass. Taken together, these data can be used to investigate whether the metastatic cancer cells included in the model have the potential to drive osteoclastogenesis and cell-mediated resorption in vitro. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Fabricating bone-like scaffolds Basic Protocol 2: Preparing cells for the humanized model of TIBD Basic Protocol 3: Crafting a 3D in vitro humanized model of TIBD.
Collapse
Affiliation(s)
- Gregory B Lowen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph P Vanderburgh
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David Florian
- Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Taylor Scott
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Julie A Rhoades Sterling
- Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Scott A Guelcher
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee.,Vanderbilt Center for Bone Biology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
3
|
Paz C, Suárez E, Gil C, Parga O. Numerical modelling of osteocyte growth on different bone tissue scaffolds. Comput Methods Biomech Biomed Engin 2021; 25:641-655. [PMID: 34459293 DOI: 10.1080/10255842.2021.1972290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The most common solution for the regeneration or replacement of damaged bones is the implantation of prostheses comprising ceramic or metallic materials. However, these implants are known to cause problems such as post-operative infections, collapse of the prosthesis, and lack of osseointegration. Consequently, bone tissue engineering was established because of the limitations of such implants. Osteogenic implants offer promising solutions for bone regeneration; however, three-dimensional scaffolds should be used as supportive structures. It is challenging to correctly design these structures and their compositions or properties to provide a microenvironment that promotes tissue regeneration and expedites bone formation. Computational fluid dynamics can be used to model the main phenomena that occur in bioreactors, such as cell metabolism, nutrient transport, and cell culture growth, or to model the influence of several key mechanisms related to the fluid medium, in particular, the wall shear stress. In this work, a new numerical bone cell growth model was developed, which considered the oxygen and nutrient consumption as well as the wall shear stress effect on cell proliferation. The model was implemented using 35 three-dimensional scaffolds of different porosities, and the effect of the main geometrical parameters involved in each scaffold type was analysed. The porosity plays an important role, however, a similar porosity did not guarantee similar shear stress or cell growth among the scaffolds. Randomised trabecular scaffolds, that more closely resembled trabecular bone, showed the highest cell growth values, so these are the best candidates for cell growth in a bioreactor.
Collapse
Affiliation(s)
- Concepción Paz
- CINTECX, Universidade de Vigo, Campus Universitario Lagoas-Marcosende, Vigo, España.,Biofluids Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Eduardo Suárez
- CINTECX, Universidade de Vigo, Campus Universitario Lagoas-Marcosende, Vigo, España.,Biofluids Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
| | - Christian Gil
- CINTECX, Universidade de Vigo, Campus Universitario Lagoas-Marcosende, Vigo, España
| | - Oscar Parga
- CINTECX, Universidade de Vigo, Campus Universitario Lagoas-Marcosende, Vigo, España
| |
Collapse
|
4
|
Lim KT, Patel DK, Dutta SD, Ganguly K. Fluid Flow Mechanical Stimulation-Assisted Cartridge Device for the Osteogenic Differentiation of Human Mesenchymal Stem Cells. MICROMACHINES 2021; 12:927. [PMID: 34442549 PMCID: PMC8398302 DOI: 10.3390/mi12080927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have the potential to differentiate into different types of mesodermal tissues. In vitro proliferation and differentiation of hMSCs are necessary for bone regeneration in tissue engineering. The present study aimed to design and develop a fluid flow mechanically-assisted cartridge device to enhance the osteogenic differentiation of hMSCs. We used the fluorescence-activated cell-sorting method to analyze the multipotent properties of hMSCs and found that the cultured cells retained their stemness potential. We also evaluated the cell viabilities of the cultured cells via water-soluble tetrazolium salt 1 (WST-1) assay under different rates of flow (0.035, 0.21, and 0.35 mL/min) and static conditions and found that the cell growth rate was approximately 12% higher in the 0.035 mL/min flow condition than the other conditions. Moreover, the cultured cells were healthy and adhered properly to the culture substrate. Enhanced mineralization and alkaline phosphatase activity were also observed under different perfusion conditions compared to the static conditions, indicating that the applied conditions play important roles in the proliferation and differentiation of hMSCs. Furthermore, we determined the expression levels of osteogenesis-related genes, including the runt-related protein 2 (Runx2), collagen type I (Col1), osteopontin (OPN), and osteocalcin (OCN), under various perfusion vis-à-vis static conditions and found that they were significantly affected by the applied conditions. Furthermore, the fluorescence intensities of OCN and OPN osteogenic gene markers were found to be enhanced in the 0.035 mL/min flow condition compared to the control, indicating that it was a suitable condition for osteogenic differentiation. Taken together, the findings of this study reveal that the developed cartridge device promotes the proliferation and differentiation of hMSCs and can potentially be used in the field of tissue engineering.
Collapse
Affiliation(s)
- Ki-Taek Lim
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
- Biomechagen Co., Ltd., Chuncheon 24341, Korea
| | - Dinesh-K. Patel
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Sayan-Deb Dutta
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| | - Keya Ganguly
- Department of Biosystems Engineering, Institute of Forest Science, Kangwon National University, Chuncheon 24341, Korea; (D.-K.P.); (S.-D.D.); (K.G.)
| |
Collapse
|
5
|
Rozila I, Azari P, Munirah S, Safwani WKZW, Pingguan-Murphy B, Chua KH. Polycaprolactone-Based Scaffolds Facilitates Osteogenic Differentiation of Human Adipose-Derived Stem Cells in a Co-Culture System. Polymers (Basel) 2021; 13:polym13040597. [PMID: 33671175 PMCID: PMC7922582 DOI: 10.3390/polym13040597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Stem cells in combination with scaffolds and bioactive molecules have made significant contributions to the regeneration of damaged bone tissues. A co-culture system can be effective in enhancing the proliferation rate and osteogenic differentiation of the stem cells. Hence, the aim of this study was to investigate the osteogenic differentiation of human adipose derived stem cells when co-cultured with human osteoblasts and seeded on polycaprolactone (PCL):hydroxyapatite (HA) scaffold; (2) Methods: Human adipose-derived stem cells (ASC) and human osteoblasts (HOB) were seeded in three different ratios of 1:2, 1:2 and 2:1 in the PCL-HA scaffolds. The osteogenic differentiation ability was evaluated based on cell morphology, proliferation rate, alkaline phosphatase (ALP) activity, calcium deposition and osteogenic genes expression levels using quantitative RT-PCR; (3) Results: The co-cultured of ASC/HOB in ratio 2:1 seeded on the PCL-HA scaffolds showed the most positive osteogenic differentiation as compared to other groups, which resulted in higher ALP activity, calcium deposition and osteogenic genes expression, particularly Runx, ALP and BSP. These genes indicate that the co-cultured ASC/HOB seeded on PCL-HA was at the early stage of osteogenic development; (4) Conclusions: The combination of co-culture system (ASC/HOB) and PCL-HA scaffolds promote osteogenic differentiation and early bone formation.
Collapse
Affiliation(s)
- Ismail Rozila
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Cyberjaya, Selangor 63000, Malaysia
| | - Pedram Azari
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sha’ban Munirah
- Department of Rehabilitation and Physiotherapy, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, Kuantan 25200, Malaysia;
| | - Wan Kamarul Zaman Wan Safwani
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Belinda Pingguan-Murphy
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (W.K.Z.W.S.); (B.P.-M.)
| | - Kien Hui Chua
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
- Correspondence: ; Tel.: +60-391458613
| |
Collapse
|
6
|
Han TTY, Flynn LE. Perfusion bioreactor culture of human adipose‐derived stromal cells on decellularized adipose tissue scaffolds enhances in vivo adipose tissue regeneration. J Tissue Eng Regen Med 2020; 14:1827-1840. [DOI: 10.1002/term.3133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/02/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Tim Tian Y. Han
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry The University of Western Ontario London Ontario Canada
| | - Lauren E. Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry The University of Western Ontario London Ontario Canada
- Department of Chemical and Biochemical Engineering, Thompson Engineering Building The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| |
Collapse
|
7
|
Size-Optimized Microspace Culture Facilitates Differentiation of Mouse Induced Pluripotent Stem Cells into Osteoid-Rich Bone Constructs. Stem Cells Int 2020; 2020:7082679. [PMID: 32508932 PMCID: PMC7244985 DOI: 10.1155/2020/7082679] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/29/2020] [Accepted: 02/15/2020] [Indexed: 01/15/2023] Open
Abstract
Microspace culture is promising for self-organization of induced pluripotent stem cells (iPSCs). However, the optimal size of microspaces for osteogenic differentiation is unclear. We hypothesized that a specific microspace size could facilitate self-organizing iPSC differentiation to form bone-like tissue in vitro. The objectives of this study were to investigate such effects of microspace size and to evaluate bone regeneration upon transplantation of the resulting osteogenic constructs. Dissociated mouse gingival fibroblast-derived iPSCs were plated in ultra-low-attachment microspace culture wells containing hundreds of U-bottom-shaped microwell spots per well to form cell aggregates in growth medium. The microwells had different aperture diameters/depths (400/560 μm (Elp400), 500/700 μm (Elp500), and 900/700 μm (Elp900)) (Kuraray; Elplasia). After 5 days of aggregation, cells were maintained in osteogenic induction medium for 35 days. Only cells in the Elp500 condition tightly aggregated and maintained high viability during osteogenic induction. After 10 days of induction, Elp500 cell constructs showed significantly higher gene expression of Runx2, Osterix, Collagen 1a1, Osteocalcin, Bone sialoprotein, and Osteopontin compared to constructs in Elp400 and Elp900. In methylene blue-counterstained von Kossa staining and Movat's pentachrome staining, only Elp500 constructs showed robust osteoid formation on day 35, with high expression of type I collagen (a major osteoid component) and osteocalcin proteins. Cell constructs were transplanted into rat calvarial bone defects, and micro-CT analysis after 3 weeks showed better bone repair with significantly higher bone mineral density in the Elp500 group compared to the Elp900 group. These results suggest that microspace size affects self-organized osteogenic differentiation of iPSCs. Elp500 microspace culture specifically induces mouse iPSCs into osteoid-rich bone-like tissue possessing high bone regeneration capacity.
Collapse
|
8
|
He T, Hausdorf J, Chevalier Y, Klar RM. Trauma induced tissue survival in vitro with a muscle-biomaterial based osteogenic organoid system: a proof of concept study. BMC Biotechnol 2020; 20:8. [PMID: 32005149 PMCID: PMC6995208 DOI: 10.1186/s12896-020-0602-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The translation from animal research into the clinical environment remains problematic, as animal systems do not adequately replicate the human in vivo environment. Bioreactors have emerged as a good alternative that can reproduce part of the human in vivo processes at an in vitro level. However, in vitro bone formation platforms primarily utilize stem cells only, with tissue based in vitro systems remaining poorly investigated. As such, the present pilot study explored the tissue behavior and cell survival capability within a new in vitro skeletal muscle tissue-based biomaterial organoid bioreactor system to maximize future bone tissue engineering prospects. RESULTS Three dimensional printed β-tricalcium phosphate/hydroxyapatite devices were either wrapped in a sheet of rat muscle tissue or first implanted in a heterotopic muscle pouch that was then excised and cultured in vitro for up to 30 days. Devices wrapped in muscle tissue showed cell death by day 15. Contrarily, devices in muscle pouches showed angiogenic and limited osteogenic gene expression tendencies with consistent TGF-ß1, COL4A1, VEGF-A, RUNX-2, and BMP-2 up-regulation, respectively. Histologically, muscle tissue degradation and fibrin release was seen being absorbed by devices acting possibly as a support for new tissue formation in the bioceramic scaffold that supports progenitor stem cell osteogenic differentiation. CONCLUSIONS These results therefore demonstrate that the skeletal muscle pouch-based biomaterial culturing system can support tissue survival over a prolonged culture period and represents a novel organoid tissue model that with further adjustments could generate bone tissue for direct clinical transplantations.
Collapse
Affiliation(s)
- Tao He
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany. .,Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jörg Hausdorf
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany
| | - Yan Chevalier
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany
| | - Roland M Klar
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital of Munich (LMU), Munich, Germany.
| |
Collapse
|
9
|
A fully automated bioreactor system for precise control of stem cell proliferation and differentiation. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Rosenberg JT, Yuan X, Helsper SN, Bagdasarian FA, Ma T, Grant SC. Effects of labeling human mesenchymal stem cells with superparamagnetic iron oxides on cellular functions and magnetic resonance contrast in hypoxic environments and long-term monitoring. Brain Circ 2018; 4:133-138. [PMID: 30450421 PMCID: PMC6187941 DOI: 10.4103/bc.bc_18_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/27/2018] [Accepted: 09/10/2018] [Indexed: 01/25/2023] Open
Abstract
Ischemia, which involves decreased blood flow to a region and a corresponding deprivation of oxygen and nutrients, can be induced as a consequence of stroke or heart attack. A prevalent disease that affects many individuals worldwide, ischemic stroke results in functional and cognitive impairments, as neural cells in the brain receive inadequate nourishment and encounter inflammation and various other detrimental toxic factors that lead to their death. Given the scarce treatments for this disease in the clinic such as the administration of tissue plasminogen activator, which is only effective in a limited time window after the occurrence of stroke, it will be necessary to develop new strategies to ameliorate or prevent stroke-induced brain damage. Cell-based therapies appear to be a promising solution for treating ischemic stroke and many other ischemia-associated and neurodegenerative maladies. Particularly, human mesenchymal stem cells (hMSCs) are of interest for cell transplantation in stroke, given their multipotency, accessibility, and reparative abilities. To determine the fate and survival of hMSC, which will be imperative for successful transplantation therapies, these cells may be monitored using magnetic resonance imaging and transfected with superparamagnetic iron oxide (SPIO), a contrast agent that facilitates the detection of these hMSCs. This review encompasses pertinent research and findings to reveal the effects of SPIO on hMSC functions in the context of transplantation in ischemic environments and over extended time periods. This paper is a review article. Referred literature in this paper has been listed in the references section. The data sets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors' experiences.
Collapse
Affiliation(s)
- Jens T Rosenberg
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Shannon N Helsper
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - F Andrew Bagdasarian
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Samuel C Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA.,The National High Magnetic Field Laboratory, CIMAR, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
11
|
Li Q, Zhang B, Kasoju N, Ma J, Yang A, Cui Z, Wang H, Ye H. Differential and Interactive Effects of Substrate Topography and Chemistry on Human Mesenchymal Stem Cell Gene Expression. Int J Mol Sci 2018; 19:E2344. [PMID: 30096912 PMCID: PMC6121573 DOI: 10.3390/ijms19082344] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
Abstract
Variations in substrate chemistry and the micro-structure were shown to have a significant effect on the biology of human mesenchymal stromal cells (hMSCs). This occurs when differences in the surface properties indirectly modulate pathways within numerous signaling networks that control cell fate. To understand how the surface features affect hMSC gene expression, we performed RNA-sequencing analysis of bone marrow-derived hMSCs cultured on tissue culture-treated polystyrene (TCP) and poly(l-lactide) (PLLA) based substrates of differing topography (Fl: flat and Fs: fibrous) and chemistry (Pr: pristine and Am: aminated). Whilst 80% of gene expression remained similar for cells cultured on test substrates, the analysis of differentially expressed genes (DEGs) revealed that surface topography significantly altered gene expression more than surface chemistry. The Fl and Fs topologies introduced opposite directional alternations in gene expression when compared to TCP control. In addition, the effect of chemical treatment interacted with that of topography in a synergistic manner with the Pr samples promoting more DEGs than Am samples in all gene ontology function groups. These findings not only highlight the significance of the culture surface on regulating the overall gene expression profile but also provide novel insights into cell-material interactions that could help further design the next-generation biomaterials to facilitate hMSC applications. At the same time, further studies are required to investigate whether or not the observations noted correlate with subsequent protein expression and functionality of cells.
Collapse
Affiliation(s)
- Qiongfang Li
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
| | - Bo Zhang
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
- Department of Engineering Science, University of Oxford, OX1 3PJ Oxford, UK.
| | - Naresh Kasoju
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| | - Jinmin Ma
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, OX1 3PJ Oxford, UK.
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| | - Hui Wang
- China National GeneBank-Shenzhen, BGI-Shenzhen, 518083 Shenzhen, China.
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
- Oxford Suzhou Centre for Advanced Research, Suzhou Industrial Park, 215123 Suzhou, China.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, UK.
| |
Collapse
|
12
|
Rogina A, Antunović M, Pribolšan L, Caput Mihalić K, Vukasović A, Ivković A, Marijanović I, Gallego Ferrer G, Ivanković M, Ivanković H. Human Mesenchymal Stem Cells Differentiation Regulated by Hydroxyapatite Content within Chitosan-Based Scaffolds under Perfusion Conditions. Polymers (Basel) 2017; 9:E387. [PMID: 30965692 PMCID: PMC6418638 DOI: 10.3390/polym9090387] [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: 07/21/2017] [Revised: 08/18/2017] [Accepted: 08/21/2017] [Indexed: 11/24/2022] Open
Abstract
The extensive need for hard tissue substituent greatly motivates development of suitable allogeneic grafts for therapeutic recreation. Different calcium phosphate phases have been accepted as scaffold's components with positive influence on osteoinduction and differentiation of human mesenchymal stem cells, in terms of their higher fraction within the graft. Nevertheless, the creation of unlimited nutrients diffusion through newly formed grafts is of great importance. The media flow accomplished by perfusion forces can provide physicochemical, and also, biomechanical stimuli for three-dimensional bone-construct growth. In the present study, the influence of a different scaffold's composition on the human mesenchymal stem cells (hMSCs) differentiation performed in a U-CUP bioreactor under perfusion conditioning was investigated. The histological and immunohistochemical analysis of cultured bony tissues, and the evaluation of osteogenic genes' expression indicate that the lower fraction of in situ formed hydroxyapatite in the range of 10⁻30% within chitosan scaffold could be preferable for bone-construct development.
Collapse
Affiliation(s)
- Anamarija Rogina
- Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, p.p.177, 10001 Zagreb, Croatia.
| | - Maja Antunović
- Faculty of Science, University of Zagreb, Horvatovac102a, 10001 Zagreb, Croatia.
| | - Lidija Pribolšan
- Faculty of Science, University of Zagreb, Horvatovac102a, 10001 Zagreb, Croatia.
| | | | - Andreja Vukasović
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Šalata 3, 10001 Zagreb, Croatia.
| | - Alan Ivković
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Šalata 3, 10001 Zagreb, Croatia.
- Department of Orthopaedic Surgery, University Hospital, Sveti Duh, 10001 Zagreb, Croatia.
| | - Inga Marijanović
- Faculty of Science, University of Zagreb, Horvatovac102a, 10001 Zagreb, Croatia.
| | - Gloria Gallego Ferrer
- Centro de Biomateriales e Ingeniería Tisular, Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
- Biomedical Research Networking centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Mariano Esquillor s/n, 50018 Zaragoza, Spain.
| | - Marica Ivanković
- Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, p.p.177, 10001 Zagreb, Croatia.
| | - Hrvoje Ivanković
- Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, p.p.177, 10001 Zagreb, Croatia.
| |
Collapse
|
13
|
Forrestal DP, Klein TJ, Woodruff MA. Challenges in engineering large customized bone constructs. Biotechnol Bioeng 2017; 114:1129-1139. [PMID: 27858993 DOI: 10.1002/bit.26222] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/18/2016] [Accepted: 10/17/2016] [Indexed: 01/22/2023]
Abstract
The ability to treat large tissue defects with customized, patient-specific scaffolds is one of the most exciting applications in the tissue engineering field. While an increasing number of modestly sized tissue engineering solutions are making the transition to clinical use, successfully scaling up to large scaffolds with customized geometry is proving to be a considerable challenge. Managing often conflicting requirements of cell placement, structural integrity, and a hydrodynamic environment supportive of cell culture throughout the entire thickness of the scaffold has driven the continued development of many techniques used in the production, culturing, and characterization of these scaffolds. This review explores a range of technologies and methods relevant to the design and manufacture of large, anatomically accurate tissue-engineered scaffolds with a focus on the interaction of manufactured scaffolds with the dynamic tissue culture fluid environment. Biotechnol. Bioeng. 2017;114: 1129-1139. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David P Forrestal
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Travis J Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, Brisbane, QLD 4059, Australia
| | - Maria A Woodruff
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, Brisbane, QLD 4059, Australia
| |
Collapse
|
14
|
Abstract
Decellularized bone matrix is gaining a lot of attention as implantable biomaterials and/or biological scaffolds for bone tissue repair, and shows good clinical performance. This chapter describes the processing techniques and characterization protocols of decellularized bone. For the applications of the decellularized bone scaffold in promoting bone repair and regeneration, we discuss some of the current advances, and highlight the advantages and disadvantages of these scaffolds. Fabrication and application of the hydrogel derived from decellularized bone for bone tissue engineering are also presented.
Collapse
Affiliation(s)
- Guobao Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, People's Republic of China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing, 400044, People's Republic of China.
| |
Collapse
|
15
|
Rosenberg JT, Yuan X, Grant S, Ma T. Tracking mesenchymal stem cells using magnetic resonance imaging. Brain Circ 2016; 2:108-113. [PMID: 30276283 PMCID: PMC6126273 DOI: 10.4103/2394-8108.192521] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 08/05/2016] [Accepted: 08/30/2016] [Indexed: 01/12/2023] Open
Abstract
Recent translational studies in the fields of tissue regeneration and cell therapy have characterized mesenchymal stem cells (MSCs) as a potentially effective and accessible measure for treating ischemic cerebral and neurodegenerative disorders such as stroke, Parkinson's disease, and amyotrophic lateral sclerosis. Developing more efficient cell tracking techniques bear the potential to optimize MSC transplantation therapies by providing a more accurate picture of the fate and area of effect of implanted cells. Currently, determining the location of transplanted MSCs involves a histological approach, but magnetic resonance imaging (MRI) presents a noninvasive paradigm that permits repeat evaluations. To visualize MSCs using MRI, the implanted cells must be treated with an intracellular contrast agent. These are commonly paramagnetic compounds, many of which are based on superparamagnetic iron oxide (SPIO) nanoparticles. Recent research has set out characterize the effects of SPIO-uptake on the cellular activity of in vitro human MSCs and the resultant influence that respective SPIO concentration has on MRI sensitivity. As these studies reveal, SPIO-uptake has no effect on the cellular processes of proliferation and differentiation while producing high contrast MRI signals. Moreover, transplantation of SPIO-labeled MSCs in animal models encouragingly showed no loss in MRI contrast, suggesting that SPIO labeling may be an appealing regime for lasting MRI detection. This study is a review article. Referred literature in this study has been listed in the reference part. The datasets supporting the conclusions of this article are available online by searching the PubMed. Some original points in this article come from the laboratory practice in our research centers and the authors’ experiences.
Collapse
Affiliation(s)
- Jens T Rosenberg
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Xuegang Yuan
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Samuel Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA.,The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
16
|
Yaghoobi M, Hashemi-Najafabadi S, Soleimani M, Vasheghani-Farahani E, Mousavi SM. Osteogenic Differentiation and Mineralization on Compact Multilayer nHA-PCL Electrospun Scaffolds in a Perfusion Bioreactor. IRANIAN JOURNAL OF BIOTECHNOLOGY 2016; 14:41-49. [PMID: 28959325 DOI: 10.15171/ijb.1382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Monolayer electrospun scaffolds have already been used in bone tissue engineering due to their high surface-tovolume ratio, interconnectivity, similarity to natural bone extracellular matrix (ECM), and simple production. OBJECTIVES The aim of this study was to evaluate the dynamic culture effect on osteogenic differentiation and mineralizationi into a compact cellular multilayer nHA-PCL electrospun construct. The dynamic culture was compared with static culture. MATERIALS AND METHODS The calcium content, alkaline phosphatase (ALP) activity and cell viability were investigated on days 3 and 7. RESULTS When the dynamic culture compared to static culture, the mineralization and ALP activity were increased in dynamic culture. After 7 days, calcium contents were 41.24 and 20.44 μg.(cm3)-1, and also normalized ALP activity were 0.32 and 0.19 U.mg-1 in dynamic and static culture, respectively. Despite decreasing the cell viability until day 7, the scanning electron microscopy (SEM) results showed that, due to higher mineralization, a larger area of the construct was covered with calcium deposition in dynamic culture. CONCLUSIONS The dynamic flow could improve ALP activity and mineralization into the compact cellular multilayer construct cultured in the perfusion bioreactor after 7 days. Fluid flow of media helped to facilitate the nutrients transportation into the construct and created uniform cellular construct with high mineralization. This construct can be applied for bone tissue engineering.
Collapse
Affiliation(s)
- Maliheh Yaghoobi
- Biomedical Engineering Group, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | | | - Masoud Soleimani
- Hematology Group, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Seyyed Mohammad Mousavi
- Biotechnology Group, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
17
|
|
18
|
Nguyen BNB, Ko H, Fisher JP. Tunable osteogenic differentiation of hMPCs in tubular perfusion system bioreactor. Biotechnol Bioeng 2016; 113:1805-13. [PMID: 26724678 DOI: 10.1002/bit.25929] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 01/24/2023]
Abstract
The use of bioreactors for bone tissue engineering has been widely investigated. While the benefits of shear stress on osteogenic differentiation are well known, the underlying effects of dynamic culture on subpopulations within a bioreactor are less evident. In this work, we explore the influence of applied flow in the tubular perfusion system (TPS) bioreactor on the osteogenic differentiation of human mesenchymal progenitor cells (hMPCs), specifically analyzing the effects of axial position along the growth chamber. TPS bioreactor experiments conducted with unidirectional flow demonstrated enhanced expression of osteogenic markers in cells cultured downstream from the inlet flow. We utilized computational fluid dynamic modeling to confirm uniform shear stress distribution on the surface of the scaffolds and along the length of the growth chamber. The concept of paracrine signaling between cell populations was validated with the use of alternating flow, which diminished the differences in osteogenic differentiation between cells cultured at the inlet and outlet of the growth chamber. After the addition of controlled release of bone morphogenic protein-2 (BMP-2) into the system, osteogenic differentiation among subpopulations along the growth chamber was augmented, yet remained homogenous. These results allow for greater understanding of axial bioreactor cultures, their microenvironment, and how well-established parameters of osteogenic differentiation affect bone tissue development. With this work, we have demonstrated the capability of tuning osteogenic differentiation of hMPCs through the application of fluid flow and the addition of exogenous growth factors. Such precise control allows for the culture of distinct subpopulation within one dynamic system for the use of complex engineered tissue constructs. Biotechnol. Bioeng. 2016;113: 1805-1813. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bao-Ngoc B Nguyen
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building (# 225), College Park, Maryland, 20742
| | - Henry Ko
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building (# 225), College Park, Maryland, 20742
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building (# 225), College Park, Maryland, 20742.
| |
Collapse
|
19
|
Sart S, Agathos SN, Li Y, Ma T. Regulation of mesenchymal stem cell 3D microenvironment: From macro to microfluidic bioreactors. Biotechnol J 2015; 11:43-57. [PMID: 26696441 DOI: 10.1002/biot.201500191] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 11/02/2015] [Accepted: 11/30/2015] [Indexed: 12/12/2022]
Abstract
Human mesenchymal stem cells (hMSCs) have emerged as an important cell type in cell therapy and tissue engineering. In these applications, maintaining the therapeutic properties of hMSCs requires tight control of the culture environments and the structural cell organizations. Bioreactor systems are essential tools to achieve these goals in the clinical-scale expansion and tissue engineering applications. This review summarizes how different bioreactors provide cues to regulate the structure and the chemico-mechanical microenvironment of hMSCs with a focus on 3D organization. In addition to conventional bioreactors, recent advances in microfluidic bioreactors as a novel approach to better control the hMSC microenvironment are also discussed. These advancements highlight the key role of bioreactor systems in preserving hMSC's functional properties by providing dynamic and temporal regulation of in vitro cellular microenvironment.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory, CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Spiros N Agathos
- Laboratory of Bioengineering, Catholic University of Louvain, Louvain-la-Neuve, Belgium
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida, USA.
| |
Collapse
|
20
|
Leijten J, Chai Y, Papantoniou I, Geris L, Schrooten J, Luyten F. Cell based advanced therapeutic medicinal products for bone repair: Keep it simple? Adv Drug Deliv Rev 2015; 84:30-44. [PMID: 25451134 DOI: 10.1016/j.addr.2014.10.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 09/18/2014] [Accepted: 10/20/2014] [Indexed: 02/08/2023]
Abstract
The development of cell based advanced therapeutic medicinal products (ATMPs) for bone repair has been expected to revolutionize the health care system for the clinical treatment of bone defects. Despite this great promise, the clinical outcomes of the few cell based ATMPs that have been translated into clinical treatments have been far from impressive. In part, the clinical outcomes have been hampered because of the simplicity of the first wave of products. In response the field has set-out and amassed a plethora of complexities to alleviate the simplicity induced limitations. Many of these potential second wave products have remained "stuck" in the development pipeline. This is due to a number of reasons including the lack of a regulatory framework that has been evolving in the last years and the shortage of enabling technologies for industrial manufacturing to deal with these novel complexities. In this review, we reflect on the current ATMPs and give special attention to novel approaches that are able to provide complexity to ATMPs in a straightforward manner. Moreover, we discuss the potential tools able to produce or predict 'goldilocks' ATMPs, which are neither too simple nor too complex.
Collapse
|
21
|
Sellgren KL, Ma T. Effects of flow configuration on bone tissue engineering using human mesenchymal stem cells in 3D chitosan composite scaffolds. J Biomed Mater Res A 2014; 103:2509-20. [DOI: 10.1002/jbm.a.35386] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 11/20/2014] [Accepted: 12/04/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Katelyn L. Sellgren
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering, Florida State University; Tallahassee Florida 32310
| | - Teng Ma
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering, Florida State University; Tallahassee Florida 32310
| |
Collapse
|
22
|
Patil S, Paul S. A comprehensive review on the role of various materials in the osteogenic differentiation of mesenchymal stem cells with a special focus on the association of heat shock proteins and nanoparticles. Cells Tissues Organs 2014; 199:81-102. [PMID: 25401759 DOI: 10.1159/000362226] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2014] [Indexed: 11/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have important roles in the area of regenerative medicine and clinical applications due to their pluripotent nature. Osteogenic differentiation of MSCs has been studied extensively using various stimulants to develop models of bone repair. There are several factors that enhance the differentiation of MSCs into bone tissues. This review focuses on the effects of various inducers on the osteoblast differentiation of MSCs at different stages of cellular development. We discuss the various growth factors, hormones, vitamins, cytokines, chemical stimulants, and mechanical forces applied in bioreactors that play an essential role in the proliferation, differentiation, and matrix mineralization of stem cells during osteogenesis. Various nanoparticles have also been used recently for the same purpose and the results are promising. Moreover, we review the role of various stresses, including thermal stress, and the subsequent involvement of heat shock proteins as inducers of the proliferation and differentiation of osteoblasts. We also report how various proteasome inhibitors have been shown to induce proliferation and osteogenic differentiation of MSCs in a number of cases. In this communication, the role of peptide-based scaffolds in osteoblast proliferation and differentiation is also reviewed. Based on the reviewed information, this article proposes novel possibilities for the enhancement of proliferation, differentiation, and migration of osteoblasts from MSCs. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Supriya Patil
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, India
| | | |
Collapse
|
23
|
Sonnaert M, Papantoniou I, Bloemen V, Kerckhofs G, Luyten FP, Schrooten J. Human periosteal-derived cell expansion in a perfusion bioreactor system: proliferation, differentiation and extracellular matrix formation. J Tissue Eng Regen Med 2014; 11:519-530. [PMID: 25186024 DOI: 10.1002/term.1951] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 05/07/2014] [Accepted: 07/16/2014] [Indexed: 12/14/2022]
Abstract
Perfusion bioreactor systems have shown to be a valuable tool for the in vitro development of three-dimensional (3D) cell-carrier constructs. Their use for cell expansion, however, has been much less explored. Since maintenance of the initial cell phenotype is essential in this process, it is imperative to obtain insight into the bioreactor-related variables determining cell fate. Therefore, this study investigated the influence of fluid flow-induced shear stress on the proliferation, differentiation and matrix deposition of human periosteal-derived cells in the absence of additional differentiation-inducing stimuli; 120 000 cells were seeded on additive manufactured 3D Ti6Al4V scaffolds and cultured for up to 28 days at different flow rates in the range 0.04-6 ml/min. DNA measurements showed, on average, a three-fold increase in cell content for all perfused conditions in comparison to static controls, whereas the magnitude of the flow rate did not have an influence. Contrast-enhanced nanofocus X-ray computed tomography showed substantial formation of an engineered neotissue in all perfused conditions, resulting in a filling (up to 70%) of the total internal void volume, and no flow rate-dependent differences were observed. The expression of key osteogenic markers, such as RunX2, OCN, OPN and Col1, did not show any significant changes in comparison to static controls after 28 days of culture, with the exception of OSX at high flow rates. We therefore concluded that, in the absence of additional osteogenic stimuli, the investigated perfusion conditions increased cell proliferation but did not significantly enhance osteogenic differentiation, thus allowing for this process to be used for cell expansion. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- M Sonnaert
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Department of Materials Engineering, Katholieke Universiteit Leuven, Belgium
| | - I Papantoniou
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Skeletal Biology and Engineering Research Centre, Katholieke Universiteit Leuven, Belgium
| | - V Bloemen
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Biomedical Engineering Research Team, Groep T, Leuven Engineering College (Association Katholieke Universiteit Leuven), Belgium
| | - G Kerckhofs
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Department of Materials Engineering, Katholieke Universiteit Leuven, Belgium.,Biomechanics Research Unit, Université de Liege, Belgium
| | - F P Luyten
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Skeletal Biology and Engineering Research Centre, Katholieke Universiteit Leuven, Belgium
| | - J Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven, Belgium.,Department of Materials Engineering, Katholieke Universiteit Leuven, Belgium
| |
Collapse
|
24
|
Analysis of Gene Expression Signatures for Osteogenic 3D Perfusion-Bioreactor Cell Cultures Based on a Multifactorial DoE Approach. Processes (Basel) 2014. [DOI: 10.3390/pr2030639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
Lambrechts T, Papantoniou I, Sonnaert M, Schrooten J, Aerts JM. Model-based cell number quantification using online single-oxygen sensor data for tissue engineering perfusion bioreactors. Biotechnol Bioeng 2014; 111:1982-92. [PMID: 24771348 DOI: 10.1002/bit.25274] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/09/2014] [Accepted: 04/15/2014] [Indexed: 01/31/2023]
Abstract
Online and non-invasive quantification of critical tissue engineering (TE) construct quality attributes in TE bioreactors is indispensable for the cost-effective up-scaling and automation of cellular construct manufacturing. However, appropriate monitoring techniques for cellular constructs in bioreactors are still lacking. This study presents a generic and robust approach to determine cell number and metabolic activity of cell-based TE constructs in perfusion bioreactors based on single oxygen sensor data in dynamic perfusion conditions. A data-based mechanistic modeling technique was used that is able to correlate the number of cells within the scaffold (R(2) = 0.80) and the metabolic activity of the cells (R(2) = 0.82) to the dynamics of the oxygen response to step changes in the perfusion rate. This generic non-destructive measurement technique is effective for a large range of cells, from as low as 1.0 × 10(5) cells to potentially multiple millions of cells, and can open-up new possibilities for effective bioprocess monitoring.
Collapse
Affiliation(s)
- T Lambrechts
- Division M3-BIORES: Measure, Model & Manage Bioresponses, KU Leuven, Heverlee, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
26
|
Expansion of human amniotic fluid stem cells in 3-dimensional fibrous scaffolds in a stirred bioreactor. Biochem Eng J 2014. [DOI: 10.1016/j.bej.2013.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Sart S, Tsai AC, Li Y, Ma T. Three-dimensional aggregates of mesenchymal stem cells: cellular mechanisms, biological properties, and applications. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:365-80. [PMID: 24168395 DOI: 10.1089/ten.teb.2013.0537] [Citation(s) in RCA: 285] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) are primary candidates in cell therapy and tissue engineering and are being tested in clinical trials for a wide range of diseases. Originally isolated and expanded as plastic adherent cells, MSCs have intriguing properties of in vitro self-assembly into three-dimensional (3D) aggregates reminiscent of skeletal condensation in vivo. Recent studies have shown that MSC 3D aggregation improved a range of biological properties, including multilineage potential, secretion of therapeutic factors, and resistance against ischemic condition. Hence, the formation of 3D MSC aggregates has been explored as a novel strategy to improve cell delivery, functional activation, and in vivo retention to enhance therapeutic outcomes. This article summarizes recent reports of MSC aggregate self-assembly, characterization of biological properties, and their applications in preclinical models. The cellular and molecular mechanisms underlying MSC aggregate formation and functional activation are discussed, and the areas that warrant further investigation are highlighted. These analyses are combined to provide perspectives for identifying the controlling mechanisms and refining the methods of aggregate fabrication and expansion for clinical applications.
Collapse
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | | | | | | |
Collapse
|
28
|
Teng S, Liu C, Krettek C, Jagodzinski M. The application of induced pluripotent stem cells for bone regeneration: current progress and prospects. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:328-39. [PMID: 24102431 DOI: 10.1089/ten.teb.2013.0301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Loss of healthy bone tissue and dysosteogenesis are still common and significant problems in clinics. Cell-based therapy using mesenchymal stem cells (MSCs) has been performed in patients for quite some time, but the inherent drawbacks of these cells, such as the reductions in proliferation rate and osteogenic differentiation potential that occur with aging, greatly limit their further application. Moreover, embryonic stem cells (ESCs) have brought new hope to osteoregenerative medicine because of their full pluripotent differentiation potential and excellent performance in bone regeneration. However, the ethical issues involved in destroying human embryos and the immune reactions that occur after transplantation are two major stumbling blocks impeding the clinical application of ESCs. Instead, induced pluripotent stem cells (iPSCs), which are ESC-like pluripotent cells that are reprogrammed from adult somatic cells using defined transcription factors, are considered a more promising source of cells for regenerative medicine because they present no ethical or immunological issues. Here, we summarize the primary technologies for generating iPSCs and the biological properties of these cells, review the current advances in iPSC-based bone regeneration and, finally, discuss the remaining challenges associated with these cells, particularly safety issues and their potential application for osteoregenerative medicine.
Collapse
Affiliation(s)
- Songsong Teng
- 1 Department of Orthopedic Trauma, Hanover Medical School (MHH) , Hanover, Germany
| | | | | | | |
Collapse
|
29
|
Liu M, Liu N, Zang R, Li Y, Yang ST. Engineering stem cell niches in bioreactors. World J Stem Cells 2013; 5:124-35. [PMID: 24179601 PMCID: PMC3812517 DOI: 10.4252/wjsc.v5.i4.124] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/05/2013] [Accepted: 07/04/2013] [Indexed: 02/06/2023] Open
Abstract
Stem cells, including embryonic stem cells, induced pluripotent stem cells, mesenchymal stem cells and amniotic fluid stem cells have the potential to be expanded and differentiated into various cell types in the body. Efficient differentiation of stem cells with the desired tissue-specific function is critical for stem cell-based cell therapy, tissue engineering, drug discovery and disease modeling. Bioreactors provide a great platform to regulate the stem cell microenvironment, known as "niches", to impact stem cell fate decision. The niche factors include the regulatory factors such as oxygen, extracellular matrix (synthetic and decellularized), paracrine/autocrine signaling and physical forces (i.e., mechanical force, electrical force and flow shear). The use of novel bioreactors with precise control and recapitulation of niche factors through modulating reactor operation parameters can enable efficient stem cell expansion and differentiation. Recently, the development of microfluidic devices and microbioreactors also provides powerful tools to manipulate the stem cell microenvironment by adjusting flow rate and cytokine gradients. In general, bioreactor engineering can be used to better modulate stem cell niches critical for stem cell expansion, differentiation and applications as novel cell-based biomedicines. This paper reviews important factors that can be more precisely controlled in bioreactors and their effects on stem cell engineering.
Collapse
Affiliation(s)
- Meimei Liu
- Meimei Liu, Ning Liu, Ru Zang, Shang-Tian Yang, William G Lowrie Department of Chemical and Biomolecular Engineering, Ohio State University, Columbus, OH 43210, United States
| | | | | | | | | |
Collapse
|
30
|
Gardel LS, Serra LA, Reis RL, Gomes ME. Use of perfusion bioreactors and large animal models for long bone tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:126-46. [PMID: 23924374 DOI: 10.1089/ten.teb.2013.0010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue engineering and regenerative medicine (TERM) strategies for generation of new bone tissue includes the combined use of autologous or heterologous mesenchymal stem cells (MSC) and three-dimensional (3D) scaffold materials serving as structural support for the cells, that develop into tissue-like substitutes under appropriate in vitro culture conditions. This approach is very important due to the limitations and risks associated with autologous, as well as allogenic bone grafiting procedures currently used. However, the cultivation of osteoprogenitor cells in 3D scaffolds presents several challenges, such as the efficient transport of nutrient and oxygen and removal of waste products from the cells in the interior of the scaffold. In this context, perfusion bioreactor systems are key components for bone TERM, as many recent studies have shown that such systems can provide dynamic environments with enhanced diffusion of nutrients and therefore, perfusion can be used to generate grafts of clinically relevant sizes and shapes. Nevertheless, to determine whether a developed tissue-like substitute conforms to the requirements of biocompatibility, mechanical stability and safety, it must undergo rigorous testing both in vitro and in vivo. Results from in vitro studies can be difficult to extrapolate to the in vivo situation, and for this reason, the use of animal models is often an essential step in the testing of orthopedic implants before clinical use in humans. This review provides an overview of the concepts, advantages, and challenges associated with different types of perfusion bioreactor systems, particularly focusing on systems that may enable the generation of critical size tissue engineered constructs. Furthermore, this review discusses some of the most frequently used animal models, such as sheep and goats, to study the in vivo functionality of bone implant materials, in critical size defects.
Collapse
Affiliation(s)
- Leandro S Gardel
- 1 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho , Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
| | | | | | | |
Collapse
|
31
|
Yeatts AB, Both SK, Yang W, Alghamdi HS, Yang F, Fisher JP, Jansen JA. In vivo bone regeneration using tubular perfusion system bioreactor cultured nanofibrous scaffolds. Tissue Eng Part A 2013; 20:139-46. [PMID: 23865551 DOI: 10.1089/ten.tea.2013.0168] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The use of bioreactors for the in vitro culture of constructs for bone tissue engineering has become prevalent as these systems may improve the growth and differentiation of a cultured cell population. Here we utilize a tubular perfusion system (TPS) bioreactor for the in vitro culture of human mesenchymal stem cells (hMSCs) and implant the cultured constructs into rat femoral condyle defects. Using nanofibrous electrospun poly(lactic-co-glycolic acid)/poly(ε-caprolactone) scaffolds, hMSCs were cultured for 10 days in vitro in the TPS bioreactor with cellular and acellular scaffolds cultured statically for 10 days as a control. After 3 and 6 weeks of in vivo culture, explants were removed and subjected to histomorphometric analysis. Results indicated more rapid bone regeneration in defects implanted with bioreactor cultured scaffolds with a new bone area of 1.23 ± 0.35 mm(2) at 21 days compared to 0.99 ± 0.43 mm(2) and 0.50 ± 0.29 mm(2) in defects implanted with statically cultured scaffolds and acellular scaffolds, respectively. At the 21 day timepoint, statistical differences (p<0.05) were only observed between defects implanted with cell containing scaffolds and the acellular control. After 42 days, however, defects implanted with TPS cultured scaffolds had the greatest new bone area with 1.72 ± 0.40 mm(2). Defects implanted with statically cultured and acellular scaffolds had a new bone area of 1.26 ± 0.43 mm(2) and 1.19 ± 0.33 mm(2), respectively. The increase in bone growth observed in defects implanted with TPS cultured scaffolds was statistically significant (p<0.05) when compared to both the static and acellular groups at this timepoint. This study demonstrates the efficacy of the TPS bioreactor to improve bone tissue regeneration and highlights the benefits of utilizing perfusion bioreactor systems to culture MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Andrew B Yeatts
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | | | | | | | | | | | | |
Collapse
|
32
|
Liu N, Zang R, Yang ST, Li Y. Stem cell engineering in bioreactors for large-scale bioprocessing. Eng Life Sci 2013. [DOI: 10.1002/elsc.201300013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Ning Liu
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Ru Zang
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Shang-Tian Yang
- William G. Lowrie Department of Chemical and Biomolecular Engineering; Ohio State University; Columbus OH USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering; Florida State University; Tallahassee FL USA
| |
Collapse
|
33
|
Ceccarelli G, Bloise N, Mantelli M, Gastaldi G, Fassina L, De Angelis MGC, Ferrari D, Imbriani M, Visai L. A comparative analysis of the in vitro effects of pulsed electromagnetic field treatment on osteogenic differentiation of two different mesenchymal cell lineages. Biores Open Access 2013; 2:283-94. [PMID: 23914335 PMCID: PMC3731679 DOI: 10.1089/biores.2013.0016] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are a promising candidate cell type for regenerative medicine and tissue engineering applications. Exposure of MSCs to physical stimuli favors early and rapid activation of the tissue repair process. In this study we investigated the in vitro effects of pulsed electromagnetic field (PEMF) treatment on the proliferation and osteogenic differentiation of bone marrow MSCs (BM-MSCs) and adipose-tissue MSCs (ASCs), to assess if both types of MSCs could be indifferently used in combination with PEMF exposure for bone tissue healing. We compared the cell viability, cell matrix distribution, and calcified matrix production in unstimulated and PEMF-stimulated (magnetic field: 2 mT, amplitude: 5 mV) mesenchymal cell lineages. After PEMF exposure, in comparison with ASCs, BM-MSCs showed an increase in cell proliferation (p<0.05) and an enhanced deposition of extracellular matrix components such as decorin, fibronectin, osteocalcin, osteonectin, osteopontin, and type-I and -III collagens (p<0.05). Calcium deposition was 1.5-fold greater in BM-MSC-derived osteoblasts (p<0.05). The immunofluorescence related to the deposition of bone matrix proteins and calcium showed their colocalization to the cell-rich areas for both types of MSC-derived osteoblast. Alkaline phosphatase activity increased nearly 2-fold (p<0.001) and its protein content was 1.2-fold higher in osteoblasts derived from BM-MSCs. The quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis revealed up-regulated transcription specific for bone sialoprotein, osteopontin, osteonectin, and Runx2, but at a higher level for cells differentiated from BM-MSCs. All together these results suggest that PEMF promotion of bone extracellular matrix deposition is more efficient in osteoblasts differentiated from BM-MSCs.
Collapse
Affiliation(s)
- Gabriele Ceccarelli
- Department of Public Health, Neuroscience, and Experimental & Forensic Medicine, University of Pavia , Pavia, Italy . ; Center for Tissue Engineering (C.I.T.), University of Pavia , Pavia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim J, Ma T. Autocrine fibroblast growth factor 2-mediated interactions between human mesenchymal stem cells and the extracellular matrix under varying oxygen tension. J Cell Biochem 2013; 114:716-27. [PMID: 23060043 DOI: 10.1002/jcb.24413] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 09/26/2012] [Indexed: 01/27/2023]
Abstract
Human mesenchymal stromal or stem cells (hMSCs) are being investigated for cell therapy in a wide range of diseases. MSCs are a potent source of trophic factors and actively remodel their immediate microenvironment through the secretion of bioactive factors in response to external stimuli such as oxygen tension. In this study, we examined the hypothesis that hypoxia influences hMSC properties in part through the regulation of extracellular milieu characterized by the extracellular matrix (ECM) matrices and the associated fibroblast growth factor-2 (FGF-2). The decellularized ECM matrices derived from hMSC culture under both hypoxic (e.g., 2% O(2)) and the standard culture (e.g., 20% O(2)) conditions have different binding capacities to the cell-secreted and exogenenous FGF-2. The reduced hMSC proliferation in the presence of FGF-2 inhibitor and the differential capacity of the decellularized ECM matrices in regulating hMSC osteogeneic and adipogenic differentiation suggest an important role of the endogenous FGF-2 in sustaining hMSC proliferation and regulating hMSC fate. Additionally, the combination of the ECM adhesion and hypoxic culture preserved hMSC viability under serum withdrawal. Together, the results suggest the synergistic effect of hypoxia and the ECM matrices in sustaining hMSC ex vivo expansion and preserving their multi-potentiality and viability under nutrient depletion. The results have important implication in optimizing hMSC expansion and delivery strategies to obtain hMSCs in sufficient quantity with required potency and to enhance survival and function upon transplantation.
Collapse
Affiliation(s)
- Junho Kim
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
| | | |
Collapse
|
35
|
Weyand B, Kasper C, Israelowitz M, Gille C, von Schroeder HP, Reimers K, Vogt PM. A differential pressure laminar flow reactor supports osteogenic differentiation and extracellular matrix formation from adipose mesenchymal stem cells in a macroporous ceramic scaffold. Biores Open Access 2013; 1:145-56. [PMID: 23515420 PMCID: PMC3559213 DOI: 10.1089/biores.2012.9901] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We present a laminar flow reactor for bone tissue engineering that was developed based on a computational fluid dynamics model. The bioreactor design permits a laminar flow field through its specific internal shape. An integrated bypass system that prevents pressure build-up through bypass openings for pressure release allows for a constant pressure environment during the changing of permeability values that are caused by cellular growth within a porous scaffold. A macroporous ceramic scaffold, composed of zirconium dioxide, was used as a test biomaterial that studies adipose stem cell behavior within a controlled three-dimensional (3D) flow and pressure environment. The topographic structure of the material provided a basis for stem cell proliferation and differentiation toward the osteogenic lineage. Dynamic culture conditions in the bioreactor supported cell viability during long-term culture and induced cell cluster formation and extra-cellular matrix deposition within the porous scaffold, though no complete closure of the pores with new-formed tissue was observed. We postulate that our system is suitable for studying fluid shear stress effects on stem cell proliferation and differentiation toward bone formation in tissue-engineered 3D constructs.
Collapse
Affiliation(s)
- Birgit Weyand
- Laboratory of Experimental Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Hannover Medical School , Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
36
|
Rosenberg JT, Sellgren KL, Sachi-Kocher A, Calixto Bejarano F, Baird MA, Davidson MW, Ma T, Grant SC. Magnetic resonance contrast and biological effects of intracellular superparamagnetic iron oxides on human mesenchymal stem cells with long-term culture and hypoxic exposure. Cytotherapy 2013; 15:307-22. [DOI: 10.1016/j.jcyt.2012.10.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 10/08/2012] [Accepted: 10/15/2012] [Indexed: 12/01/2022]
|
37
|
Macro and microfluidic flows for skeletal regenerative medicine. Cells 2012; 1:1225-45. [PMID: 24710552 PMCID: PMC3901127 DOI: 10.3390/cells1041225] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/07/2012] [Accepted: 12/04/2012] [Indexed: 11/16/2022] Open
Abstract
Fluid flow has a great potential as a cell stimulatory tool for skeletal regenerative medicine, because fluid flow-induced bone cell mechanotransduction in vivo plays a critical role in maintaining healthy bone homeostasis. Applications of fluid flow for skeletal regenerative medicine are reviewed at macro and microscale. Macroflow in two dimensions (2D), in which flow velocity varies along the normal direction to the flow, has explored molecular mechanisms of bone forming cell mechanotransduction responsible for flow-regulated differentiation, mineralized matrix deposition, and stem cell osteogenesis. Though 2D flow set-ups are useful for mechanistic studies due to easiness in in situ and post-flow assays, engineering skeletal tissue constructs should involve three dimensional (3D) flows, e.g., flow through porous scaffolds. Skeletal tissue engineering using 3D flows has produced promising outcomes, but 3D flow conditions (e.g., shear stress vs. chemotransport) and scaffold characteristics should further be tailored. Ideally, data gained from 2D flows may be utilized to engineer improved 3D bone tissue constructs. Recent microfluidics approaches suggest a strong potential to mimic in vivo microscale interstitial flows in bone. Though there have been few microfluidics studies on bone cells, it was demonstrated that microfluidic platform can be used to conduct high throughput screening of bone cell mechanotransduction behavior under biomimicking flow conditions.
Collapse
|
38
|
Hunter KT, Ma T. In vitroevaluation of hydroxyapatite-chitosan-gelatin composite membrane in guided tissue regeneration. J Biomed Mater Res A 2012; 101:1016-25. [DOI: 10.1002/jbm.a.34396] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/10/2012] [Accepted: 07/25/2012] [Indexed: 11/11/2022]
|
39
|
Kim J, Ma T. Regulation of autocrine fibroblast growth factor-2 signaling by perfusion flow in 3D human mesenchymal stem cell constructs. Biotechnol Prog 2012; 28:1384-8. [PMID: 22848034 DOI: 10.1002/btpr.1604] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/18/2012] [Indexed: 02/06/2023]
Abstract
Perfusion bioreactor systems play a crucial role in mitigating nutrient limitation as well as providing biomechanical stimuli and redistributing regulatory macromolecules that influence human mesenchymal stem cells (hMSC) fate in three-dimensional (3D) scaffolds. As fibroblast growth factor-2 (FGF-2) is known to regulate hMSC phenotype, understanding the role of autocrine FGF-2 signaling in the 3D construct under the different perfusion flow provides important insight into an optimal bioreactor design. To investigate FGF-2 signaling inhibition in hMSC cultured in the porous poly(ethylene terephthalate) (PET) scaffolds perfused under two flow configurations, PD173074, an FGFR1 inhibitor, was added in growth media after 7 day of pre-culture and its impact on hMSC proliferation and clonogenicity during the subsequent 7 days of cultivation was analyzed. Compared with control constructs in growth media, the addition of PD173074 resulted in significant reduction in hMSC proliferation and colony formation in both constructs with a more dramatic reduction in the parallel flow constructs. The results demonstrate that autocrine FGF-2 plays a significant role in 3D scaffold and suggest modulation of the perfusion flow in the bioreactor as a strategy to influence autocrine actions and cell fate in the 3D scaffold.
Collapse
Affiliation(s)
- Junho Kim
- Dept of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
| | | |
Collapse
|
40
|
Kim J, Ma T. Bioreactor strategy in bone tissue engineering: pre-culture and osteogenic differentiation under two flow configurations. Tissue Eng Part A 2012; 18:2354-64. [PMID: 22690750 DOI: 10.1089/ten.tea.2011.0674] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Since robust osteogenic differentiation and mineralization are integral to the engineering of bone constructs, understanding the impact of the cellular microenvironments on human mesenchymal stem cell (hMSCs) osteogenic differentiation is crucial to optimize bioreactor strategy. Two perfusion flow conditions were utilized in order to understand the impact of the flow configuration on hMSC construct development during both pre-culture (PC) in growth media and its subsequent osteogenic induction (OI). The media in the in-house perfusion bioreactor was controlled to perfuse either around (termed parallel flow [PF]) the construct surfaces or penetrate through the construct (termed transverse flow [TF]) for 7 days of the PC followed by 7 days of the OI. The flow configuration during the PC not only changed growth kinetics but also influenced cell distribution and potency of osteogenic differentiation and mineralization during the subsequent OI. While shear stress resulted from the TF stimulated cell proliferation during PC, the convective removal of de novo extracellular matrix (ECM) proteins and growth factors (GFs) reduced cell proliferation on OI. In contrast, the effective retention of de novo ECM proteins and GFs in the PC constructs under the PF maintained cell proliferation under the OI but resulted in localized cell aggregations, which influenced their osteogenic differentiation. The results revealed the contrasting roles of the convective flow as a mechanical stimulus, the redistribution of the cells and macromolecules in 3D constructs, and their divergent impacts on cellular events, leading to bone construct formation. The results suggest that the modulation of the flow configuration in the perfusion bioreactor is an effective strategy that regulates the construct properties and maximizes the functional outcome.
Collapse
Affiliation(s)
- Junho Kim
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida 32310, USA
| | | |
Collapse
|