1
|
Sutyagina OI, Beilin AK, Vorotelyak EA, Vasiliev AV. Immortalization Reversibility in the Context of Cell Therapy Biosafety. Int J Mol Sci 2023; 24:7738. [PMID: 37175444 PMCID: PMC10178325 DOI: 10.3390/ijms24097738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Immortalization (genetically induced prevention of replicative senescence) is a promising approach to obtain cellular material for cell therapy or for bio-artificial organs aimed at overcoming the problem of donor material shortage. Immortalization is reversed before cells are used in vivo to allow cell differentiation into the mature phenotype and avoid tumorigenic effects of unlimited cell proliferation. However, there is no certainty that the process of de-immortalization is 100% effective and that it does not cause unwanted changes in the cell. In this review, we discuss various approaches to reversible immortalization, emphasizing their advantages and disadvantages in terms of biosafety. We describe the most promising approaches in improving the biosafety of reversibly immortalized cells: CRISPR/Cas9-mediated immortogene insertion, tamoxifen-mediated self-recombination, tools for selection of successfully immortalized cells, using a decellularized extracellular matrix, and ensuring post-transplant safety with the use of suicide genes. The last process may be used as an add-on for previously existing reversible immortalized cell lines.
Collapse
Affiliation(s)
- Oksana I. Sutyagina
- N.K. Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Laboratory of Cell Biology, Vavilov Str. 26, 119334 Moscow, Russia
| | | | | | | |
Collapse
|
2
|
Xu M, Chen G, Dong Y, Xiang S, Xue M, Liu Y, Song H, Song H, Wang Y. Stable expression of a truncated TLX variant drives differentiation of induced pluripotent stem cells into self-renewing neural stem cells for production of extracellular vesicles. Stem Cell Res Ther 2022; 13:436. [PMID: 36056423 PMCID: PMC9438273 DOI: 10.1186/s13287-022-03131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background Neural stem cells (NSCs)-derived extracellular vesicles (EVs) possess great potential in treating severe neurological and cerebrovascular diseases, as they carry the modulatory and regenerative ingredients of NSCs. Induced pluripotent stem cells (iPSCs)-derived NSCs culture represents a sustainable source of therapeutic EVs. However, there exist two major challenges in obtaining a scalable culture of NSCs for high-efficiency EVs production: (1) the heterogeneity of iPSC-derived NSCs culture impairs the production of high-quality EVs and (2) the intrinsic propensity of neuronal or astroglial differentiation of NSCs during prolonged culturing reduces the number of NSCs for preparing EVs. A NSCs strain that is amenable to stable self-renewal and proliferation is thus greatly needed for scalable and long-term culture. Methods Various constructs of the genes encoding the orphan nuclear receptor NR2E1 (TLX) were stably transfected in iPSCs, which were subsequently cultured in a variety of differentiation media for generation of iNSCsTLX. Transcriptomic and biomarker profile of iNSCsTLX were investigated. In particular, the positivity ratios of Sox2/Nestin and Musashi/Vimentin were used to gauge the homogeneity of the iNSCsTLX culture. The iNSCs expressing a truncated version of TLX (TLX-TP) was expanded for up to 45 passages, after which its neuronal differentiation potential and EV activity were evaluated. Results Stable expression of TLX-TP could confer the iPSCs with rapid and self-driven differentiation into NSCs through stable passaging up to 225 days. The long-term culture of NSCs maintained the highly homogenous expression of NSC-specific biomarkers and potential of neuronal differentiation. EVs harvested from the TLX-expressing NSCs cultures exhibited anti-inflammatory and neuroprotective activities. Conclusions iPSC-derived NSCs stably expressing TLX-TP is a promising cell line for scalable production of EVs, which should be further exploited for therapeutic development in neurological treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03131-4.
Collapse
Affiliation(s)
- Mingzhi Xu
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Gang Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Yanan Dong
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Shensi Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Miaomiao Xue
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Yongxue Liu
- Anti-Radiation Medical Laboratory, Beijing Institute of Radiation Medicine, Beijing, 100039, China
| | - Haijing Song
- Emergency Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, China.
| | - Haifeng Song
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Yi Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences(Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, 102206, China.
| |
Collapse
|
3
|
Eleftheriadou D, Berg M, Phillips JB, Shipley RJ. A combined experimental and computational framework to evaluate the behavior of therapeutic cells for peripheral nerve regeneration. Biotechnol Bioeng 2022; 119:1980-1996. [PMID: 35445744 PMCID: PMC9323509 DOI: 10.1002/bit.28105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 11/08/2022]
Abstract
Recent studies have explored the potential of tissue‐mimetic scaffolds in encouraging nerve regeneration. One of the major determinants of the regenerative success of cellular nerve repair constructs (NRCs) is the local microenvironment, particularly native low oxygen conditions which can affect implanted cell survival and functional performance. In vivo, cells reside in a range of environmental conditions due to the spatial gradients of nutrient concentrations that are established. Here we evaluate in vitro the differences in cellular behavior that such conditions induce, including key biological features such as oxygen metabolism, glucose consumption, cell death, and vascular endothelial growth factor secretion. Experimental measurements are used to devise and parameterize a mathematical model that describes the behavior of the cells. The proposed model effectively describes the interactions between cells and their microenvironment and could in the future be extended, allowing researchers to compare the behavior of different therapeutic cells. Such a combinatorial approach could be used to accelerate the clinical translation of NRCs by identifying which critical design features should be optimized when fabricating engineered nerve repair conduits.
Collapse
Affiliation(s)
- D Eleftheriadou
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| | - M Berg
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| | - J B Phillips
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX
| | - R J Shipley
- Centre for Nerve Engineering, University College London, London, WC1E 6B.,Department of Mechanical Engineering, University College London, London, WC1E 7JE
| |
Collapse
|
4
|
Kamiyama Y, Naritomi Y, Moriya Y, Yamamoto S, Kitahashi T, Maekawa T, Yahata M, Hanada T, Uchiyama A, Noumaru A, Koga Y, Higuchi T, Ito M, Komatsu H, Miyoshi S, Kimura S, Umeda N, Fujita E, Tanaka N, Sugita T, Takayama S, Kurogi A, Yasuda S, Sato Y. Biodistribution studies for cell therapy products: Current status and issues. Regen Ther 2021; 18:202-216. [PMID: 34307798 PMCID: PMC8282960 DOI: 10.1016/j.reth.2021.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Information on the biodistribution (BD) of cell therapy products (CTPs) is essential for prediction and assessment of their efficacy and toxicity profiles in non-clinical and clinical studies. To conduct BD studies, it is necessary to understand regulatory requirements, implementation status, and analytical methods. This review aimed at surveying international and Japanese trends concerning the BD study for CTPs and the following subjects were investigated, which were considered particularly important: 1) comparison of guidelines to understand the regulatory status of BD studies in a global setting; 2) case studies of the BD study using databases to understand its current status in cell therapy; 3) case studies on quantitative polymerase chain reaction (qPCR) used primarily in non-clinical BD studies for CTPs; and 4) survey of imaging methods used for non-clinical and clinical BD studies. The results in this review will be a useful resource for implementing BD studies.
Collapse
Affiliation(s)
- Yoshiteru Kamiyama
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yoichi Naritomi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yuu Moriya
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Tsukasa Kitahashi
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Toshihiko Maekawa
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Masahiro Yahata
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, Japan
| | - Takeshi Hanada
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo.Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, Japan
| | - Asako Uchiyama
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Kagoshima, Japan
| | - Akari Noumaru
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Yoshiyuki Koga
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Tomoaki Higuchi
- Non-clinical Development, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Masahiko Ito
- Tsukuba Research Institute, BoZo Research Center Inc., 8 Okubo, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Komatsu
- Science BD Department, CMIC Pharma Science Co., Ltd., 1-1-1 Shibaura, Minato-ku, Tokyo, Japan
| | - Sosuke Miyoshi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Sadaaki Kimura
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Nobuhiro Umeda
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Eriko Fujita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Naoko Tanaka
- Evaluation Center, Terumo Corporation, 1500 Inokuchi, Nakai-machi, Ashigarakami-gun, Kanagawa, Japan
| | - Taku Sugita
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Satoru Takayama
- Cell Therapy Technology, Healthcare R&D Center, Asahi Kasei Corporation, 2-1 Samejima, Fuji-Shi, Shizuoka, Japan
| | - Akihiko Kurogi
- Regenerative Medicine Research & Planning Division, ROHTO Pharmaceutical Co., Ltd., Osaka, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| |
Collapse
|
5
|
Gazze SA, Thomas SJ, Garcia-Parra J, James DW, Rees P, Marsh-Durban V, Corteling R, Gonzalez D, Conlan RS, Francis LW. High content, quantitative AFM analysis of the scalable biomechanical properties of extracellular vesicles. NANOSCALE 2021; 13:6129-6141. [PMID: 33729236 DOI: 10.1039/d0nr09235e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Extracellular vesicles (EVs) are studied extensively as natural biomolecular shuttles and for their diagnostic and therapeutic potential. This exponential rise in interest has highlighted the need for highly robust and reproducible approaches for EV characterisation. Here we optimise quantitative nanomechanical tools and demonstrate the advantages of EV population screening by atomic force microscopy (AFM). Our high-content informatics analytical tools are made available for use by the EV community for widespread, standardised determination of structural stability. Ultracentrifugation (UC) and sonication, the common mechanical techniques used for EV isolation and loading respectively, are used to demonstrate the utility of optimised PeakForce-Quantitative Nano Mechanics (PF-QNM) analysis. EVs produced at an industrial scale exhibited biochemical and biomechanical alterations after exposure to these common techniques. UC resulted in slight increases in physical dimensions, and decreased EV adhesion concurrent with a decrease in CD63 content. Sonicated EVs exhibited significantly reduced levels of CD81, a decrease in size, increased Young's modulus and decreased adhesive force. These biomechanical and biochemical changes highlight the effect of EV sample preparation techniques on critical properties linked to EV cellular uptake and biological function. PF-QNM offers significant additional information about the structural information of EVs following their purification and downstream processing, and the analytical tools will ensure consistency of analysis of AFM data by the EV community, as this technique continues to become more widely implemented.
Collapse
Affiliation(s)
- Salvatore Andrea Gazze
- Reproductive Biology and Gynaecological Oncology Group, Institute for Life Science 2, Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Luzzi S, Crovace AM, Del Maestro M, Giotta Lucifero A, Elbabaa SK, Cinque B, Palumbo P, Lombardi F, Cimini A, Cifone MG, Crovace A, Galzio R. The cell-based approach in neurosurgery: ongoing trends and future perspectives. Heliyon 2019; 5:e02818. [PMID: 31844735 PMCID: PMC6889232 DOI: 10.1016/j.heliyon.2019.e02818] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/11/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Examination of the current trends and future perspectives of the cell-based therapies in neurosurgery. METHODS A PubMed/MEDLINE-based systematic review has been performed combining the main Medical Subject Headings (MeSH) regarding the cell- and tissue-based therapies with the "Brain", "Spinal Cord", "Spine" and "Skull" MeSH terms. Only articles in English published in the last 10 years and pertinent to neurosurgery have been selected. RESULTS A total of 1,173 relevant articles have been chosen. Somatic cells and gene-modification technologies have undergone the greatest development. Immunotherapies and gene therapies have been tested for the cure of glioblastoma, stem cells mainly for brain and spinal cord traumatic injuries. Stem cells have also found a rationale in the treatment of the cranial and spinal bony defects, and of the intervertebral disc degeneration, as well.Most of the completed or ongoing trials concerning the cell-based therapies in neurosurgery are on phase 2. Future perspectives involve the need to overcome issues related to immunogenicity, oncogenicity and routes for administration. Refinement and improvement of vector design and delivery are required within the gene therapies. CONCLUSION The last decade has been characterised by a progressive evolution of neurosurgery from a purely mechanical phase to a new biological one. This trend has followed the rapid and parallel development of translational medicine and nanotechnologies.The introduction of new technologies, the optimisation of the already existing ones, and the reduction of costs are among the main challenges of the foreseeable future.
Collapse
Affiliation(s)
- Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| | - Alberto Maria Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Mattia Del Maestro
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
- PhD School in Experimental Medicine, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
| | - Samer K. Elbabaa
- Pediatric Neurosurgery, Pediatric Neuroscience Center of Excellence, Arnold Palmer Hospital for Children, 1222 S. Orange Avenue, 2nd Floor, MP 154, Orlando, FL, 32806, USA
| | - Benedetta Cinque
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Paola Palumbo
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Francesca Lombardi
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Annamaria Cimini
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Maria Grazia Cifone
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Building Delta 6, via Coppito, L'Aquila, 67100, Italy
| | - Antonio Crovace
- Department of Emergency and Organ Transplantation, University of Bari "Aldo Moro", Piazza G. Cesare, 11 – Policlinico di Bari, Bari, 70124, Italy
| | - Renato Galzio
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Polo Didattico "Cesare Brusotti", Viale Brambilla, 74, Pavia, 27100, Italy
- Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Viale C. Golgi, 19, Pavia, 27100, Italy
| |
Collapse
|
7
|
Santiago-Toledo G, Georgiou M, Dos Reis J, Roberton VH, Valinhas A, Wood RC, Phillips JB, Mason C, Li D, Li Y, Sinden JD, Choi D, Jat PS, Wall IB. Generation of c-MycER TAM-transduced human late-adherent olfactory mucosa cells for potential regenerative applications. Sci Rep 2019; 9:13190. [PMID: 31519924 PMCID: PMC6744411 DOI: 10.1038/s41598-019-49315-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 07/31/2019] [Indexed: 12/17/2022] Open
Abstract
Human olfactory mucosa cells (hOMCs) have been transplanted to the damaged spinal cord both pre-clinically and clinically. To date mainly autologous cells have been tested. However, inter-patient variability in cell recovery and quality, and the fact that the neuroprotective olfactory ensheathing cell (OEC) subset is difficult to isolate, means an allogeneic hOMC therapy would be an attractive "off-the-shelf" alternative. The aim of this study was to generate a candidate cell line from late-adherent hOMCs, thought to contain the OEC subset. Primary late-adherent hOMCs were transduced with a c-MycERTAM gene that enables cell proliferation in the presence of 4-hydroxytamoxifen (4-OHT). Two c-MycERTAM-derived polyclonal populations, PA5 and PA7, were generated and expanded. PA5 cells had a normal human karyotype (46, XY) and exhibited faster growth kinetics than PA7, and were therefore selected for further characterisation. PA5 hOMCs express glial markers (p75NTR, S100ß, GFAP and oligodendrocyte marker O4), neuronal markers (nestin and ß-III-tubulin) and fibroblast-associated markers (CD90/Thy1 and fibronectin). Co-culture of PA5 cells with a neuronal cell line (NG108-15) and with primary dorsal root ganglion (DRG) neurons resulted in significant neurite outgrowth after 5 days. Therefore, c-MycERTAM-derived PA5 hOMCs have potential as a regenerative therapy for neural cells.
Collapse
Affiliation(s)
| | - Melanie Georgiou
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
| | - Joana Dos Reis
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
| | - Victoria H Roberton
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
| | - Ana Valinhas
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
| | - Rachael C Wood
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
- Aston Medical Research Institute and School of Life & Health Sciences, Aston University, Birmingham, B4 7ET, UK
| | - James B Phillips
- Department of Pharmacology, UCL School of Pharmacy, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1E 6BT, UK
| | - Chris Mason
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK
- AVROBIO Inc, Cambridge, MA 02139, USA
| | - Daqing Li
- Department of Neurosurgery, National Hospital for Neurology & Neurosurgery, London, WC1N 3BG, UK
| | - Ying Li
- Department of Neurosurgery, National Hospital for Neurology & Neurosurgery, London, WC1N 3BG, UK
| | - John D Sinden
- UCL Centre for Nerve Engineering, London, WC1E 6BT, UK
- ReNeuron Limited, Pencoed, Bridgend, CF35 5HY, UK
| | - David Choi
- UCL Centre for Nerve Engineering, London, WC1E 6BT, UK
- Department of Neurosurgery, National Hospital for Neurology & Neurosurgery, London, WC1N 3BG, UK
| | - Parmjit S Jat
- MRC Prion Unit at UCL, Institute of Prion Diseases, London, W1W 7FF, UK
| | - Ivan B Wall
- Department of Biochemical Engineering, University College London, London, WC1H 0AH, UK.
- Aston Medical Research Institute and School of Life & Health Sciences, Aston University, Birmingham, B4 7ET, UK.
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
8
|
Dabrowski A, Robinson TJ, Felling RJ. Promoting Brain Repair and Regeneration After Stroke: a Plea for Cell-Based Therapies. Curr Neurol Neurosci Rep 2019; 19:5. [PMID: 30712068 DOI: 10.1007/s11910-019-0920-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW After decades of hype, cell-based therapies are emerging into the clinical arena for the purposes of promoting recovery after stroke. In this review, we discuss the most recent science behind the role of cell-based therapies in ischemic stroke and the efforts to translate these therapies into human clinical trials. RECENT FINDINGS Preclinical data support numerous beneficial effects of cell-based therapies in both small and large animal models of ischemic stroke. These benefits are driven by multifaceted mechanisms promoting brain repair through immunomodulation, trophic support, circuit reorganization, and cell replacement. Cell-based therapies offer tremendous potential for improving outcomes after stroke through multimodal support of brain repair. Based on recent clinical trials, cell-based therapies appear both feasible and safe in all phases of stroke. Ongoing translational research and clinical trials will further refine these therapies and have the potential to transform the approach to stroke recovery and rehabilitation.
Collapse
Affiliation(s)
- Ania Dabrowski
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Thomas J Robinson
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA
| | - Ryan J Felling
- Department of Neurology, Johns Hopkins School of Medicine, 200 N. Wolfe Street, Suite 2158, Baltimore, MD, 21287, USA.
| |
Collapse
|
9
|
Zhao LR, Willing A. Enhancing endogenous capacity to repair a stroke-damaged brain: An evolving field for stroke research. Prog Neurobiol 2018; 163-164:5-26. [PMID: 29476785 PMCID: PMC6075953 DOI: 10.1016/j.pneurobio.2018.01.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/11/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Stroke represents a severe medical condition that causes stroke survivors to suffer from long-term and even lifelong disability. Over the past several decades, a vast majority of stroke research targets neuroprotection in the acute phase, while little work has been done to enhance stroke recovery at the later stage. Through reviewing current understanding of brain plasticity, stroke pathology, and emerging preclinical and clinical restorative approaches, this review aims to provide new insights to advance the research field for stroke recovery. Lifelong brain plasticity offers the long-lasting possibility to repair a stroke-damaged brain. Stroke impairs the structural and functional integrity of entire brain networks; the restorative approaches containing multi-components have great potential to maximize stroke recovery by rebuilding and normalizing the stroke-disrupted entire brain networks and brain functioning. The restorative window for stroke recovery is much longer than previously thought. The optimal time for brain repair appears to be at later stage of stroke rather than the earlier stage. It is expected that these new insights will advance our understanding of stroke recovery and assist in developing the next generation of restorative approaches for enhancing brain repair after stroke.
Collapse
Affiliation(s)
- Li-Ru Zhao
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Alison Willing
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
10
|
Borlongan CV. Preliminary Reports of Stereotaxic Stem Cell Transplants in Chronic Stroke Patients. Mol Ther 2018; 24:1710-1711. [PMID: 27818493 DOI: 10.1038/mt.2016.186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Cesar V Borlongan
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, USA
| |
Collapse
|
11
|
Napoli E, Borlongan CV. Stem Cell Recipes of Bone Marrow and Fish: Just What the Stroke Doctors Ordered. Stem Cell Rev Rep 2017; 13:192-197. [PMID: 28064388 DOI: 10.1007/s12015-016-9716-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stem cell therapy for stroke has advanced from the laboratory to the clinic, but remains as an experimental treatment. Two lines of transplant regimens have emerged, namely the "early bird" peripheral injections in subacute stroke patients and the "late night" direct intracerebral treatments in chronic stroke patients. Autologous bone marrow-derived stem cells, which only required minimal manipulations during graft cell preparation, gained fast-track entry into the clinic, while gene modified stem cells necessitated overcoming more stringent regulatory criteria before they were approved for clinical use. Safety of the stem cell therapy can be declared from these clinical trials, but efficacy warrants further investigations. Here, we offer insights into the translation of cell therapy from the laboratory to the clinic, in the hopes that highlighting the lessons we learned from this experience will guide the optimization of functional outcomes of future clinical trials of stem cell therapy for stroke.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, University of California Davis, Davis, CA, 95616, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
12
|
Wei L, Wei ZZ, Jiang MQ, Mohamad O, Yu SP. Stem cell transplantation therapy for multifaceted therapeutic benefits after stroke. Prog Neurobiol 2017; 157:49-78. [PMID: 28322920 PMCID: PMC5603356 DOI: 10.1016/j.pneurobio.2017.03.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/30/2017] [Accepted: 03/05/2017] [Indexed: 02/06/2023]
Abstract
One of the exciting advances in modern medicine and life science is cell-based neurovascular regeneration of damaged brain tissues and repair of neuronal structures. The progress in stem cell biology and creation of adult induced pluripotent stem (iPS) cells has significantly improved basic and pre-clinical research in disease mechanisms and generated enthusiasm for potential applications in the treatment of central nervous system (CNS) diseases including stroke. Endogenous neural stem cells and cultured stem cells are capable of self-renewal and give rise to virtually all types of cells essential for the makeup of neuronal structures. Meanwhile, stem cells and neural progenitor cells are well-known for their potential for trophic support after transplantation into the ischemic brain. Thus, stem cell-based therapies provide an attractive future for protecting and repairing damaged brain tissues after injury and in various disease states. Moreover, basic research on naïve and differentiated stem cells including iPS cells has markedly improved our understanding of cellular and molecular mechanisms of neurological disorders, and provides a platform for the discovery of novel drug targets. The latest advances indicate that combinatorial approaches using cell based therapy with additional treatments such as protective reagents, preconditioning strategies and rehabilitation therapy can significantly improve therapeutic benefits. In this review, we will discuss the characteristics of cell therapy in different ischemic models and the application of stem cells and progenitor cells as regenerative medicine for the treatment of stroke.
Collapse
Affiliation(s)
- Ling Wei
- Laboratories of Stem Cell Biology and Regenerative Medicine, Department of Neurology, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zheng Z Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael Qize Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Osama Mohamad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shan Ping Yu
- Laboratories of Stem Cell Biology and Regenerative Medicine, Department of Neurology, Experimental Research Center and Neurological Disease Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
13
|
Sinden JD, Hicks C, Stroemer P, Vishnubhatla I, Corteling R. Human Neural Stem Cell Therapy for Chronic Ischemic Stroke: Charting Progress from Laboratory to Patients. Stem Cells Dev 2017; 26:933-947. [PMID: 28446071 PMCID: PMC5510676 DOI: 10.1089/scd.2017.0009] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic disability after stroke represents a major unmet neurologic need. ReNeuron's development of a human neural stem cell (hNSC) therapy for chronic disability after stroke is progressing through early clinical studies. A Phase I trial has recently been published, showing no safety concerns and some promising signs of efficacy. A single-arm Phase II multicenter trial in patients with stable upper-limb paresis has recently completed recruitment. The hNSCs administrated are from a manufactured, conditionally immortalized hNSC line (ReNeuron's CTX0E03 or CTX), generated with c-mycERTAM technology. This technology has enabled CTX to be manufactured at large scale under cGMP conditions, ensuring sufficient supply to meets the demands of research, clinical development, and, eventually, the market. CTX has key pro-angiogenic, pro-neurogenic, and immunomodulatory characteristics that are mechanistically important in functional recovery poststroke. This review covers the progress of CTX cell therapy from its laboratory origins to the clinic, concluding with a look into the late stage clinical future.
Collapse
|
14
|
Liska MG, Crowley MG, Borlongan CV. Regulated and Unregulated Clinical Trials of Stem Cell Therapies for Stroke. Transl Stroke Res 2017; 8:93-103. [PMID: 28127687 DOI: 10.1007/s12975-017-0522-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Michael G Liska
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Marci G Crowley
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
15
|
Kalladka D, Sinden J, Pollock K, Haig C, McLean J, Smith W, McConnachie A, Santosh C, Bath PM, Dunn L, Muir KW. Human neural stem cells in patients with chronic ischaemic stroke (PISCES): a phase 1, first-in-man study. Lancet 2016; 388:787-96. [PMID: 27497862 DOI: 10.1016/s0140-6736(16)30513-x] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND CTX0E03 is an immortalised human neural stem-cell line from which a drug product (CTX-DP) was developed for allogeneic therapy. Dose-dependent improvement in sensorimotor function in rats implanted with CTX-DP 4 weeks after middle cerebral artery occlusion stroke prompted investigation of the safety and tolerability of this treatment in stroke patients. METHODS We did an open-label, single-site, dose-escalation study. Men aged 60 years or older with stable disability (National Institutes of Health Stroke Scale [NIHSS] score ≥6 and modified Rankin Scale score 2-4) 6-60 months after ischaemic stroke were implanted with single doses of 2 million, 5 million, 10 million, or 20 million cells by stereotactic ipsilateral putamen injection. Clinical and brain imaging data were collected over 2 years. The primary endpoint was safety (adverse events and neurological change). This trial is registered with ClinicalTrials.gov, number NCT01151124. FINDINGS 13 men were recruited between September, 2010, and January, 2013, of whom 11 (mean age 69 years, range 60-82) received CTX-DP. Median NIHSS score before implantation was 7 (IQR 6-8) and the mean time from stroke was 29 (SD 14) months. Three men had subcortical infarcts only and seven had right-hemisphere infarcts. No immunological or cell-related adverse events were seen. Other adverse events were related to the procedure or comorbidities. Hyperintensity around the injection tracts on T2-weighted fluid-attenuation inversion recovery MRI was seen in five patients. At 2 years, improvement in NIHSS score ranged from 0 to 5 (median 2) points. INTERPRETATION Single intracerebral doses of CTX-DP up to 20 million cells induced no adverse events and were associated with improved neurological function. Our observations support further investigation of CTX-DP in stroke patients. FUNDING ReNeuron Limited.
Collapse
Affiliation(s)
- Dheeraj Kalladka
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - John Sinden
- ReNeuron Ltd, Surrey Research Park, Guildford, Surrey, UK
| | | | - Caroline Haig
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - John McLean
- Department of Neuroradiology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Wilma Smith
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Celestine Santosh
- Department of Neuroradiology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Laurence Dunn
- Neurosurgery, Institute of Neurological Sciences, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Keith W Muir
- Institute of Neuroscience and Psychology, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK.
| |
Collapse
|
16
|
Affiliation(s)
- Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
17
|
Semo M, Haamedi N, Stevanato L, Carter D, Brooke G, Young M, Coffey P, Sinden J, Patel S, Vugler A. Efficacy and Safety of Human Retinal Progenitor Cells. Transl Vis Sci Technol 2016; 5:6. [PMID: 27486556 PMCID: PMC4959814 DOI: 10.1167/tvst.5.4.6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/11/2016] [Indexed: 01/08/2023] Open
Abstract
PURPOSE We assessed the long-term efficacy and safety of human retinal progenitor cells (hRPC) using established rodent models. METHODS Efficacy of hRPC was tested initially in Royal College of Surgeons (RCS) dystrophic rats immunosuppressed with cyclosporine/dexamethasone. Due to adverse effects of dexamethasone, this drug was omitted from a subsequent dose-ranging study, where different hRPC doses were tested for their ability to preserve visual function (measured by optokinetic head tracking) and retinal structure in RCS rats at 3 to 6 months after grafting. Safety of hRPC was assessed by subretinal transplantation into wild type (WT) rats and NIH-III nude mice, with analysis at 3 to 6 and 9 months after grafting, respectively. RESULTS The optimal dose of hRPC for preserving visual function/retinal structure in dystrophic rats was 50,000 to 100,000 cells. Human retinal progenitor cells integrated/survived in dystrophic and WT rat retina up to 6 months after grafting and expressed nestin, vimentin, GFAP, and βIII tubulin. Vision and retinal structure remained normal in WT rats injected with hRPC and there was no evidence of tumors. A comparison between dexamethasone-treated and untreated dystrophic rats at 3 months after grafting revealed an unexpected reduction in the baseline visual acuity of dexamethasone-treated animals. CONCLUSIONS Human retinal progenitor cells appear safe and efficacious in the preclinical models used here. TRANSLATIONAL RELEVANCE Human retinal progenitor cells could be deployed during early stages of retinal degeneration or in regions of intact retina, without adverse effects on visual function. The ability of dexamethasone to reduce baseline visual acuity in RCS dystrophic rats has important implications for the interpretation of preclinical and clinical cell transplant studies.
Collapse
Affiliation(s)
- Ma'ayan Semo
- Department of Ocular Biology and Therapeutics, UCL-Institute of Ophthalmology, London, UK
| | - Nasrin Haamedi
- Department of Ocular Biology and Therapeutics, UCL-Institute of Ophthalmology, London, UK
| | | | - David Carter
- Department of Ocular Biology and Therapeutics, UCL-Institute of Ophthalmology, London, UK
| | | | - Michael Young
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, MA, USA
| | - Peter Coffey
- Department of Ocular Biology and Therapeutics, UCL-Institute of Ophthalmology, London, UK
| | | | | | - Anthony Vugler
- Department of Ocular Biology and Therapeutics, UCL-Institute of Ophthalmology, London, UK
| |
Collapse
|
18
|
Mount NM, Ward SJ, Kefalas P, Hyllner J. Cell-based therapy technology classifications and translational challenges. Philos Trans R Soc Lond B Biol Sci 2016; 370:20150017. [PMID: 26416686 PMCID: PMC4634004 DOI: 10.1098/rstb.2015.0017] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cell therapies offer the promise of treating and altering the course of diseases which cannot be addressed adequately by existing pharmaceuticals. Cell therapies are a diverse group across cell types and therapeutic indications and have been an active area of research for many years but are now strongly emerging through translation and towards successful commercial development and patient access. In this article, we present a description of a classification of cell therapies on the basis of their underlying technologies rather than the more commonly used classification by cell type because the regulatory path and manufacturing solutions are often similar within a technology area due to the nature of the methods used. We analyse the progress of new cell therapies towards clinical translation, examine how they are addressing the clinical, regulatory, manufacturing and reimbursement requirements, describe some of the remaining challenges and provide perspectives on how the field may progress for the future.
Collapse
Affiliation(s)
| | - Stephen J Ward
- Cell Therapy Catapult, Guy's Hospital, London SE1 9RT, UK
| | - Panos Kefalas
- Cell Therapy Catapult, Guy's Hospital, London SE1 9RT, UK
| | - Johan Hyllner
- Cell Therapy Catapult, Guy's Hospital, London SE1 9RT, UK Division of Biotechnology, IFM, Linköping University, Linköping 581 83, Sweden
| |
Collapse
|
19
|
Anderson GW, Deans PJM, Taylor RDT, Raval P, Chen D, Lowder H, Murkerji S, Andreae LC, Williams BP, Srivastava DP. Characterisation of neurons derived from a cortical human neural stem cell line CTX0E16. Stem Cell Res Ther 2015; 6:149. [PMID: 26296747 PMCID: PMC4546258 DOI: 10.1186/s13287-015-0136-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/29/2015] [Accepted: 07/17/2015] [Indexed: 01/11/2023] Open
Abstract
Introduction Conditionally immortalised human neural progenitor cells (hNPCs) represent a robust source of native neural cells to investigate physiological mechanisms in both health and disease. However, in order to recognise the utility of such cells, it is critical to determine whether they retain characteristics of their tissue of origin and generate appropriate neural cell types upon differentiation. To this end, we have characterised the conditionally immortalised, cortically-derived, human NPC line, CTX0E16, investigating the molecular and cellular phenotype of differentiated neurons to determine whether they possess characteristics of cortical glutamatergic neurons. Methods Differentiated CTX0E16 cells were characterised by assessing expression of several neural fates markers, and examination of developing neuronal morphology. Expression of neurotransmitter receptors, signalling proteins and related proteins were assessed by q- and RT-PCR and complemented by Ca2+ imaging, electrophysiology and assessment of ERK signalling in response to neurotransmitter ligand application. Finally, differentiated neurons were assessed for their ability to form putative synapses and to respond to activity-dependent stimulation. Results Differentiation of CTX0E16 hNPCs predominately resulted in the generation of neurons expressing markers of cortical and glutamatergic (excitatory) fate, and with a typical polarized neuronal morphology. Gene expression analysis confirmed an upregulation in the expression of cortical, glutamatergic and signalling proteins following differentiation. CTX0E16 neurons demonstrated Ca2+ and ERK1/2 responses following exogenous neurotransmitter application, and after 6 weeks displayed spontaneous Ca2+ transients and electrophysiological properties consistent with that of immature neurons. Differentiated CTX0E16 neurons also expressed a range of pre- and post-synaptic proteins that co-localized along distal dendrites, and moreover, displayed structural plasticity in response to modulation of neuronal activity. Conclusions Taken together, these findings demonstrate that the CTX0E16 hNPC line is a robust source of cortical neurons, which display functional properties consistent with a glutamatergic phenotype. Thus CTX0E16 neurons can be used to study cortical cell function, and furthermore, as these neurons express a range of disease-associated genes, they represent an ideal platform with which to investigate neurodevelopmental mechanisms in native human cells in health and disease. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0136-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Greg W Anderson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - P J Michael Deans
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Ruth D T Taylor
- MRC Centre for Developmental Neurobiology, King's College London, London, SE5 8AF, UK.
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Ding Chen
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Harrison Lowder
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Srishti Murkerji
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Laura C Andreae
- MRC Centre for Developmental Neurobiology, King's College London, London, SE5 8AF, UK.
| | - Brenda P Williams
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, SE5 8AF, UK.
| |
Collapse
|
20
|
Kalladka D, Muir KW. Brain repair: cell therapy in stroke. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2014; 7:31-44. [PMID: 24627643 PMCID: PMC3937183 DOI: 10.2147/sccaa.s38003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stroke affects one in every six people worldwide, and is the leading cause of adult disability. Some spontaneous recovery is usual but of limited extent, and the mechanisms of late recovery are not completely understood. Endogenous neurogenesis in humans is thought to contribute to repair, but its extent is unknown. Exogenous cell therapy is promising as a means of augmenting brain repair, with evidence in animal stroke models of cell migration, survival, and differentiation, enhanced endogenous angiogenesis and neurogenesis, immunomodulation, and the secretion of trophic factors by stem cells from a variety of sources, but the potential mechanisms of action are incompletely understood. In the animal models of stroke, both mesenchymal stem cells (MSCs) and neural stem cells (NSCs) improve functional recovery, and MSCs reduce the infarct volume when administered acutely, but the heterogeneity in the choice of assessment scales, publication bias, and the possible confounding effects of immunosuppressants make the comparison of effects across cell types difficult. The use of adult-derived cells avoids the ethical issues around embryonic cells but may have more restricted differentiation potential. The use of autologous cells avoids rejection risk, but the sources are restricted, and culture expansion may be necessary, delaying treatment. Allogeneic cells offer controlled cell numbers and immediate availability, which may have advantages for acute treatment. Early clinical trials of both NSCs and MSCs are ongoing, and clinical safety data are emerging from limited numbers of selected patients. Ongoing research to identify prognostic imaging markers may help to improve patient selection, and the novel imaging techniques may identify biomarkers of recovery and the mechanism of action for cell therapies.
Collapse
Affiliation(s)
- Dheeraj Kalladka
- Institute of Neuroscience and Psychology, University of Glasgow, Southern General Hospital, Glasgow, United Kingdom
| | - Keith W Muir
- Institute of Neuroscience and Psychology, University of Glasgow, Southern General Hospital, Glasgow, United Kingdom
| |
Collapse
|
21
|
Drury-Stewart D, Song M, Mohamad O, Guo Y, Gu X, Chen D, Wei L. Highly efficient differentiation of neural precursors from human embryonic stem cells and benefits of transplantation after ischemic stroke in mice. Stem Cell Res Ther 2013; 4:93. [PMID: 23928330 PMCID: PMC3854684 DOI: 10.1186/scrt292] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 07/26/2013] [Indexed: 02/07/2023] Open
Abstract
Introduction Ischemic stroke is a leading cause of death and disability, but treatment options are severely limited. Cell therapy offers an attractive strategy for regenerating lost tissues and enhancing the endogenous healing process. In this study, we investigated the use of human embryonic stem cell-derived neural precursors as a cell therapy in a murine stroke model. Methods Neural precursors were derived from human embryonic stem cells by using a fully adherent SMAD inhibition protocol employing small molecules. The efficiency of neural induction and the ability of these cells to further differentiate into neurons were assessed by using immunocytochemistry. Whole-cell patch-clamp recording was used to demonstrate the electrophysiological activity of human embryonic stem cell-derived neurons. Neural precursors were transplanted into the core and penumbra regions of a focal ischemic stroke in the barrel cortex of mice. Animals received injections of bromodeoxyuridine to track regeneration. Neural differentiation of the transplanted cells and regenerative markers were measured by using immunohistochemistry. The adhesive removal test was used to determine functional improvement after stroke and intervention. Results After 11 days of neural induction by using the small-molecule protocol, over 95% of human embryonic stem-derived cells expressed at least one neural marker. Further in vitro differentiation yielded cells that stained for mature neuronal markers and exhibited high-amplitude, repetitive action potentials in response to depolarization. Neuronal differentiation also occurred after transplantation into the ischemic cortex. A greater level of bromodeoxyuridine co-localization with neurons was observed in the penumbra region of animals receiving cell transplantation. Transplantation also improved sensory recovery in transplant animals over that in control animals. Conclusions Human embryonic stem cell-derived neural precursors derived by using a highly efficient small-molecule SMAD inhibition protocol can differentiate into electrophysiologically functional neurons in vitro. These cells also differentiate into neurons in vivo, enhance regenerative activities, and improve sensory recovery after ischemic stroke.
Collapse
|
22
|
Park JS, Chang DY, Kim JH, Jung JH, Park J, Kim SH, Lee YD, Kim SS, Suh-Kim H. Retrovirus-mediated transduction of a cytosine deaminase gene preserves the stemness of mesenchymal stem cells. Exp Mol Med 2013; 45:e10. [PMID: 23429359 PMCID: PMC3584665 DOI: 10.1038/emm.2013.21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) have emerged as attractive cellular vehicles to deliver therapeutic genes for ex-vivo therapy of diverse diseases; this is, in part, because they have the capability to migrate into tumor or lesion sites. Previously, we showed that MSCs could be utilized to deliver a bacterial cytosine deaminase (CD) suicide gene to brain tumors. Here we assessed whether transduction with a retroviral vector encoding CD gene altered the stem cell property of MSCs. MSCs were transduced at passage 1 and cultivated up to passage 11. We found that proliferation and differentiation potentials, chromosomal stability and surface antigenicity of MSCs were not altered by retroviral transduction. The results indicate that retroviral vectors can be safely utilized for delivery of suicide genes to MSCs for ex-vivo therapy. We also found that a single retroviral transduction was sufficient for sustainable expression up to passage 10. The persistent expression of the transduced gene indicates that transduced MSCs provide a tractable and manageable approach for potential use in allogeneic transplantation.
Collapse
Affiliation(s)
- Jin Sung Park
- Department of Anatomy, Ajou University School of Medicine, Suwon, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Somarelli JA, Schaeffer D, Bosma R, Bonano VI, Sohn JW, Kemeny G, Ettyreddy A, Garcia-Blanco MA. Fluorescence-based alternative splicing reporters for the study of epithelial plasticity in vivo. RNA (NEW YORK, N.Y.) 2013; 19:116-127. [PMID: 23185039 PMCID: PMC3527723 DOI: 10.1261/rna.035097.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 10/22/2012] [Indexed: 05/31/2023]
Abstract
Alternative splicing generates a vast diversity of protein isoforms from a limited number of protein-coding genes, with many of the isoforms possessing unique, and even contrasting, functions. Fluorescence-based splicing reporters have the potential to facilitate studies of alternative splicing at the single-cell level and can provide valuable information on phenotypic transitions in almost real time. Fibroblast growth factor receptor 2 (FGFR2) pre-mRNA is alternatively spliced to form the epithelial-specific and mesenchymal-specific IIIb and IIIc isoforms, respectively, which are useful markers of epithelial-mesenchymal transitions (EMT). We have used our knowledge of FGFR2 splicing regulation to develop a fluorescence-based reporter system to visualize exon IIIc regulation in vitro and in vivo. Here we show the application of this reporter system to the study of EMT in vitro in cell culture and in vivo in transgenic mice harboring these splicing constructs. In explant studies, the reporters revealed that FGFR2 isoform switching is not required for keratinocyte migration during cutaneous wound closure. Our results demonstrate the value of the splicing reporters as tools to study phenotypic transitions and cell fates at single cell resolution. Moreover, our data suggest that keratinocytes migrate efficiently in the absence of a complete EMT.
Collapse
Affiliation(s)
| | - Daneen Schaeffer
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Reggie Bosma
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Vivian I. Bonano
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
- University Program in Genetics and Genomics
| | - Jang Wook Sohn
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Gabor Kemeny
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Abhinav Ettyreddy
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
| | - Mariano A. Garcia-Blanco
- Center for RNA Biology
- Department of Molecular Genetics and Microbiology
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
24
|
Hassani Z, O'Reilly J, Pearse Y, Stroemer P, Tang E, Sinden J, Price J, Thuret S. Human neural progenitor cell engraftment increases neurogenesis and microglial recruitment in the brain of rats with stroke. PLoS One 2012. [PMID: 23185625 PMCID: PMC3503964 DOI: 10.1371/journal.pone.0050444] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Main Objectives Stem cell transplantation is to date one of the most promising therapies for chronic ischemic stroke. The human conditionally immortalised neural stem cell line, CTX0E03, has demonstrable efficacy in a rodent model of stroke and is currently in clinical trials. Nonetheless, the mechanisms by which it promotes brain repair are not fully characterised. This study investigated the cellular events occurring after CTX0E03 transplantation in the brains of rats that underwent ischemic stroke. Methods We focused on the endogenous proliferative activity of the host brain in response to cell transplantation and determined the identity of the proliferating cells using markers for young neurons (doublecortin, Dcx) and microglia (CD11b). So as to determine the chronology of events occurring post-transplantation, we analysed the engrafted brains one week and four weeks post-transplantation. Results We observed a significantly greater endogenous proliferation in the striatum of ischemic brains receiving a CTX0E03 graft compared to vehicle-treated ischemic brains. A significant proportion of these proliferative cells were found to be Dcx+ striatal neuroblasts. Further, we describe an enhanced immune response after CTX0E03 engraftment, as shown by a significant increase of proliferating CD11b+ microglial cells. Conclusions Our study demonstrates that few Dcx+ neuroblasts are proliferative in normal conditions, and that this population of proliferative neuroblasts is increased in response to stroke. We further show that CTX0E03 transplantation after stroke leads to the maintenance of this proliferative activity. Interestingly, the preservation of neuronal proliferative activity upon CTX0E03 transplantation is preceded and accompanied by a high rate of proliferating microglia. Our study suggests that microglia might mediate in part the effect of CTX0E03 transplantation on neuronal proliferation in ischemic stroke conditions.
Collapse
Affiliation(s)
- Zahra Hassani
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Joanna O'Reilly
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Yewande Pearse
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
| | - Paul Stroemer
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - Ellen Tang
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - John Sinden
- ReNeuron Group plc, Guildford, Surrey, United Kingdom
| | - Jack Price
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
- * E-mail: (JP); (ST)
| | - Sandrine Thuret
- Centre for the Cellular Basis of Behaviour, The James Black Centre, King's College London, Institute of Psychiatry, London, United Kingdom
- * E-mail: (JP); (ST)
| |
Collapse
|
25
|
Hicks C, Stevanato L, Stroemer RP, Tang E, Richardson S, Sinden JD. In vivo and in vitro characterization of the angiogenic effect of CTX0E03 human neural stem cells. Cell Transplant 2012; 22:1541-52. [PMID: 23067568 DOI: 10.3727/096368912x657936] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
CTX0E03 is a human neural stem cell line previously reported to reduce sensory motor deficits in a middle cerebral artery occlusion (MCAo) model of stroke. The objective of this study was to investigate if CTX0E03 treatment promotes angiogenesis. As stroke leads to damage of the vasculature in the brain, angiogenesis may contribute to the functional recovery. To test this hypothesis, the angiogenic activity of CTX0E03 was assessed both in vitro and in vivo. In vitro, CTX0E03 expression of trophic and proangiogenic factors was determined by real-time RT-PCR, Western blot, and ELISA, and its angiogenic activity was investigated in well-established angiogenesis assays. In vivo, angiogenesis was investigated in naive mice and MCAo rat brain and was evaluated by immunohistochemistry (IHC) using Von Willebrand factor (VWF), a marker of blood vessel formation, and BrdU/CD31 double labeling in naive mice only. In vitro results showed that CTX0E03-conditioned medium and coculture significantly increased total tubule formation compared with controls (p=0.002 and p=0.0008, respectively). Furthermore, CTX0E03 cells were found to be in direct association with the tubules by ICC. In vivo CTX0E03-treated brains demonstrated a significant increase in areas occupied by VWF-positive microvessels compared with vehicle-treated naive mice (two-way ANOVA, Interaction p<0.05, Treatment p<0.0001, Time p<0.0) and MCAo rat (p=0.001 unpaired t test, Welch's correction). CTX0E03-treated naive mouse brains showed an increase in BrdU/CD31 colabeling. In conclusion, in vitro CTX0E03 cells express proangiogenic factors and may promote angiogenesis by both release of paracrine factors and direct physical interaction. Furthermore, in vivo CTX0E03-treated rodent brains exhibited a significant increase in microvessels at the site of implantation compared with vehicle-injected groups. Taken together these data suggest that CTX0E03 cell therapy may provide significant benefit to stroke patients through upregulation of angiogenesis in the ischemic brain.
Collapse
Affiliation(s)
- Caroline Hicks
- ReNeuron Limited, Surrey Research Park, Guildford, Surrey, UK
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Stroke, for some years now the neglected major indication in the pharmaceutical development cupboard, has recently become one of the hot areas for stem cell therapy development. This is driven by better understanding of potential therapeutic opportunities both in the acute and chronic phases and the launch of a series of new early phase clinical trials in a number of countries, driven by positive data in relevant animal models. In addition, the impetus for stem cell product development is motivated by patient demand, with thousands of victims seeking unproven treatments abroad. This article looks at the many challenges facing the development of a stem cell therapy for stroke. These range from product characterization and banking, through nonclinical safety and efficacy to the regulatory requirements for starting patient trials and beyond to maximizing value from carefully designed efficacy trials.
Collapse
Affiliation(s)
| | - Keith W. Muir
- Institute of Neuroscience and Psychology, University of Glasgow, Scotland, UK
| |
Collapse
|
27
|
Abstract
The prospects for stem cell-derived therapy in stroke look promising, with a myriad of cell therapy products developed from brain, blood, bone marrow, and adipose tissue in early clinical development. Eight clinical trials have now reported final results, and several are currently registered recruiting patients or pending to start. Products passing the safety hurdle are recruiting patients for large efficacy studies. Besides identifying the most appropriate cell type, other issues to resolve include optimal timing for intervention, optimal delivery route, cell dose, patient selection, relevant clinical endpoints, and monitoring for effectiveness, to advance cell therapy through the hurdles of clinical research. In this chapter, we present the products and strategies used in the current cell therapy trials in ischemic stroke, provide an update on relevant preclinical research, and discuss the vital developments still needed to advance their clinical application as a future therapeutic option.
Collapse
Affiliation(s)
- John D Sinden
- ReNeuron Limited, Surrey Research Park, Guildford, Surrey, UK.
| | | | | |
Collapse
|
28
|
Mejía-Toiber J, Castillo CG, Giordano M. Strategies for the Development of Cell Lines for Ex Vivo Gene Therapy in the Central Nervous System. Cell Transplant 2011; 20:983-1001. [DOI: 10.3727/096368910x546599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Disorders of the central nervous system (CNS) as a result of trauma or ischemic or neurodegenerative processes still pose a challenge for modern medicine. Due to the complexity of the CNS, and in spite of the advances in the knowledge of its anatomy, pharmacology, and molecular and cellular biology, treatments for these diseases are still limited. The development of cell lines as a source for transplantation into the damaged CNS (cell therapy), and more recently their genetic modification to favor the expression and delivery of molecules with therapeutic potential (ex vivo gene therapy), are some of the techniques used in search of novel restorative strategies. This article reviews the different approaches that have been used and perfected during the last decade to generate cell lines and their use in experimental models of neuronal damage, and evaluates the prospects of applying these methods to treat CNS disorders.
Collapse
Affiliation(s)
- Jana Mejía-Toiber
- Laboratorio de Plasticidad Neuronal, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro, Mexico
| | - Claudia G. Castillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico
| | - Magda Giordano
- Laboratorio de Plasticidad Neuronal, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro, Mexico
| |
Collapse
|
29
|
|