1
|
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: 0.3] [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
|
2
|
Assis JLD, Fernandes AM, Aniceto BS, Fernandes da Costa PP, Banchio C, Girardini J, Vieyra A, Valverde RRHF, Einicker‐Lamas M. Sphingosine 1‐Phosphate Prevents Human Embryonic Stem Cell Death Following Ischemic Injury. EUR J LIPID SCI TECH 2022. [DOI: 10.1002/ejlt.202200019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Juliane L. de Assis
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Aline M. Fernandes
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Bárbara S. Aniceto
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Pedro P. Fernandes da Costa
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Claudia Banchio
- Instituto de Biologia Molecular y Celular de Rosário Rosário Argentina
| | - Javier Girardini
- Instituto de Biologia Molecular y Celular de Rosário Rosário Argentina
| | - Adalberto Vieyra
- Laboratório de Físico‐Química Biológica Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Rafael R. H. F. Valverde
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| | - Marcelo Einicker‐Lamas
- Laboratório de Biomembranas Instituto de Biofísica Carlos Chagas Filho–Universidade Federal do Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
3
|
Wang Z, Zuo F, Liu Q, Wu X, Du Q, Lei Y, Wu Z, Lin H. Comparative Study of Human Pluripotent Stem Cell-Derived Endothelial Cells in Hydrogel-Based Culture Systems. ACS OMEGA 2021; 6:6942-6952. [PMID: 33748608 PMCID: PMC7970572 DOI: 10.1021/acsomega.0c06187] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial cells (ECs) are promising cell sources for drug discovery, tissue engineering, and studying or treating vascular diseases. However, hPSC-ECs derived from different culture methods display different phenotypes. Herein, we made a detailed comparative study of hPSC-ECs from three different culture systems (e.g., 2D, 3D PNIPAAm-PEG hydrogel, and 3D alginate hydrogel cultures) based on our previous reports. We expanded hPSCs and differentiated them into ECs in three culture systems. Both 3D hydrogel systems could mimic an in vivo physiologically relevant microenvironment to protect cells from shear force and prevent cell agglomeration, leading to a high culture efficiency and a high volumetric yield. We demonstrated that hPSC-ECs produced from both hydrogel systems had similar results as 2D-ECs. The transcriptome analysis showed that PEG-ECs and alginate-ECs displayed a functional phenotype due to their higher gene expressions in vasculature development, extracellular matrix, angiogenesis, and glycolysis, while 2D-ECs showed a proliferative phenotype due to their higher gene expressions in cell proliferation. Taken together, both PEG- and alginate-hydrogel systems will significantly advance the applications of hPSC-ECs in various biomedical fields.
Collapse
Affiliation(s)
- Zhanqi Wang
- Department
of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Fuxing Zuo
- Department
of Neurosurgery, National Cancer Center/National Clinical Research
Center for Cancer/Cancer Hospital, Chinese
Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qing Liu
- Department
of Obstetrics, Beijing Obstetrics and Gynecology
Hospital Capital Medical University, Beijing 100006, China
| | - Xuesheng Wu
- Department
of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qian Du
- Department
of Biological Systems Engineering, University
of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Yuguo Lei
- Department
of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Zhangmin Wu
- Department
of Vascular Surgery, Beijing Anzhen Hospital of Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Haishuang Lin
- Department
of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
4
|
Torizal FG, Horiguchi I, Sakai Y. Physiological Microenvironmental Conditions in Different Scalable Culture Systems for Pluripotent Stem Cell Expansion and Differentiation. Open Biomed Eng J 2019. [DOI: 10.2174/1874120701913010041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human Pluripotent Stem Cells (PSCs) are a valuable cell type that has a wide range of biomedical applications because they can differentiate into many types of adult somatic cell. Numerous studies have examined the clinical applications of PSCs. However, several factors such as bioreactor design, mechanical stress, and the physiological environment have not been optimized. These factors can significantly alter the pluripotency and proliferation properties of the cells, which are important for the mass production of PSCs. Nutritional mass transfer and oxygen transfer must be effectively maintained to obtain a high yield. Various culture systems are currently available for optimum cell propagation by maintaining the physiological conditions necessary for cell cultivation. Each type of culture system using a different configuration with various advantages and disadvantages affecting the mechanical conditions in the bioreactor, such as shear stress. These factors make it difficult to preserve the cellular viability and pluripotency of PSCs. Additional limitations of the culture system for PSCs must also be identified and overcome to maintain the culture conditions and enable large-scale expansion and differentiation of PSCs. This review describes the different physiological conditions in the various culture systems and recent developments in culture technology for PSC expansion and differentiation.
Collapse
|
5
|
Al Abbar A, Nordin N, Ghazalli N, Abdullah S. Generation of induced pluripotent stem cells by a polycistronic lentiviral vector in feeder- and serum- free defined culture. Tissue Cell 2018; 55:13-24. [DOI: 10.1016/j.tice.2018.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/08/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022]
|
6
|
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: 67] [Impact Index Per Article: 8.4] [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
|
7
|
Kim MH, Matsubara Y, Fujinaga Y, Kino-Oka M. A Simple and Robust Method for Culturing Human-Induced Pluripotent Stem Cells in an Undifferentiated State Using Botulinum Hemagglutinin. Biotechnol J 2017; 13. [PMID: 29027750 DOI: 10.1002/biot.201700384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/24/2017] [Indexed: 01/18/2023]
Abstract
Clinical and industrial applications of human-induced pluripotent stem cells (hiPSCs) is hindered by the lack of robust culture strategies capable of sustaining a culture in an undifferentiated state. Here, a simple and robust hiPSC-culture-propagation strategy incorporating botulinum hemagglutinin (HA)-mediated selective removal of cells deviating from an undifferentiated state is developed. After HA treatment, cell-cell adhesion is disrupted, and deviated cells detached from the central region of the colony to subsequently form tight monolayer colonies following prolonged incubation. The authors find that the temporal and dose-dependent activity of HA regulated deviated-cell removal and recoverability after disruption of cell-cell adhesion in hiPSC colonies. The effects of HA are confirmed under all culture conditions examined, regardless of hiPSC line and feeder-dependent or -free culture conditions. After routine application of our HA-treatment paradigm for serial passages, hiPSCs maintains expression of pluripotent markers and readily forms embryoid bodies expressing markers for all three germ-cell layers. This method enables highly efficient culturing of hiPSCs and use of entire undifferentiated portions without having to pick deviated cells manually. This simple and readily reproducible culture strategy is a potentially useful tool for improving the robust and scalable maintenance of undifferentiated hiPSC cultures.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Matsubara
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yukako Fujinaga
- Laboratory for Infection Cell Biology, International Research Centre for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
8
|
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: 521] [Impact Index Per Article: 65.1] [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
|
9
|
Ko JY, Oh HJ, Lee J, Im GI. Nanotopographic Influence on the In Vitro Behavior of Induced Pluripotent Stem Cells. Tissue Eng Part A 2017; 24:595-606. [PMID: 28726546 DOI: 10.1089/ten.tea.2017.0144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
While the influence of nanotopography on stem cell behavior has been extensively investigated on adult stem cells, far fewer studies have investigated the interaction of induced pluripotent stem cells (iPSCs) with various nanotopographical patterns. The purpose of this study was to identify nanopatterns that can influence the stemness and proliferation, as well as the adhesive properties in iPSCs, and thereby explore the feasibility of applying these nano-features for regenerative medicine. Three kinds of nanopatterns were fabricated from polydimethylsiloxane membranes, irregular patterned membrane (IPM), groove patterned membrane (GPM), and postpatterned membrane (PPM), in addition to flat patterned membrane (FPM) which did not have any nanotopographic features and was used as the control pattern. On the surfaces of GPM or PPM, iPSCs showed tendency for aggregation and did not spread out well at passage 1. However, with continued passaging (P6, P10), the tendency to form aggregates was greatly reduced. While iPSCs cultured on GPM and PPM had low population doubling time values compared with FPM and IPM at P1, the differences disappeared in later passages. The expression of the cell proliferation marker Ki67 in iPSCs gradually decreased with continued passaging in cells cultured on FPM and IPM, but not in those cultured on GPM and PPM. The expression of Oct3/4 and Nanog, marker of stemness, was significantly higher on GPM and PPM than on FPM at P6 and P10. At P5, numerous filopodia were demonstrated in the peripheral attachments of iPSC colonies on FPM and IPM, while GPM and PPM generally had globular appearance. The expression of the focal adhesion (FA) molecules α-actinin, vinculin, phalloidin, or FA kinase was significantly greater on GPM and PPM than on FPM and IPM at P6 or P10. In conclusion, continued passaging on regular nanopatterns, including groove- and post-forms, was effective in maintaining an undifferentiated state and proliferation of iPSCs and also in increasing the expression of FA molecules.
Collapse
Affiliation(s)
- Ji-Yun Ko
- 1 Department of Orthopaedics, Dongguk University Ilsan Hospital , Goyang, Republic of Korea
| | - Hyun-Jik Oh
- 2 Department of Biomedical Engineering, College of Health Science, Korea University , Seoul, Republic of Korea.,3 MicroFIT R&BD Institute , Gyeonggi-do, Republic of Korea
| | - Jimin Lee
- 1 Department of Orthopaedics, Dongguk University Ilsan Hospital , Goyang, Republic of Korea
| | - Gun-Il Im
- 1 Department of Orthopaedics, Dongguk University Ilsan Hospital , Goyang, Republic of Korea
| |
Collapse
|
10
|
Cell fiber-based three-dimensional culture system for highly efficient expansion of human induced pluripotent stem cells. Sci Rep 2017; 7:2850. [PMID: 28588295 PMCID: PMC5460280 DOI: 10.1038/s41598-017-03246-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells are a potentially powerful cellular resource for application in regenerative medicine. Because such applications require large numbers of human pluripotent stem cell-derived cells, a scalable culture system of human pluripotent stem cell needs to be developed. Several suspension culture systems for human pluripotent stem cell expansion exist; however, it is difficult to control the thickness of cell aggregations in these systems, leading to increased cell death likely caused by limited diffusion of gases and nutrients into the aggregations. Here, we describe a scalable culture system using the cell fiber technology for the expansion of human induced pluripotent stem (iPS) cells. The cells were encapsulated and cultured within the core region of core-shell hydrogel microfibers, resulting in the formation of rod-shaped or fiber-shaped cell aggregations with sustained thickness and high viability. By encapsulating the cells with type I collagen, we demonstrated a long-term culture of the cells by serial passaging at a high expansion rate (14-fold in four days) while retaining its pluripotency. Therefore, our culture system could be used for large-scale expansion of human pluripotent stem cells for use in regenerative medicine.
Collapse
|
11
|
|
12
|
He X. Microscale Biomaterials with Bioinspired Complexity of Early Embryo Development and in the Ovary for Tissue Engineering and Regenerative Medicine. ACS Biomater Sci Eng 2016; 3:2692-2701. [PMID: 29367949 DOI: 10.1021/acsbiomaterials.6b00540] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tissue engineering and regenerative medicine (TERM) are attracting more and more attention for treating various diseases in modern medicine. Various biomaterials including hydrogels and scaffolds have been developed to prepare cells (particularly stem cells) and tissues under 3D conditions for TERM applications. Although these biomaterials are usually homogeneous in early studies, effort has been made recently to generate biomaterials with the spatiotemporal complexities present in the native milieu of the specific cells and tissues under investigation. In this communication, the microfluidic and coaxial electrospray approaches that we used for generating microscale biomaterials with the spatial complexity of both pre-hatching embryos and ovary in the female reproductive system were introduced. This is followed by an overview of our recent work on applying the resultant bioinspired biomaterials for cultivation of normal and cancer stem cells, regeneration of cardiac tissue, and culture of ovarian follicles. The cardiac regeneration studies show the importance of using different biomaterials to engineer stem cells at different stages (i.e., in vitro culture versus in vivo implantation) for tissue regeneration. All the studies demonstrate the merit of accounting for bioinspired complexities in engineering cells and tissues for TERM applications.
Collapse
Affiliation(s)
- Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA.,Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
13
|
Microcarrier-based platforms for in vitro expansion and differentiation of human pluripotent stem cells in bioreactor culture systems. J Biotechnol 2016; 234:71-82. [PMID: 27480342 DOI: 10.1016/j.jbiotec.2016.07.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 01/15/2023]
Abstract
Human pluripotent stem cells (hPSC) have attracted a great attention as an unlimited source of cells for cell therapies and other in vitro biomedical applications such as drug screening, toxicology assays and disease modeling. The implementation of scalable culture platforms for the large-scale production of hPSC and their derivatives is mandatory to fulfill the requirement of obtaining large numbers of cells for these applications. Microcarrier technology has been emerging as an effective approach for the large scale ex vivo hPSC expansion and differentiation. This review presents recent achievements in hPSC microcarrier-based culture systems and discusses the crucial aspects that influence the performance of these culture platforms. Recent progress includes addressing chemically-defined culture conditions for manufacturing of hPSC and their derivatives, with the development of xeno-free media and microcarrier coatings to meet good manufacturing practice (GMP) quality requirements. Finally, examples of integrated platforms including hPSC expansion and directed differentiation to specific lineages are also presented in this review.
Collapse
|
14
|
Enhanced Reprogramming Efficiency and Kinetics of Induced Pluripotent Stem Cells Derived from Human Duchenne Muscular Dystrophy. PLOS CURRENTS 2015; 7. [PMID: 26579330 PMCID: PMC4638229 DOI: 10.1371/currents.md.a77c2f0516a8cb4809ffad5963342905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The generation of disease-specific induced pluripotent stem cells (iPSCs) holds a great promise for understanding disease mechanisms and for drug screening. Recently, patient-derived iPSCs, containing identical genetic anomalies of the patient, have offered a breakthrough approach to studying Duchenne muscular dystrophy (DMD), a fatal disease caused by the mutation in the dystrophin gene. However, development of scalable and high fidelity DMD-iPSCs is hampered by low reprogramming efficiency, the addition of expensive growth factors and slow kinetics of disease-specific fibroblasts. Here, we show an efficient generation of DMD-iPSCs on bFGF secreting human foreskin fibroblast feeders (I-HFF) by employing single polycistronic lentiviral vector for delivering of transcription factors to DMD patient-specific fibroblast cells. Using this method, DMD-iPSCs generated on I-HFF feeders displayed pluripotent characteristics and disease genotype with improved reprogramming efficiency and kinetics over to mouse feeders. Moreover, we were able to maintain disease-specific iPSCs without additional supplementation of bFGF on I-HFF feeders. Our findings offer improvements in the generation of DMD-iPSCs and will facilitate in understanding of pathological mechanisms and screening of safer drugs for clinical intervention. Key Words: Duchenne Muscular Dystrophy, Reprogramming, Induced pluripotent Stem Cells, Immortalized Human Feeder, Basic Fibroblast Growth Factor, Stem Cell Cassette
Collapse
|
15
|
Rao W, Huang H, Wang H, Zhao S, Dumbleton J, Zhao G, He X. Nanoparticle-mediated intracellular delivery enables cryopreservation of human adipose-derived stem cells using trehalose as the sole cryoprotectant. ACS APPLIED MATERIALS & INTERFACES 2015; 7:5017-28. [PMID: 25679454 PMCID: PMC4734639 DOI: 10.1021/acsami.5b00655] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
In this study, pH responsive genipin-cross-linked Pluronic F127-chitosan nanoparticles (GNPs) was synthesized to encapsulate trehalose for intracellular delivery to cryopreserve primary human adipose-derived stem cells (hADSCs). Trehalose is a disaccharide of glucose used by lower organisms to survive extreme cold in nature and has been used to cryopreserve various biomacromolecules. However, it does not enter mammalian cells because of its highly hydrophilic nature, and has only been used in combination with other cell-penetrating cryoprotectants (such as dimethyl sulfoxide, DMSO) to cryopreserve mammalian cells. Our data show that trehalose can be efficiently encapsulated in our GNPs for intracellular delivery, which enables cryopreservation of primary hADSCs using the nontoxic sugar as the sole cryoprotectant. This capability is important because the conventional approach of cryopreserving mammalian cells using highly toxic (at body temperature) cell-penetrating cryoprotectants requires multistep washing of the cryopreserved cells to remove the toxic cryoprotectant for further use, which is time-consuming and associated with significant cell loss (∼10% during each washing step). By contrast, the trehalose-cryopreserved cells can be used without washing, which should greatly facilitate the wide application of the burgeoning cell-based medicine.
Collapse
Affiliation(s)
- Wei Rao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Haishui Huang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Mechanical Engineering, The Ohio State University, Columbus, OH 43210, US
| | - Hai Wang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Shuting Zhao
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jenna Dumbleton
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Gang Zhao
- Centre for Biomedical Engineering, 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, US
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
16
|
Fitzpatrick LE, McDevitt TC. Cell-derived matrices for tissue engineering and regenerative medicine applications. Biomater Sci 2015; 3:12-24. [PMID: 25530850 PMCID: PMC4270054 DOI: 10.1039/c4bm00246f] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The development and application of decellularized extracellular matrices (ECM) has grown rapidly in the fields of cell biology, tissue engineering and regenerative medicine in recent years. Similar to decellularized tissues and whole organs, cell-derived matrices (CDMs) represent bioactive, biocompatible materials consisting of a complex assembly of fibrillar proteins, matrix macromolecules and associated growth factors that often recapitulate, at least to some extent, the composition and organization of native ECM microenvironments. The unique ability to engineer CDMs de novo based on cell source and culture methods makes them an attractive alternative to conventional allogeneic and xenogeneic tissue-derived matrices that are currently harvested from cadaveric sources, suffer from inherent heterogeneity, and have limited ability for customization. Although CDMs have been investigated for a number of biomedical applications, including adhesive cell culture substrates, synthetic scaffold coatings, and tissue engineered products, such as heart valves and vascular grafts, the state of the field is still at a relatively nascent stage of development. In this review, we provide an overview of the various applications of CDM and discuss successes to date, current limitations and future directions.
Collapse
Affiliation(s)
| | - Todd C. McDevitt
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Finosh GT, Jayabalan M. Hybrid amphiphilic bimodal hydrogels having mechanical and biological recognition characteristics for cardiac tissue engineering. RSC Adv 2015. [DOI: 10.1039/c5ra04448k] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Tissue engineering strategies rely on the favourable microniche scaffolds for 3D cell growth.
Collapse
Affiliation(s)
- G. T. Finosh
- Sree Chitra Tirunal Institute for Medical Sciences and Technology
- Polymer Science Division
- Thiruvananthapuram-695 012
- India
| | - M. Jayabalan
- Sree Chitra Tirunal Institute for Medical Sciences and Technology
- Polymer Science Division
- Thiruvananthapuram-695 012
- India
| |
Collapse
|
18
|
Pettinato G, Vanden Berg-Foels WS, Zhang N, Wen X. ROCK inhibitor is not required for embryoid body formation from singularized human embryonic stem cells. PLoS One 2014; 9:e100742. [PMID: 25365581 PMCID: PMC4217711 DOI: 10.1371/journal.pone.0100742] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/30/2014] [Indexed: 12/18/2022] Open
Abstract
We report a technology to form human embryoid bodies (hEBs) from singularized human embryonic stem cells (hESCs) without the use of the p160 rho-associated coiled-coil kinase inhibitor (ROCKi) or centrifugation (spin). hEB formation was tested under four conditions: +ROCKi/+spin, +ROCKi/-spin, -ROCKi/+spin, and -ROCKi/-spin. Cell suspensions of BG01V/hOG and H9 hESC lines were pipetted into non-adherent hydrogel substrates containing defined microwell arrays. hEBs of consistent size and spherical geometry can be formed in each of the four conditions, including the -ROCKi/-spin condition. The hEBs formed under the -ROCKi/-spin condition differentiated to develop the three embryonic germ layers and tissues derived from each of the germ layers. This simplified hEB production technique offers homogeneity in hEB size and shape to support synchronous differentiation, elimination of the ROCKi xeno-factor and rate-limiting centrifugation treatment, and low-cost scalability, which will directly support automated, large-scale production of hEBs and hESC-derived cells needed for clinical, research, or therapeutic applications.
Collapse
Affiliation(s)
- Giuseppe Pettinato
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Wendy S. Vanden Berg-Foels
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
- Department of Craniofacial Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Ning Zhang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
- * E-mail: (NZ); (XW)
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
- Department of Craniofacial Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Institute for Biomedical Engineering and Nano Science (iNANO), Shanghai East Hospital, Tongji Medical School, Tongji University, Shanghai, People's Republic of China
- * E-mail: (NZ); (XW)
| |
Collapse
|
19
|
Lei Y, Jeong D, Xiao J, Schaffer DV. Developing Defined and Scalable 3D Culture Systems for Culturing Human Pluripotent Stem Cells at High Densities. Cell Mol Bioeng 2014; 7:172-183. [PMID: 25419247 DOI: 10.1007/s12195-014-0333-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) - including embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) - are very promising candidates for cell therapies, tissue engineering, high throughput pharmacology screens, and toxicity testing. These applications require large numbers of high quality cells; however, scalable production of human pluripotent stem cells and their derivatives at a high density and under well-defined conditions has been a challenge. We recently reported a simple, efficient, fully defined, scalable, and good manufacturing practice (GMP) compatible 3D culture system based on a thermoreversible hydrogel for hPSC expansion and differentiation. Here, we describe additional design rationale and characterization of this system. For instance, we have determined that culturing hPSCs as a suspension in a liquid medium can exhibit lower volumetric yields due to cell agglomeration and possible shear force-induced cell loss. By contrast, using hydrogels as 3D scaffolds for culturing hPSCs reduces aggregation and may insulate from shear forces. Additionally, hydrogel-based 3D culture systems can support efficient hPSC expansion and differentiation at a high density if compatible with hPSC biology. Finally, there are considerable opportunities for future development to further enhance hydrogel-based 3D culture systems for producing hPSCs and their progeny.
Collapse
Affiliation(s)
- Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Daeun Jeong
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Jifang Xiao
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA ; Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA ; California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, California, 94720, USA ; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
20
|
A novel perfused rotary bioreactor for cardiomyogenesis of embryonic stem cells. Biotechnol Lett 2014; 36:947-60. [PMID: 24652542 DOI: 10.1007/s10529-014-1456-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
Developments in bioprocessing technology play an important role for overcoming challenges in cardiac tissue engineering. To this end, our laboratory has developed a novel rotary perfused bioreactor for supporting three-dimensional cardiac tissue engineering. The dynamic culture environments provided by our novel perfused rotary bioreactor and/or the high-aspect rotating vessel produced constructs with higher viability and significantly higher cell numbers (up to 4 × 10(5) cells/bead) than static tissue culture flasks. Furthermore, cells in the perfused rotary bioreactor showed earlier gene expressions of cardiac troponin-T, α- and β-myosin heavy chains with higher percentages of cardiac troponin-I-positive cells and better uniformity of sacromeric α-actinin expression. A dynamic and perfused environment, as provided by this bioreactor, provides a superior culture performance in cardiac differentiation for embryonic stem cells particularly for larger 3D constructs.
Collapse
|
21
|
|
22
|
A fully defined and scalable 3D culture system for human pluripotent stem cell expansion and differentiation. Proc Natl Acad Sci U S A 2013; 110:E5039-48. [PMID: 24248365 DOI: 10.1073/pnas.1309408110] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, are promising for numerous biomedical applications, such as cell replacement therapies, tissue and whole-organ engineering, and high-throughput pharmacology and toxicology screening. Each of these applications requires large numbers of cells of high quality; however, the scalable expansion and differentiation of hPSCs, especially for clinical utilization, remains a challenge. We report a simple, defined, efficient, scalable, and good manufacturing practice-compatible 3D culture system for hPSC expansion and differentiation. It employs a thermoresponsive hydrogel that combines easy manipulation and completely defined conditions, free of any human- or animal-derived factors, and entailing only recombinant protein factors. Under an optimized protocol, the 3D system enables long-term, serial expansion of multiple hPSCs lines with a high expansion rate (~20-fold per 5-d passage, for a 10(72)-fold expansion over 280 d), yield (~2.0 × 10(7) cells per mL of hydrogel), and purity (~95% Oct4+), even with single-cell inoculation, all of which offer considerable advantages relative to current approaches. Moreover, the system enabled 3D directed differentiation of hPSCs into multiple lineages, including dopaminergic neuron progenitors with a yield of ~8 × 10(7) dopaminergic progenitors per mL of hydrogel and ~80-fold expansion by the end of a 15-d derivation. This versatile system may be useful at numerous scales, from basic biological investigation to clinical development.
Collapse
|
23
|
Gibson TM, Gersbach CA. The role of single-cell analyses in understanding cell lineage commitment. Biotechnol J 2013; 8:397-407. [PMID: 23520130 DOI: 10.1002/biot.201200201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/08/2013] [Accepted: 02/26/2013] [Indexed: 12/18/2022]
Abstract
The study of cell lineage commitment is critical for improving our understanding of tissue development and regeneration, and for realizing stem cell-based therapies and engineered tissue replacements. Recently, the discovery of an unanticipated degree of variability in fundamental biological processes, including divergent responses of genetically identical cells to various stimuli, has provided mechanistic insight into cellular decision making and the collective behavior of cell populations. Therefore, the study of lineage commitment with single-cell resolution could provide greater knowledge of cellular differentiation mechanisms and the influence of noise on cellular processes. This will require the adoption of new technologies for single-cell analysis as traditional methods typically measure average values of bulk population behavior. This review discusses the recent developments in methods for analyzing the behavior of individual cells, and how these approaches are leading to a deeper understanding and better control of cellular decision making.
Collapse
Affiliation(s)
- Tyler M Gibson
- Department of Biomedical Engineering, Duke University, Durham, NC 27708-0281, USA
| | | |
Collapse
|
24
|
Mooney BM, Raof NA, Li Y, Xie Y. Convergent mechanisms in pluripotent stem cells and cancer: Implications for stem cell engineering. Biotechnol J 2013; 8:408-19. [DOI: 10.1002/biot.201200202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/03/2012] [Accepted: 01/02/2013] [Indexed: 12/24/2022]
|
25
|
Ratcliffe E, Glen KE, Naing MW, Williams DJ. Current status and perspectives on stem cell-based therapies undergoing clinical trials for regenerative medicine: case studies. Br Med Bull 2013; 108:73-94. [PMID: 24200742 DOI: 10.1093/bmb/ldt034] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Apart from haematopoietic stem cell transplantation for haematological disorders many stem cell-based therapies are experimental. However, with only 12 years between human embryonic stem cell isolation and the first clinical trial, development of stem cell products for regenerative medicine has been rapid and numerous clinical trials have begun to investigate their therapeutic potential. SOURCE OF DATA This review summarizes key clinical trial data, current and future perspectives on stem cell-based products undergoing clinical trials, based on literature search and author research. AREAS OF AGREEMENT It is widely recognized that the ability to stimulate stem cell differentiation into specialized cells for use as cellular therapies will revolutionize health care and offer major hope for numerous diseases for which there are limited or no therapeutic options. AREAS OF CONTROVERSY Stem cell-based products are unique and cover a large range of disorders to be treated; therefore, there is significant potential for variation in cell source, type, processing manipulation, the bioprocessing approach and scalability, the cost and purity of manufacture, final product quality and mode of action. As such there are gaps in regulatory and manufacturing frameworks and technologies, only a small number of products are currently within late phase clinical trials and few products have achieved commercialization. GROWING POINTS Recent developments are encouraging acceleration through the difficulties encountered en route to clinical trials and commercialization of stem cell therapies. AREAS TIMELY FOR DEVELOPING RESEARCH The field is growing year on year with the first clinical trial using induced pluripotent stem cells anticipated by end 2013.
Collapse
Affiliation(s)
- Elizabeth Ratcliffe
- Healthcare Engineering Research Group, Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire LE11 3TU, UK
| | | | | | | |
Collapse
|
26
|
Jeon K, Oh HJ, Lim H, Kim JH, Lee DH, Lee ER, Park BH, Cho SG. Self-renewal of embryonic stem cells through culture on nanopattern polydimethylsiloxane substrate. Biomaterials 2012; 33:5206-20. [DOI: 10.1016/j.biomaterials.2012.04.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 04/01/2012] [Indexed: 10/28/2022]
|
27
|
Carlson AL, Florek CA, Kim JJ, Neubauer T, Moore JC, Cohen RI, Kohn J, Grumet M, Moghe PV. Microfibrous substrate geometry as a critical trigger for organization, self-renewal, and differentiation of human embryonic stem cells within synthetic 3-dimensional microenvironments. FASEB J 2012; 26:3240-51. [PMID: 22542683 DOI: 10.1096/fj.11-192732] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Substrates used to culture human embryonic stem cells (hESCs) are typically 2-dimensional (2-D) in nature, with limited ability to recapitulate in vivo-like 3-dimensional (3-D) microenvironments. We examined critical determinants of hESC self-renewal in poly-d-lysine-pretreated synthetic polymer-based substrates with variable microgeometries, including planar 2-D films, macroporous 3-D sponges, and microfibrous 3-D fiber mats. Completely synthetic 2-D substrates and 3-D macroporous scaffolds failed to retain hESCs or support self-renewal or differentiation. However, synthetic microfibrous geometries made from electrospun polymer fibers were found to promote cell adhesion, viability, proliferation, self-renewal, and directed differentiation of hESCs in the absence of any exogenous matrix proteins. Mechanistic studies of hESC adhesion within microfibrous scaffolds indicated that enhanced cell confinement in such geometries increased cell-cell contacts and altered colony organization. Moreover, the microfibrous scaffolds also induced hESCs to deposit and organize extracellular matrix proteins like laminin such that the distribution of laminin was more closely associated with the cells than the Matrigel treatment, where the laminin remained associated with the coated fibers. The production of and binding to laminin was critical for formation of viable hESC colonies on synthetic fibrous scaffolds. Thus, synthetic substrates with specific 3-D microgeometries can support hESC colony formation, self-renewal, and directed differentiation to multiple lineages while obviating the stringent needs for complex, exogenous matrices. Similar scaffolds could serve as tools for developmental biology studies in 3-D and for stem cell differentiation in situ and transplantation using defined humanized conditions.
Collapse
Affiliation(s)
- Aaron L Carlson
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Volz KS, Miljan E, Khoo A, Cooke JP. Development of pluripotent stem cells for vascular therapy. Vascul Pharmacol 2012; 56:288-96. [PMID: 22387745 DOI: 10.1016/j.vph.2012.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/02/2012] [Accepted: 02/18/2012] [Indexed: 12/21/2022]
Abstract
Peripheral arterial disease (PAD) is characterized by reduced limb blood flow due to arterial obstruction. Current treatment includes surgical or endovascular procedures, the failure of which may result in amputation of the affected limb. An emerging therapeutic approach is cell therapy to enhance angiogenesis and tissue survival. Small clinical trials of adult progenitor cell therapies have generated promising results, although large randomized clinical trials using well-defined cells have not been performed. Intriguing pre-clinical studies have been performed using vascular cells derived from human embryonic stem cells (hESC) or human induced pluripotent stem cells (hiPSCs). In particular, hiPSC-derived vascular cells may be a superior approach for vascular regeneration. The regulatory roadmap to the clinic will be arduous, but achievable with further understanding of the reprogramming and differentiation processes; with meticulous attention to quality control; and perseverance.
Collapse
Affiliation(s)
- Katharina S Volz
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, United States
| | | | | | | |
Collapse
|
29
|
Valamehr B, Tsutsui H, Ho CM, Wu H. Developing defined culture systems for human pluripotent stem cells. Regen Med 2012; 6:623-34. [PMID: 21916597 DOI: 10.2217/rme.11.54] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human pluripotent stem cells hold promising potential in many therapeutics applications including regenerative medicine and drug discovery. Over the past three decades, embryonic stem cell research has illustrated that embryonic stem cells possess two important and distinct properties: the ability to continuously self-renew and the ability to differentiate into all specialized cell types. In this article, we will discuss the continuing evolution of human pluripotent stem cell culture by examining requirements needed for the maintenance of self-renewal in vitro. We will also elaborate on the future direction of the field toward generating a robust and completely defined culture system, which has brought forth collaborations amongst biologists and engineers. As human pluripotent stem cell research progresses towards identifying solutions for debilitating diseases, it will be critical to establish a defined, reproducible and scalable culture system to meet the requirements of these clinical applications.
Collapse
Affiliation(s)
- Bahram Valamehr
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles, CA, USA
| | | | | | | |
Collapse
|
30
|
Teo A, Mantalaris A, Lim M. Hydrodynamics and bioprocess considerations in designing bioreactors for cardiac tissue engineering. ACTA ACUST UNITED AC 2012. [DOI: 10.7243/2050-1218-1-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
31
|
Want AJ, Nienow AW, Hewitt CJ, Coopman K. Large-scale expansion and exploitation of pluripotent stem cells for regenerative medicine purposes: beyond the T flask. Regen Med 2012; 7:71-84. [DOI: 10.2217/rme.11.101] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Human pluripotent stem cells will likely be a significant part of the regenerative medicine-driven healthcare revolution. In order to realize this potential, culture processes must be standardized, scalable and able to produce clinically relevant cell numbers, whilst maintaining critical biological functionality. This review comprises a broad overview of important bioprocess considerations, referencing the development of biopharmaceutical processes in an effort to learn from current best practice in the field. Particular focus is given to the recent efforts to grow human pluripotent stem cells in microcarrier or aggregate suspension culture, which would allow geometric expansion of productive capacity were it to be fully realized. The potential of these approaches is compared with automation of traditional T-flask culture, which may provide a cost-effective platform for low-dose, low-incidence conditions or autologous therapies. This represents the first step in defining the full extent of the challenges facing bioprocess engineers in the exploitation of large-scale human pluripotent stem cell manufacture.
Collapse
Affiliation(s)
- Andrew J Want
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | - Alvin W Nienow
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
- Centre for Bioprocess Engineering, Department of Chemical Engineering, University of Birmingham, B15 2TT, UK
| | - Christopher J Hewitt
- Centre for Biological Engineering, Department of Chemical Engineering, Loughborough University, Leicestershire, LE11 3TU, UK
| | | |
Collapse
|
32
|
Azarin SM, Lian X, Larson EA, Popelka HM, de Pablo JJ, Palecek SP. Modulation of Wnt/β-catenin signaling in human embryonic stem cells using a 3-D microwell array. Biomaterials 2011; 33:2041-9. [PMID: 22177620 DOI: 10.1016/j.biomaterials.2011.11.070] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 11/25/2011] [Indexed: 12/27/2022]
Abstract
Intercellular interactions in the cell microenvironment play a critical role in determining cell fate, but the effects of these interactions on pathways governing human embryonic stem cell (hESC) behavior have not been fully elucidated. We and others have previously reported that 3-D culture of hESCs affects cell fates, including self-renewal and differentiation to a variety of lineages. Here we have used a microwell culture system that produces 3-D colonies of uniform size and shape to provide insight into the effect of modulating cell-cell contact on canonical Wnt/β-catenin signaling in hESCs. Canonical Wnt signaling has been implicated in both self-renewal and differentiation of hESCs, and competition for β-catenin between the Wnt pathway and cadherin-mediated cell-cell interactions impacts various developmental processes, including the epithelial-mesenchymal transition. Our results showed that hESCs cultured in 3-D microwells exhibited higher E-cadherin expression than cells on 2-D substrates. The increase in E-cadherin expression in microwells was accompanied by a downregulation of Wnt signaling, as evidenced by the lack of nuclear β-catenin and downregulation of Wnt target genes. Despite this reduction in Wnt signaling in microwell cultures, embryoid bodies (EBs) formed from hESCs cultured in microwells exhibited higher levels of Wnt signaling than EBs from hESCs cultured on 2-D substrates. Furthermore, the Wnt-positive cells within EBs showed upregulation of genes associated with cardiogenesis. These results demonstrate that modulation of intercellular interactions impacts Wnt/β-catenin signaling in hESCs.
Collapse
Affiliation(s)
- Samira M Azarin
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
33
|
Rodrigues CAV, Fernandes TG, Diogo MM, da Silva CL, Cabral JMS. Stem cell cultivation in bioreactors. Biotechnol Adv 2011; 29:815-29. [PMID: 21726624 DOI: 10.1016/j.biotechadv.2011.06.009] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 06/11/2011] [Accepted: 06/12/2011] [Indexed: 12/22/2022]
Abstract
Cell-based therapies have generated great interest in the scientific and medical communities, and stem cells in particular are very appealing for regenerative medicine, drug screening and other biomedical applications. These unspecialized cells have unlimited self-renewal capacity and the remarkable ability to produce mature cells with specialized functions, such as blood cells, nerve cells or cardiac muscle. However, the actual number of cells that can be obtained from available donors is very low. One possible solution for the generation of relevant numbers of cells for several applications is to scale-up the culture of these cells in vitro. This review describes recent developments in the cultivation of stem cells in bioreactors, particularly considerations regarding critical culture parameters, possible bioreactor configurations, and integration of novel technologies in the bioprocess development stage. We expect that this review will provide updated and detailed information focusing on the systematic production of stem cell products in compliance with regulatory guidelines, while using robust and cost-effective approaches.
Collapse
Affiliation(s)
- Carlos A V Rodrigues
- Department of Bioengineering and Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Technical University of Lisbon, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | | | | | | | | |
Collapse
|
34
|
Kumar A, Pati NT, Sarin SK. Use of stem cells for liver diseases-current scenario. J Clin Exp Hepatol 2011; 1:17-26. [PMID: 25755306 PMCID: PMC3940313 DOI: 10.1016/s0973-6883(11)60114-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/04/2011] [Indexed: 12/12/2022] Open
Abstract
End-stage liver disease and liver failure are major health problems worldwide leading to high mortality and morbidity and high healthcare costs. Currently, orthotropic liver transplantation is the only effective treatment available to the patients of end-stage liver disease. However, a serious shortage of liver donors, high cost, and risk of organ rejection are the major obstacles to liver transplantation. Because of the ability of stem cells for differentiation into any tissue type, they have huge potential in therapy of various end-stage or degenerative diseases and traumatic injuries. Stem cell therapy has the potential to provide a valuable adjunct and alternative to liver transplantation and has immense potential in the management of end stage liver disease and liver failure. Stem cell therapy can be mediated by either a direct contribution to the functional hepatocyte population with embryonic, induced pluripotent, or adult stem cells or by promotion of endogenous regenerative processes with bone marrow-derived stem cells. Initial translational studies have been encouraging and have suggested improved liver function in advanced chronic liver disease and enhanced liver regeneration after portal vein embolization and partial hepatic resection. Stem cells infusion in cirrhotic patients has improved liver parameters and could form a viable bridge to transplantation. The present review summarizes basic of stem cell biology relevant to clinicians and an update on recent advances on the management of liver diseases using stem cells.
Collapse
Key Words
- AFP, alpha (α)-fetoprotein
- BM, bone marrow
- EPCAM, epithelial cell adhesion molecule
- ES, embryonic stem
- FSCs, fetal stem cells
- HPC, hepatic progenitor cells
- HSC, hematopoietic stem cells
- Hepatocyte transplantation
- ICAM, intercellular adhesion molecule
- MSCs, mesenchymal stem cells
- NCAM, neural cell adhesion molecule
- UCB, umbilical cord blood
- hAECs, human amniotic epithelial cells
- iPSCs, induced pluripotent stem cells
- liver transplantation
- stem cell
Collapse
Affiliation(s)
- Ashish Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- Address for correspondence: Dr Ashish Kumar MD DM, Associate Professor, Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), D-1, Vasant Kunj, New Delhi-110070, India
| | - Nirupama Trehan Pati
- Department of Research, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
35
|
Szebényi K, Erdei Z, Péntek A, Sebe A, Orbán TI, Sarkadi B, Apáti Á. Human pluripotent stem cells in pharmacological and toxicological screening: new perspectives for personalized medicine. Per Med 2011; 8:347-364. [DOI: 10.2217/pme.11.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human stem cells provide an important novel tool for generating in vitro pharmacological and toxicological test systems. In the development of new targeted therapies, as well as in critical safety issues, including hepato-, neuro- and cardio-toxicity, animal-based tests are mostly unsatisfactory, whereas the use of in vitro model systems is limited by the unavailability of relevant human tissues. Human embryonic stem cell lines may fill this gap and offer an advantage over primary cultures as well as tissue-derived (adult) stem cells. Human embryonic stem cells represent an unlimited source for the production of differentiated somatic progenies and allow various stable genetic manipulations. As a new opening in personalized medicine test systems, the generation of induced pluripotent stem cell lines and their derivatives can provide patient- and disease-specific cellular assays for drug development and safety assessments. This article reviews promising human stem cell applications in pharmacological and toxicological screenings, focusing on the implications for personalized medicine.
Collapse
Affiliation(s)
- Kornélia Szebényi
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
| | - Zsuzsa Erdei
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
| | - Adrienn Péntek
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
| | - Attila Sebe
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
- Department of Biochemistry & Molecular Biology, Medical & Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Tamás I Orbán
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
| | - Balázs Sarkadi
- Membrane Research Group, Hungarian Academy of Sciences & National Blood Center, 1113 Budapest, Diószegi u. 64, Hungary
| | | |
Collapse
|
36
|
GROα regulates human embryonic stem cell self-renewal or adoption of a neuronal fate. Differentiation 2011; 81:222-32. [PMID: 21396766 DOI: 10.1016/j.diff.2011.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/05/2011] [Indexed: 11/21/2022]
Abstract
Previously we reported that feeders formed from human placental fibroblasts (hPFs) support derivation and long-term self-renewal of human embryonic stem cells (hESCs) under serum-free conditions. Here, we show, using antibody array and ELISA platforms, that hPFs secrete ∼6-fold higher amounts of the CXC-type chemokine, GROα, than IMR 90, a human lung fibroblast line, which does not support hESC growth. Furthermore, immunocytochemistry and immunoblot approaches revealed that hESCs express CXCR, a GROα receptor. We used this information to develop defined culture medium for feeder-free propagation of hESCs in an undifferentiated state. Cells passaged as small aggregates and maintained in the GROα-containing medium had a normal karyotype, expressed pluripotency markers, and exhibited apical-basal polarity, i.e., had the defining features of pluripotent hESCs. They also differentiated into the three primary (embryonic) germ layers and formed teratomas in immunocompromised mice. hESCs cultured as single cells in the GROα-containing medium also had a normal karyotype, but they downregulated markers of pluripotency, lost apical-basal polarity, and expressed markers that are indicative of the early stages of neuronal differentiation-βIII tubulin, vimentin, radial glial protein, and nestin. These data support our hypothesis that establishing and maintaining cell polarity is essential for the long-term propagation of hESCs in an undifferentiated state and that disruption of cell-cell contacts can trigger adoption of a neuronal fate.
Collapse
|
37
|
Ratcliffe E, Thomas RJ, Williams DJ. Current understanding and challenges in bioprocessing of stem cell-based therapies for regenerative medicine. Br Med Bull 2011; 100:137-55. [PMID: 21852279 DOI: 10.1093/bmb/ldr037] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND A novel manufacturing industry is emerging to translate unique cellular therapy bioprocesses to robust, scaled manufacturing production for successful clinical translation. SOURCE OF DATA This review summarizes key translational issues, and current and future perspectives to improve translation of cell-based therapy bioprocessing, based on literature search and author research. AREAS OF AGREEMENT It is widely recognized that cell-based therapies could revolutionize health care for a range of diseases, and that there are gaps in the overarching framework and technologies to generate clinical success. AREAS OF CONTROVERSY There is limited understanding of how to fulfil requirements as regulatory and manufacturing guidelines are incomplete and few have achieved commercialization. GROWING POINTS Recent developments are encouraging adoption of automation and quality engineering approaches for bioprocessing of cell-based therapies. AREAS TIMELY FOR DEVELOPING RESEARCH Include technology development to improve the cost and purity of manufacture and final product quality.
Collapse
Affiliation(s)
- Elizabeth Ratcliffe
- Centre for Biological Engineering, Wolfson School of Mechanical and Manufacturing Engineering, Loughborough University, Loughborough, LE11 3TU, UK.
| | | | | |
Collapse
|
38
|
Prabhakaran MP, Venugopal J, Ghasemi-Mobarakeh L, Kai D, Jin G, Ramakrishna S. Stem Cells and Nanostructures for Advanced Tissue Regeneration. BIOMEDICAL APPLICATIONS OF POLYMERIC NANOFIBERS 2011. [DOI: 10.1007/12_2011_113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
39
|
Rodríguez-Serrano F, Alvarez P, Caba O, Picón M, Marchal JA, Perán M, Prados J, Melguizo C, Rama AR, Boulaiz H, Aránega A. Promotion of human adipose-derived stem cell proliferation mediated by exogenous nucleosides. Cell Biol Int 2010; 34:917-924. [PMID: 20522021 DOI: 10.1042/cbi20100227] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Adult stem cells are becoming the best option for regenerative medicine because they have low tumourigenic potential and permit autologous transplantation, even without in vitro culture. Our objectives were to evaluate the effects of exogenous nucleosides on the proliferation of hASCs (human adipose-derived stem cells), with or without co-treatment with 5-aza (5-azacytidine), and to analyse the expression of lamin A/C during cardiomyocyte differentiation of these cells. We isolated hASCs from human lipoaspirates that were positive for mesenchymal stem cell markers. We found that 5-aza induces a dose-dependent inhibition of hASC proliferation [IC50 (inhibitory concentration 50): 5.37 microM], whereas exogenous nucleosides significantly promote the proliferation of hASCs and partially revert the antiproliferative effect of the drug. Multipotentiality of isolated hASCs was confirmed by adipogenic, osteogenic and cardiomyogenic induction. 5-Aza-induced cells expressed cardiac troponins I and T and myosin light chain 2, myocardial markers that were directly correlated with lamin A/C expression. Our results support the importance of the nucleoside supplementation of media to improve conditions for the expansion and maintenance of hASCs in culture. In addition, the quantification of lamin A/C expression appears to be a good marker for the characterization of cardiomyocyte differentiation of stem cells that has rarely been used.
Collapse
|
40
|
Smith JA, Ndoye AMN, Geary K, Lisanti MP, Igoucheva O, Daniel R. A role for the Werner syndrome protein in epigenetic inactivation of the pluripotency factor Oct4. Aging Cell 2010; 9:580-91. [PMID: 20477760 PMCID: PMC2910250 DOI: 10.1111/j.1474-9726.2010.00585.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Werner syndrome (WS) is an autosomal recessive disorder, the hallmarks of which are premature aging and early onset of neoplastic diseases (Orren, 2006; Bohr, 2008). The gene, whose mutation underlies the WS phenotype, is called WRN. The protein encoded by the WRN gene, WRNp, has DNA helicase activity (Gray et al., 1997; Orren, 2006; Bohr, 2008; Opresko, 2008). Extensive evidence suggests that WRNp plays a role in DNA replication and DNA repair (Chen et al., 2003; Hickson, 2003; Orren, 2006; Turaga et al., 2007; Bohr, 2008). However, WRNp function is not yet fully understood. In this study, we show that WRNp is involved in de novo DNA methylation of the promoter of the Oct4 gene, which encodes a crucial stem cell transcription factor. We demonstrate that WRNp localizes to the Oct4 promoter during retinoic acid-induced differentiation of human pluripotent cells and associates with the de novo methyltransferase Dnmt3b in the chromatin of differentiating pluripotent cells. Depletion of WRNp does not affect demethylation of lysine 4 of the histone H3 at the Oct4 promoter, nor methylation of lysine 9 of H3, but it blocks the recruitment of Dnmt3b to the promoter and results in the reduced methylation of CpG sites within the Oct4 promoter. The lack of DNA methylation was associated with continued, albeit greatly reduced, Oct4 expression in WRN-deficient, retinoic acid-treated cells, which resulted in attenuated differentiation. The presented results reveal a novel function of WRNp and demonstrate that WRNp controls a key step in pluripotent stem cell differentiation.
Collapse
Affiliation(s)
- Johanna A. Smith
- Division of Infectious Diseases - Center for Human Virology, and Jefferson Center for Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, U.S.A
| | - Abibatou M. N. Ndoye
- Division of Infectious Diseases - Center for Human Virology, and Jefferson Center for Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, U.S.A
| | - Kyla Geary
- Division of Infectious Diseases - Center for Human Virology, and Jefferson Center for Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, U.S.A
| | - Michael P. Lisanti
- Department of Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, U.S.A
| | - Olga Igoucheva
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, U.S.A
| | - René Daniel
- Division of Infectious Diseases - Center for Human Virology, and Jefferson Center for Stem Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, U.S.A
| |
Collapse
|
41
|
Fernandes AM, Meletti T, Guimarães R, Stelling MP, Marinho PAN, Valladão AS, Rehen SK. Worldwide Survey of Published Procedures to Culture Human Embryonic Stem Cells. Cell Transplant 2010; 19:509-23. [DOI: 10.3727/096368909x485067] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Since their derivation 11 years ago, human embryonic stem (hES) cells have become a powerful tool in both basic biomedical research and developmental biology. Their capacity for self-renewal and differentiation into any tissue type has also brought interest from fields such as cell therapy and drug screening. We conducted an extensive analysis of 750 papers (51% of the total published about hES cells between 1998 and 2008) to present a spectrum of hES cell research including culture protocols developed worldwide. This review may stimulate discussions about the importance of having unvarying methods to culture hES cells, in order to facilitate comparisons among data obtained by research groups elsewhere, especially concerning preclinical studies. Moreover, the description of the most widely used cell lines, reagents, and procedures adopted internationally will help newcomers on deciding the best strategies for starting their own studies. Finally, the results will contribute with the efforts of stem cell researchers on comparing the performance of different aspects related to hES cell culture methods.
Collapse
Affiliation(s)
- A. M. Fernandes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - T. Meletti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - R. Guimarães
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - M. P. Stelling
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - P. A. N. Marinho
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Engenharia Química/COPPE, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - A. S. Valladão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - S. K. Rehen
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Steiner D, Khaner H, Cohen M, Even-Ram S, Gil Y, Itsykson P, Turetsky T, Idelson M, Aizenman E, Ram R, Berman-Zaken Y, Reubinoff B. Derivation, propagation and controlled differentiation of human embryonic stem cells in suspension. Nat Biotechnol 2010; 28:361-4. [PMID: 20351691 DOI: 10.1038/nbt.1616] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 02/16/2010] [Indexed: 11/09/2022]
Abstract
Undifferentiated human embryonic stem cells (hESCs) are currently propagated on a relatively small scale as monolayer colonies. Culture of hESCs as floating aggregates is widely used for induction of differentiation into embryoid bodies. Here we show that hESC lines can be derived from floating inner cell masses in suspension culture conditions that do not involve feeder cells or microcarriers. This culture system supports prolonged propagation of the pluripotent stem cells as floating clusters without their differentiation into embryoid bodies. HESCs cultivated as aggregates in suspension maintain the expression of pluripotency markers and can differentiate into progeny of the three germ layers both in vitro and in vivo. We further show the controlled differentiation of hESC clusters in suspension into neural spheres. These results pave the way for large-scale expansion and controlled differentiation of hESCs in suspension, which would be valuable in basic and applied research.
Collapse
|
43
|
Abstract
The use of human pluripotent stem cells, including embryonic and induced pluripotent stem cells, in therapeutic applications will require the development of robust, scalable culture technologies for undifferentiated cells. Advances made in large-scale cultures of other mammalian cells will facilitate expansion of undifferentiated human embryonic stem cells (hESCs), but challenges specific to hESCs will also have to be addressed, including development of defined, humanized culture media and substrates, monitoring spontaneous differentiation and heterogeneity in the cultures, and maintaining karyotypic integrity in the cells. This review will describe our current understanding of environmental factors that regulate hESC self-renewal and efforts to provide these cues in various scalable bioreactor culture systems.
Collapse
Affiliation(s)
- Samira M. Azarin
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison, 1415 Engineering Drive, Madison, WI 53706
- WiCell Research Institute, P.O. Box 7365, Madison, WI 53707
| | - Sean P. Palecek
- To whom correspondence should be addressed. 1415 Engineering Drive Madison, WI 53706 Ph: 608-262-8931. Fax: 608-262-5434
| |
Collapse
|
44
|
Kung JWC, Currie IS, Forbes SJ, Ross JA. Liver development, regeneration, and carcinogenesis. J Biomed Biotechnol 2010; 2010:984248. [PMID: 20169172 PMCID: PMC2821627 DOI: 10.1155/2010/984248] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Accepted: 11/12/2009] [Indexed: 02/06/2023] Open
Abstract
The identification of putative liver stem cells has brought closer the previously separate fields of liver development, regeneration, and carcinogenesis. Significant overlaps in the regulation of these processes are now being described. For example, studies in embryonic liver development have already provided the basis for directed differentiation of human embryonic stem cells and induced pluripotent stem cells into hepatocyte-like cells. As a result, the understanding of the cell biology of proliferation and differentiation in the liver has been improved. This knowledge can be used to improve the function of hepatocyte-like cells for drug testing, bioartificial livers, and transplantation. In parallel, the mechanisms regulating cancer cell biology are now clearer, providing fertile soil for novel therapeutic approaches. Recognition of the relationships between development, regeneration, and carcinogenesis, and the increasing evidence for the role of stem cells in all of these areas, has sparked fresh enthusiasm in understanding the underlying molecular mechanisms and has led to new targeted therapies for liver cirrhosis and primary liver cancers.
Collapse
Affiliation(s)
- Janet W C Kung
- Tissue Injury and Repair Group, Medical Research Council Centre for Regenerative Medicine, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK.
| | | | | | | |
Collapse
|
45
|
Mason C, Dunnill P. Assessing the value of autologous and allogeneic cells for regenerative medicine. Regen Med 2010; 4:835-53. [PMID: 19903003 DOI: 10.2217/rme.09.64] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The advantages and disadvantages of autologous and allogeneic human cells for regenerative medicine are summarized. The comparison of relative advantages includes: ease and cost of treating large numbers of patients, the speed of availability of therapy and the differing complexity of the development pathways. The comparison of relative disadvantages deals with issues such as variability of source material, the risks of cell abnormality and of viral and prion contamination, and the sensitive issues surrounding use of embryo-derived cells. From the comparisons, several potentially decisive issues are drawn out, such as possible immune response and teratoma formation, the impact of patents and the virtues of hospital versus industry-centered development.
Collapse
Affiliation(s)
- Chris Mason
- Advanced Centre for Biochemical Engineering, University College London, London, UK.
| | | |
Collapse
|
46
|
Directed differentiation of hematopoietic precursors and functional osteoclasts from human ES and iPS cells. Blood 2010; 115:2769-76. [PMID: 20065292 DOI: 10.1182/blood-2009-07-234690] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The directed differentiation of human pluripotent stem cells offers the unique opportunity to generate a broad spectrum of human cell types and tissues for transplantation, drug discovery, and studying disease mechanisms. Here, we report the stepwise generation of bone-resorbing osteoclasts from human embryonic and induced pluripotent stem cells. Generation of a primitive streak-like population in embryoid bodies, followed by specification to hematopoiesis and myelopoiesis by vascular endothelial growth factor and hematopoietic cytokines in serum-free media, yielded a precursor population enriched for cells expressing the monocyte-macrophage lineage markers CD14, CD18, CD11b, and CD115. When plated in monolayer culture in the presence of macrophage colony-stimulating factor and receptor activator of nuclear factor-kappaB ligand (RANKL), these precursors formed large, multinucleated osteoclasts that expressed tartrate-resistant acid phosphatase and were capable of resorption. No tartrate-resistant acid phosphatase-positive multinucleated cells or resorption pits were observed in the absence of RANKL. Molecular analyses confirmed the expression of the osteoclast marker genes NFATc1, cathepsin K, and calcitonin receptor in a RANKL-dependent manner, and confocal microscopy demonstrated the coexpression of the alphavbeta3 integrin, cathepsin K and F-actin rings characteristic of active osteoclasts. Generating hematopoietic and osteoclast populations from human embryonic and induced pluripotent stem cells will be invaluable for understanding embryonic bone development and postnatal bone disease.
Collapse
|
47
|
Engineering integrin signaling for promoting embryonic stem cell self-renewal in a precisely defined niche. Biomaterials 2009; 31:1219-26. [PMID: 19926127 DOI: 10.1016/j.biomaterials.2009.10.054] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 10/25/2009] [Indexed: 11/22/2022]
Abstract
We present development and use of a 3D synthetic extracellular matrix (ECM) analog with integrin-specific adhesion ligands to characterize the microenvironmental influences in embryonic stem cell (ESC) self-renewal. Transcriptional analysis of 24 integrin subunits followed by confirmation at the translational and functional levels suggested that integrins alpha(5)beta(1), alpha(v)beta(5), alpha(6)beta(1) and alpha(9)beta(1) play important roles in maintenance of stemness in undifferentiated mouse ESCs. Using the well-defined matrix as a tool to activate integrins alpha(5)beta(1) plus alpha(v)beta(5), alpha(6)beta(1) and alpha(9)beta(1), individually and in combination, differential integrin activation was demonstrated to exert exquisite control over ESC fate decisions. Simultaneous ligation of these four integrin heterodimers promoted self-renewal, as evidence by prolonged SSEA-1, Oct4 and Nanog expression, and induced Akt1 kinase signaling along with translational regulation of other stemness-related genes. The biofunctional network we have designed based on this knowledge may be useful as a defined niche for regulating ESC pluripotency through selective cell-matrix interactions, and the method we present may be more generally useful for probing matrix interactions in stem cell self-renewal and differentiation.
Collapse
|
48
|
Wang X, Lin G, Martins-Taylor K, Zeng H, Xu RH. Inhibition of caspase-mediated anoikis is critical for basic fibroblast growth factor-sustained culture of human pluripotent stem cells. J Biol Chem 2009; 284:34054-64. [PMID: 19828453 DOI: 10.1074/jbc.m109.052290] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Apoptosis and proliferation are two dynamically and tightly regulated processes that together maintain the homeostasis of renewable tissues. Anoikis is a subtype of apoptosis induced by detachment of adherent cells from the extracellular matrix. By using the defined mTeSR1 medium and collecting freshly detached cells, we found here that human pluripotent stem (PS) cells including embryonic stem (ES) cells and induced pluripotent stem cells are subject to constant anoikis in culture, which is escalated in the absence of basic fibroblast growth factor (bFGF). Withdrawal of bFGF also promotes apoptosis and differentiation of the remaining adherent cells without affecting their cell cycle progression. Insulin-like growth factor 2 (IGF2) has previously been reported to act downstream of FGF signaling to support self-renewal of human ES cells. However, we found that IGF2 cannot substitute bFGF in the TeSR1-supported culture, although endogenous IGF signaling is required to sustain self-renewal of human ES cells. On the other hand, all of the bFGF withdrawal effects observed here can be markedly prevented by the caspase inhibitor z-VAD-FMK. We further demonstrated that the bFGF-repressed anoikis is dependent on activation of ERK and AKT and associated with inhibition of Bcl-2-interacting mediator of cell death and the caspase-ROCK1-myosin signaling. Anoikis is independent of pre-detachment apoptosis and differentiation of the cells. Because previous studies of human PS cells have been focused on attached cells, our findings revealed a neglected role of bFGF in sustaining self-renewal of human PS cells: preventing them from anoikis via inhibition of caspase activation.
Collapse
Affiliation(s)
- Xiaofang Wang
- University of Connecticut Stem Cell Institute, Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | | | | | |
Collapse
|
49
|
Lim SH, Mao HQ. Electrospun scaffolds for stem cell engineering. Adv Drug Deliv Rev 2009; 61:1084-96. [PMID: 19647024 DOI: 10.1016/j.addr.2009.07.011] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 07/16/2009] [Indexed: 11/24/2022]
Abstract
Stem cells interact with and respond to a myriad of signals emanating from their extracellular microenvironment. The ability to harness the regenerative potential of stem cells via a synthetic matrix has promising implications for regenerative medicine. Electrospun fibrous scaffolds can be prepared with high degree of control over their structure creating highly porous meshes of ultrafine fibers that resemble the extracellular matrix topography, and are amenable to various functional modifications targeted towards enhancing stem cell survival and proliferation, directing specific stem cell fates, or promoting tissue organization. The feasibility of using such a scaffold platform to present integrated topographical and biochemical signals that are essential to stem cell manipulation has been demonstrated. Future application of this versatile scaffold platform to human embryonic and induced pluripotent stem cells for functional tissue repair and regeneration will further expand its potential for regenerative therapies.
Collapse
|
50
|
Abstract
Stem cell fate is influenced by a number of factors and interactions that require robust control for safe and effective regeneration of functional tissue. Coordinated interactions with soluble factors, other cells, and extracellular matrices define a local biochemical and mechanical niche with complex and dynamic regulation that stem cells sense. Decellularized tissue matrices and synthetic polymer niches are being used in the clinic, and they are also beginning to clarify fundamental aspects of how stem cells contribute to homeostasis and repair, for example, at sites of fibrosis. Multifaceted technologies are increasingly required to produce and interrogate cells ex vivo, to build predictive models, and, ultimately, to enhance stem cell integration in vivo for therapeutic benefit.
Collapse
Affiliation(s)
- Dennis E Discher
- Biophysical Engineering and Nanobiopolymers Laboratory, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|