1
|
Barrett L, Coopman K. Cell microencapsulation techniques for cancer modelling and drug discovery. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:345-354. [PMID: 38829715 DOI: 10.1080/21691401.2024.2359996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Cell encapsulation into spherical microparticles is a promising bioengineering tool in many fields, including 3D cancer modelling and pre-clinical drug discovery. Cancer microencapsulation models can more accurately reflect the complex solid tumour microenvironment than 2D cell culture and therefore would improve drug discovery efforts. However, these microcapsules, typically in the range of 1 - 5000 µm in diameter, must be carefully designed and amenable to high-throughput production. This review therefore aims to outline important considerations in the design of cancer cell microencapsulation models for drug discovery applications and examine current techniques to produce these. Extrusion (dripping) droplet generation and emulsion-based techniques are highlighted and their suitability to high-throughput drug screening in terms of tumour physiology and ease of scale up is evaluated.
Collapse
Affiliation(s)
- Lisa Barrett
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| | - Karen Coopman
- Department of Chemical Engineering, School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, UK
| |
Collapse
|
2
|
Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and Considerations of Preclinical Development for iPSC-Based Myogenic Cell Therapy. Cells 2024; 13:596. [PMID: 38607035 PMCID: PMC11011706 DOI: 10.3390/cells13070596] [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: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.
Collapse
Affiliation(s)
- Congshan Sun
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| | - Carlo Serra
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mahendra Rao
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| |
Collapse
|
3
|
Ortiz Silva NA, Denis S, Vergnaud J, Hillaireau H. Controlled hydrogel-based encapsulation of macrophages determines cell survival and functionality upon cryopreservation. Int J Pharm 2024; 650:123491. [PMID: 37806508 DOI: 10.1016/j.ijpharm.2023.123491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
The development of novel cell-based therapies has increased the necessity to improve the long-term storage of cells. The current method of cryopreservation is far from optimal, causing ice-associated mechanical and osmotic damage to sensitive cells. Cell encapsulation is emerging as a new strategy to overcome those current limitations; however, few data are applicable to slow freezing, with conflicting results and multiple experimental conditions. The objective of this research work was to evaluate the impact of capsule size and encapsulation method on cell survival and functionality after a conventional freezing protocol. To this end, cells were encapsulated in alginate beads of different sizes, spanning the range of 200-2000 µm thanks to multiple extrusion techniques and conditions, and further cryopreserved using a slow cooling rate (-1°C/min) and 10 % DMSO as cryoprotectant. Our data show that there is a strong correlation between bead size and cell survival after a slow cooling cryopreservation process, with cell viabilities ranging from 7 to 70 % depending on the capsule size, with the smallest capsules (230 µm) achieving the highest level of survival. The obtained results indicate that the beads' diameter, rather than their morphology or the technique used, plays a significant role in the post-thawing cell survival and functionality. These results show that a fine control of cell encapsulation in alginate hydrogels is required when it comes to overcoming the current limitations of long-term preservation techniques by slow cooling.
Collapse
Affiliation(s)
| | - Stéphanie Denis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Juliette Vergnaud
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Hervé Hillaireau
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France.
| |
Collapse
|
4
|
Netsrithong R, Garcia-Perez L, Themeli M. Engineered T cells from induced pluripotent stem cells: from research towards clinical implementation. Front Immunol 2024; 14:1325209. [PMID: 38283344 PMCID: PMC10811463 DOI: 10.3389/fimmu.2023.1325209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived T (iT) cells represent a groundbreaking frontier in adoptive cell therapies with engineered T cells, poised to overcome pivotal limitations associated with conventional manufacturing methods. iPSCs offer an off-the-shelf source of therapeutic T cells with the potential for infinite expansion and straightforward genetic manipulation to ensure hypo-immunogenicity and introduce specific therapeutic functions, such as antigen specificity through a chimeric antigen receptor (CAR). Importantly, genetic engineering of iPSC offers the benefit of generating fully modified clonal lines that are amenable to rigorous safety assessments. Critical to harnessing the potential of iT cells is the development of a robust and clinically compatible production process. Current protocols for genetic engineering as well as differentiation protocols designed to mirror human hematopoiesis and T cell development, vary in efficiency and often contain non-compliant components, thereby rendering them unsuitable for clinical implementation. This comprehensive review centers on the remarkable progress made over the last decade in generating functional engineered T cells from iPSCs. Emphasis is placed on alignment with good manufacturing practice (GMP) standards, scalability, safety measures and quality controls, which constitute the fundamental prerequisites for clinical application. In conclusion, the focus on iPSC as a source promises standardized, scalable, clinically relevant, and potentially safer production of engineered T cells. This groundbreaking approach holds the potential to extend hope to a broader spectrum of patients and diseases, leading in a new era in adoptive T cell therapy.
Collapse
Affiliation(s)
- Ratchapong Netsrithong
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Laura Garcia-Perez
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Maria Themeli
- Department of Hematology, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
5
|
Tabata Y, Joanna I, Higuchi A. Stem cell culture and differentiation in 3-D scaffolds. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:109-127. [PMID: 37678968 DOI: 10.1016/bs.pmbts.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Conventional two-dimensional (2-D) cultivation are easy to utilize for human pluripotent stem (hPS) cell cultivation in standard techniques and are important for analysis or development of the signal pathways to keep pluripotent state of hPS cells cultivated on 2-D cell culture materials. However, the most efficient protocol to prepare hPS cells is the cell culture in a three dimensional (3-D) cultivation unit because huge numbers of hPS cells should be utilized in clinical treatment. Some 3-D cultivation strategies for hPS cells are considered: (a) microencapsulated cell cultivation in suspended hydrogels, (b) cell cultivation on microcarriers (MCs), (c) cell cultivation on self-aggregated spheroid [cell aggregates; embryoid bodies (EBs) and organoids], (d) cell cultivation on microfibers or nanofibers, and (e) cell cultivation in macroporous scaffolds. These cultivation ways are described in this chapter.
Collapse
Affiliation(s)
- Yasuhiko Tabata
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kawara-cho, Shogoin, Sakyo-ku, Kyoto, Japan.
| | - Idaszek Joanna
- Division of Materials Design, Faculty of Materials Science and Engineering, Warsaw University of Technology, Woloska Street, Warsaw, Poland
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan; State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China.
| |
Collapse
|
6
|
Cohen PJR, Luquet E, Pletenka J, Leonard A, Warter E, Gurchenkov B, Carrere J, Rieu C, Hardouin J, Moncaubeig F, Lanero M, Quelennec E, Wurtz H, Jamet E, Demarco M, Banal C, Van Liedekerke P, Nassoy P, Feyeux M, Lefort N, Alessandri K. Engineering 3D micro-compartments for highly efficient and scale-independent expansion of human pluripotent stem cells in bioreactors. Biomaterials 2023; 295:122033. [PMID: 36764194 DOI: 10.1016/j.biomaterials.2023.122033] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Human pluripotent stem cells (hPSCs) have emerged as the most promising cellular source for cell therapies. To overcome the scale-up limitations of classical 2D culture systems, suspension cultures have been developed to meet the need for large-scale culture in regenerative medicine. Despite constant improvements, current protocols that use microcarriers or generate cell aggregates only achieve moderate amplification performance. Here, guided by reports showing that hPSCs can self-organize in vitro into cysts reminiscent of the epiblast stage in embryo development, we developed a physio-mimetic approach for hPSC culture. We engineered stem cell niche microenvironments inside microfluidics-assisted core-shell microcapsules. We demonstrate that lumenized three-dimensional colonies significantly improve viability and expansion rates while maintaining pluripotency compared to standard hPSC culture platforms such as 2D cultures, microcarriers, and aggregates. By further tuning capsule size and culture conditions, we scale up this method to industrial-scale stirred tank bioreactors and achieve an unprecedented hPSC amplification rate of 277-fold in 6.5 days. In brief, our findings indicate that our 3D culture system offers a suitable strategy both for basic stem cell biology experiments and for clinical applications.
Collapse
Affiliation(s)
- Philippe J R Cohen
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France.
| | | | | | | | | | | | | | | | | | | | | | - Eddy Quelennec
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France
| | | | | | | | - Celine Banal
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | - Paul Van Liedekerke
- Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, F-75012, Paris, France
| | - Pierre Nassoy
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France; Institut D'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France
| | | | - Nathalie Lefort
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | | |
Collapse
|
7
|
Alginates Combined with Natural Polymers as Valuable Drug Delivery Platforms. Mar Drugs 2022; 21:md21010011. [PMID: 36662184 PMCID: PMC9861938 DOI: 10.3390/md21010011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Alginates (ALG) have been used in biomedical and pharmaceutical technologies for decades. ALG are natural polymers occurring in brown algae and feature multiple advantages, including biocompatibility, low toxicity and mucoadhesiveness. Moreover, ALG demonstrate biological activities per se, including anti-hyperlipidemic, antimicrobial, anti-reflux, immunomodulatory or anti-inflammatory activities. ALG are characterized by gelling ability, one of the most frequently utilized properties in the drug form design. ALG have numerous applications in pharmaceutical technology that include micro- and nanoparticles, tablets, mucoadhesive dosage forms, wound dressings and films. However, there are some shortcomings, which impede the development of modified-release dosage forms or formulations with adequate mechanical strength based on pure ALG. Other natural polymers combined with ALG create great potential as drug carriers, improving limitations of ALG matrices. Therefore, in this paper, ALG blends with pectins, chitosan, gelatin, and carrageenans were critically reviewed.
Collapse
|
8
|
Wang O, Han L, Lin H, Tian M, Zhang S, Duan B, Chung S, Zhang C, Lian X, Wang Y, Lei Y. Fabricating 3-dimensional human brown adipose microtissues for transplantation studies. Bioact Mater 2022; 22:518-534. [PMID: 36330162 PMCID: PMC9619153 DOI: 10.1016/j.bioactmat.2022.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
Transplanting cell cultured brown adipocytes (BAs) represents a promising approach to prevent and treat obesity (OB) and its associated metabolic disorders, including type 2 diabetes mellitus (T2DM). However, transplanted BAs have a very low survival rate in vivo. The enzymatic dissociation during the harvest of fully differentiated BAs also loses significant cells. There is a critical need for novel methods that can avoid cell death during cell preparation, transplantation, and in vivo. Here, we reported that preparing BAs as injectable microtissues could overcome the problem. We found that 3D culture promoted BA differentiation and UCP-1 expression, and the optimal initial cell aggregate size was 100 μm. The microtissues could be produced at large scales via 3D suspension assisted with a PEG hydrogel and could be cryopreserved. Fabricated microtissues could survive in vivo for long term. They alleviated body weight and fat gain and improved glucose tolerance and insulin sensitivity in high-fat diet (HFD)-induced OB and T2DM mice. Transplanted microtissues impacted multiple organs, secreted protein factors, and influenced the secretion of endogenous adipokines. To our best knowledge, this is the first report on fabricating human BA microtissues and showing their safety and efficacy in T2DM mice. The proposal of transplanting fabricated BA microtissues, the microtissue fabrication method, and the demonstration of efficacy in T2DM mice are all new. Our results show that engineered 3D human BA microtissues have considerable advantages in product scalability, storage, purity, safety, dosage, survival, and efficacy.
Collapse
Affiliation(s)
- Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, NE, USA
- Biomedical Engineering Program, University of Nebraska-Lincoln, NE, USA
| | - Li Han
- Department of Biomedical Engineering, Pennsylvania State University, PA, USA
| | - Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, NE, USA
| | - Mingmei Tian
- China Novartis Institutes for BioMedical Research Co., Ltd., Beijing, China
| | - Shuyang Zhang
- Department of Chemistry, University of Nebraska-Lincoln, NE, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Soonkyu Chung
- Department of Nutrition, University of Massachusetts, Amherst, MA, USA
| | - Chi Zhang
- School of Biological Science, University of Nebraska-Lincoln, NE, USA
| | - Xiaojun Lian
- Department of Biomedical Engineering, Pennsylvania State University, PA, USA
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University, PA, USA
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, NE, USA
- Department of Biomedical Engineering, Pennsylvania State University, PA, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, PA, USA
- Corresponding author. The Pennsylvania State University, PA, USA.
| |
Collapse
|
9
|
Xie X, Zhou X, Liu T, Zhong Z, Zhou Q, Iqbal W, Xie Q, Wei C, Zhang X, Chang TMS, Sun P. Direct Differentiation of Human Embryonic Stem Cells to 3D Functional Hepatocyte-like Cells in Alginate Microencapsulation Sphere. Cells 2022; 11:3134. [PMID: 36231094 PMCID: PMC9562699 DOI: 10.3390/cells11193134] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/30/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The lack of a stable source of hepatocytes is one of major limitations in hepatocyte transplantation and clinical applications of a bioartificial liver. Human embryonic stem cells (hESCs) with a high degree of self-renewal and totipotency are a potentially limitless source of a variety of cell lineages, including hepatocytes. Many techniques have been developed for effective differentiation of hESCs into functional hepatocyte-like cells. However, the application of hESC-derived hepatocyte-like cells (hESC-Heps) in the clinic has been constrained by the low yield of fully differentiated cells, small-scale culture, difficulties in harvesting, and immunologic graft rejection. To resolve these shortcomings, we developed a novel 3D differentiation system involving alginate-microencapsulated spheres to improve current hepatic differentiation, providing ready-to-use hESC-Heps. METHODS In this study, we used alginate microencapsulation technology to differentiate human embryonic stem cells into hepatocyte-like cells (hESC-Heps). Hepatic markers of hESC-Heps were examined by qPCR and Western blotting, and hepatic functions of hESC-Heps were evaluated by indocyanine-green uptake and release, and ammonia removal. RESULTS The maturity and hepatic functions of the hESC-Heps derived from this 3D system were better than those derived from 2D culture. Hepatocyte-enriched genes, such as HNF4α, AFP, and ALB, were expressed at higher levels in 3D hESC-Heps than in 2D hESC-Heps. 3D hESC-Heps could metabolize indocyanine green and had better capacity to scavenge ammonia. In addition, the 3D sodium alginate hydrogel microspheres could block viral entry into the microspheres, and thus protect hESC-Heps in 3D microspheres from viral infection. CONCLUSION We developed a novel 3D differentiation system for differentiating hESCs into hepatocyte-like cells by using alginate microcapsules.
Collapse
Affiliation(s)
- Xiaoling Xie
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- Guangdong Chaozhou Health Vocational College, Chaozhou 521000, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Xiaoling Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Tingdang Liu
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Zhiqian Zhong
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Qi Zhou
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Waqas Iqbal
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Qingdong Xie
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Chiju Wei
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Xin Zhang
- Laboratory of Molecular Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Thomas Ming Swi Chang
- Artificial Cells & Organs Research Centre, Departments of Physiology, Medicine & Biomedical Engineering, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Pingnan Sun
- Stem Cell Research Center, Shantou University Medical College, Shantou 515041, China
- The Center for Reproductive Medicine, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
10
|
Hasturk O, Smiley JA, Arnett M, Sahoo JK, Staii C, Kaplan DL. Cytoprotection of Human Progenitor and Stem Cells through Encapsulation in Alginate Templated, Dual Crosslinked Silk and Silk-Gelatin Composite Hydrogel Microbeads. Adv Healthc Mater 2022; 11:e2200293. [PMID: 35686928 PMCID: PMC9463115 DOI: 10.1002/adhm.202200293] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/28/2022] [Indexed: 01/27/2023]
Abstract
Susceptibility of mammalian cells against harsh processing conditions limit their use in cell transplantation and tissue engineering applications. Besides modulation of the cell microenvironment, encapsulation of mammalian cells within hydrogel microbeads attract attention for cytoprotection through physical isolation of the encapsulated cells. The hydrogel formulations used for cell microencapsulation are largely dominated by ionically crosslinked alginate (Alg), which suffer from low structural stability under physiological culture conditions and poor cell-matrix interactions. Here the fabrication of Alg templated silk and silk/gelatin composite hydrogel microspheres with permanent or on-demand cleavable enzymatic crosslinks using simple and cost-effective centrifugation-based droplet processing are demonstrated. The composite microbeads display structural stability under ion exchange conditions with improved mechanical properties compared to ionically crosslinked Alg microspheres. Human mesenchymal stem and neural progenitor cells are successfully encapsulated in the composite beads and protected against environmental factors, including exposure to polycations, extracellular acidosis, apoptotic cytokines, ultraviolet (UV) irradiation, anoikis, immune recognition, and particularly mechanical stress. The microbeads preserve viability, growth, and differentiation of encapsulated stem and progenitor cells after extrusion in viscous polyethylene oxide solution through a 27-gauge fine needle, suggesting potential applications in injection-based delivery and three-dimensional bioprinting of mammalian cells with higher success rates.
Collapse
Affiliation(s)
- Onur Hasturk
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Jordan A. Smiley
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Miles Arnett
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
11
|
Heydarzadeh S, Kia SK, Boroomand S, Hedayati M. Recent Developments in Cell Shipping Methods. Biotechnol Bioeng 2022; 119:2985-3006. [PMID: 35898166 DOI: 10.1002/bit.28197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/09/2022] [Accepted: 07/17/2022] [Indexed: 11/11/2022]
Abstract
As opposed to remarkable advances in the cell therapy industry, researches reveal inexplicable difficulties associated with preserving and post-thawing cell death. Post cryopreservation apoptosis is a common occurrence that has attracted the attention of scientists to use apoptosis inhibitors. Transporting cells without compromising their survival and function is crucial for any experimental cell-based therapy. Preservation of cells allows the safe transportation of cells between distances and improves quality control testing in clinical and research applications. The vitality of transported cells is used to evaluate the efficacy of transportation strategies. For many decades, the conventional global methods of cell transfer were not only expensive but also challenging and had adverse effects. The first determination of some projects is optimizing cell survival after cryopreservation. The new generation of cryopreservation science wishes to find appropriate and alternative methods for cell transportation to ship viable cells at an ambient temperature without dry ice or in media-filled flasks. The diversity of cell therapies demands new cell shipping methodologies and cryoprotectants. In this review, we tried to summarize novel improved cryopreservation methods and alternatives to cryopreservation with safe and viable cell shipping at ambient temperature, including dry preservation, hypothermic preservation, gel-based methods, encapsulation methods, fibrin microbeads, and osmolyte solution compositions. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shabnam Heydarzadeh
- Department of Biochemistry, School of Biological Sciences, Falavarjan Branch Islamic Azad University, Isfahan, Iran.,Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sima Kheradmand Kia
- Laboratory for Red Blood Cell Diagnostics, Sanquin, Amsterdam, The Netherlands
| | - Seti Boroomand
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mehdi Hedayati
- Djavad Mowafaghian Centre for Brain Health, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Choi D, Gwon K, Hong HJ, Baskaran H, Calvo-Lozano O, Gonzalez-Suarez AM, Park K, de Hoyos-Vega JM, Lechuga LM, Hong J, Stybayeva G, Revzin A. Coating Bioactive Microcapsules with Tannic Acid Enhances the Phenotype of the Encapsulated Pluripotent Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10.1021/acsami.2c06783. [PMID: 35658394 PMCID: PMC10314364 DOI: 10.1021/acsami.2c06783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Human pluripotent stem cells (hPSCs) may be differentiated into any adult cell type and therefore hold incredible promise for cell therapeutics and disease modeling. There is increasing interest in three-dimensional (3D) hPSC culture because of improved differentiation outcomes and potential for scale up. Our team has recently described bioactive heparin (Hep)-containing core-shell microcapsules that promote rapid aggregation of stem cells into spheroids and may also be loaded with growth factors for the local and sustained delivery to the encapsulated cells. In this study, we explored the possibility of further modulating bioactivity of microcapsules through the use of an ultrathin coating composed of tannic acid (TA). Deposition of the TA film onto model substrates functionalized with Hep and poly(ethylene glycol) was characterized by ellipsometry and atomic force microscopy. Furthermore, the presence of the TA coating was observed to increase the amount of basic fibroblast growth factor (bFGF) incorporation by up to twofold and to extend its release from 5 to 7 days. Most significantly, TA-microcapsules loaded with bFGF induced higher levels of pluripotency expression compared to uncoated microcapsules containing bFGF. Engineered microcapsules described here represent a new stem cell culture approach that enables 3D cultivation and relies on local delivery of inductive cues.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Olalla Calvo-Lozano
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBERBBN and BIST, Barcelona 08193, Spain
| | - Alan M Gonzalez-Suarez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Kyungtae Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Jose M de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Laura M Lechuga
- Nanobiosensors and Bioanalytical Applications Group (NanoB2A), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC, CIBERBBN and BIST, Barcelona 08193, Spain
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
13
|
Tan LS, Chen JT, Lim LY, Teo AKK. Manufacturing clinical-grade human induced pluripotent stem cell-derived beta cells for diabetes treatment. Cell Prolif 2022; 55:e13232. [PMID: 35474596 PMCID: PMC9357357 DOI: 10.1111/cpr.13232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/26/2022] [Accepted: 03/28/2022] [Indexed: 12/25/2022] Open
Abstract
The unlimited proliferative capacity of human pluripotent stem cells (hPSCs) fortifies it as one of the most attractive sources for cell therapy application in diabetes. In the past two decades, vast research efforts have been invested in developing strategies to differentiate hPSCs into clinically suitable insulin‐producing endocrine cells or functional beta cells (β cells). With the end goal being clinical translation, it is critical for hPSCs and insulin‐producing β cells to be derived, handled, stored, maintained and expanded with clinical compliance. This review focuses on the key processes and guidelines for clinical translation of human induced pluripotent stem cell (hiPSC)‐derived β cells for diabetes cell therapy. Here, we discuss the (1) key considerations of manufacturing clinical‐grade hiPSCs, (2) scale‐up and differentiation of clinical‐grade hiPSCs into β cells in clinically compliant conditions and (3) mandatory quality control and product release criteria necessitated by various regulatory bodies to approve the use of the cell‐based products.
Collapse
Affiliation(s)
- Lay Shuen Tan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juin Ting Chen
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lillian Yuxian Lim
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
14
|
Zhou P, Qin L, Ge Z, Xie B, Huang H, He F, Ma S, Ren L, Shi J, Pei S, Dong G, Qi Y, Lan F. Design of chemically defined synthetic substrate surfaces for the in vitro maintenance of human pluripotent stem cells: A review. J Biomed Mater Res B Appl Biomater 2022; 110:1968-1990. [PMID: 35226397 DOI: 10.1002/jbm.b.35034] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential of long-term self-renewal and differentiation into nearly all cell types in vitro. Prior to the downstream applications, the design of chemically defined synthetic substrates for the large-scale proliferation of quality-controlled hPSCs is critical. Although great achievements have been made, Matrigel and recombinant proteins are still widely used in the fundamental research and clinical applications. Therefore, much effort is still needed to improve the performance of synthetic substrates in the culture of hPSCs, realizing their commercial applications. In this review, we summarized the design of reported synthetic substrates and especially their limitations in terms of cell culture. Moreover, much attention was paid to the development of promising peptide displaying surfaces. Besides, the biophysical regulation of synthetic substrate surfaces as well as the three-dimensional culture systems were described.
Collapse
Affiliation(s)
- Ping Zhou
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Zhangjie Ge
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Biyao Xie
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Hongxin Huang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Fei He
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Shengqin Ma
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lina Ren
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiamin Shi
- Department of Laboratory Animal Centre, Changzhi Medical College, Changzhi, China
| | - Suying Pei
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Genxi Dong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Shenzhen, China
| |
Collapse
|
15
|
Domenici G, Trindade G, Estrada MF, Cartaxo AL, Alves PM, André S, Brito C. Patient-Derived Breast Cancer Tissue Cultures for Anti-Endocrine Drug Assays. Methods Mol Biol 2022; 2535:11-31. [PMID: 35867219 DOI: 10.1007/978-1-0716-2513-2_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Breast cancer is a complex and heterogeneous pathology, characterized by a variety of histological and molecular phenotypes. The majority of the breast cancers express the estrogen receptor alpha (ER), which plays a pivotal role in the pathobiology of the disease and are therefore classified as ER-positive (ER+). In fact, targeting of the ER signaling pathway is the main therapeutic strategy for ER+ breast cancer. Despite the success of endocrine therapy, intrinsic and acquired resistance are reported in 30-50% of the ER+ breast cancers. However, the mechanisms underlying ER heterogeneity and therapeutic resistance are far from being fully disclosed, and efficacious clinical strategies to overcome resistance are still pending. One of the hurdles in studying ER+ breast cancer resistance is related with the scarcity of experimental models that can recapitulate ER heterogeneity and signaling. This is the case of ER+ breast cancer cell models, typically based on cells derived from metastasis, which also fail to recapitulate tumor complexity. Primary cultures of patient-derived breast cancer cells are difficult to establish, and generally characterized by stromal fibroblasts overgrowth and rapid loss of phenotypic and molecular traits of the tumor cells, including ER expression. Ex vivo cultures of breast cancer tissue have been reported to retain the tissue architecture, with preservation of the tumor microenvironment (TME) and ER expression for short periods of time.Given the cumulating evidence on the role of the TME in sustaining ER+ tumor cells, we hypothesized that TME preservation in culture would favor the long-term retention of ER expression and signaling. We employed alginate encapsulation to provide a supporting scaffold to breast cancer tissue microstructures, coupled to dynamic culture to improve the lifespan of the culture by avoiding diffusional limitations. In this chapter, we provide a detailed description of this culture methodology, which has been previously published by our group (Cartaxo et al., J Exp Clin Cancer Res 39:161, 2020), based on electrostatically driven breast cancer tissue encapsulation in alginate, coupled to culture under agitation in a defined culture medium. We also describe challenge of the ex vivo model with an ER activator and inhibitors (anti-endocrine drugs) and a gene expression endpoint of drug response using reverse transcription PCR-based analysis of three distinct genes downstream of ER.
Collapse
Affiliation(s)
- Giacomo Domenici
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Gonçalo Trindade
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Marta F Estrada
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana Luísa Cartaxo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Saudade André
- IPOLFG, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
16
|
Preparation of Alginate-Based Biomaterials and Their Applications in Biomedicine. Mar Drugs 2021; 19:md19050264. [PMID: 34068547 PMCID: PMC8150954 DOI: 10.3390/md19050264] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
Alginates are naturally occurring polysaccharides extracted from brown marine algae and bacteria. Being biocompatible, biodegradable, non-toxic and easy to gel, alginates can be processed into various forms, such as hydrogels, microspheres, fibers and sponges, and have been widely applied in biomedical field. The present review provides an overview of the properties and processing methods of alginates, as well as their applications in wound healing, tissue repair and drug delivery in recent years.
Collapse
|
17
|
Gryshkov O, Mutsenko V, Tarusin D, Khayyat D, Naujok O, Riabchenko E, Nemirovska Y, Danilov A, Petrenko AY, Glasmacher B. Coaxial Alginate Hydrogels: From Self-Assembled 3D Cellular Constructs to Long-Term Storage. Int J Mol Sci 2021; 22:3096. [PMID: 33803546 PMCID: PMC8003018 DOI: 10.3390/ijms22063096] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
Alginate as a versatile naturally occurring biomaterial has found widespread use in the biomedical field due to its unique features such as biocompatibility and biodegradability. The ability of its semipermeable hydrogels to provide a favourable microenvironment for clinically relevant cells made alginate encapsulation a leading technology for immunoisolation, 3D culture, cryopreservation as well as cell and drug delivery. The aim of this work is the evaluation of structural properties and swelling behaviour of the core-shell capsules for the encapsulation of multipotent stromal cells (MSCs), their 3D culture and cryopreservation using slow freezing. The cells were encapsulated in core-shell capsules using coaxial electrospraying, cultured for 35 days and cryopreserved. Cell viability, metabolic activity and cell-cell interactions were analysed. Cryopreservation of MSCs-laden core-shell capsules was performed according to parameters pre-selected on cell-free capsules. The results suggest that core-shell capsules produced from the low viscosity high-G alginate are superior to high-M ones in terms of stability during in vitro culture, as well as to solid beads in terms of promoting formation of viable self-assembled cellular structures and maintenance of MSCs functionality on a long-term basis. The application of 0.3 M sucrose demonstrated a beneficial effect on the integrity of capsules and viability of formed 3D cell assemblies, as compared to 10% dimethyl sulfoxide (DMSO) alone. The proposed workflow from the preparation of core-shell capsules with self-assembled cellular structures to the cryopreservation appears to be a promising strategy for their off-the-shelf availability.
Collapse
Affiliation(s)
- Oleksandr Gryshkov
- Institute for Multiphase Processes, Leibniz University Hannover, An der Universität 1, Building 8143, 30823 Garbsen, Germany; (V.M.); (D.K.); (B.G.)
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| | - Vitalii Mutsenko
- Institute for Multiphase Processes, Leibniz University Hannover, An der Universität 1, Building 8143, 30823 Garbsen, Germany; (V.M.); (D.K.); (B.G.)
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| | - Dmytro Tarusin
- Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavsky Street, 61015 Kharkiv, Ukraine; (D.T.); (Y.N.); (A.Y.P.)
| | - Diaa Khayyat
- Institute for Multiphase Processes, Leibniz University Hannover, An der Universität 1, Building 8143, 30823 Garbsen, Germany; (V.M.); (D.K.); (B.G.)
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| | - Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany;
| | - Ekaterina Riabchenko
- Institute for Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia; (E.R.); (A.D.)
| | - Yuliia Nemirovska
- Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavsky Street, 61015 Kharkiv, Ukraine; (D.T.); (Y.N.); (A.Y.P.)
| | - Arseny Danilov
- Institute for Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia; (E.R.); (A.D.)
| | - Alexander Y. Petrenko
- Institute for Problems of Cryobiology and Cryomedicine of the National Academy of Sciences of Ukraine, 23 Pereyaslavsky Street, 61015 Kharkiv, Ukraine; (D.T.); (Y.N.); (A.Y.P.)
| | - Birgit Glasmacher
- Institute for Multiphase Processes, Leibniz University Hannover, An der Universität 1, Building 8143, 30823 Garbsen, Germany; (V.M.); (D.K.); (B.G.)
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| |
Collapse
|
18
|
Almeida HV, Tenreiro MF, Louro AF, Abecasis B, Santinha D, Calmeiro T, Fortunato E, Ferreira L, Alves PM, Serra M. Human Extracellular-Matrix Functionalization of 3D hiPSC-Based Cardiac Tissues Improves Cardiomyocyte Maturation. ACS APPLIED BIO MATERIALS 2021; 4:1888-1899. [PMID: 35014458 DOI: 10.1021/acsabm.0c01490] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human induced pluripotent stem cells (hiPSC) possess significant therapeutic potential due to their high self-renewal capability and potential to differentiate into specialized cells such as cardiomyocytes. However, generated hiPSC-derived cardiomyocytes (hiPSC-CM) are still immature, with phenotypic and functional features resembling the fetal rather than their adult counterparts, which limits their application in cell-based therapies, in vitro cardiac disease modeling, and drug cardiotoxicity screening. Recent discoveries have demonstrated the potential of the extracellular matrix (ECM) as a critical regulator in development, homeostasis, and injury of the cardiac microenvironment. Within this context, this work aimed to assess the impact of human cardiac ECM in the phenotype and maturation features of hiPSC-CM. Human ECM was isolated from myocardium tissue through a physical decellularization approach. The cardiac tissue decellularization process reduced DNA content significantly while maintaining ECM composition in terms of sulfated glycosaminoglycans (s-GAG) and collagen content. These ECM particles were successfully incorporated in three-dimensional (3D) hiPSC-CM aggregates (CM+ECM) with no impact on viability and metabolic activity throughout 20 days in 3D culture conditions. Also, CM+ECM aggregates displayed organized and longer sarcomeres, with improved calcium handling when compared to hiPSC-CM aggregates. This study shows that human cardiac ECM functionalization of hiPSC-based cardiac tissues improves cardiomyocyte maturation. The knowledge generated herein provides essential insights to streamline the application of ECM in the development of hiPSC-based therapies targeting cardiac diseases.
Collapse
Affiliation(s)
- Henrique V Almeida
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Miguel F Tenreiro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F Louro
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Bernardo Abecasis
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Deolinda Santinha
- CNC, Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004-517 Coimbra, Portugal.,Faculdade de Medicina, Universidade de Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Tomás Calmeiro
- CENIMAT
- i3N, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Elvira Fortunato
- CENIMAT
- i3N, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Lino Ferreira
- CNC, Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004-517 Coimbra, Portugal.,Faculdade de Medicina, Universidade de Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
19
|
McKee C, Brown C, Bakshi S, Walker K, Govind CK, Chaudhry GR. Transcriptomic Analysis of Naïve Human Embryonic Stem Cells Cultured in Three-Dimensional PEG Scaffolds. Biomolecules 2020; 11:E21. [PMID: 33379237 PMCID: PMC7824559 DOI: 10.3390/biom11010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022] Open
Abstract
Naïve human embryonic stem cells (ESCs) are characterized by improved viability, proliferation, and differentiation capacity in comparison to traditionally derived primed human ESCs. However, currently used two-dimensional (2-D) cell culture techniques fail to mimic the three-dimensional (3-D) in vivo microenvironment, altering morphological and molecular characteristics of ESCs. Here, we describe the use of 3-D self-assembling scaffolds that support growth and maintenance of the naïve state characteristics of ESC line, Elf1. Scaffolds were formed via a Michael addition reaction upon the combination of two 8-arm polyethylene glycol (PEG) polymers functionalized with thiol (PEG-8-SH) and acrylate (PEG-8-Acr) end groups. 3-D scaffold environment maintained the naïve state and supported the long-term growth of ESCs. RNA-sequencing demonstrated significant changes in gene expression profiles between 2-D and 3-D grown cells. Gene ontology analysis revealed upregulation of biological processes involved in the regulation of transcription and translation, extracellular matrix organization, and chromatin remodeling in 3-D grown cells. 3-D culture conditions also induced upregulation of genes associated with Wnt and focal adhesion signaling, while p53 signaling pathway associated genes were downregulated. Our findings, for the first time, provide insight into the possible mechanisms of self-renewal of naïve ESCs stimulated by the transduction of mechanical signals from the 3-D microenvironment.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Christina Brown
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Shreeya Bakshi
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Keegan Walker
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - Chhabi K. Govind
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| | - G. Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (C.M.); (C.B.); (S.B.); (K.W.); (C.K.G.)
- OU-WB Institute for Stem Cell and Regenerative Medicine, Rochester, MI 48309, USA
| |
Collapse
|
20
|
Abecasis B, Canhão PGM, Almeida HV, Calmeiro T, Fortunato E, Gomes-Alves P, Serra M, Alves PM. Toward a Microencapsulated 3D hiPSC-Derived in vitro Cardiac Microtissue for Recapitulation of Human Heart Microenvironment Features. Front Bioeng Biotechnol 2020; 8:580744. [PMID: 33224931 PMCID: PMC7674657 DOI: 10.3389/fbioe.2020.580744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/14/2020] [Indexed: 12/28/2022] Open
Abstract
The combination of cardiomyocytes (CM) and non-myocyte cardiac populations, such as endothelial cells (EC), and mesenchymal cells (MC), has been shown to be critical for recapitulation of the human heart tissue for in vitro cell-based modeling. However, most of the current engineered cardiac microtissues still rely on either (i) murine/human limited primary cell sources, (ii) animal-derived and undefined hydrogels/matrices with batch-to-batch variability, or (iii) culture systems with low compliance with pharmacological high-throughput screenings. In this work, we explored a culture platform based on alginate microencapsulation and suspension culture systems to develop three-dimensional (3D) human cardiac microtissues, which entails the co-culture of human induced pluripotent stem cell (hiPSC) cardiac derivatives including aggregates of hiPSC–CM and single cells of hiPSC–derived EC and MC (hiPSC–EC+MC). We demonstrate that the 3D human cardiac microtissues can be cultured for 15 days in dynamic conditions while maintaining the viability and phenotype of all cell populations. Noteworthy, we show that hiPSC–EC+MC survival was promoted by the co-culture with hiPSC–CM as compared to the control single-cell culture. Additionally, the presence of the hiPSC–EC+MC induced changes in the physical properties of the biomaterial, as observed by an increase in the elastic modulus of the cardiac microtissue when compared to the hiPSC–CM control culture. Detailed characterization of the 3D cardiac microtissues revealed that the crosstalk between hiPSC–CM, hiPSC–EC+MC, and extracellular matrix induced the maturation of hiPSC–CM. The cardiac microtissues displayed functional calcium signaling and respond to known cardiotoxins in a dose-dependent manner. This study is a step forward on the development of novel 3D cardiac microtissues that recapitulate features of the human cardiac microenvironment and is compliant with the larger numbers needed in preclinical research for toxicity assessment and disease modeling.
Collapse
Affiliation(s)
- Bernardo Abecasis
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Pedro G M Canhão
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Henrique V Almeida
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Tomás Calmeiro
- CENIMAT
- i3N, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Elvira Fortunato
- CENIMAT
- i3N, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Patrícia Gomes-Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
21
|
Montero P, Flandes-Iparraguirre M, Musquiz S, Pérez Araluce M, Plano D, Sanmartín C, Orive G, Gavira JJ, Prosper F, Mazo MM. Cells, Materials, and Fabrication Processes for Cardiac Tissue Engineering. Front Bioeng Biotechnol 2020; 8:955. [PMID: 32850768 PMCID: PMC7431658 DOI: 10.3389/fbioe.2020.00955] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease is the number one killer worldwide, with myocardial infarction (MI) responsible for approximately 1 in 6 deaths. The lack of endogenous regenerative capacity, added to the deleterious remodelling programme set into motion by myocardial necrosis, turns MI into a progressively debilitating disease, which current pharmacological therapy cannot halt. The advent of Regenerative Therapies over 2 decades ago kick-started a whole new scientific field whose aim was to prevent or even reverse the pathological processes of MI. As a highly dynamic organ, the heart displays a tight association between 3D structure and function, with the non-cellular components, mainly the cardiac extracellular matrix (ECM), playing both fundamental active and passive roles. Tissue engineering aims to reproduce this tissue architecture and function in order to fabricate replicas able to mimic or even substitute damaged organs. Recent advances in cell reprogramming and refinement of methods for additive manufacturing have played a critical role in the development of clinically relevant engineered cardiovascular tissues. This review focuses on the generation of human cardiac tissues for therapy, paying special attention to human pluripotent stem cells and their derivatives. We provide a perspective on progress in regenerative medicine from the early stages of cell therapy to the present day, as well as an overview of cellular processes, materials and fabrication strategies currently under investigation. Finally, we summarise current clinical applications and reflect on the most urgent needs and gaps to be filled for efficient translation to the clinical arena.
Collapse
Affiliation(s)
- Pilar Montero
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - María Flandes-Iparraguirre
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
| | - Saioa Musquiz
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
| | - María Pérez Araluce
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country – UPV/EHU, Vitoria-Gasteiz, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- University Institute for Regenerative Medicine and Oral Implantology – UIRMI (UPV/EHU – Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, Singapore, Singapore
| | - Juan José Gavira
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Cardiology Department, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Manuel M. Mazo
- Regenerative Medicine Program, Cima Universidad de Navarra, Foundation for Applied Medical Research, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Hematology and Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
22
|
Koh B, Sulaiman N, Fauzi MB, Law JX, Ng MH, Idrus RBH, Yazid MD. Three dimensional microcarrier system in mesenchymal stem cell culture: a systematic review. Cell Biosci 2020; 10:75. [PMID: 32518618 PMCID: PMC7271456 DOI: 10.1186/s13578-020-00438-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/27/2020] [Indexed: 01/09/2023] Open
Abstract
Stem cell-based regenerative medicine is a promising approach for tissue reconstruction. However, a large number of cells are needed in a typical clinical study, where conventional monolayer cultures might pose a limitation for scale-up. The purpose of this review was to systematically assess the application of microcarriers in Mesenchymal Stem Cell cultures. A comprehensive search was conducted in Medline via Ebscohost, Pubmed, and Scopus, and relevant studies published between 2015 and 2019 were selected. The literature search identified 53 related studies, but only 14 articles met the inclusion criteria. These include 7 utilised commercially available microcarriers, while the rest were formulated based on different surface characteristics, all of which are discussed in this review. Current applications of microcarriers were focused on MSC expansion and induction of MSCs into different lineages. These studies demonstrated that MSCs could proliferate in a microcarrier culture system in-fold compared to monolayer cultures, and the culture system could simulate a three-dimensional environment which induces cell differentiation. However, detailed studies are still required before this system were to be adapted into the scale of GMP manufacturing.
Collapse
Affiliation(s)
- Benson Koh
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Nadiah Sulaiman
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Mh Busra Fauzi
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia.,Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Khatab S, Leijs MJ, van Buul G, Haeck J, Kops N, Nieboer M, Bos PK, Verhaar JAN, Bernsen M, van Osch GJVM. MSC encapsulation in alginate microcapsules prolongs survival after intra-articular injection, a longitudinal in vivo cell and bead integrity tracking study. Cell Biol Toxicol 2020; 36:553-570. [PMID: 32474743 PMCID: PMC7661423 DOI: 10.1007/s10565-020-09532-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSC) are promising candidates for use as a biological therapeutic. Since locally injected MSC disappear within a few weeks, we hypothesize that efficacy of MSC can be enhanced by prolonging their presence. Previously, encapsulation in alginate was suggested as a suitable approach for this purpose. We found no differences between the two alginate types, alginate high in mannuronic acid (High M) and alginate high in guluronic acid (High G), regarding MSC viability, MSC immunomodulatory capability, or retention of capsule integrity after subcutaneous implantation in immune competent rats. High G proved to be more suitable for production of injectable beads. Firefly luciferase-expressing rat MSC were used to track MSC viability. Encapsulation in high G alginate prolonged the presence of metabolically active allogenic MSC in immune competent rats with monoiodoacetate-induced osteoarthritis for at least 8 weeks. Encapsulation of human MSC for local treatment by intra-articular injection did not significantly influence the effect on pain, synovial inflammation, or cartilage damage in this disease model. MSC encapsulation in alginate allows for an injectable approach which prolongs the presence of viable cells subcutaneously or in an osteoarthritic joint. Further fine tuning of alginate formulation and effective dosage for might be required in order to improve therapeutic efficacy depending on the target disease. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Sohrab Khatab
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Maarten J Leijs
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Gerben van Buul
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Joost Haeck
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Nicole Kops
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Michael Nieboer
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - P Koen Bos
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Jan A N Verhaar
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Monique Bernsen
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
- Department of Otorhinolaryngology, Erasmus MC University Medical Center Rotterdam, Wytemaweg 80, 3015, CN, Rotterdam, the Netherlands.
| |
Collapse
|
24
|
Liu W, Deng C, Godoy-Parejo C, Zhang Y, Chen G. Developments in cell culture systems for human pluripotent stem cells. World J Stem Cells 2019; 11:968-981. [PMID: 31768223 PMCID: PMC6851012 DOI: 10.4252/wjsc.v11.i11.968] [Citation(s) in RCA: 8] [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: 03/26/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are important resources for cell-based therapies and pharmaceutical applications. In order to realize the potential of hPSCs, it is critical to develop suitable technologies required for specific applications. Most hPSC technologies depend on cell culture, and are critically influenced by culture medium composition, extracellular matrices, handling methods, and culture platforms. This review summarizes the major technological advances in hPSC culture, and highlights the opportunities and challenges in future therapeutic applications.
Collapse
Affiliation(s)
- Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
25
|
Khanal S, Bhattarai SR, Sankar J, Bhandari RK, Macdonald JM, Bhattarai N. Nano-fibre Integrated Microcapsules: A Nano-in-Micro Platform for 3D Cell Culture. Sci Rep 2019; 9:13951. [PMID: 31562351 PMCID: PMC6765003 DOI: 10.1038/s41598-019-50380-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/09/2019] [Indexed: 01/25/2023] Open
Abstract
Nano-in-micro (NIM) system is a promising approach to enhance the performance of devices for a wide range of applications in disease treatment and tissue regeneration. In this study, polymeric nanofibre-integrated alginate (PNA) hydrogel microcapsules were designed using NIM technology. Various ratios of cryo-ground poly (lactide-co-glycolide) (PLGA) nanofibres (CPN) were incorporated into PNA hydrogel microcapsule. Electrostatic encapsulation method was used to incorporate living cells into the PNA microcapsules (~500 µm diameter). Human liver carcinoma cells, HepG2, were encapsulated into the microcapsules and their physio-chemical properties were studied. Morphology, stability, and chemical composition of the PNA microcapsules were analysed by light microscopy, fluorescent microscopy, scanning electron microscopy (SEM), Fourier-Transform Infrared spectroscopy (FTIR), and thermogravimetric analysis (TGA). The incorporation of CPN caused no significant changes in the morphology, size, and chemical structure of PNA microcapsules in cell culture media. Among four PNA microcapsule products (PNA-0, PNA-10, PNA-30, and PNA-50 with size 489 ± 31 µm, 480 ± 40 µm, 473 ± 51 µm and 464 ± 35 µm, respectively), PNA-10 showed overall suitability for HepG2 growth with high cellular metabolic activity, indicating that the 3D PNA-10 microcapsule could be suitable to maintain better vitality and liver-specific metabolic functions. Overall, this novel design of PNA microcapsule and the one-step method of cell encapsulation can be a versatile 3D NIM system for spontaneous generation of organoids with in vivo like tissue architectures, and the system can be useful for numerous biomedical applications, especially for liver tissue engineering, cell preservation, and drug toxicity study.
Collapse
Affiliation(s)
- Shalil Khanal
- 0000 0001 0287 4439grid.261037.1Department of Applied Science and Technology, North Carolina A&T State University, Greensboro, NC USA ,0000 0001 0287 4439grid.261037.1Department of Chemical, Biological, and Bioengineering, North Carolina A&T State University, Greensboro, NC USA
| | - Shanta R. Bhattarai
- 0000 0001 0287 4439grid.261037.1Department of Chemistry, North Carolina A&T State University, Greensboro, NC USA ,0000 0001 0287 4439grid.261037.1Department of Biology, North Carolina A&T State University, Greensboro, NC USA ,0000 0001 0671 255Xgrid.266860.cDepartment of Biology, University of North Carolina Greensboro, Greensboro, NC USA
| | - Jagannathan Sankar
- 0000 0001 0287 4439grid.261037.1Department of Mechanical Engineering, North Carolina A&T State University, Greensboro, NC USA
| | - Ramji K. Bhandari
- 0000 0001 0671 255Xgrid.266860.cDepartment of Biology, University of North Carolina Greensboro, Greensboro, NC USA
| | - Jeffrey M. Macdonald
- 0000 0001 1034 1720grid.410711.2Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC USA
| | - Narayan Bhattarai
- 0000 0001 0287 4439grid.261037.1Department of Chemical, Biological, and Bioengineering, North Carolina A&T State University, Greensboro, NC USA
| |
Collapse
|
26
|
Hasturk O, Kaplan DL. Cell armor for protection against environmental stress: Advances, challenges and applications in micro- and nanoencapsulation of mammalian cells. Acta Biomater 2019; 95:3-31. [PMID: 30481608 PMCID: PMC6534491 DOI: 10.1016/j.actbio.2018.11.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Unlike unicellular organisms and plant cells surrounded with a cell wall, naked plasma membranes of mammalian cells make them more susceptible to environmental stresses encountered during in vitro biofabrication and in vivo cell therapy applications. Recent advances in micro- and nanoencapsulation of single mammalian cells provide an effective strategy to isolate cells from their surroundings and protect them against harsh environmental conditions. Microemulsification and droplet-based microfluidics have enabled researchers to encapsulate single cells within a variety of microscale hydrogel materials with a range of biochemical and mechanical properties and functionalities including enhanced cell-matrix interactions or on-demand degradation. In addition to microcapsules, nanocoatings of various organic and inorganic substances on mammalian cells have allowed for the formation of protective shells. A wide range of synthetic and natural polymers, minerals and supramolecular metal-organic complexes have been deposited as nanolayers on the cells via electrostatic interactions, receptor-ligand binding, non-specific interactions, and in situ polymerization/crosslinking. Here, current strategies in encapsulation of single mammalian cells along with challenges and advances are reviewed. Protection of encapsulated stem cells, fibroblasts, red and white blood cells and cancer cells against harsh in vitro and in vivo conditions including anoikis, UV radiation, physical forces, proteolytic enzymes and immune clearance are discussed. STATEMENT OF SIGNIFICANCE: The mechanical fragility of the plasma membrane and susceptibility to extracellular biochemical factors due to the lack of a physical barrier like a tough cell wall or exoskeleton make mammalian cells extra sensitive to harsh environmental conditions. This sensitively, in turn, limits the ex vivo storage, handling and manipulation of mammalian cells, as well as their in vivo applications. Environmental stresses such as exposure to UV, reactive chemicals and mechanical stress during biofabrication processes like 3D bioprinting can often compromise cell viability and function. Micro- and nanoencapsulation of single mammalian cells in protective shells have emerged as promising approaches to isolate cells from their surroundings and enhance resistance against perturbations in conditions during regenerative medicine and tissue engineering applications. In this review, the current state of art of single cell encapsulation strategies and the challenges associated with these technologies are discussed in detail. This is followed by the review of the protection provided by cell armor against a range of harsh in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
27
|
Stem cells in Osteoporosis: From Biology to New Therapeutic Approaches. Stem Cells Int 2019; 2019:1730978. [PMID: 31281368 PMCID: PMC6589256 DOI: 10.1155/2019/1730978] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis is a systemic disease that affects the skeleton, causing reduction of bone density and mass, resulting in destruction of bone microstructure and increased risk of bone fractures. Since osteoporosis is a disease affecting the elderly and the aging of the world's population is constantly increasing, it is expected that the incidence of osteoporosis and its financial burden on the insurance systems will increase continuously and there is a need for more understanding this condition in order to prevent and/or treat it. At present, available drug therapy for osteoporosis primarily targets the inhibition of bone resorption and agents that promote bone mineralization, designed to slow disease progression. Safe and predictable pharmaceutical means to increase bone formation have been elusive. Stem cell therapy of osteoporosis, as a therapeutic strategy, offers the promise of an increase in osteoblast differentiation and thus reversing the shift towards bone resorption in osteoporosis. This review is focused on the current views regarding the implication of the stem cells in the cellular and physiologic mechanisms of osteoporosis and discusses data obtained from stem cell-based therapies of osteoporosis in experimental animal models and the possibility of their future application in clinical trials.
Collapse
|
28
|
McNamara MC, Sharifi F, Okuzono J, Montazami R, Hashemi NN. Microfluidic Manufacturing of Alginate Fibers with Encapsulated Astrocyte Cells. ACS APPLIED BIO MATERIALS 2019; 2:1603-1613. [DOI: 10.1021/acsabm.9b00022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Pereira J, Ferraretto X, Patrat C, Meddahi-Pellé A. Dextran-Based Hydrogel as a New Tool for BALB/c 3T3 Cell Cryopreservation Without Dimethyl Sulfoxide. Biopreserv Biobank 2019; 17:2-10. [DOI: 10.1089/bio.2018.0034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Jessica Pereira
- INSERM, UMR S-1148, LVTS, CHU Xavier Bichat, Paris, France
- Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
| | - Xavier Ferraretto
- INSERM, UMR S-1148, LVTS, CHU Xavier Bichat, Paris, France
- Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
- Department of Reproductive Biology, AP-HP, Bichat-Claude Bernard Hospital, Paris, France
| | - Catherine Patrat
- Department of Reproductive Biology, AP-HP, Bichat-Claude Bernard Hospital, Paris, France
- Université Paris-Diderot, Paris, France
| | - Anne Meddahi-Pellé
- INSERM, UMR S-1148, LVTS, CHU Xavier Bichat, Paris, France
- Université Paris 13, Sorbonne Paris Cité, Villetaneuse, France
| |
Collapse
|
30
|
Chui CY, Bonilla-Brunner A, Seifert J, Contera S, Ye H. Atomic force microscopy-indentation demonstrates that alginate beads are mechanically stable under cell culture conditions. J Mech Behav Biomed Mater 2019; 93:61-69. [PMID: 30772703 DOI: 10.1016/j.jmbbm.2019.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/18/2018] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
Abstract
Alginate microbeads are extensively used in tissue engineering as microcarriers and cell encapsulation vessels. In this study, we used atomic force microscopy (AFM) based indentation using 20 µm colloidal probes to assess the local reduced elastic modulus (E * ) using a novel method to detect the contact point based on the principle of virtual work, to measure microbead mechanical stability under cell culture conditions for 2 weeks. The bead diameter and swelling were assessed in parallel. Alginate beads swelled up to 150% of their original diameter following addition of cell culture media. The diameter eventually stabilized from day 2 onwards. This behaviour was mirrored in E * where a significant decrease was observed at the start of the culture period before stabilization was observed at ~ 2.1 kPa. Furthermore, the mechanical properties of freeze dried alginate beads after re-swelling them in culture media were measured. These beads displayed vastly different structural and mechanical properties compared those that did not go through the freeze drying process, with around 125% swelling and a significantly higher E * at values over 3 kPa.
Collapse
Affiliation(s)
- Chih-Yao Chui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus off Roosevelt Drive, OX3 7DQ, United Kingdom
| | - Andrea Bonilla-Brunner
- Clarendon Laboratory, Department of Physics, University of Oxford, Parks Road, OX1 3PU, United Kingdom
| | - Jacob Seifert
- Clarendon Laboratory, Department of Physics, University of Oxford, Parks Road, OX1 3PU, United Kingdom; Department of Plant Sciences, University of Oxford, South Parks Road, OX1 3RB, United Kingdom
| | - Sonia Contera
- Clarendon Laboratory, Department of Physics, University of Oxford, Parks Road, OX1 3PU, United Kingdom.
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus off Roosevelt Drive, OX3 7DQ, United Kingdom.
| |
Collapse
|
31
|
Lee SY, George JH, Nagel DA, Ye H, Kueberuwa G, Seymour LW. Optogenetic control of iPS cell-derived neurons in 2D and 3D culture systems using channelrhodopsin-2 expression driven by the synapsin-1 and calcium-calmodulin kinase II promoters. J Tissue Eng Regen Med 2019; 13:369-384. [PMID: 30550638 PMCID: PMC6492196 DOI: 10.1002/term.2786] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 09/04/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
Abstract
Development of an optogenetically controllable human neural network model in three-dimensional (3D) cultures can provide an investigative system that is more physiologically relevant and better able to mimic aspects of human brain function. Light-sensitive neurons were generated by transducing channelrhodopsin-2 (ChR2) into human induced pluripotent stem cell (hiPSC) derived neural progenitor cells (Axol) using lentiviruses and cell-type specific promoters. A mixed population of human iPSC-derived cortical neurons, astrocytes and progenitor cells were obtained (Axol-ChR2) upon neural differentiation. Pan-neuronal promoter synapsin-1 (SYN1) and excitatory neuron-specific promoter calcium-calmodulin kinase II (CaMKII) were used to drive reporter gene expression in order to assess the differentiation status of the targeted cells. Expression of ChR2 and characterisation of subpopulations in differentiated Axol-ChR2 cells were evaluated using flow cytometry and immunofluorescent staining. These cells were transferred from 2D culture to 3D alginate hydrogel functionalised with arginine-glycine-aspartate (RGD) and small molecules (Y-27632). Improved RGD-alginate hydrogel was physically characterised and assessed for cell viability to serve as a generic 3D culture system for human pluripotent stem cells (hPSCs) and neuronal cells. Prior to cell encapsulation, neural network activities of Axol-ChR2 cells and primary neurons were investigated using calcium imaging. Results demonstrate that functional activities were successfully achieved through expression of ChR2- by both the CaMKII and SYN1 promoters. The RGD-alginate hydrogel system supports the growth of differentiated Axol-ChR2 cells whilst allowing detection of ChR2 expression upon light stimulation. This allows precise and non-invasive control of human neural networks in 3D.
Collapse
Affiliation(s)
- Si-Yuen Lee
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK.,Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Julian H George
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - David A Nagel
- School of Life and Health Sciences, University of Aston, Birmingham, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Oxford, UK
| | - Gray Kueberuwa
- Department of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Leonard W Seymour
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Laminin as a Potent Substrate for Large-Scale Expansion of Human Induced Pluripotent Stem Cells in a Closed Cell Expansion System. Stem Cells Int 2019; 2019:9704945. [PMID: 30805013 PMCID: PMC6362483 DOI: 10.1155/2019/9704945] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/28/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022] Open
Abstract
The number of high-quality cells required for engineering an adult human-sized bioartificial organ is greater than one billion. Until the emergence of induced pluripotent stem cells (iPSCs), autologous cell sources of this magnitude and with the required complexity were not available. Growing this number of cells in a traditional 2D cell culture system requires extensive time, resources, and effort and does not always meet clinical requirements. The use of a closed cell culture system is an efficient and clinically applicable method that can be used to expand cells under controlled conditions. We aimed to use the Quantum Cell Expansion System (QES) as an iPSC monolayer-based expansion system. Human iPSCs were expanded (up to 14-fold) using the QES on two different coatings (laminin 521 (LN521) and vitronectin (VN)), and a karyotype analysis was performed. The cells were characterized for spontaneous differentiation and pluripotency by RT-PCR and flow cytometry. Our results demonstrated that the QES provides the necessary environment for exponential iPSC growth, reaching 689.75 × 106 ± 86.88 × 106 in less than 7 days using the LN521 coating with a population doubling level of 3.80 ± 0.19. The same result was not observed when VN was used as a coating. The cells maintained normal karyotype (46-XX), expressed pluripotency markers (OCT4, NANOG, LIN28, SOX2, REX1, DPPA4, NODAL, TDGFb, TERT3, and GDF), and expressed high levels of OCT4, SOX2, NANOG, SSEA4, TRA1-60, and TRA1-81. Spontaneous differentiation into ectoderm (NESTIN, TUBB3, and NEFH), mesoderm (MSX1, BMP4, and T), and endoderm (GATA6, AFP, and SOX17) lineages was detected by RT-PCR with both coating systems. We conclude that the QES maintains the stemness of iPSCs and is a promising platform to provide the number of cells necessary to recellularize small human-sized organ scaffolds for clinical purposes.
Collapse
|
33
|
Samaras JJ, Abecasis B, Serra M, Ducci A, Micheletti M. Impact of hydrodynamics on iPSC-derived cardiomyocyte differentiation processes. J Biotechnol 2018; 287:18-27. [DOI: 10.1016/j.jbiotec.2018.07.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 10/28/2022]
|
34
|
The effect of alginate-gelatin encapsulation on the maturation of human myelomonocytic cell line U937. J Tissue Eng Regen Med 2018; 13:25-35. [DOI: 10.1002/term.2765] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/02/2018] [Accepted: 10/18/2018] [Indexed: 01/04/2023]
|
35
|
Rodrigues AL, Rodrigues CAV, Gomes AR, Vieira SF, Badenes SM, Diogo MM, Cabral JM. Dissolvable Microcarriers Allow Scalable Expansion And Harvesting Of Human Induced Pluripotent Stem Cells Under Xeno‐Free Conditions. Biotechnol J 2018; 14:e1800461. [DOI: 10.1002/biot.201800461] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- André L. Rodrigues
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
- The Discoveries Centre for Regenerative and Precision MedicineLisbon CampusInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Carlos A. V. Rodrigues
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
- The Discoveries Centre for Regenerative and Precision MedicineLisbon CampusInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Ana R. Gomes
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
- The Discoveries Centre for Regenerative and Precision MedicineLisbon CampusInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Sara F. Vieira
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Sara M. Badenes
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Maria M. Diogo
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
- The Discoveries Centre for Regenerative and Precision MedicineLisbon CampusInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| | - Joaquim M.S. Cabral
- Department of Bioengineering and iBB‐Institute for Bioengineering and BiosciencesInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
- The Discoveries Centre for Regenerative and Precision MedicineLisbon CampusInstituto Superior TécnicoUniversidade de Lisboa1049‐001 LisboaPortugal
| |
Collapse
|
36
|
Zhang C, Zhou Y, Zhang L, Wu L, Chen Y, Xie D, Chen W. Hydrogel Cryopreservation System: An Effective Method for Cell Storage. Int J Mol Sci 2018; 19:E3330. [PMID: 30366453 PMCID: PMC6274795 DOI: 10.3390/ijms19113330] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/12/2018] [Accepted: 10/20/2018] [Indexed: 12/27/2022] Open
Abstract
At present, living cells are widely used in cell transplantation and tissue engineering. Many efforts have been made aiming towards the use of a large number of living cells with high activity and integrated functionality. Currently, cryopreservation has become well-established and is effective for the long-term storage of cells. However, it is still a major challenge to inhibit cell damage, such as from solution injury, ice injury, recrystallization and osmotic injury during the thawing process, and the cytotoxicity of cryoprotectants. Hence, this review focused on different novel gel cryopreservation systems. Natural polymer hydrogel cryopreservation, the synthetic polymer hydrogel cryopreservation system and the supramolecular hydrogel cryopreservation system were presented, respectively. Due to the unique three-dimensional network structure of the hydrogel, these hydrogel cryopreservation systems have the advantages of excellent biocompatibility for natural polymer hydrogel cryopreservation systems, designability for synthetic polymer hydrogel cryopreservation systems, and versatility for supramolecular hydrogel cryopreservation systems. To some extent, the different hydrogel cryopreservation methods can confine ice crystal growth and decrease the change rates of osmotic shock in cell encapsulation systems. It is notable that the cryopreservation of complex cells and tissues is demanded in future clinical research and therapy, and depends on the linkage of different methods.
Collapse
Affiliation(s)
- Chaocan Zhang
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Youliang Zhou
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Li Zhang
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Lili Wu
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Yanjun Chen
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Dong Xie
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| | - Wanyu Chen
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
37
|
Chui CY, Odeleye A, Nguyen L, Kasoju N, Soliman E, Ye H. Electrosprayed genipin cross-linked alginate-chitosan microcarriers for ex vivo expansion of mesenchymal stem cells. J Biomed Mater Res A 2018; 107:122-133. [PMID: 30256517 DOI: 10.1002/jbm.a.36539] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 11/06/2022]
Abstract
Mesenchymal stem cells (MSCs) are potential therapeutic candidates, owing to their ability to differentiate into several cell types. However, the gap between availability and demand of MSCs requires alternative expansion methods from 2D culture flasks. Microcarriers are a promising approach for MSC expansion due to their high surface area-to-volume ratio. However, current commercial microcarriers do not provide the highest cell yield due to low cell attachment efficiencies and difficulty in cell detachment. This study developed a hydrogel-based microcarrier from genipin cross-linked alginate-chitosan beads. Alginate beads were produced by electrospraying before being coated with chitosan and cross-linked in genipin. The degree of cross-linking was determined through fluorescence reading of the genipin-chitosan conjugates. MSCs cultured on these microcarriers had a 26% higher cell attachment and twice the proliferation rate compared to the commercial microcarrier Cytodex 1. Cells easily detached from the hydrogel beads and did not require extended incubation periods or intense agitation during cell harvest. There was no significant difference in gene expression between the two microcarriers for the positive MSC surface markers CD-90, CD-105, and CD-73 as well as showing either low or no signal for negative MSC surface markers CD-45 and CD-34. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 122-133, 2019.
Collapse
Affiliation(s)
- Chih-Yao Chui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Akinlolu Odeleye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Linh Nguyen
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Naresh Kasoju
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Erfan Soliman
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| | - Hua Ye
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
38
|
Choe G, Park J, Park H, Lee JY. Hydrogel Biomaterials for Stem Cell Microencapsulation. Polymers (Basel) 2018; 10:E997. [PMID: 30960922 PMCID: PMC6403586 DOI: 10.3390/polym10090997] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
Stem cell transplantation has been recognized as a promising strategy to induce the regeneration of injured and diseased tissues and sustain therapeutic molecules for prolonged periods in vivo. However, stem cell-based therapy is often ineffective due to low survival, poor engraftment, and a lack of site-specificity. Hydrogels can offer several advantages as cell delivery vehicles, including cell stabilization and the provision of tissue-like environments with specific cellular signals; however, the administration of bulk hydrogels is still not appropriate to obtain safe and effective outcomes. Hence, stem cell encapsulation in uniform micro-sized hydrogels and their transplantation in vivo have recently garnered great attention for minimally invasive administration and the enhancement of therapeutic activities of the transplanted stem cells. Several important methods for stem cell microencapsulation are described in this review. In addition, various natural and synthetic polymers, which have been employed for the microencapsulation of stem cells, are reviewed in this article.
Collapse
Affiliation(s)
- Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| | - Junha Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea.
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea.
| |
Collapse
|
39
|
A fully defined static suspension culture system for large-scale human embryonic stem cell production. Cell Death Dis 2018; 9:892. [PMID: 30166524 PMCID: PMC6117302 DOI: 10.1038/s41419-018-0863-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/16/2018] [Accepted: 06/21/2018] [Indexed: 12/30/2022]
Abstract
Human embryonic stem cells (hESCs) play an important role in regenerative medicine due to their potential to differentiate into various functional cells. However, the conventional adherent culture system poses challenges to mass production of high-quality hESCs. Though scientists have made many attempts to establish a robust and economical hESC suspension culture system, there are existing limitations, including suboptimal passage methods and shear force caused by dynamic stirring. Here, we report on an efficient large-scale culture system, which enables long-term, GMP grade, single-cell inoculation, and serial expansion of hESCs with a yield of about 1.5 × 109 cells per 1.5-L culture, while maintaining good pluripotency. The suspension culture system was enlarged gradually from a 100-mm dish to a 1.8-L culture bag with methylcellulose involvement to avoid sphere fusion. Under the optimal experimental protocol, this 3D system resolves current problems that limit mass production and clinical application of hESCs, and thus can be used in commercial-level hESC production for cell therapy and pharmaceutics screening in the future.
Collapse
|
40
|
Intra-Articular Injection of Alginate-Microencapsulated Adipose Tissue-Derived Mesenchymal Stem Cells for the Treatment of Osteoarthritis in Rabbits. Stem Cells Int 2018; 2018:2791632. [PMID: 30046312 PMCID: PMC6038583 DOI: 10.1155/2018/2791632] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/17/2018] [Accepted: 05/20/2018] [Indexed: 12/14/2022] Open
Abstract
We investigated the effects of intra-articular injections of alginate-microencapsulated adipose tissue-derived mesenchymal stem cells (ASCs) during osteoarthritis (OA) development in a rabbit model of anterior cruciate ligament transection (ACLT). We induced OA in mature New Zealand white rabbits by bilateral ACLT. Stifle joints were categorised into four groups according to intra-articular injection materials. Alginate microbeads and microencapsulated ASCs were prepared using the vibrational nozzle technology. Two weeks after ACLT, the rabbits received three consecutive weekly intra-articular injections of 0.9% NaCl, alginate microbeads, ASCs, or microencapsulated ASCs, into each joint. Nine weeks after ACLT, we euthanised the rabbits and collected bilateral femoral condyles for macroscopic, histological, and immunohistochemical analyses. Macroscopic evaluation using the modified OA Research Society International (OARSI) score and total cartilage damage score showed that cartilage degradation on the femoral condyle was relatively low in the microencapsulated-ASC group. Histological analysis of the lateral femoral condyles indicated that microencapsulated ASCs had significant chondroprotective effects. Immunohistochemically, the expression of MMP-13 after the articular cartilage damage was relatively low in the microencapsulated-ASC-treated stifle joints. During the development of experimental OA, as compared to ASCs alone, intra-articular injection of microencapsulated ASCs significantly decreased the progression and extent of OA.
Collapse
|
41
|
Ekerdt BL, Fuentes CM, Lei Y, Adil MM, Ramasubramanian A, Segalman RA, Schaffer DV. Thermoreversible Hyaluronic Acid-PNIPAAm Hydrogel Systems for 3D Stem Cell Culture. Adv Healthc Mater 2018; 7:e1800225. [PMID: 29717823 PMCID: PMC6289514 DOI: 10.1002/adhm.201800225] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/27/2018] [Indexed: 12/20/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer considerable potential for biomedical applications including drug screening and cell replacement therapies. Clinical translation of hPSCs requires large quantities of high quality cells, so scalable methods for cell culture are needed. However, current methods are limited by scalability, the use of animal-derived components, and/or low expansion rates. A thermoresponsive 3D hydrogel for scalable hPSC expansion and differentiation into several defined lineages is recently reported. This system would benefit from increased control over material properties to further tune hPSC behavior, and here a scalable 3D biomaterial with the capacity to tune both the chemical and the mechanical properties is demonstrated to promote hPSC expansion under defined conditions. This 3D biomaterial, comprised of hyaluronic acid and poly(N-isopropolyacrylamide), has thermoresponsive properties that readily enable mixing with cells at low temperatures, physical encapsulation within the hydrogel upon elevation at 37 °C, and cell recovery upon cooling and reliquefaction. After optimization, the resulting biomaterial supports hPSC expansion over long cell culture periods while maintaining cell pluripotency. The capacity to modulate the mechanical and chemical properties of the hydrogel provides a new avenue to expand hPSCs for future therapeutic application.
Collapse
Affiliation(s)
- Barbara L. Ekerdt
- Department of Chemical and Biolomolecular Engineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
| | - Christina M. Fuentes
- Department of Bioengineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, 207 Othmer, University of Nebraska - Lincoln, Lincoln, NE 68588, USA
| | - Maroof M. Adil
- Department of Chemical and Biolomolecular Engineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
| | - Anusuya Ramasubramanian
- Department of Bioengineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
| | - Rachel A. Segalman
- Department of Chemical Engineering, 3333 Engineering IIUniversity of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - David V. Schaffer
- Department of Chemical and Biolomolecular Engineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
- Department of Bioengineering, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
- Department of Molecular and Cell Biology, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
- The Helen Wills Neuroscience Institute, 274 Stanley Hall University of California, Berkeley, Berkeley, CA, USA,
| |
Collapse
|
42
|
Naqvi SM, Gansau J, Buckley CT. Priming and cryopreservation of microencapsulated marrow stromal cells as a strategy for intervertebral disc regeneration. ACTA ACUST UNITED AC 2018; 13:034106. [PMID: 29380742 DOI: 10.1088/1748-605x/aaab7f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A challenge in using stromal cells for intervertebral disc (IVD) regeneration is their limited differentiation capacity in vivo without exogenous growth factor (GF) supplementation. Priming of stromal cells prior to transplantation may offer a feasible strategy to overcome this limitation. Furthermore, the ability to cryopreserve cells could help alleviate logistical issues associated with storage and transport. With these critical translational challenges in mind, we aimed to develop a strategy involving priming and subsequent cryopreservation of microencapsulated bone marrow stromal cells (BMSCs). In phase one, we utilised the electrohydrodynamic atomisation process to fabricate BMSC-encapsulated microcapsules that were primed with TGF-β3 for 14 d after which they were cultured for a further 21 d under basal or GF supplemented media conditions. Results showed that priming induced differentiation of BMSC microcapsules such that they synthesised significant amounts of sGAG (61.9 ± 2.0 μg and 55.3 ± 6.1 μg for low and high cell densities) and collagen (24.4 ± 1.9 μg and 55.3 ± 4.6 μg for low and high cell densities) in continued culture without GF supplementation compared to Unprimed microcapsules. Phase two of this work assessed the extracellular matrix forming capacity of Primed BMSC microcapsules over 21 d after cryopreservation. Notably, primed and cryopreserved BMSCs successfully retained the ability to synthesise both sGAG (24.8 ± 2.7 μg and 75.1 ± 11.6 μg for low and high cell densities) and collagen (26.4 ± 7.8 μg and 93.1 ± 10.2 μg for low and high cell densities) post-cryopreservation. These findings demonstrate the significant potential of priming and cryopreservation approaches for IVD repair and could possibly open new horizons for pre-designed, 'off-the-shelf' injectable therapeutics.
Collapse
Affiliation(s)
- Syeda M Naqvi
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland. School of Engineering, Trinity College Dublin, Ireland
| | | | | |
Collapse
|
43
|
Li Q, Lin H, Du Q, Liu K, Wang O, Evans C, Christian H, Zhang C, Lei Y. Scalable and physiologically relevant microenvironments for human pluripotent stem cell expansion and differentiation. Biofabrication 2018; 10:025006. [PMID: 29319535 DOI: 10.1088/1758-5090/aaa6b5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human pluripotent stem cells (hPSCs) are required in large numbers for various biomedical applications. However, the scalable and cost-effective culturing of high quality hPSCs and their derivatives remains very challenging. Here, we report a novel and physiologically relevant 3D culture system (called the AlgTube cell culture system) for hPSC expansion and differentiation. With this system, cells are processed into and cultured in microscale alginate hydrogel tubes that are suspended in the cell culture medium in a culture vessel. The hydrogel tubes protect cells from hydrodynamic stresses in the culture vessel and limit the cell mass smaller than 400 μm in diameter to ensure efficient mass transport, creating cell-friendly microenvironments for growing cells. This system is simple, scalable, highly efficient, defined and compatible with the current good manufacturing practices. Under optimized culture conditions, the AlgTubes enabled long-term culture of hPSCs (>10 passages, >50 days) with high cell viability, high growth rate (1000-fold expansion over 10 days per passage), high purity (>95% Oct4+) and high yield (5.0 × 108 cells ml-1), all of which offer considerable advantages compared to current approaches. Moreover, the AlgTubes enabled directed differentiation of hPSCs into various tissue cells. This system can be readily scaled to support research from basic biological study to clinical development and the future industry-scale production.
Collapse
Affiliation(s)
- Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America. Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Neuro-differentiated Ntera2 cancer stem cells encapsulated in alginate beads: First evidence of biological functionality. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 81:32-38. [DOI: 10.1016/j.msec.2017.07.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 07/07/2017] [Accepted: 07/19/2017] [Indexed: 12/29/2022]
|
45
|
Zhao G, Liu X, Zhu K, He X. Hydrogel Encapsulation Facilitates Rapid-Cooling Cryopreservation of Stem Cell-Laden Core-Shell Microcapsules as Cell-Biomaterial Constructs. Adv Healthc Mater 2017; 6:10.1002/adhm.201700988. [PMID: 29178480 PMCID: PMC5729581 DOI: 10.1002/adhm.201700988] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/30/2017] [Indexed: 01/08/2023]
Abstract
Core-shell structured stem cell microencapsulation in hydrogel has wide applications in tissue engineering, regenerative medicine, and cell-based therapies because it offers an ideal immunoisolative microenvironment for cell delivery and 3D culture. Long-term storage of such microcapsules as cell-biomaterial constructs by cryopreservation is an enabling technology for their wide distribution and ready availability for clinical transplantation. However, most of the existing studies focus on cryopreservation of single cells or cells in microcapsules without a core-shell structure (i.e., hydrogel beads). The goal of this study is to achieve cryopreservation of stem cells encapsulated in core-shell microcapsules as cell-biomaterial constructs or biocomposites. To this end, a capillary microfluidics-based core-shell alginate hydrogel encapsulation technology is developed to produce porcine adipose-derived stem cell-laden microcapsules for vitreous cryopreservation with very low concentration (2 mol L-1 ) of cell membrane penetrating cryoprotective agents (CPAs) by suppressing ice formation. This may provide a low-CPA and cost-effective approach for vitreous cryopreservation of "ready-to-use" stem cell-biomaterial constructs, facilitating their off-the-shelf availability and widespread applications.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Xiaoli Liu
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Kaixuan Zhu
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
46
|
Finding the design space of a filtration-based operation for the concentration of human pluripotent stem cells. J Memb Sci 2017. [DOI: 10.1016/j.memsci.2017.08.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
47
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 479] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
48
|
Kilbride P, Lamb S, Gibbons S, Bundy J, Erro E, Selden C, Fuller B, Morris J. Cryopreservation and re-culture of a 2.3 litre biomass for use in a bioartificial liver device. PLoS One 2017; 12:e0183385. [PMID: 28841674 PMCID: PMC5572048 DOI: 10.1371/journal.pone.0183385] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 07/28/2017] [Indexed: 12/29/2022] Open
Abstract
For large and complex tissue engineered constructs to be available on demand, long term storage using methods, such as cryopreservation, are essential. This study optimised parameters such as excess media concentration and warming rates and used the findings to enable the successful cryopreservation of 2.3 litres of alginate encapsulated liver cell spheroids. This volume of biomass is typical of those required for successful treatment of Acute Liver Failure using our Bioartificial Liver Device. Adding a buffer of medium above the biomass, as well as slow (0.6°C/min) warming rates was found to give the best results, so long as the warming through the equilibrium melting temperature was rapid. After 72 h post thaw-culture, viable cell number, glucose consumption, lactate production, and alpha-fetoprotein production had recovered to pre-freeze values in the 2.3 litre biomass (1.00 ± 0.05, 1.19 ± 0.10, 1.23 ± 0.18, 2.03 ± 0.04 per ml biomass of the pre-cryopreservation values respectively). It was also shown that further improvements in warming rates of the biomass could reduce recovery time to < 48 h. This is the first example of a biomass of this volume being successfully cryopreserved in a single cassette and re-cultured. It demonstrates that a bioartificial liver device can be cryopreserved, and has wider applications to scale-up large volume cryopreservation.
Collapse
Affiliation(s)
- Peter Kilbride
- Asymptote, General Electric Healthcare, Cambridge, United Kingdom
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London, United Kingdom
- * E-mail:
| | - Stephen Lamb
- Asymptote, General Electric Healthcare, Cambridge, United Kingdom
| | - Stephanie Gibbons
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London, United Kingdom
| | - James Bundy
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London, United Kingdom
| | - Eloy Erro
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London, United Kingdom
| | - Clare Selden
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London, United Kingdom
| | - Barry Fuller
- Department of Surgery, Royal Free Hospital Campus, University College London, London, United Kingdom
| | - John Morris
- Asymptote, General Electric Healthcare, Cambridge, United Kingdom
| |
Collapse
|
49
|
Li Y, Li L, Chen ZN, Gao G, Yao R, Sun W. Engineering-derived approaches for iPSC preparation, expansion, differentiation and applications. Biofabrication 2017; 9:032001. [DOI: 10.1088/1758-5090/aa7e9a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Kilbride P, Mahbubani KT, Saeb-Parsy K, Morris GJ. Engaging Cold to Upregulate Cell Proliferation in Alginate-Encapsulated Liver Spheroids. Tissue Eng Part C Methods 2017; 23:455-464. [PMID: 28727981 DOI: 10.1089/ten.tec.2017.0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
For many years, the impact of hyper- and hypothermia on mammalian cells has been examined. With the exception of short, low temperature storage, which has uses in areas such as preservation for transplantation or regenerative medicine, advantages for the use of low temperature treatment in hepatocytes have not been previously reported. We have observed that alginate-encapsulated HepG2 liver spheroids that are cryopreserved or experience a cold reduction in temperature (≤10°C) for periods between 1 and 90 min display an enhanced cell proliferation during culture 7-16 days post-treatment compared with untreated samples. Following 8-12 days post-treatment, alginate-encapsulated liver spheroids experienced a cell density of 1.71 ± 0.35 times that of control samples (p < 0.001). This effect occurred in samples with a variety of cold treatments. This low temperature treatment offers a simple method to rapidly increase cell proliferation rates for extended culture systems, such as bioartificial liver devices. This would allow the manufacture of required biomass more rapidly, and to a higher cell density, reducing final required biomass volume. This could enable bioartificial liver devices to be prepared more cheaply, making them a more cost effective treatment.
Collapse
Affiliation(s)
| | - Krishnaa T Mahbubani
- 2 Department of Surgery, University of Cambridge , Cambridge, United Kingdom .,3 Cambridge NIHR Biomedical Research Centre , Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- 2 Department of Surgery, University of Cambridge , Cambridge, United Kingdom .,3 Cambridge NIHR Biomedical Research Centre , Cambridge, United Kingdom
| | | |
Collapse
|