1
|
Rajalekshmi R, Agrawal DK. Synergistic potential of stem cells and microfluidics in regenerative medicine. Mol Cell Biochem 2025; 480:1481-1493. [PMID: 39285093 PMCID: PMC11842489 DOI: 10.1007/s11010-024-05108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/27/2024] [Indexed: 02/21/2025]
Abstract
Regenerative medicine has immense potential to revolutionize healthcare by using regenerative capabilities of stem cells. Microfluidics, a cutting-edge technology, offers precise control over cellular microenvironments. The integration of these two fields provides a deep understanding of stem cell behavior and enables the development of advanced therapeutic strategies. This critical review explores the use of microfluidic systems to culture and differentiate stem cells with precision. We examined the use of microfluidic platforms for controlled nutrient supply, mechanical stimuli, and real-time monitoring, providing an unprecedented level of detail in studying cellular responses. The convergence of stem cells and microfluidics holds immense promise for tissue repair, regeneration, and personalized medicine. It offers a unique opportunity to revolutionize the approach to regenerative medicine, facilitating the development of advanced therapeutic strategies and enhancing healthcare outcomes.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
2
|
Yehya H, Raudins S, Padmanabhan R, Jensen J, Bukys MA. Addressing bioreactor hiPSC aggregate stability, maintenance and scaleup challenges using a design of experiment approach. Stem Cell Res Ther 2024; 15:191. [PMID: 38956608 PMCID: PMC11218057 DOI: 10.1186/s13287-024-03802-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Stem cell-derived therapies hold the potential for treatment of regenerative clinical indications. Static culture has a limited ability to scale up thus restricting its use. Suspension culturing can be used to produce target cells in large quantities, but also presents challenges related to stress and aggregation stability. METHODS Utilizing a design of experiments (DoE) approach in vertical wheel bioreactors, we evaluated media additives that have versatile properties. The additives evaluated are Heparin sodium salt (HS), polyethylene glycol (PEG), poly (vinyl alcohol) (PVA), Pluronic F68 and dextran sulfate (DS). Multiple response variables were chosen to assess cell growth, pluripotency maintenance and aggregate stability in response to the additive inputs, and mathematical models were generated and tuned for maximal predictive power. RESULTS Expansion of iPSCs using 100 ml vertical wheel bioreactor assay for 4 days on 19 different media combinations resulted in models that can optimize pluripotency, stability, and expansion. The expansion optimization resulted in the combination of PA, PVA and PEG with E8. This mixture resulted in an expansion doubling time that was 40% shorter than that of E8 alone. Pluripotency optimizer highlighted the importance of adding 1% PEG to the E8 medium. Aggregate stability optimization that minimizes aggregate fusion in 3D culture indicated that the interaction of both Heparin and PEG can limit aggregation as well as increase the maintenance capacity and expansion of hiPSCs, suggesting that controlling fusion is a critical parameter for expansion and maintenance. Validation of optimized solution on two cell lines in bioreactors with decreased speed of 40 RPM, showed consistency and prolonged control over aggregates that have high frequency of pluripotency markers of OCT4 and SOX2 (> 90%). A doubling time of around 1-1.4 days was maintained after passaging as clumps in the optimized medium. Controlling aggregate fusion allowed for a decrease in bioreactor speed and therefore shear stress exerted on the cells in a large-scale expansion. CONCLUSION This study resulted in a control of aggregate size within suspension cultures, while informing about concomitant state control of the iPSC state. Wider application of this approach can address media optimization complexity and bioreactor scale-up challenges.
Collapse
Affiliation(s)
- Haneen Yehya
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
- Cleveland State University, 2121 Euclid Ave, Cleveland, OH, 44115, USA
| | - Sofija Raudins
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | | | - Jan Jensen
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA
| | - Michael A Bukys
- Trailhead Biosystems, 23215 Commerce Park, Beachwood, OH, 44122, USA.
| |
Collapse
|
3
|
de Janon A, Mantalaris A, Panoskaltsis N. Three-Dimensional Human Bone Marrow Organoids for the Study and Application of Normal and Abnormal Hematoimmunopoiesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:895-904. [PMID: 36947817 PMCID: PMC7614371 DOI: 10.4049/jimmunol.2200836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/18/2023] [Indexed: 03/24/2023]
Abstract
Hematoimmunopoiesis takes place in the adult human bone marrow (BM), which is composed of heterogeneous niches with complex architecture that enables tight regulation of homeostatic and stress responses. There is a paucity of representative culture systems that recapitulate the heterogeneous three-dimensional (3D) human BM microenvironment and that can endogenously produce soluble factors and extracellular matrix that deliver culture fidelity for the study of both normal and abnormal hematopoiesis. Native BM lymphoid populations are also poorly represented in current in vitro and in vivo models, creating challenges for the study and treatment of BM immunopathology. BM organoid models leverage normal 3D organ structure to recreate functional niche microenvironments. Our focus herein is to review the current state of the art in the use of 3D BM organoids, focusing on their capacities to recreate critical quality attributes of the in vivo BM microenvironment for the study of human normal and abnormal hematopoiesis.
Collapse
Affiliation(s)
- Alejandro de Janon
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
| | - Athanasios Mantalaris
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Ireland
- National Institute for Bioprocessing Research and Training, Ireland
| | - Nicki Panoskaltsis
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, The Georgia Institute of Technology, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Ireland
- Department of Haematology, St. James’s Hospital Dublin, Ireland
| |
Collapse
|
4
|
Akulo KA, Adali T, Moyo MTG, Bodamyali T. Intravitreal Injectable Hydrogels for Sustained Drug Delivery in Glaucoma Treatment and Therapy. Polymers (Basel) 2022; 14:polym14122359. [PMID: 35745935 PMCID: PMC9230531 DOI: 10.3390/polym14122359] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/11/2022] Open
Abstract
Glaucoma is extensively treated with topical eye drops containing drugs. However, the retention time of the loaded drugs and the in vivo bioavailability of the drugs are highly influenced before reaching the targeted area sufficiently, due to physiological and anatomical barriers of the eye, such as rapid nasolacrimal drainage. Poor intraocular penetration and frequent administration may also cause ocular cytotoxicity. A novel approach to overcome these drawbacks is the use of injectable hydrogels administered intravitreously for sustained drug delivery to the target site. These injectable hydrogels are used as nanocarriers to intimately interact with specific diseased ocular tissues to increase the therapeutic efficacy and drug bioavailability of the anti-glaucomic drugs. The human eye is very delicate, and is sensitive to contact with any foreign body material. However, natural biopolymers are non-reactive, biocompatible, biodegradable, and lack immunogenic and inflammatory responses to the host whenever they are incorporated in drug delivery systems. These favorable biomaterial properties have made them widely applicable in biomedical applications, with minimal adversity. This review highlights the importance of using natural biopolymer-based intravitreal hydrogel drug delivery systems for glaucoma treatment over conventional methods.
Collapse
Affiliation(s)
- Kassahun Alula Akulo
- Department of Biomedical Engineering, Faculty of Engineering, Near East University, Mersin 10, Lefkoşa 99138, Turkey; (K.A.A.); (M.T.G.M.)
- Tissue Engineering and Biomaterials Research Center, Near East University, Mersin 10, Lefkoşa 99138, Turkey
| | - Terin Adali
- Department of Biomedical Engineering, Faculty of Engineering, Near East University, Mersin 10, Lefkoşa 99138, Turkey; (K.A.A.); (M.T.G.M.)
- Tissue Engineering and Biomaterials Research Center, Near East University, Mersin 10, Lefkoşa 99138, Turkey
- Nanotechnology Research Center, Sabanci University SUNUM, Istanbul 34956, Turkey
- Correspondence:
| | - Mthabisi Talent George Moyo
- Department of Biomedical Engineering, Faculty of Engineering, Near East University, Mersin 10, Lefkoşa 99138, Turkey; (K.A.A.); (M.T.G.M.)
- Tissue Engineering and Biomaterials Research Center, Near East University, Mersin 10, Lefkoşa 99138, Turkey
| | - Tulin Bodamyali
- Department of Pathology, Faculty of Medicine, Girne American University, Mersin 10, Girne 99428, Turkey;
| |
Collapse
|
5
|
Cha JM, Hwang YS, Kang DK, Lee J, Cooper ES, Mantalaris A. Development of a Novel Perfusion Rotating Wall Vessel Bioreactor with Ultrasound Stimulation for Mass-Production of Mineralized Tissue Constructs. Tissue Eng Regen Med 2022; 19:739-754. [PMID: 35532736 PMCID: PMC9294093 DOI: 10.1007/s13770-022-00447-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/06/2022] [Accepted: 02/20/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND As stem cells are considered a promising cell source for tissue engineering, many culture strategies have been extensively studied to generate in vitro stem cell-based tissue constructs. However, most approaches using conventional tissue culture plates are limited by the lack of biological relevance in stem cell microenvironments required for neotissue formation. In this study, a novel perfusion rotating wall vessel (RWV) bioreactor was developed for mass-production of stem cell-based 3D tissue constructs. METHODS An automated RWV bioreactor was fabricated, which is capable of controlling continuous medium perfusion, highly efficient gas exchange with surrounding air, as well as low-intensity pulsed ultrasound (LIPUS) stimulation. Embryonic stem cells encapsulated in alginate/gelatin hydrogel were cultured in the osteogenic medium by using our bioreactor system. Cellular viability, growth kinetics, and osteogenesis/mineralization were thoroughly evaluated, and culture media were profiled at real time. The in vivo efficacy was examined by a rabbit cranial defect model. RESULTS Our bioreactor successfully maintained the optimal culture environments for stem cell proliferation, osteogenic differentiation, and mineralized tissue formation during the culture period. The mineralized tissue constructs produced by our bioreactor demonstrated higher void filling efficacy in the large bone defects compared to the group implanted with hydrogel beads only. In addition, the LIPUS modules mounted on our bioreactor successfully reached higher mineralization of the tissue constructs compared to the groups without LIPUS stimulation. CONCLUSION This study suggests an effective biomanufacturing strategy for mass-production of implantable mineralized tissue constructs from stem cells that could be applicable to future clinical practice.
Collapse
|
6
|
Induced pluripotency in the context of stem cell expansion bioprocess development, optimization, and manufacturing: a roadmap to the clinic. NPJ Regen Med 2021; 6:72. [PMID: 34725374 PMCID: PMC8560749 DOI: 10.1038/s41536-021-00183-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 10/07/2021] [Indexed: 11/09/2022] Open
Abstract
The translation of laboratory-scale bioprocess protocols and technologies to industrial scales and the application of human induced pluripotent stem cell (hiPSC) derivatives in clinical trials globally presents optimism for the future of stem-cell products to impact healthcare. However, while many promising therapeutic approaches are being tested in pre-clinical studies, hiPSC-derived products currently account for a small fraction of active clinical trials. The complexity and volatility of hiPSCs present several bioprocessing challenges, where the goal is to generate a sufficiently large, high-quality, homogeneous population for downstream differentiation-the derivatives of which must retain functional efficacy and meet regulatory safety criteria in application. It is argued herein that one of the major challenges currently faced in improving the robustness of routine stem-cell biomanufacturing is in utilizing continuous, meaningful assessments of molecular and cellular characteristics from process to application. This includes integrating process data with biological characteristic and functional assessment data to model the interplay between variables in the search for global optimization strategies. Coupling complete datasets with relevant computational methods will contribute significantly to model development and automation in achieving process robustness. This overarching approach is thus crucially important in realizing the potential of hiPSC biomanufacturing for transformation of regenerative medicine and the healthcare industry.
Collapse
|
7
|
Tan B, Gan S, Wang X, Liu W, Li X. Applications of 3D bioprinting in tissue engineering: advantages, deficiencies, improvements, and future perspectives. J Mater Chem B 2021; 9:5385-5413. [PMID: 34124724 DOI: 10.1039/d1tb00172h] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Over the past decade, 3D bioprinting technology has progressed tremendously in the field of tissue engineering in its ability to fabricate individualized biological constructs with precise geometric designability, which offers us the capability to bridge the divergence between engineered tissue constructs and natural tissues. In this work, we first review the current widely used 3D bioprinting approaches, cells, and materials. Next, the updated applications of this technique in tissue engineering, including bone tissue, cartilage tissue, vascular grafts, skin, neural tissue, heart tissue, liver tissue and lung tissue, are briefly introduced. Then, the prominent advantages of 3D bioprinting in tissue engineering are summarized in detail: rapidly prototyping the customized structure, delivering cell-laden materials with high precision in space, and engineering with a highly controllable microenvironment. The current technical deficiencies of 3D bioprinted constructs in terms of mechanical properties and cell behaviors are afterward illustrated, as well as corresponding improvements. Finally, we conclude with future perspectives about 3D bioprinting in tissue engineering.
Collapse
Affiliation(s)
- Baosen Tan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Shaolei Gan
- Jiangxi Borayer Biotech Co., Ltd, Nanchang 330052, China
| | - Xiumei Wang
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Wenyong Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| |
Collapse
|
8
|
Park YL, Park K, Cha JM. 3D-Bioprinting Strategies Based on In Situ Bone-Healing Mechanism for Vascularized Bone Tissue Engineering. MICROMACHINES 2021; 12:mi12030287. [PMID: 33800485 PMCID: PMC8000586 DOI: 10.3390/mi12030287] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Over the past decades, a number of bone tissue engineering (BTE) approaches have been developed to address substantial challenges in the management of critical size bone defects. Although the majority of BTE strategies developed in the laboratory have been limited due to lack of clinical relevance in translation, primary prerequisites for the construction of vascularized functional bone grafts have gained confidence owing to the accumulated knowledge of the osteogenic, osteoinductive, and osteoconductive properties of mesenchymal stem cells and bone-relevant biomaterials that reflect bone-healing mechanisms. In this review, we summarize the current knowledge of bone-healing mechanisms focusing on the details that should be embodied in the development of vascularized BTE, and discuss promising strategies based on 3D-bioprinting technologies that efficiently coalesce the abovementioned main features in bone-healing systems, which comprehensively interact during the bone regeneration processes.
Collapse
Affiliation(s)
- Ye Lin Park
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon 22012, Korea;
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon 22012, Korea
| | - Kiwon Park
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon 22012, Korea;
- Correspondence: (K.P.); (J.M.C.); Tel.: +82-32-835-8685 (K.P.); +82-32-835-8686 (J.M.C.)
| | - Jae Min Cha
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon 22012, Korea;
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon 22012, Korea
- Correspondence: (K.P.); (J.M.C.); Tel.: +82-32-835-8685 (K.P.); +82-32-835-8686 (J.M.C.)
| |
Collapse
|
9
|
Nanofibrous polyester-polypeptide block copolymer scaffolds with high porosity and controlled degradation promote cell adhesion, proliferation and differentiation. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.109647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
10
|
Hoyle HW, Smith LA, Williams RJ, Przyborski SA. Applications of novel bioreactor technology to enhance the viability and function of cultured cells and tissues. Interface Focus 2020; 10:20190090. [PMID: 32194933 DOI: 10.1098/rsfs.2019.0090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
As the field of tissue engineering continues to advance rapidly, so too does the complexity of cell culture techniques used to generate in vitro tissue constructs, with the overall aim of mimicking the in vivo microenvironment. This complexity typically comes at a cost with regards to the size of the equipment required and associated expenses. We have developed a small, low-cost bioreactor system which overcomes some of the issues of typical bioreactor systems while retaining a suitable scale for the formation of complex tissues. Herein, we have tested this system with three cell populations/tissues: the culture of hepatocellular carcinoma cells, where an improved structure and basic metabolic function is seen; the culture of human pluripotent stem cells, in which the cultures can form more heterogeneous tissues resembling the in vivo teratoma and ex vivo liver tissue slices, in which improved maintenance of cellular viability is seen over the 3 days tested. This system has the flexibility to be used for a variety of further uses and has the potential to provide a more accessible alternative to current bioreactor technologies.
Collapse
Affiliation(s)
- H W Hoyle
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - L A Smith
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - R J Williams
- Department of Engineering, Durham University, South Road, Durham DH1 3LE, UK
| | - S A Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK.,Reprocell Europe Ltd, NETPark Incubator, Thomas Wright Way, Sedgefield TS21 3FD, UK
| |
Collapse
|
11
|
Malhotra N. Bioreactors Design, Types, Influencing Factors and Potential Application in Dentistry. A Literature Review. Curr Stem Cell Res Ther 2019; 14:351-366. [DOI: 10.2174/1574888x14666190111105504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/26/2018] [Accepted: 12/27/2018] [Indexed: 11/22/2022]
Abstract
Objectives:A variety of bioreactors and related approaches have been applied to dental tissues as their use has become more essential in the field of regenerative dentistry and dental tissue engineering. The review discusses the various types of bioreactors and their potential application in dentistry.Methods:Review of the literature was conducted using keywords (and MeSH) like Bioreactor, Regenerative Dentistry, Fourth Factor, Stem Cells, etc., from the journals published in English. All the searched abstracts, published in indexed journals were read and reviewed to further refine the list of included articles. Based on the relevance of abstracts pertaining to the manuscript, full-text articles were assessed.Results:Bioreactors provide a prerequisite platform to create, test, and validate the biomaterials and techniques proposed for dental tissue regeneration. Flow perfusion, rotational, spinner-flask, strain and customize-combined bioreactors have been applied for the regeneration of bone, periodontal ligament, gingiva, cementum, oral mucosa, temporomandibular joint and vascular tissues. Customized bioreactors can support cellular/biofilm growth as well as apply cyclic loading. Center of disease control & dip-flow biofilm-reactors and micro-bioreactor have been used to evaluate the biological properties of dental biomaterials, their performance assessment and interaction with biofilms. Few case reports have also applied the concept of in vivo bioreactor for the repair of musculoskeletal defects and used customdesigned bioreactor (Aastrom) to repair the defects of cleft-palate.Conclusions:Bioreactors provide a sterile simulated environment to support cellular differentiation for oro-dental regenerative applications. Also, bioreactors like, customized bioreactors for cyclic loading, biofilm reactors (CDC & drip-flow), and micro-bioreactor, can assess biological responses of dental biomaterials by simultaneously supporting cellular or biofilm growth and application of cyclic stresses.
Collapse
|
12
|
Baakdhah T, van der Kooy D. Expansion of retinal stem cells and their progeny using cell microcarriers in a bioreactor. Biotechnol Prog 2019; 35:e2800. [DOI: 10.1002/btpr.2800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/10/2019] [Accepted: 02/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Tahani Baakdhah
- Institute of Medical ScienceUniversity of Toronto Toronto Ontario Canada
| | - Derek van der Kooy
- Institute of Medical ScienceUniversity of Toronto Toronto Ontario Canada
- Department of Molecular GeneticsUniversity of Toronto Toronto Ontario Canada
| |
Collapse
|
13
|
Al-Bagdadi FA, Barona HM, Martinez-Ceballos E, Yao S. Ultrastructure Morphological Characterization of Different Passages of Rat Dental Follicle Stem Cells at In vitro Culture. J Microsc Ultrastruct 2019; 7:57-64. [PMID: 31293886 PMCID: PMC6585478 DOI: 10.4103/jmau.jmau_44_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Introduction: Stem cells play important roles in tissue renewal and repair. Tissue-derived stem cells have been demonstrated for their applications in tissue engineering and regenerative medicine. Expansion of primary stem cells isolated from tissues to a large quantity through in vitro culture is needed for application of the stem cells. However, it is known that tissue stem cells commonly reduce or lose their stemness properties during in vitro culture. In this study, we assessed ultrastructural changes of rat dental follicle stem cells (DFSCs) during in vitro culture. It is our attempt to explain the loss of stemness properties in cultured tissue-stem cells at the ultrastructural level. Method: DFSCs was isolated from first molars of Sprague Dawley rat pups and cultured in medium consisting of alpha-MEM plus 20% FBS. Cells were passaged at 1 to 3 ratio at 90% confluence, and collected at passages 3, 6, 7 and 9 for assessment of ultrastructure morphology by transmission electron microscopy. Results: Of the four passages (3, 6, 7, and 9) examined, dilated rough endoplasmic reticulum (RER) was abundant in Passage 3 but less so in Passages 6, 7, and 9. The dilated RER contained lipid in Passages 3, 7, and 9. The mono- and polyribosomes in Passages 3 and 6 were located between the mitochondria and the RER. Mono- and polyribosomes were abundant in Passage 7, although mainly monoribosomes were present in Passage 9. Membrane-bound glycogen granules were in vacuoles bulging off the cells in Passage 3. Some glycogen granules were grouped in the periphery of a stem cell in Passage 9. Nuclei shapes were irregular and mainly euchromatic in Passages 6, 7, and 9. The mitochondria were dark and scarce in Passage 9; irregular, small, and dark in Passage 7; and small and rounded in Passage 6, and they were spread in the cytoplasm away from the nucleus in Passage 3. Cell contacts were seen in Passages 6, 7, and 9. The ultrastructure morphology of the examined DFScs was not very different from the morphology criteria of the undifferentiated cells. Large vacuoles in Passage 3 were mainly at the periphery of the cell, with the small vacuoles in the cell center. Small vacuoles were scattered in the cell center of Passage 6 and the larger ones were observed at the cell's periphery. Conclusions: We observed the following ultrastructural changes: decreases of fine cell cytoplasmic processes, dilated cytoplasmic vacuoles, cytoplasmic pinocytotic vesicles, and nuclear heterochromatin with increasing cell passage number. Conversely, mean ratios of lipid globules, nuclear euchromatin, irregular nuclear shape, and cell contact between cells were increased with passage number. The observations may suggest an increase in committed cells among the population after long-term culture of DFSCs.
Collapse
Affiliation(s)
- Fakhri A Al-Bagdadi
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Humberto M Barona
- Department of Mathematics, Southern University and A and M College, Baton Rouge, LA, USA
| | | | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
14
|
Bunpetch V, Wu H, Zhang S, Ouyang H. From "Bench to Bedside": Current Advancement on Large-Scale Production of Mesenchymal Stem Cells. Stem Cells Dev 2018; 26:1662-1673. [PMID: 28934885 DOI: 10.1089/scd.2017.0104] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the primary cell source in cell therapy and regenerative medicine due to its extraordinary self-renewing capacity and multilineage differentiation potential. Clinical trials involving MSCs are being conducted in a range of human diseases and the number of registered cases is continuously increasing. However, a wide gap exists between the number of MSCs obtainable from the donor site and the number required for implantation to damage tissues, and also between MSC scalability and MSC phenotype stability. The clinical translation of MSCs necessitates a scalable expansion bioprocess for the biomanufacturing of therapeutically qualified cells. This review presents current achievements for expansion of MSCs. Issues involving culture condition modification, bioreactor systems, as well as microcarrier and scaffold platforms for optimal MSC systems are discussed. Most importantly, the gap between current MSC expansion and clinical application, as well as outbreak directions for the future are discussed. The present systemic review will bring new insights into future large-scale MSC expansion and clinical application.
Collapse
Affiliation(s)
- Varitsara Bunpetch
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Haoyu Wu
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Shufang Zhang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hongwei Ouyang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China .,4 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou, China .,5 Department of Sports Medicine, School of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
15
|
Marcucio RS, Qin L, Alsberg E, Boerckel JD. Reverse engineering development: Crosstalk opportunities between developmental biology and tissue engineering. J Orthop Res 2017; 35:2356-2368. [PMID: 28660712 DOI: 10.1002/jor.23636] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
The fields of developmental biology and tissue engineering have been revolutionized in recent years by technological advancements, expanded understanding, and biomaterials design, leading to the emerging paradigm of "developmental" or "biomimetic" tissue engineering. While developmental biology and tissue engineering have long overlapping histories, the fields have largely diverged in recent years at the same time that crosstalk opportunities for mutual benefit are more salient than ever. In this perspective article, we will use musculoskeletal development and tissue engineering as a platform on which to discuss these emerging crosstalk opportunities and will present our opinions on the bright future of these overlapping spheres of influence. The multicellular programs that control musculoskeletal development are rapidly becoming clarified, represented by shifting paradigms in our understanding of cellular function, identity, and lineage specification during development. Simultaneously, advancements in bioartificial matrices that replicate the biochemical, microstructural, and mechanical properties of developing tissues present new tools and approaches for recapitulating development in tissue engineering. Here, we introduce concepts and experimental approaches in musculoskeletal developmental biology and biomaterials design and discuss applications in tissue engineering as well as opportunities for tissue engineering approaches to inform our understanding of fundamental biology. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2356-2368, 2017.
Collapse
Affiliation(s)
- Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania
| | - Eben Alsberg
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, Ohio
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania.,Department of Bioengineering, University of Pennslyvania, Philadelphia, Pennsylvania.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
16
|
Yu C, Kornmuller A, Brown C, Hoare T, Flynn LE. Decellularized adipose tissue microcarriers as a dynamic culture platform for human adipose-derived stem/stromal cell expansion. Biomaterials 2016; 120:66-80. [PMID: 28038353 DOI: 10.1016/j.biomaterials.2016.12.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/22/2016] [Accepted: 12/15/2016] [Indexed: 12/12/2022]
Abstract
With the goal of designing a clinically-relevant expansion strategy for human adipose-derived stem/stromal cells (ASCs), methods were developed to synthesize porous microcarriers derived purely from human decellularized adipose tissue (DAT). An electrospraying approach was applied to generate spherical DAT microcarriers with an average diameter of 428 ± 41 μm, which were soft, compliant, and stable in long-term culture without chemical crosslinking. Human ASCs demonstrated enhanced proliferation on the DAT microcarriers relative to commercially-sourced Cultispher-S microcarriers within a spinner culture system over 1 month. ASC immunophenotype was maintained post expansion, with a trend for reduced expression of the cell adhesion receptors CD73, CD105, and CD29 under dynamic conditions. Upregulation of the early lineage-specific genes PPARγ, LPL, and COMP was observed in the ASCs expanded on the DAT microcarriers, but the cells retained their multilineage differentiation capacity. Comparison of adipogenic and osteogenic differentiation in 2-D cultures prepared with ASCs pre-expanded on the DAT microcarriers or Cultispher-S microcarriers revealed similar adipogenic and enhanced osteogenic marker expression in the DAT microcarrier group, which had undergone a higher population fold change. Further, histological staining results suggested a more homogeneous differentiation response in the ASCs expanded on the DAT microcarriers as compared to either Cultispher-S microcarriers or tissue culture polystyrene. A pilot chondrogenesis study revealed higher levels of chondrogenic gene and protein expression in the ASCs expanded on the DAT microcarriers relative to all other groups, including the baseline controls. Overall, this study demonstrates the promise of applying dynamic culture with tissue-specific DAT microcarriers as a means of deriving regenerative cell populations.
Collapse
Affiliation(s)
- Claire Yu
- Department of Chemical Engineering, Queen's University, 19 Division St., Kingston, ON, K7L 3N6, Canada; Human Mobility Research Center, Kingston General Hospital, 76 Stuart St., Kingston, ON, K7L 2V7, Canada
| | - Anna Kornmuller
- Biomedical Engineering Graduate Program, Claudette MacKay Lassonde Pavilion, The University of Western Ontario, London, ON, N6A 5B9, Canada
| | - Cody Brown
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L7, Canada
| | - Lauren E Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, N6A 5C1, Canada; Department of Chemical and Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, ON, N6A 5B9, Canada.
| |
Collapse
|
17
|
Huang L, Xu F, Guo B, Ma J, Zhao J. Morphological study of dynamic culture of thermosensitive collagen hydrogel in constructing tissue engineering complex. Bioengineered 2016; 7:266-73. [PMID: 27459597 DOI: 10.1080/21655979.2016.1197741] [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: 10/21/2022] Open
Abstract
ABSTACT The purpose of this study is to research the morphologies and functional characteristics of the cell-scaffold complex in vitro constructed under dynamic culture conditions. BMSCs were isolated from the long bones of Fischer344 rats, and performed in vitro amplification to the third generation as seed cells, together with thermosensitive collagen hydrogel (TCH) as cell adhesion matrix, and poly-L-lactic acid (PLLA) as scaffold, to construct cell-scaffold complex. The cell-scaffold complexes in the experiment group and the control group were then performed dynamic culture and static culture. After 7 d of in vitro culture, the complexes in the 2 groups were performed gross observation and SEM; meanwhile, the total DNA content in the complex was detected on D0,1,3, and 7 of culture. After cultured using these 2 ways, collagen could both wrap the PLLA scaffold, forming dense film-like structures on the PLLA surface. The total DNA contents in the cell-scaffold complex of the experiment group on D1,3, and 7 were significantly higher than the control group (P < 0.05). Compared with D0, the total DNA contents on D1,3, and 7 in both groups were gradually increased, but only the total DNA contents on D7 showed statistically significant difference than D0 (P < 0.05). TCH -PLLA fiber joint-constructed complex extracellular matrix had good biocompatibility, and dynamic culture could promote the distribution of BMSCs on the surface and inside the structure, thus promoting cell proliferation, so it could be used for the in vitro construction of tissue engineering complex.
Collapse
Affiliation(s)
- Lanfeng Huang
- a Department of Orthopedics , The Second Hospital of Jilin University , Changchun , China
| | - Feixiang Xu
- a Department of Orthopedics , The Second Hospital of Jilin University , Changchun , China
| | - Bin Guo
- a Department of Orthopedics , The Second Hospital of Jilin University , Changchun , China
| | - Jianchao Ma
- a Department of Orthopedics , The Second Hospital of Jilin University , Changchun , China
| | - Jinsong Zhao
- b Department of Ophthalmology , The Second Hospital of Jilin University , Changchun , China
| |
Collapse
|
18
|
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: 0.3] [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
|
19
|
Wuchter P, Saffrich R, Giselbrecht S, Nies C, Lorig H, Kolb S, Ho AD, Gottwald E. Microcavity arrays as an in vitro model system of the bone marrow niche for hematopoietic stem cells. Cell Tissue Res 2016; 364:573-584. [PMID: 26829941 DOI: 10.1007/s00441-015-2348-8] [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: 09/15/2015] [Accepted: 12/10/2015] [Indexed: 12/28/2022]
Abstract
In previous studies human mesenchymal stromal cells (MSCs) maintained the "stemness" of human hematopoietic progenitor cells (HPCs) through direct cell-cell contact in two-dimensional co-culture systems. We establish a three-dimensional (3D) co-culture system based on a custom-made chip, the 3(D)-KITChip, as an in vitro model system of the human hematopoietic stem cell niche. This array of up to 625 microcavities, with 300 μm size in each orientation, was inserted into a microfluidic bioreactor. The microcavities of the 3(D)-KITChip were inoculated with human bone marrow MSCs together with umbilical cord blood HPCs. MSCs used the microcavities as a scaffold to build a complex 3D mesh. HPCs were distributed three-dimensionally inside this MSC network and formed ß-catenin- and N-cadherin-based intercellular junctions to the surrounding MSCs. Using RT(2)-PCR and western blots, we demonstrate that a proportion of HPCs maintained the expression of CD34 throughout a culture period of 14 days. In colony-forming unit assays, the hematopoietic stem cell plasticity remained similar after 14 days of bioreactor co-culture, whereas monolayer co-cultures showed increasing signs of HPC differentiation and loss of stemness. These data support the notion that the 3D microenvironment created within the microcavity array preserves vital stem cell functions of HPCs more efficiently than conventional co-culture systems.
Collapse
Affiliation(s)
- Patrick Wuchter
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany. .,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.
| | - Rainer Saffrich
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Stefan Giselbrecht
- HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.,Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Cordula Nies
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Hanna Lorig
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Stephanie Kolb
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Anthony D Ho
- Department of Medicine V, Heidelberg University, 69120, Heidelberg, Germany.,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany
| | - Eric Gottwald
- Institute for Biological Interfaces-5, Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Karlsruhe, Germany. .,HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University and Karlsruhe Institute of Technology, Heidelberg and Karlsruhe, Germany.
| |
Collapse
|
20
|
Fotia C, Massa A, Boriani F, Baldini N, Granchi D. Prolonged exposure to hypoxic milieu improves the osteogenic potential of adipose derived stem cells. J Cell Biochem 2016; 116:1442-53. [PMID: 25648991 DOI: 10.1002/jcb.25106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/27/2015] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSC) have been widely used in orthopedics for several applications. Conventionally, MSC are maintained under 21% O2 which does not reflect the real O2 tension in vivo. Recently, it was reported that different O2 conditions can give different cellular responses. Here, we investigated whether prolonged exposure to hypoxia affects the osteogenic differentiation of adipose-derived stem cells (ASC). ASC from six individuals were cultured under "low" (2-3%) or "air" (21%) oxygen tensions, either without or with osteogenic stimuli. The effect of the O2 tension was evaluated on cell proliferation, surface antigens, stemness and bone-related genes expression, alkaline phosphatase activity (ALP), mineralization activity, and release of osteogenic growth factors. Without differentiating stimuli, hypoxia favored ASC proliferation, reduced the number of CD184+ and CD34+ cells, and preserved the expression of NANOG and SOX2. The combination of hypoxia and osteogenic medium induced a high proliferation rate, a rapid and more pronounced mineralization activity, a higher expression of genes related to the MSC differentiation, a higher release of mitogenic growth factors (bFGF, PDGF-BB), and the decrease in TGF-β secretion, an inhibitor of the early stage of the osteoblast differentiation. We demonstrated that hypoxia acts dually, favoring ASC proliferation and the maintenance of the stemness in the absence of osteogenic stimuli, but inducing the differentiation in a bone-like microenvironment. In conclusion, prolonged cell culture in hypoxic microenvironment represents a proper method to modulate the stem cell function that may be used in several applications, for example, studies on bone pathophysiology or bone-tissue engineering.
Collapse
Affiliation(s)
- Caterina Fotia
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Annamaria Massa
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Filippo Boriani
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Nicola Baldini
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
21
|
Mohammadpour H, Pourfathollah AA, Nikougoftar Zarif M, Hashemi SM. Increasing proliferation of murine adipose tissue-derived mesenchymal stem cells by TNF-α plus IFN-γ. Immunopharmacol Immunotoxicol 2015; 38:68-76. [DOI: 10.3109/08923973.2015.1115519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
22
|
Davies OG, Cooper PR, Shelton RM, Smith AJ, Scheven BA. Isolation of adipose and bone marrow mesenchymal stem cells using CD29 and CD90 modifies their capacity for osteogenic and adipogenic differentiation. J Tissue Eng 2015; 6:2041731415592356. [PMID: 26380065 PMCID: PMC4555348 DOI: 10.1177/2041731415592356] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells isolated from rats are frequently used for tissue engineering research. However, considerable differences have been identified between rat mesenchymal stem cells and those derived from humans, and no defined panel of markers currently exists for the isolation of these cells. The aim of this study was to examine the effects of cell sorting for CD29+/CD90+ cells from rat adipose and bone marrow tissues on their differentiation and expression of stem cell–associated genes. Flow cytometry showed 66% and 78% CD29+/CD90+ positivity within passage 1 of adipose and bone marrow cultures, respectively. CD29+/CD90+ cells showed a reduction in both osteogenic and adipogenic differentiation when compared with unsorted cells, as determined by alizarin red and Oil Red-O staining, respectively. These findings could not entirely be explained by fluorescence-activated cell sorting–induced cell injury as sort recovery was only modestly affected in adipose-derived cells. Maintaining cells in fluorescence-activated cell sorting buffer did not affect adipose-derived cell viability, but a significant (p < 0.05) reduction was found in bone marrow–derived cell viability. Additionally, CD29+/CD90+ selection was associated with a significant decrease in the expression of Lin28, Sox2, Nanog and CD73 in adipose-derived cell cultures, whereas differences in stem cell–associated gene expression were not observed in sorted bone marrow–derived cell cultures. In summary, this study demonstrated that fluorescence-activated cell sorting had differential effects on adipose-derived cells and bone marrow–derived cells, and both CD29+/CD90+ cells displayed a significantly reduced capacity for osteogenic/adipogenic differentiation. In conclusion, we identify that maintaining heterogeneity within the mesenchymal stem cell population may be important for optimal differentiation.
Collapse
Affiliation(s)
- Owen G Davies
- School of Dentistry, University of Birmingham, Birmingham, UK
- Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Paul R Cooper
- School of Dentistry, University of Birmingham, Birmingham, UK
| | | | - Anthony J Smith
- School of Dentistry, University of Birmingham, Birmingham, UK
| | - Ben A Scheven
- School of Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
23
|
Maguire G, Friedman P. Systems biology approach to developing S 2RM-based “systems therapeutics” and naturally induced pluripotent stem cells. World J Stem Cells 2015; 7:745-756. [PMID: 26029345 PMCID: PMC4444614 DOI: 10.4252/wjsc.v7.i4.745] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/25/2014] [Accepted: 03/18/2015] [Indexed: 02/06/2023] Open
Abstract
The degree to, and the mechanisms through, which stem cells are able to build, maintain, and heal the body have only recently begun to be understood. Much of the stem cell’s power resides in the release of a multitude of molecules, called stem cell released molecules (SRM). A fundamentally new type of therapeutic, namely “systems therapeutic”, can be realized by reverse engineering the mechanisms of the SRM processes. Recent data demonstrates that the composition of the SRM is different for each type of stem cell, as well as for different states of each cell type. Although systems biology has been successfully used to analyze multiple pathways, the approach is often used to develop a small molecule interacting at only one pathway in the system. A new model is emerging in biology where systems biology is used to develop a new technology acting at multiple pathways called “systems therapeutics”. A natural set of healing pathways in the human that uses SRM is instructive and of practical use in developing systems therapeutics. Endogenous SRM processes in the human body use a combination of SRM from two or more stem cell types, designated as S2RM, doing so under various state dependent conditions for each cell type. Here we describe our approach in using state-dependent SRM from two or more stem cell types, S2RM technology, to develop a new class of therapeutics called “systems therapeutics.” Given the ubiquitous and powerful nature of innate S2RM-based healing in the human body, this “systems therapeutic” approach using S2RM technology will be important for the development of anti-cancer therapeutics, antimicrobials, wound care products and procedures, and a number of other therapeutics for many indications.
Collapse
|
24
|
Seifarth V, Gossmann M, Janke HP, Grosse JO, Becker C, Heschel I, Artmann GM, Temiz Artmann A. Development of a Bioreactor to Culture Tissue Engineered Ureters Based on the Application of Tubular OPTIMAIX 3D Scaffolds. Urol Int 2015; 95:106-13. [DOI: 10.1159/000368419] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/16/2014] [Indexed: 11/19/2022]
Abstract
Regenerative medicine, tissue engineering and biomedical research give hope to many patients who need bio-implants. Tissue engineering applications have already been developed based on bioreactors. Physiological ureter implants, however, do not still function sufficiently, as they represent tubular hollow structures with very specific cellular structures and alignments consisting of several cell types. The aim of this study was to a develop a new bioreactor system based on seamless, collagenous, tubular OPTIMAIX 3D prototype sponge as scaffold material for ex-vivo culturing of a tissue engineered ureter replacement for future urological applications. Particular emphasis was given to a great extent to mimic the physiological environment similar to the in vivo situation of a ureter. NIH-3T3 fibroblasts, C2C12, Urotsa and primary genitourinary tract cells were applied as co-cultures on the scaffold and the penetration of cells into the collagenous material was followed. By the end of this study, the bioreactor was functioning, physiological parameter as temperature and pH and the newly developed BIOREACTOR system is applicable to tubular scaffold materials with different lengths and diameters. The automatized incubation system worked reliably. The tubular OPTIMAIX 3D sponge was a suitable scaffold material for tissue engineering purposes and co-cultivation procedures.
Collapse
|
25
|
Antanavičiūtė I, Ereminienė E, Vysockas V, Račkauskas M, Skipskis V, Rysevaitė K, Treinys R, Benetis R, Jurevičius J, Skeberdis VA. Exogenous connexin43-expressing autologous skeletal myoblasts ameliorate mechanical function and electrical activity of the rabbit heart after experimental infarction. Int J Exp Pathol 2014; 96:42-53. [PMID: 25529770 DOI: 10.1111/iep.12109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 10/26/2014] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction is one of the major causes of mortality worldwide. For regeneration of the rabbit heart after experimentally induced infarction we used autologous skeletal myoblasts (SMs) due to their high proliferative potential, resistance to ischaemia and absence of immunological and ethical concerns. The cells were characterized with muscle-specific and myogenic markers. Cell transplantation was performed by injection of cell suspension (0.5 ml) containing approximately 6 million myoblasts into the infarction zone. The animals were divided into four groups: (i) no injection; (ii) sham injected; (iii) injected with wild-type SMs; and (iv) injected with SMs expressing connexin43 fused with green fluorescent protein (Cx43EGFP). Left ventricular ejection fraction (LVEF) was evaluated by 2D echocardiography in vivo before infarction, when myocardium has stabilized after infarction, and 3 months after infarction. Electrical activity in the healthy and infarction zones of the heart was examined ex vivo in Langendorff-perfused hearts by optical mapping using di-4-ANEPPS, a potential sensitive fluorescent dye. We demonstrate that SMs in the coculture can couple electrically not only to abutted but also to remote acutely isolated allogenic cardiac myocytes through membranous tunnelling tubes. The beneficial effect of cellular therapy on LVEF and electrical activity was observed in the group of animals injected with Cx43EGFP-expressing SMs. L-type Ca(2+) current amplitude was approximately fivefold smaller in the isolated SMs compared to healthy myocytes suggesting that limited recovery of LVEF may be related to inadequate expression or function of L-type Ca(2+) channels in transplanted differentiating SMs.
Collapse
Affiliation(s)
- Ieva Antanavičiūtė
- Institute of Cardiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Li Y, Liu M, Yang ST. Dendritic cells derived from pluripotent stem cells: Potential of large scale production. World J Stem Cells 2014; 6:1-10. [PMID: 24567783 PMCID: PMC3927009 DOI: 10.4252/wjsc.v6.i1.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/23/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, are promising sources for hematopoietic cells due to their unlimited growth capacity and the pluripotency. Dendritic cells (DCs), the unique immune cells in the hematopoietic system, can be loaded with tumor specific antigen and used as vaccine for cancer immunotherapy. While autologous DCs from peripheral blood are limited in cell number, hPSC-derived DCs provide a novel alternative cell source which has the potential for large scale production. This review summarizes recent advances in differentiating hPSCs to DCs through the intermediate stage of hematopoietic stem cells. Step-wise growth factor induction has been used to derive DCs from hPSCs either in suspension culture of embryoid bodies (EBs) or in co-culture with stromal cells. To fulfill the clinical potential of the DCs derived from hPSCs, the bioprocess needs to be scaled up to produce a large number of cells economically under tight quality control. This requires the development of novel bioreactor systems combining guided EB-based differentiation with engineered culture environment. Hence, recent progress in using bioreactors for hPSC lineage-specific differentiation is reviewed. In particular, the potential scale up strategies for the multistage DC differentiation and the effect of shear stress on hPSC differentiation in bioreactors are discussed in detail.
Collapse
|