51
|
Nucci LP, Silva HR, Giampaoli V, Mamani JB, Nucci MP, Gamarra LF. Stem cells labeled with superparamagnetic iron oxide nanoparticles in a preclinical model of cerebral ischemia: a systematic review with meta-analysis. Stem Cell Res Ther 2015; 6:27. [PMID: 25889904 PMCID: PMC4425914 DOI: 10.1186/s13287-015-0015-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 11/26/2014] [Accepted: 02/23/2015] [Indexed: 12/17/2022] Open
Abstract
Introduction Although there is an increase in clinical trials assessing the efficacy of cell therapy in structural and functional regeneration after stroke, there are not enough data in the literature describing the best cell type to be used, the best route, and also the best nanoparticle to analyze these stem cells in vivo. This review analyzed published data on superparamagnetic iron oxide nanoparticle (SPION)-labeled stem cells used for ischemic stroke therapy. Method We performed a systematic review and meta-analysis of data from experiments testing the efficacy of cellular treatment with SPION versus no treatment to improve behavioral or modified neural scale outcomes in animal models of stroke by the Cochrane Collaboration and indexed in EMBASE, PubMed, and Web of Science since 2000. To test the impact of study quality and design characteristics, we used random-effects meta-regression. In addition, trim and fill were used to assess publication bias. Results The search retrieved 258 articles. After application of the inclusion criteria, 24 reports published between January 2000 and October 2014 were selected. These 24 articles were analyzed for nanoparticle characteristics, stem cell types, and efficacy in animal models. Conclusion This study highlights the therapeutic role of stem cells in stroke and emphasizes nanotechnology as an important tool for monitoring stem cell migration to the affected neurological locus.
Collapse
Affiliation(s)
- Leopoldo P Nucci
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Universidade Federal de São Paulo, Rua Sena Madureira, 1500 - Vila Clementino, 04021-001, São Paulo-SP, Brazil.
| | - Helio R Silva
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Santa Casa Misericórdia de São Paulo, Dr. Cesario Motta Junior, 61 - Vila Buarque, 01221-020, São Paulo-SP, Brazil.
| | - Viviana Giampaoli
- Instituto de Matemática e Estatística, Universidade de São Paulo, Rua do Matão 1010 - Cidade Universitária, 05508-090, São Paulo-SP, Brazil.
| | - Javier B Mamani
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil.
| | - Mariana P Nucci
- LIM44, Universidade de São Paulo, Rua Dr Éneas de Carvalho Aguiar, 255 - Cerqueira César, 05403-000, São Paulo-SP, Brazil.
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, Av. Albert Einstein, 627/701, Morumbi, CEP: 05651-901, São Paulo, Brazil. .,Universidade Federal de São Paulo, Rua Sena Madureira, 1500 - Vila Clementino, 04021-001, São Paulo-SP, Brazil. .,Santa Casa Misericórdia de São Paulo, Dr. Cesario Motta Junior, 61 - Vila Buarque, 01221-020, São Paulo-SP, Brazil.
| |
Collapse
|
52
|
Tang YH, Ma YY, Zhang ZJ, Wang YT, Yang GY. Opportunities and challenges: stem cell-based therapy for the treatment of ischemic stroke. CNS Neurosci Ther 2015; 21:337-47. [PMID: 25676164 DOI: 10.1111/cns.12386] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 01/01/2023] Open
Abstract
Stem cell-based therapy for ischemic stroke has been widely explored in animal models and provides strong evidence of benefits. In this review, we summarize the types of stem cells, various delivery routes, and tracking tools for stem cell therapy of ischemic stroke. MSCs, EPCs, and NSCs are the most explored cell types for ischemic stroke treatment. Although the mechanisms of stem cell-based therapies are not fully understood, the most possible functions of the transplanted cells are releasing growth factors and regulating microenvironment through paracrine mechanism. Clinical application of stem cell-based therapy is still in its infancy. The next decade of stem cell research in stroke field needs to focus on combining different stem cells and different imaging modalities to fully explore the potential of this therapeutic avenue: from bench to bedside and vice versa.
Collapse
Affiliation(s)
- Yao-Hui Tang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
53
|
Yan Y, Sart S, Calixto Bejarano F, Muroski ME, Strouse GF, Grant SC, Li Y. Cryopreservation of embryonic stem cell-derived multicellular neural aggregates labeled with micron-sized particles of iron oxide for magnetic resonance imaging. Biotechnol Prog 2015; 31:510-21. [PMID: 25905549 DOI: 10.1002/btpr.2049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 12/14/2014] [Indexed: 12/19/2022]
Abstract
Magnetic resonance imaging (MRI) provides an effective approach to track labeled pluripotent stem cell (PSC)-derived neural progenitor cells (NPCs) for neurological disorder treatments after cell labeling with a contrast agent, such as an iron oxide derivative. Cryopreservation of pre-labeled neural cells, especially in three-dimensional (3D) structure, can provide a uniform cell population and preserve the stem cell niche for the subsequent applications. In this study, the effects of cryopreservation on PSC-derived multicellular NPC aggregates labeled with micron-sized particles of iron oxide (MPIO) were investigated. These NPC aggregates were labeled prior to cryopreservation because labeling thawed cells can be limited by inefficient intracellular uptake, variations in labeling efficiency, and increased culture time before use, minimizing their translation to clinical settings. The results indicated that intracellular MPIO incorporation was retained after cryopreservation (70-80% labeling efficiency), and MPIO labeling had little adverse effects on cell recovery, proliferation, cytotoxicity and neural lineage commitment post-cryopreservation. MRI analysis showed comparable detectability for the MPIO-labeled cells before and after cryopreservation indicated by T2 and T2* relaxation rates. Cryopreserving MPIO-labeled 3D multicellular NPC aggregates can be applied in in vivo cell tracking studies and lead to more rapid translation from preservation to clinical implementation.
Collapse
Affiliation(s)
- Yuanwei Yan
- Dept. of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL
| | | | | | | | | | | | | |
Collapse
|
54
|
Sart S, Bejarano FC, Baird MA, Yan Y, Rosenberg JT, Ma T, Grant SC, Li Y. Intracellular labeling of mouse embryonic stem cell–derived neural progenitor aggregates with micron-sized particles of iron oxide. Cytotherapy 2015; 17:98-111. [DOI: 10.1016/j.jcyt.2014.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/28/2014] [Accepted: 09/16/2014] [Indexed: 12/21/2022]
|
55
|
Insulin-producing cells from embryonic stem cells rescues hyperglycemia via intra-spleen migration. Sci Rep 2014; 4:7586. [PMID: 25533571 PMCID: PMC4274503 DOI: 10.1038/srep07586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/24/2014] [Indexed: 02/07/2023] Open
Abstract
Implantation of embryonic stem cells (ESC)-derived insulin-producing cells has been extensively investigated for treatment of diabetes in animal models. However, the in vivo behavior and migration of transplanted cells in diabetic models remains unclear. Here we investigated the location and migration of insulin-producing cells labeled with superparamagnetic iron oxide (SPIO) using a dynamic MRI tracking method. SPIO labeled cells showed hypointense signal under the kidney subcapsules of diabetic mice on MRI, and faded gradually over the visiting time. However, new hypointense signal appeared in the spleen 1 week after transplantation, and became obvious with the time prolongation. Further histological examination proved the immigrated cells were insulin and C-peptide positive cells which were evenly distributed throughout the spleen. These intra-spleen insulin-producing cells maintained their protective effects against hyperglycemia in vivo, and these effects were reversed upon spleen removal. Transplantation of insulin-producing cells through spleen acquired an earlier blood glucose control as compared with that through kidney subcapsules. In summary, our data demonstrate that insulin-producing cells transplanted through kidney subcapsules were not located in situ but migrated into spleen, and rescues hyperglycemia in diabetic models. MRI may provide a novel tracking method for preclinical cell transplantation therapy of diabetes continuously and non-invasively.
Collapse
|
56
|
Abstract
Although cellular therapies hold great promise for the treatment of human disease, results from several initial clinical trials have not shown a level of efficacy required for their use as a first line therapy. Here we discuss how in vivo molecular imaging has helped identify barriers to clinical translation and potential strategies that may contribute to successful transplantation and improved outcomes, with a focus on cardiovascular and neurological diseases. We conclude with a perspective on the future role of molecular imaging in defining safety and efficacy for clinical implementation of stem cell therapies.
Collapse
|
57
|
Duffy BA, Weitz AJ, Lee JH. In vivo imaging of transplanted stem cells in the central nervous system. Curr Opin Genet Dev 2014; 28:83-8. [PMID: 25461455 DOI: 10.1016/j.gde.2014.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/14/2014] [Indexed: 12/11/2022]
Abstract
In vivo imaging is increasingly being utilized in studies investigating stem cell-based treatments for neurological disorders. Direct labeling is used in preclinical and clinical studies to track the fate of transplanted cells. To further determine cell viability, experimental studies are able to take advantage of reporter gene technologies. Structural and functional brain imaging can also be used alongside cell imaging as biomarkers of treatment efficacy. Furthermore, it is possible that new imaging techniques could be used to monitor functional integration of stem cell-derived cells with the host nervous system. In this review, we examine recent developments in these areas and identify promising directions for future research at the interface of stem cell therapies and neuroimaging.
Collapse
Affiliation(s)
- Ben A Duffy
- Department of Neurology & Neurological Sciences, Stanford University, CA 94305, USA
| | - Andrew J Weitz
- Department of Bioengineering, Stanford University, CA 94305, USA
| | - Jin Hyung Lee
- Department of Neurology & Neurological Sciences, Stanford University, CA 94305, USA; Department of Bioengineering, Stanford University, CA 94305, USA; Department of Neurosurgery, Stanford University, CA 94305, USA; Department of Electrical Engineering, Stanford University, CA 94305, USA.
| |
Collapse
|
58
|
Pomeshchik Y, Puttonen KA, Kidin I, Ruponen M, Lehtonen S, Malm T, Åkesson E, Hovatta O, Koistinaho J. Transplanted Human Induced Pluripotent Stem Cell-Derived Neural Progenitor Cells Do Not Promote Functional Recovery of Pharmacologically Immunosuppressed Mice With Contusion Spinal Cord Injury. Cell Transplant 2014; 24:1799-812. [PMID: 25203632 DOI: 10.3727/096368914x684079] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Improved functional recovery after spinal cord injury by transplantation of induced pluripotent stem cell-derived neural stem/progenitor cells (iPSC-NPCs) has been reported. However, beneficial effects of iPSC-based therapy have so far been produced mostly using genetically immunodeficient rodents. Because of the long time required for generation and characterization of iPSCs, the use of autologous iPSCs for treating patients with acute spinal cord injury (SCI) is not feasible. Therefore, it is of utmost importance to investigate the effect of iPSC-based therapy on functional recovery after SCI using pharmacologically immunosuppressed, immunocompetent animal models. Here we studied the functional outcome following subacute transplantation of human iPSC-derived NPCs into contused mouse spinal cord when tacrolimus was used as an immunosuppressive agent. We show that human iPSC-derived NPCs transplanted into pharmacologically immunosuppressed C57BL/6J mice exhibited poor long-term survival and failed to improve functional recovery after SCI as measured by Basso Mouse Scale (BMS) for locomotion and CatWalk gait analysis when compared to vehicle-treated animals. The scarce effect of iPSC-based therapy observed in the current study may be attributable to insufficient immunosuppressive effect, provided by monotherapy with tacrolimus in combination with immunogenicity of transplanted cells and complex microenvironment of the injured spinal cord. Our results highlight the importance of extensive preclinical studies of transplanted cells before the clinical application of iPSC-based cell therapy is achieved.
Collapse
Affiliation(s)
- Yuriy Pomeshchik
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Aswendt M, Adamczak J, Tennstaedt A. A review of novel optical imaging strategies of the stroke pathology and stem cell therapy in stroke. Front Cell Neurosci 2014; 8:226. [PMID: 25177269 PMCID: PMC4132298 DOI: 10.3389/fncel.2014.00226] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/22/2014] [Indexed: 12/17/2022] Open
Abstract
Transplanted stem cells can induce and enhance functional recovery in experimental stroke. Invasive analysis has been extensively used to provide detailed cellular and molecular characterization of the stroke pathology and engrafted stem cells. But post mortem analysis is not appropriate to reveal the time scale of the dynamic interplay between the cell graft, the ischemic lesion and the endogenous repair mechanisms. This review describes non-invasive imaging techniques which have been developed to provide complementary in vivo information. Recent advances were made in analyzing simultaneously different aspects of the cell graft (e.g., number of cells, viability state, and cell fate), the ischemic lesion (e.g., blood-brain-barrier consistency, hypoxic, and necrotic areas) and the neuronal and vascular network. We focus on optical methods, which permit simple animal preparation, repetitive experimental conditions, relatively medium-cost instrumentation and are performed under mild anesthesia, thus nearly under physiological conditions. A selection of recent examples of optical intrinsic imaging, fluorescence imaging and bioluminescence imaging to characterize the stroke pathology and engrafted stem cells are discussed. Special attention is paid to novel optimal reporter genes/probes for genetic labeling and tracking of stem cells and appropriate transgenic animal models. Requirements, advantages and limitations of these imaging platforms are critically discussed and placed into the context of other non-invasive techniques, e.g., magnetic resonance imaging and positron emission tomography, which can be joined with optical imaging in multimodal approaches.
Collapse
Affiliation(s)
| | | | - Annette Tennstaedt
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, KölnGermany
| |
Collapse
|
60
|
Gonzales-Portillo GS, Reyes S, Aguirre D, Pabon MM, Borlongan CV. Stem cell therapy for neonatal hypoxic-ischemic encephalopathy. Front Neurol 2014; 5:147. [PMID: 25161645 PMCID: PMC4130306 DOI: 10.3389/fneur.2014.00147] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 07/22/2014] [Indexed: 11/27/2022] Open
Abstract
Treatments for neonatal hypoxic-ischemic encephalopathy (HIE) have been limited. The aim of this paper is to offer translational research guidance on stem cell therapy for neonatal HIE by examining clinically relevant animal models, practical stem cell sources, safety and efficacy of endpoint assays, as well as a general understanding of modes of action of this cellular therapy. In order to do so, we discuss the clinical manifestations of HIE, highlighting its overlapping pathologies with stroke and providing insights on the potential of cell therapy currently investigated in stroke, for HIE. To this end, we draw guidance from recommendations outlined in stem cell therapeutics as an emerging paradigm for stroke or STEPS, which have been recently modified to Baby STEPS to cater for the “neonatal” symptoms of HIE. These guidelines recognized that neonatal HIE exhibit distinct disease symptoms from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, new information about recent clinical trials and insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with HIE.
Collapse
Affiliation(s)
| | - Stephanny Reyes
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Daniela Aguirre
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Mibel M Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| |
Collapse
|
61
|
Alvarim LT, Nucci LP, Mamani JB, Marti LC, Aguiar MF, Silva HR, Silva GS, Nucci-da-Silva MP, DelBel EA, Gamarra LF. Therapeutics with SPION-labeled stem cells for the main diseases related to brain aging: a systematic review. Int J Nanomedicine 2014; 9:3749-70. [PMID: 25143726 PMCID: PMC4137998 DOI: 10.2147/ijn.s65616] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The increase in clinical trials assessing the efficacy of cell therapy for structural and functional regeneration of the nervous system in diseases related to the aging brain is well known. However, the results are inconclusive as to the best cell type to be used or the best methodology for the homing of these stem cells. This systematic review analyzed published data on SPION (superparamagnetic iron oxide nanoparticle)-labeled stem cells as a therapy for brain diseases, such as ischemic stroke, Parkinson’s disease, amyotrophic lateral sclerosis, and dementia. This review highlights the therapeutic role of stem cells in reversing the aging process and the pathophysiology of brain aging, as well as emphasizing nanotechnology as an important tool to monitor stem cell migration in affected regions of the brain.
Collapse
Affiliation(s)
- Larissa T Alvarim
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | | | | | | | - Marina F Aguiar
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Helio R Silva
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | | | | | - Elaine A DelBel
- Universidade de São Paulo-Faculdade de Odontologia de Ribeirão Preto, São Paulo, Brazil ; NAPNA-Núcleo de Apoio a Pesquisa em Neurociências Aplicadas, São Paulo, Brazil
| | - Lionel F Gamarra
- Hospital Israelita Albert Einstein, São Paulo, Brazil ; Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil ; Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| |
Collapse
|
62
|
Zhou C, Huang Z, Li P, Li W, Liu Y, Li C, Liu Z, Wang X, Wan P, Wang Z. Safety and Efficacy of Embryonic Stem Cell Microenvironment in a Leukemia Mouse Model. Stem Cells Dev 2014; 23:1741-54. [DOI: 10.1089/scd.2013.0585] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Affiliation(s)
- Chenjing Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zheqian Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Panlong Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Chaoyang Li
- State Key Laboratory of Ophthalmology, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Pengxia Wan
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhichong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
63
|
Stem cell therapy for acute cerebral injury: what do we know and what will the future bring? Curr Opin Neurol 2014; 26:617-25. [PMID: 24136128 DOI: 10.1097/wco.0000000000000023] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW The central nervous system has limited capacity for regeneration after acute and chronic injury. An attractive approach to stimulate neural plasticity in the brain is to transplant stem cells in order to restore function. Here, we discuss potential mechanisms of action, current knowledge and future perspectives of clinical stem cell research for stroke and traumatic brain injury. RECENT FINDINGS Preclinical data using various models suggest stem cell therapy to be a promising therapeutic avenue. Progress has been made in elucidating the mechanism of action of various cell types used, shifting the hypothesis from neural replacement to enhancing endogenous repair processes. Translation of these findings in clinical trials is currently being pursued with emphasis on both safety as well as efficacy. SUMMARY Clinical trials are currently recruiting patients in phase I and II trials to gain more insight in the therapeutic potential of stem cells in acute cerebral injury. A close interplay between results of these clinical trials and more extensive basic research is essential for future trial design, choosing the optimal transplantation strategy and selecting the right patients.
Collapse
|
64
|
Shinozuka K, Dailey T, Tajiri N, Ishikawa H, Kaneko Y, Borlongan CV. Stem cell transplantation for neuroprotection in stroke. Brain Sci 2014; 3:239-61. [PMID: 24147217 PMCID: PMC3800120 DOI: 10.3390/brainsci3010239] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cell-based therapies for stroke have expanded substantially over the last decade. The diversity of embryonic and adult tissue sources provides researchers with the ability to harvest an ample supply of stem cells. However, the optimal conditions of stem cell use are still being determined. Along this line of the need for optimization studies, we discuss studies that demonstrate effective dose, timing, and route of stem cells. We recognize that stem cell derivations also provide uniquely individual difficulties and limitations in their therapeutic applications. This review will outline the current knowledge, including benefits and challenges, of the many current sources of stem cells for stroke therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Cesar V. Borlongan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-813-974-3988; Fax: +1-813-974-3078
| |
Collapse
|
65
|
Khalili MA, Sadeghian-Nodoushan F, Fesahat F, Mir-Esmaeili SM, Anvari M, Hekmati-Moghadam SH. Mesenchymal stem cells improved the ultrastructural morphology of cerebral tissues after subarachnoid hemorrhage in rats. Exp Neurobiol 2014; 23:77-85. [PMID: 24737942 PMCID: PMC3984959 DOI: 10.5607/en.2014.23.1.77] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/24/2014] [Accepted: 02/24/2014] [Indexed: 01/19/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) causes widespread disruption in the cerebral architecture.The process of SAH is complicated and many people lose their lives or become disabled after injury. Mesenchymal stem cells (MSCs) are considered as good candidate for repair of cerebral damage. The aim was to assess the ultrastructural changes in the rat cerebral tissue after intravenous transplantation of MSCs. Female Wistar rats (8 per group) weighing 275~300 g were assigned to control (SAH+PBS) and experimental groups (SAH+MSCs).The samples from middle cerebral arterial wall and parietal cerebral tissue were prepared for transmission electron microscopy (TEM) according to standard protocol. Fine architectures of the vessel wall, including the contraction of the inner layer, smooth muscle layer,as well as neural cells were observed after SAH. Cerebral arterial wall and cortex, including neuronal and glial cells were injured post SAH. But, administration of MSCs improved the structural integrity of cerebral tissues. Changes were much more balanced with their relative improvement in some areas. The role of MSCs for repairing the injured cerebral tissues post experimental SAH was approved by electron microscopy.
Collapse
Affiliation(s)
- Mohammad Ali Khalili
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Sadeghian-Nodoushan
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. ; Stem Cell Biology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farzaneh Fesahat
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mohsen Mir-Esmaeili
- Yazd ACECR Higher Education Institute, Yazd, Iran. ; Department of Biology, Faculty of Science, Sistan & Baluchestan University, Zahedan, Iran
| | - Morteza Anvari
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | |
Collapse
|
66
|
Bernau K, Lewis CM, Petelinsek AM, Benink HA, Zimprich CA, Meyerand ME, Suzuki M, Svendsen CN. In vivo tracking of human neural progenitor cells in the rat brain using bioluminescence imaging. J Neurosci Methods 2014; 228:67-78. [PMID: 24675049 DOI: 10.1016/j.jneumeth.2014.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND Stem cell therapies appear promising for treating certain neurodegenerative disorders and molecular imaging methods that track these cells in vivo could answer some key questions regarding their survival and migration. Bioluminescence imaging (BLI), which relies on luciferase expression in these cells, has been used for this purpose due to its high sensitivity. NEW METHOD In this study, we employ BLI to track luciferase-expressing human neural progenitor cells (hNPC(Luc2)) in the rat striatum long-term. RESULTS We show that hNPC(Luc2) are detectable in the rat striatum. Furthermore, we demonstrate that using this tracking method, surviving grafts can be detected in vivo for up to 12 weeks, while those that were rejected do not produce bioluminescence signal. We also demonstrate the ability to discern hNPC(Luc2) contralateral migration. COMPARISON WITH EXISTING METHODS Some of the advantages of BLI compared to other imaging methods used to track progenitor/stem cells include its sensitivity and specificity, low background signal and ability to distinguish surviving grafts from rejected ones over the long term while the blood-brain barrier remains intact. CONCLUSIONS These new findings may be useful in future preclinical applications developing cell-based treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Ksenija Bernau
- University of Wisconsin-Madison, 4325a Veterinary Medicine Building, 2015 Linden Dr., Madison, WI 53706, USA.
| | - Christina M Lewis
- University of Wisconsin-Madison, 1005 Wisconsin Institute for Medical Research, 1111 Highland Ave., Madison, WI 53705, USA.
| | - Anna M Petelinsek
- University of Wisconsin-Madison, 4325a Veterinary Medicine Building, 2015 Linden Dr., Madison, WI 53706, USA.
| | - Hélène A Benink
- Promega Corporation, 2800 Woods Hollow Rd., Fitchburg, WI 53711, USA.
| | - Chad A Zimprich
- Promega Corporation, 2800 Woods Hollow Rd., Fitchburg, WI 53711, USA.
| | - M Elizabeth Meyerand
- University of Wisconsin-Madison, 1129 Wisconsin Institute for Medical Research, 1111 Highland Ave., Madison, WI 53705, USA.
| | - Masatoshi Suzuki
- University of Wisconsin-Madison, 4124 Veterinary Medicine Building, 2015 Linden Dr., Madison, WI 53706, USA.
| | - Clive N Svendsen
- University of Wisconsin-Madison, 5009 Wisconsin Institute for Medical Research, 1111 Highland Ave., Madison, WI 53705, USA.
| |
Collapse
|
67
|
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
|
68
|
Sart S, Bejarano FC, Yan Y, Grant SC, Li Y. Labeling pluripotent stem cell-derived neural progenitors with iron oxide particles for magnetic resonance imaging. Methods Mol Biol 2014; 1283:43-52. [PMID: 25304204 DOI: 10.1007/7651_2014_123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Due to the unlimited proliferation capacity and the unique differentiation ability of pluripotent stem cells (PSCs), including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), large numbers of PSC-derived cell products are in demand for applications in drug screening, disease modeling, and especially cell therapy. In stem cell-based therapy, tracking transplanted cells with magnetic resonance imaging (MRI) has emerged as a powerful technique to reveal cell survival and distribution. This chapter illustrated the basic steps of labeling PSC-derived neural progenitors (NPs) with micron-sized particles of iron oxide (MPIO, 0.86 μm) for MRI analysis. The protocol described PSC expansion and differentiation into NPs, and the labeling of the derived cells either after replating on adherent surface or in suspension. The labeled cells can be analyzed using in vitro MRI analysis. The methods presented here can be easily adapted for cell labeling in cell processing facilities under current Good Manufacturing Practices (cGMP). The iron oxide-labeled NPs can be used for cellular monitoring of in vitro cultures and in vivo transplantation.
Collapse
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, 2525 Pottsdamer St, Tallahassee, FL, 32310, USA
| | | | | | | | | |
Collapse
|
69
|
Daadi MM, Hu S, Klausner J, Li Z, Sofilos M, Sun G, Wu JC, Steinberg GK. Imaging neural stem cell graft-induced structural repair in stroke. Cell Transplant 2013; 22:881-92. [PMID: 23044338 DOI: 10.3727/096368912x656144] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Stem cell therapy ameliorates motor deficits in experimental stroke model. Multimodal molecular imaging enables real-time longitudinal monitoring of infarct location, size, and transplant survival. In the present study, we used magnetic resonance imaging (MRI) and positron emission tomography (PET) to track the infarct evolution,tissue repair, and the fate of grafted cells. We genetically engineered embryonic stem cell-derived neural stem cells (NSCs) with a triple fusion reporter gene to express monomeric red fluorescence protein and herpes simplex virus-truncated thymidine kinase for multimodal molecular imaging and SPIO labeled for MRI. The infarct size as well as fate and function of grafted cells were tracked in real time for 3 months using MRI and PET. We report that grafted NSCs reduced the infarct size in animals with less than 0.1 cm(3) initial infarct in a dose-dependent manner, while larger stroke was not amenable to such beneficial effects. PET imaging revealed increased metabolic activity in grafted animals and visualized functioning grafted cells in vivo. Immunohistopathological analysis demonstrated that, after a 3-month survival period, grafted NSCs dispersed in the stroke-lesioned parenchyma and differentiated into neurons, astrocytes, and oligodendrocytes. Longitudinal multimodal imaging provides insights into time course dose-dependent interactions between NSC grafts and structural changes in infarcted tissue.
Collapse
Affiliation(s)
- Marcel M Daadi
- Department of Neurosurgery, Stanford Stroke Center and Stanford Institute for Neuro-Innovationand Translational Neurosciences, Stanford, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Boehm-Sturm P, Aswendt M, Minassian A, Michalk S, Mengler L, Adamczak J, Mezzanotte L, Löwik C, Hoehn M. A multi-modality platform to image stem cell graft survival in the naïve and stroke-damaged mouse brain. Biomaterials 2013; 35:2218-26. [PMID: 24355489 DOI: 10.1016/j.biomaterials.2013.11.085] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 11/27/2013] [Indexed: 02/08/2023]
Abstract
Neural stem cell implantations have been extensively investigated for treatment of brain diseases such as stroke. In order to follow the localization and functional status of cells after implantation noninvasive imaging is essential. Therefore, we developed a comprehensive multi-modality platform for in vivo imaging of graft localization, density, and survival using 19F magnetic resonance imaging in combination with bioluminescence imaging. We quantitatively analyzed cell graft survival over the first 4 weeks after transplantation in both healthy and stroke-damaged mouse brain and correlated our findings of graft vitality with the host innate immune response. The multi-modality imaging platform will help to improve cell therapy also in context other than stroke and to gain indispensable information for clinical translation.
Collapse
Affiliation(s)
- Philipp Boehm-Sturm
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Markus Aswendt
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Anuka Minassian
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Stefanie Michalk
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Luam Mengler
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Joanna Adamczak
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany
| | - Laura Mezzanotte
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clemens Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mathias Hoehn
- In-Vivo-NMR Laboratory, Max-Planck-Institute for Neurological Research in Cologne, Cologne, Germany; Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
71
|
Liu X, Ye R, Yan T, Yu SP, Wei L, Xu G, Fan X, Jiang Y, Stetler RA, Liu G, Chen J. Cell based therapies for ischemic stroke: from basic science to bedside. Prog Neurobiol 2013; 115:92-115. [PMID: 24333397 DOI: 10.1016/j.pneurobio.2013.11.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/11/2013] [Accepted: 11/26/2013] [Indexed: 12/20/2022]
Abstract
Cell therapy is emerging as a viable therapy to restore neurological function after stroke. Many types of stem/progenitor cells from different sources have been explored for their feasibility and efficacy for the treatment of stroke. Transplanted cells not only have the potential to replace the lost circuitry, but also produce growth and trophic factors, or stimulate the release of such factors from host brain cells, thereby enhancing endogenous brain repair processes. Although stem/progenitor cells have shown a promising role in ischemic stroke in experimental studies as well as initial clinical pilot studies, cellular therapy is still at an early stage in humans. Many critical issues need to be addressed including the therapeutic time window, cell type selection, delivery route, and in vivo monitoring of their migration pattern. This review attempts to provide a comprehensive synopsis of preclinical evidence and clinical experience of various donor cell types, their restorative mechanisms, delivery routes, imaging strategies, future prospects and challenges for translating cell therapies as a neurorestorative regimen in clinical applications.
Collapse
Affiliation(s)
- Xinfeng Liu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Ruidong Ye
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Neurology, Tianjin General Hospital, Tianjin University School of Medicine, Tianjin, China
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xinying Fan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
72
|
Panizzo RA, Gadian DG, Sowden JC, Wells JA, Lythgoe MF, Ferretti P. Monitoring ferumoxide-labelled neural progenitor cells and lesion evolution by magnetic resonance imaging in a model of cell transplantation in cerebral ischaemia. F1000Res 2013; 2:252. [PMID: 24715962 PMCID: PMC3962009 DOI: 10.12688/f1000research.2-252.v2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2014] [Indexed: 01/05/2023] Open
Abstract
Efficacy of neural stem/progenitor cell (NPC) therapies after cerebral ischaemia could be better evaluated by monitoring
in vivo migration and distribution of cells post-engraftment in parallel with analysis of lesion volume and functional recovery. Magnetic resonance imaging (MRI) is ideally placed to achieve this, but still poses several challenges. We show that combining the ferumoxide MRI contrast agent Endorem with protamine sulphate (FePro) improves iron oxide uptake in cells compared to Endorem alone and is non-toxic. Hence FePro complex is a better contrast agent than Endorem for monitoring NPCs. FePro complex-labelled NPCs proliferated and differentiated normally
in vitro, and upon grafting into the brain 48 hours post-ischaemia they were detected
in vivo by MRI. Imaging over four weeks showed the development of a confounding endogenous hypointense contrast evolution at later timepoints within the lesioned tissue. This was at least partly due to accumulation within the lesion of macrophages and endogenous iron. Neither significant NPC migration, assessed by MRI and histologically, nor a reduction in the ischaemic lesion volume was observed in NPC-grafted brains. Crucially, while MRI provides reliable information on engrafted cell location early after an ischaemic insult, pathophysiological changes to ischaemic lesions can interfere with cellular imaging at later timepoints.
Collapse
Affiliation(s)
- Rachael A Panizzo
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - David G Gadian
- Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jane C Sowden
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Jack A Wells
- UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - Mark F Lythgoe
- Imaging and Biophysics Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK ; UCL Centre for Advanced Biomedical Imaging, Department of Medicine, University College London, London, WC1E 6DD, UK
| | - Patrizia Ferretti
- Developmental Biology Unit, UCL Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
73
|
Multimodality molecular imaging of stem cells therapy for stroke. BIOMED RESEARCH INTERNATIONAL 2013; 2013:849819. [PMID: 24222920 PMCID: PMC3816035 DOI: 10.1155/2013/849819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 08/21/2013] [Indexed: 12/03/2022]
Abstract
Stem cells have been proposed as a promising therapy for treating stroke. While several studies have demonstrated the therapeutic benefits of stem cells, the exact mechanism remains elusive. Molecular imaging provides the possibility of the visual representation of biological processes at the cellular and molecular level. In order to facilitate research efforts to understand the stem cells therapeutic mechanisms, we need to further develop means of monitoring these cells noninvasively, longitudinally and repeatedly. Because of tissue depth and the blood-brain barrier (BBB), in vivo imaging of stem cells therapy for stroke has unique challenges. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including magnetic resonance imaging (MRI), nuclear medicine imaging, and optical imaging (OI). Each of the imaging techniques has advantages and drawbacks. Finally, we describe multimodality imaging strategies as a more comprehensive and potential method to monitor transplanted stem cells for stroke.
Collapse
|
74
|
Puttonen KA, Ruponen M, Kauppinen R, Wojciechowski S, Hovatta O, Koistinaho J. Improved Method of Producing Human Neural Progenitor Cells of High Purity and in Large Quantities from Pluripotent Stem Cells for Transplantation Studies. Cell Transplant 2013; 22:1753-66. [DOI: 10.3727/096368912x658764] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Transplantation of human neural progenitor cells (hNPCs) is a promising therapeutic approach for various diseases of the central nervous system (CNS). Reliable testing of hNPC transplantation in animal models of neurological diseases requires that these cells can be produced in sufficient amounts, show consistent homogeneity as a neural cell population, and be reliably labeled for in vivo tracking. In addition, the cells should be characterized as being at the optimal state of differentiation favoring successful engraftment. Here, we show that high numbers of purified hNPCs can be produced from human embryonic stem cells (hESCs) by manually selecting specifically sized and shaped spheres followed by fluorescence-activated cell sorting based on the relative cell size. In addition, we report that labeling of hNPCs with ultra-small superparamagnetic iron oxide (USPIO) particles does not affect the cellular morphology or growth. More importantly, we show that the transduction with lentiviral vector encoding green fluorescent protein (GFP) decreases the neurality of the cell population. We conclude that our cost-effective protocol of generating hNPCs is widely applicable for preclinical studies on CNS disorders. This improved method of producing large quantities of high-purity hNPCs maybe useful also when generating hNPCs from human induced pluripotent stem (hiPS) cell lines. However, caution should be used when lenti-GFP transduction is applied for hNPC labeling.
Collapse
Affiliation(s)
- Katja A. Puttonen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marika Ruponen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Riitta Kauppinen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sara Wojciechowski
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Outi Hovatta
- Department of Biotechnology and Molecular Medicine, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Jari Koistinaho
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
75
|
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
|
76
|
MRI detection of nonproliferative tumor cells in lymph node metastases using iron oxide particles in a mouse model of breast cancer. Transl Oncol 2013; 6:347-54. [PMID: 23730415 DOI: 10.1593/tlo.13121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 01/28/2023] Open
Abstract
Cell tracking with magnetic resonance imaging (MRI) and iron nanoparticles is commonly used to monitor the fate of implanted cells in preclinical disease models. Few studies have employed these methods to study cancer cells because proliferative iron-labeled cancer cells will lose the label as they divide. In this study, we evaluate the potential for retention of the iron nanoparticle label, and resulting MRI signal, to serve as a marker for slowly dividing cancer cells. Green fluorescent protein-transfected MDA-MB-231 breast cancer cells were labeled with red fluorescent micron-sized superparamagnetic iron oxide (MPIO) nanoparticles. Cells were examined in vitro at multiple time points after labeling by staining for iron-labeled cells and by flow cytometric detection of the fluorescent MPIO. Severe combined immune deficiency (SCID) mice were implanted with 5 x 10(5) MPIO-labeled or unlabeled cells in the mammary fat pad and MRI was performed weekly until 28 days after injection. Microscopy was performed to validate MRI. In vitro assays revealed a very small percentage of cells that retained MPIO at 14 days after labeling. Regions of signal loss were observed in MRI of primary tumors that developed from iron-labeled cancer cells. Small focal regions of signal loss were detected in images of the axillary and brachial nodes in six of eight mice, at day 14 or later, with microscopy confirming the presence of iron-labeled cancer cells. Our data suggest an interesting role for cell tracking with iron particles since label retention leads to persistent signal void, allowing proliferative status to be determined.
Collapse
|
77
|
Dutta S, Singh G, Sreejith S, Mamidi MK, Husin JM, Datta I, Pal R, Das AK. Cell therapy: the final frontier for treatment of neurological diseases. CNS Neurosci Ther 2013; 19:5-11. [PMID: 23253099 DOI: 10.1111/cns.12027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/21/2012] [Accepted: 09/25/2012] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases are devastating because they cause increasing loss of cognitive and physical functions and affect an estimated 1 billion individuals worldwide. Unfortunately, no drugs are currently available to halt their progression, except a few that are largely inadequate. This mandates the search of new treatments for these progressively degenerative diseases. Neural stem cells (NSCs) have been successfully isolated, propagated, and characterized from the adult brains of mammals, including humans. The confirmation that neurogenesis occurs in the adult brain via NSCs opens up fresh avenues for treating neurological problems. The proof-of-concept studies demonstrating the neural differentiation capacity of stem cells both in vitro and in vivo have raised widespread enthusiasm toward cell-based interventions. It is anticipated that cell-based neurogenic drugs may reverse or compensate for deficits associated with neurological diseases. The increasing interest of the private sector in using human stem cells in therapeutics is evidenced by launching of several collaborative clinical research activities between Pharma giants and research institutions or small start-up companies. In this review, we discuss the major developments that have taken place in this field to position stem cells as a prospective candidate drug for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Susmita Dutta
- Clinical Sciences, International Medical University, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Rosado-de-Castro PH, Pimentel-Coelho PM, da Fonseca LMB, de Freitas GR, Mendez-Otero R. The rise of cell therapy trials for stroke: review of published and registered studies. Stem Cells Dev 2013; 22:2095-111. [PMID: 23509917 DOI: 10.1089/scd.2013.0089] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stroke is the second leading cause of death and the third leading cause of disability worldwide. Approximately 16 million first-ever strokes occur each year, leading to nearly 6 million deaths. Nevertheless, currently, very few therapeutic options are available. Cell therapies have been applied successfully in different hematological diseases, and are currently being investigated for treating ischemic heart disease, with promising results. Recent preclinical studies have indicated that cell therapies may provide structural and functional benefits after stroke. However, the effects of these treatments are not yet fully understood and are the subject of continuing investigation. Meanwhile, different clinical trials for stroke, the majority of them small, nonrandomized, and uncontrolled, have been reported, and their results indicate that cell therapy seems safe and feasible in these conditions. In the last 2 years, the number of published and registered trials has dramatically increased. Here, we review the main findings available in the field, with emphasis on the clinical results. Moreover, we address some of the questions that have been raised to date, to improve future studies.
Collapse
|
79
|
Pabon MM, Borlongan CV. ADVANCES IN THE CELL-BASED TREATMENT OF NEONATAL HYPOXIC-ISCHEMIC BRAIN INJURY. FUTURE NEUROLOGY 2013; 8:193-203. [PMID: 23565051 DOI: 10.2217/fnl.12.85] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stem cell therapy for adult stroke has reached limited clinical trials. Here, we provide translational research guidance on stem cell therapy for neonatal hypoxic-ischemic brain injury requiring a careful consideration of clinically relevant animal models, feasible stem cell sources, and validated safety and efficacy endpoint assays, as well as a general understanding of modes of action of this cellular therapy. To this end, we refer to existing translational guidelines, in particular the recommendations outlined in the consortium of academicians, industry partners and regulators called Stem cell Therapeutics as an Emerging Paradigm for Stroke or STEPS. Although the STEPS guidelines are directed at enhancing the successful outcome of cell therapy in adult stroke, we highlight overlapping pathologies between adult stroke and neonatal hypoxic-ischemic brain injury. We are, however, cognizant that the neonatal hypoxic-ischemic brain injury displays disease symptoms distinct from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Mibel M Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, Florida 33612 USA
| | | |
Collapse
|
80
|
Garcia I, Huang L, Ung K, Arenkiel BR. Tracing synaptic connectivity onto embryonic stem cell-derived neurons. Stem Cells 2013; 30:2140-51. [PMID: 22996827 DOI: 10.1002/stem.1185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Transsynaptic circuit tracing using genetically modified rabies virus (RV) is an emerging technology for identifying synaptic connections between neurons. Complementing this methodology, it is now possible to assay the basic molecular and cellular properties of neuronal lineages derived from embryonic stem cells (ESCs) in vitro, and these properties are under intense investigation toward devising cell replacement therapies. Here, we report the generation of a novel mouse ESC (mESC) line that harbors the genetic elements to allow RV-mediated transsynaptic circuit tracing in ESC-derived neurons and their synaptic networks. To facilitate transsynaptic tracing, we have engineered a new reporter allele by introducing cDNA encoding tdTomato, the Rabies-G glycoprotein, and the avian TVA receptor into the ROSA26 locus by gene targeting. We demonstrate high-efficiency differentiation of these novel mESCs into functional neurons, show their capacity to synaptically connect with primary neuronal cultures as evidenced by immunohistochemistry and electrophysiological recordings, and show their ability to act as source cells for presynaptic tracing of neuronal networks in vitro and in vivo. Together, our data highlight the potential for using genetically engineered stem cells to investigate fundamental mechanisms of synapse and circuit formation with unambiguous identification of presynaptic inputs onto neuronal populations of interest.
Collapse
Affiliation(s)
- Isabella Garcia
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
81
|
Ren H, Chen J, Wang Y, Zhang S, Zhang B. Intracerebral neural stem cell transplantation improved the auditory of mice with presbycusis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:230-41. [PMID: 23330008 PMCID: PMC3544227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 11/27/2012] [Indexed: 06/01/2023]
Abstract
Stem cell-based regenerative therapy is a potential cellular therapeutic strategy for patients with incurable brain diseases. Embryonic neural stem cells (NSCs) represent an attractive cell source in regenerative medicine strategies in the treatment of diseased brains. Here, we assess the capability of intracerebral embryonic NSCs transplantation for C57BL/6J mice with presbycusis in vivo. Morphology analyses revealed that the neuronal rate of apoptosis was lower in the aged group (10 months of age) but not in the young group (2 months of age) after NSCs transplantation, while the electrophysiological data suggest that the Auditory Brain Stem Response (ABR) threshold was significantly decreased in the aged group at 2 weeks and 3 weeks after transplantation. By contrast, there was no difference in the aged group at 4 weeks post-transplantation or in the young group at any time post-transplantation. Furthermore, immunofluorescence experiments showed that NSCs differentiated into neurons that engrafted and migrated to the brain, even to sites of lesions. Together, our results demonstrate that NSCs transplantation improve the auditory of C57BL/6J mice with presbycusis.
Collapse
Affiliation(s)
- Hongmiao Ren
- Department of Otolaryngology Head and Neck Surgery, Institute of Surgery Research & Daping Hospital, Third Military Medical UniversityChongqing 400042, China
- Institute of Otolaryngology of Chongqing, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Jichuan Chen
- Department of Otolaryngology Head and Neck Surgery, Institute of Surgery Research & Daping Hospital, Third Military Medical UniversityChongqing 400042, China
- Institute of Otolaryngology of Chongqing, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Yinan Wang
- Department of Otolaryngology Head and Neck Surgery, Institute of Surgery Research & Daping Hospital, Third Military Medical UniversityChongqing 400042, China
- Institute of Otolaryngology of Chongqing, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Shichang Zhang
- Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| | - Bo Zhang
- Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical UniversityChongqing 400042, China
| |
Collapse
|
82
|
Gan Y, Jing Z, Stetler RA, Cao G. Gene delivery with viral vectors for cerebrovascular diseases. Front Biosci (Elite Ed) 2013; 5:188-203. [PMID: 23276981 PMCID: PMC5516729 DOI: 10.2741/e607] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Recent achievements in the understanding of molecular events involved in the pathogenesis of central nervous system (CNS) injury have made gene transfer a promising approach for various neurological disorders, including cerebrovascular diseases. However, special obstacles, including the post-mitotic nature of neurons and the blood-brain barrier (BBB), constitute key challenges for gene delivery to the CNS. Despite the various limitations in current gene delivery systems, a spectrum of viral vectors has been successfully used to deliver genes to the CNS. Furthermore, recent advancements in vector engineering have improved the safety and delivery of viral vectors. Numerous viral vector-based clinical trials for neurological disorders have been initiated. This review will summarize the current implementation of viral gene delivery in the context of cerebrovascular diseases including ischemic stroke, hemorrhagic stroke and subarachnoid hemorrhage (SAH). In particular, we will discuss the potentially feasible ways in which viral vectors can be manipulated and exploited for use in neural delivery and therapy.
Collapse
Affiliation(s)
- Yu Gan
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Zheng Jing
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - R. Anne Stetler
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Guodong Cao
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, U.S.A
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| |
Collapse
|
83
|
Isolation and purification of self-renewable human neural stem cells for cell therapy in experimental model of ischemic stroke. Methods Mol Biol 2013; 1059:157-67. [PMID: 23934842 DOI: 10.1007/978-1-62703-574-3_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human embryonic stem cells (hESCs) are pluripotent with a strong self-renewable ability making them a virtually unlimited source of neural cells for structural repair in neurological disorders. Currently, hESCs are one of the most promising cell sources amenable for commercialization of off-shelf cell therapy products. However, along with this strong proliferative capacity of hESCs comes the tumorigenic potential of these cells after transplantation. Thus, the isolation and purification of a homogeneous, population of neural stem cells (hNSCs) are of paramount importance to avoid tumor formation in the host brain. This chapter describes the isolation, neuralization, and long-term perpetuation of hNSCs derived from hESCs through use of specific mitogenic growth factors and the preparation of hNSCs for transplantation in an experimental model of stroke. Additionally, we describe methods to analyze the stroke and size of grafts using magnetic resonance imaging and Osirix software, and neuroanatomical tracing procedures to study axonal remodeling after stroke and cell transplantation.
Collapse
|
84
|
Mine Y, Tatarishvili J, Oki K, Monni E, Kokaia Z, Lindvall O. Grafted human neural stem cells enhance several steps of endogenous neurogenesis and improve behavioral recovery after middle cerebral artery occlusion in rats. Neurobiol Dis 2012; 52:191-203. [PMID: 23276704 DOI: 10.1016/j.nbd.2012.12.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/30/2012] [Accepted: 12/18/2012] [Indexed: 12/12/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) in subventricular zone (SVZ) produce new striatal neurons during several months after stroke, which may contribute to recovery. Intracerebral grafts of NSPCs can exert beneficial effects after stroke through neuronal replacement, trophic actions, neuroprotection, and modulation of inflammation. Here we have explored whether human fetal striatum-derived NSPC-grafts influence striatal neurogenesis and promote recovery in stroke-damaged brain. T cell-deficient rats were subjected to 1h middle cerebral artery occlusion (MCAO). Human fetal NSPCs or vehicle were implanted into ipsilateral striatum 48 h after MCAO, animals were assessed behaviorally, and perfused at 6 or 14 weeks. Grafted human NSPCs survived in all rats, and a subpopulation had differentiated to neuroblasts or mature neurons at 6 and 14 weeks. Numbers of proliferating cells in SVZ and new migrating neuroblasts and mature neurons were higher, and numbers of activated microglia/macrophages were lower in the ischemic striatum of NSPC-grafted compared to vehicle-injected group both at 6 and 14 weeks. A fraction of grafted NSPCs projected axons from striatum to globus pallidus. The NSPC-grafted rats showed improved functional recovery in stepping and cylinder tests from 6 and 12 weeks, respectively. Our data show, for the first time, that intrastriatal implants of human fetal NSPCs exert a long-term enhancement of several steps of striatal neurogensis after stroke. The grafts also suppress striatal inflammation and ameliorate neurological deficits. Our findings support the idea that combination of NSPC transplantation and stimulation of neurogenesis from endogenous NSPCs may become a valuable strategy for functional restoration after stroke.
Collapse
Affiliation(s)
- Yutaka Mine
- Laboratory of Stem Cells and Restorative Neurology, University Hospital, SE-221 84 Lund, Sweden
| | | | | | | | | | | |
Collapse
|
85
|
Manley NC, Steinberg GK. Tracking stem cells for cellular therapy in stroke. Curr Pharm Des 2012; 18:3685-93. [PMID: 22571604 DOI: 10.2174/138161212802002643] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 03/06/2012] [Indexed: 01/06/2023]
Abstract
Stem cell transplantation has emerged as a promising treatment strategy for stroke. The development of effective ways to monitor transplanted stem cells is essential to understand how stem cell transplantation enhances stroke recovery and ultimately will be an indispensable tool for advancing stem cell therapy to the clinic. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including optical imaging, magnetic resonance imaging (MRI), and positron emission tomography (PET), with emphasis on the benefits and drawbacks of each imaging approach. Key considerations such as the potential impact of each tracking system on stem cell function, as well as its relative applicability to humans are discussed. Finally, we describe multi-modal imaging strategies as a more comprehensive method to track transplanted stem cells in the stroke-injured brain.
Collapse
Affiliation(s)
- Nathan C Manley
- Department of Neurosurgery, Stanford Stroke Center and Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University School of Medicine, 300 Pasteur Drive Stanford, California, CA 94305-5327, USA
| | | |
Collapse
|
86
|
Garcia I, Kim C, Arenkiel BR. Genetic strategies to investigate neuronal circuit properties using stem cell-derived neurons. Front Cell Neurosci 2012; 6:59. [PMID: 23264761 PMCID: PMC3524522 DOI: 10.3389/fncel.2012.00059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 11/30/2012] [Indexed: 01/28/2023] Open
Abstract
The mammalian brain is anatomically and functionally complex, and prone to diverse forms of injury and neuropathology. Scientists have long strived to develop cell replacement therapies to repair damaged and diseased nervous tissue. However, this goal has remained unrealized for various reasons, including nascent knowledge of neuronal development, the inability to track and manipulate transplanted cells within complex neuronal networks, and host graft rejection. Recent advances in embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) technology, alongside novel genetic strategies to mark and manipulate stem cell-derived neurons, now provide unprecedented opportunities to investigate complex neuronal circuits in both healthy and diseased brains. Here, we review current technologies aimed at generating and manipulating neurons derived from ESCs and iPSCs toward investigation and manipulation of complex neuronal circuits, ultimately leading to the design and development of novel cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Isabella Garcia
- Program in Developmental Biology, Baylor College of Medicine Houston, TX, USA ; Medical Scientist Training Program, Baylor College of Medicine Houston, TX, USA
| | | | | |
Collapse
|
87
|
Oki K, Tatarishvili J, Wood J, Koch P, Wattananit S, Mine Y, Monni E, Tornero D, Ahlenius H, Ladewig J, Brüstle O, Lindvall O, Kokaia Z. Human-induced pluripotent stem cells form functional neurons and improve recovery after grafting in stroke-damaged brain. Stem Cells 2012; 30:1120-33. [PMID: 22495829 DOI: 10.1002/stem.1104] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reprogramming of adult human somatic cells to induced pluripotent stem cells (iPSCs) is a novel approach to produce patient-specific cells for autologous transplantation. Whether such cells survive long-term, differentiate to functional neurons, and induce recovery in the stroke-injured brain are unclear. We have transplanted long-term self-renewing neuroepithelial-like stem cells, generated from adult human fibroblast-derived iPSCs, into the stroke-damaged mouse and rat striatum or cortex. Recovery of forepaw movements was observed already at 1 week after transplantation. Improvement was most likely not due to neuronal replacement but was associated with increased vascular endothelial growth factor levels, probably enhancing endogenous plasticity. Transplanted cells stopped proliferating, could survive without forming tumors for at least 4 months, and differentiated to morphologically mature neurons of different subtypes. Neurons in intrastriatal grafts sent axonal projections to the globus pallidus. Grafted cells exhibited electrophysiological properties of mature neurons and received synaptic input from host neurons. Our study provides the first evidence that transplantation of human iPSC-derived cells is a safe and efficient approach to promote recovery after stroke and can be used to supply the injured brain with new neurons for replacement.
Collapse
Affiliation(s)
- Koichi Oki
- Laboratory of Neural Stem Cell Biology and Therapy, University Hospital, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Paspala SA, Vishwakarma SK, Murthy TV, Rao TN, Khan AA. Potential role of stem cells in severe spinal cord injury: current perspectives and clinical data. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2012; 5:15-27. [PMID: 24198535 PMCID: PMC3781762 DOI: 10.2147/sccaa.s28477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem cell transplantation for spinal cord injury (SCI) along with new pharmacotherapy research offers the potential to restore function and ease the associated social and economic burden in the years ahead. Various sources of stem cells have been used in the treatment of SCI, but the most convincing results have been obtained with neural progenitor cells in preclinical models. Although the use of cell-based transplantation strategies for the repair of chronic SCI remains the long sought after holy grail, these approaches have been to date the most successful when applied in the subacute phase of injury. Application of cell-based strategies for the repair and regeneration of the chronically injured spinal cord will require a combinational strategy that may need to include approaches to overcome the effects of the glial scar, inhibitory molecules, and use of tissue engineering strategies to bridge the lesion. Nonetheless, cell transplantation strategies are promising, and it is anticipated that the Phase I clinical trials of some form of neural stem cell-based approach in SCI will commence very soon.
Collapse
Affiliation(s)
- Syed Ab Paspala
- PAN Research Foundation, CARE, Hyderabad, India ; The Institute of Medical Sciences, Hyderabad, India
| | | | | | | | | |
Collapse
|
89
|
Walter J, Hausmann S, Drepper T, Puls M, Eggert T, Dihné M. Flavin mononucleotide-based fluorescent proteins function in mammalian cells without oxygen requirement. PLoS One 2012; 7:e43921. [PMID: 22984451 PMCID: PMC3439463 DOI: 10.1371/journal.pone.0043921] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/27/2012] [Indexed: 11/23/2022] Open
Abstract
Usage of the enhanced green fluorescent protein (eGFP) in living mammalian cells is limited to aerobic conditions due to requirement of oxygen during chromophore formation. Since many diseases or disease models are associated with acute or chronic hypoxia, eGFP-labeling of structures of interest in experimental studies might be unreliable leading to biased results. Thus, a chromophore yielding a stable fluorescence under hypoxic conditions is desirable. The fluorescence of flavin mononucleotide (FMN)-based fluorescent proteins (FbFPs) does not require molecular oxygen. Recently, the advantages of FbFPs for several bacterial strains and yeasts were described, specifically, their usage as a real time fluorescence marker in bacterial expression studies and their ability of chromophore formation under anaerobic conditions. Our objective was to verify if FbFPs also function in mammalian cells in order to potentially broaden the repertoire of chromophores with ones that can reliably be used in mammalian studies under hypoxic conditions. In the present study, we demonstrate for the first time, that FbFPs can be expressed in different mammalian cells, among them murine neural stem cells during proliferative and differentiated stages. Fluorescence intensities were comparable to eGFP. In contrast to eGFP, the FbFP fluorescence did not decrease when cells were exposed to defined hypoxic conditions neither in proliferating nor in differentiated cells. Thus, FbFPs can be regarded as an alternative to eGFP in studies that target cellular structures which are exposed to hypoxic conditions.
Collapse
Affiliation(s)
- Janine Walter
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Baden-Württemberg, Germany.
| | | | | | | | | | | |
Collapse
|
90
|
Polentes J, Jendelova P, Cailleret M, Braun H, Romanyuk N, Tropel P, Brenot M, Itier V, Seminatore C, Baldauf K, Turnovcova K, Jirak D, Teletin M, Côme J, Tournois J, Reymann K, Sykova E, Viville S, Onteniente B. Human induced pluripotent stem cells improve stroke outcome and reduce secondary degeneration in the recipient brain. Cell Transplant 2012; 21:2587-602. [PMID: 22889472 DOI: 10.3727/096368912x653228] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are a most appealing source for cell replacement therapy in acute brain lesions. We evaluated the potential of hiPSC therapy in stroke by transplanting hiPSC-derived neural progenitor cells (NPCs) into the postischemic striatum. Grafts received host tyrosine hydroxylase-positive afferents and contained developing interneurons and homotopic GABAergic medium spiny neurons that, with time, sent axons to the host substantia nigra. Grafting reversed stroke-induced somatosensory and motor deficits. Grafting also protected the host substantia nigra from the atrophy that follows disruption of reciprocal striatonigral connections. Graft innervation by tyrosine hydoxylase fibers, substantia nigra protection, and somatosensory functional recovery were early events, temporally dissociated from the slow maturation of GABAergic neurons in the grafts and innervation of substantia nigra. This suggests that grafted hiPSC-NPCs initially exert trophic effects on host brain structures, which precede integration and potential pathway reconstruction. We believe that transplantation of NPCs derived from hiPSCs can provide useful interventions to limit the functional consequences of stroke through both neuroprotective effects and reconstruction of impaired pathways.
Collapse
|
91
|
Abe K, Yamashita T, Takizawa S, Kuroda S, Kinouchi H, Kawahara N. Stem cell therapy for cerebral ischemia: from basic science to clinical applications. J Cereb Blood Flow Metab 2012; 32:1317-31. [PMID: 22252239 PMCID: PMC3390814 DOI: 10.1038/jcbfm.2011.187] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent stem cell technology provides a strong therapeutic potential not only for acute ischemic stroke but also for chronic progressive neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis with neuroregenerative neural cell replenishment and replacement. In addition to resident neural stem cell activation in the brain by neurotrophic factors, bone marrow stem cells (BMSCs) can be mobilized by granulocyte-colony stimulating factor for homing into the brain for both neurorepair and neuroregeneration in acute stroke and neurodegenerative diseases in both basic science and clinical settings. Exogenous stem cell transplantation is also emerging into a clinical scene from bench side experiments. Early clinical trials of intravenous transplantation of autologous BMSCs are showing safe and effective results in stroke patients. Further basic sciences of stem cell therapy on a neurovascular unit and neuroregeneration, and further clinical advancements on scaffold technology for supporting stem cells and stem cell tracking technology such as magnetic resonance imaging, single photon emission tomography or optical imaging with near-infrared could allow stem cell therapy to be applied in daily clinical applications in the near future.
Collapse
Affiliation(s)
- Koji Abe
- Department of Neurology, Okayama University Medical School, Okayama, Japan.
| | | | | | | | | | | |
Collapse
|
92
|
Reekmans K, Praet J, Daans J, Reumers V, Pauwels P, Van der Linden A, Berneman ZN, Ponsaerts P. Current challenges for the advancement of neural stem cell biology and transplantation research. Stem Cell Rev Rep 2012; 8:262-78. [PMID: 21537994 DOI: 10.1007/s12015-011-9266-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transplantation of neural stem cells (NSC) is hoped to become a promising primary or secondary therapy for the treatment of various neurodegenerative disorders of the central nervous system (CNS), as demonstrated by multiple pre-clinical animal studies in which functional recovery has already been demonstrated. However, for NSC therapy to be successful, the first challenge will be to define a transplantable cell population. In the first part of this review, we will briefly discuss the main features of ex vivo culture and characterisation of NSC. Next, NSC grafting itself may not only result in the regeneration of lost tissue, but more importantly has the potential to improve functional outcome through many bystander mechanisms. In the second part of this review, we will briefly discuss several pre-clinical studies that contributed to a better understanding of the therapeutic potential of NSC grafts in vivo. However, while many pre-clinical animal studies mainly report on the clinical benefit of NSC grafting, little is known about the actual in vivo fate of grafted NSC. Therefore, the third part of this review will focus on non-invasive imaging techniques for monitoring cellular grafts in the brain under in vivo conditions. Finally, as NSC transplantation research has evolved during the past decade, it has become clear that the host micro-environment itself, either in healthy or injured condition, is an important player in defining success of NSC grafting. The final part of this review will focus on the host environmental influence on survival, migration and differentiation of grafted NSC.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Kapucu FE, Tanskanen JMA, Mikkonen JE, Ylä-Outinen L, Narkilahti S, Hyttinen JAK. Burst analysis tool for developing neuronal networks exhibiting highly varying action potential dynamics. Front Comput Neurosci 2012; 6:38. [PMID: 22723778 PMCID: PMC3378047 DOI: 10.3389/fncom.2012.00038] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 05/30/2012] [Indexed: 01/15/2023] Open
Abstract
In this paper we propose a firing statistics based neuronal network burst detection algorithm for neuronal networks exhibiting highly variable action potential dynamics. Electrical activity of neuronal networks is generally analyzed by the occurrences of spikes and bursts both in time and space. Commonly accepted analysis tools employ burst detection algorithms based on predefined criteria. However, maturing neuronal networks, such as those originating from human embryonic stem cells (hESCs), exhibit highly variable network structure and time-varying dynamics. To explore the developing burst/spike activities of such networks, we propose a burst detection algorithm which utilizes the firing statistics based on interspike interval (ISI) histograms. Moreover, the algorithm calculates ISI thresholds for burst spikes as well as for pre-burst spikes and burst tails by evaluating the cumulative moving average (CMA) and skewness of the ISI histogram. Because of the adaptive nature of the proposed algorithm, its analysis power is not limited by the type of neuronal cell network at hand. We demonstrate the functionality of our algorithm with two different types of microelectrode array (MEA) data recorded from spontaneously active hESC-derived neuronal cell networks. The same data was also analyzed by two commonly employed burst detection algorithms and the differences in burst detection results are illustrated. The results demonstrate that our method is both adaptive to the firing statistics of the network and yields successful burst detection from the data. In conclusion, the proposed method is a potential tool for analyzing of hESC-derived neuronal cell networks and thus can be utilized in studies aiming to understand the development and functioning of human neuronal networks and as an analysis tool for in vitro drug screening and neurotoxicity assays.
Collapse
Affiliation(s)
- Fikret E Kapucu
- Department of Biomedical Engineering, Tampere University of Technology Tampere, Finland
| | | | | | | | | | | |
Collapse
|
94
|
De Vocht N, Reekmans K, Bergwerf I, Praet J, Hoornaert C, Le Blon D, Daans J, Berneman Z, Van der Linden A, Ponsaerts P. Multimodal imaging of stem cell implantation in the central nervous system of mice. J Vis Exp 2012:e3906. [PMID: 22733218 DOI: 10.3791/3906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the past decade, stem cell transplantation has gained increasing interest as primary or secondary therapeutic modality for a variety of diseases, both in preclinical and clinical studies. However, to date results regarding functional outcome and/or tissue regeneration following stem cell transplantation are quite diverse. Generally, a clinical benefit is observed without profound understanding of the underlying mechanism(s). Therefore, multiple efforts have led to the development of different molecular imaging modalities to monitor stem cell grafting with the ultimate aim to accurately evaluate survival, fate and physiology of grafted stem cells and/or their micro-environment. Changes observed in one or more parameters determined by molecular imaging might be related to the observed clinical effect. In this context, our studies focus on the combined use of bioluminescence imaging (BLI), magnetic resonance imaging (MRI) and histological analysis to evaluate stem cell grafting. BLI is commonly used to non-invasively perform cell tracking and monitor cell survival in time following transplantation, based on a biochemical reaction where cells expressing the Luciferase-reporter gene are able to emit light following interaction with its substrate (e.g. D-luciferin). MRI on the other hand is a non-invasive technique which is clinically applicable and can be used to precisely locate cellular grafts with very high resolution, although its sensitivity highly depends on the contrast generated after cell labeling with an MRI contrast agent. Finally, post-mortem histological analysis is the method of choice to validate research results obtained with non-invasive techniques with highest resolution and sensitivity. Moreover end-point histological analysis allows us to perform detailed phenotypic analysis of grafted cells and/or the surrounding tissue, based on the use of fluorescent reporter proteins and/or direct cell labeling with specific antibodies. In summary, we here visually demonstrate the complementarities of BLI, MRI and histology to unravel different stem cell- and/or environment-associated characteristics following stem cell grafting in the CNS of mice. As an example, bone marrow-derived stromal cells, genetically engineered to express the enhanced Green Fluorescent Protein (eGFP) and firefly Luciferase (fLuc), and labeled with blue fluorescent micron-sized iron oxide particles (MPIOs), will be grafted in the CNS of immune-competent mice and outcome will be monitored by BLI, MRI and histology (Figure 1).
Collapse
|
95
|
|
96
|
Gorelik M, Janowski M, Galpoththawela C, Rifkin R, Levy M, Lukomska B, Kerr DA, Bulte JWM, Walczak P. Noninvasive monitoring of immunosuppressive drug efficacy to prevent rejection of intracerebral glial precursor allografts. Cell Transplant 2012; 21:2149-57. [PMID: 22508097 DOI: 10.3727/096368912x636911] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The development of cell-based therapies opens up new avenues for treating a myriad of diseases of the central nervous system (CNS). While significant effort is being directed toward development of patient-specific, autologous transplantable cells, at present, the majority of cell transplantation studies performed clinically utilize allografts. In this context, the issue of graft rejection and immunoprotection is of key importance. In this study, we transplanted mouse glial-restricted progenitors into immunodeficient, immunocompetent, and immunosuppressed mice and monitored their survival noninvasively using bioluminescence imaging (BLI). With the use of serial BLI, we evaluated both the prevalence and dynamics of cell rejection. We demonstrate that allografts in immunocompetent mice were rejected at a rate of 69.2% (n = 13) indicating that graft tolerance is possible even without immunosuppression. Immunosuppression using a combination of rapamycin and FK506 or cyclosporin failed to fully protect the grafts. FK506 and rapamycin treatment resulted in a slight improvement of immunoprotection (22.2% rejected, n = 9) compared to cyclosporin A (55.6% rejected, n = 9); however, the difference was not significant. Notably, immunohistochemistry revealed leukocytes infiltrating the graft area in both rejecting and nonrejecting immunocompetent animals, but not in immunodeficient animals. The induction of an inflammatory process, even in surviving allografts, has implications for their long-term survival and functionality.
Collapse
Affiliation(s)
- Michael Gorelik
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Praet J, Reekmans K, Lin D, De Vocht N, Bergwerf I, Tambuyzer B, Daans J, Hens N, Goossens H, Pauwels P, Berneman Z, Van der Linden A, Ponsaerts P. Cell type-associated differences in migration, survival, and immunogenicity following grafting in CNS tissue. Cell Transplant 2012; 21:1867-81. [PMID: 22472278 DOI: 10.3727/096368912x636920] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell transplantation has been suggested to display several neuroprotective and/or neuroregenerative effects in animal models of central nervous system (CNS) trauma. However, while most studies report on clinical observations, currently little is known regarding the actual fate of the cell populations grafted and whether or how the brain's innate immune system, mainly directed by activated microglia and astrocytes, interacts with autologous cellular implants. In this study, we grafted well-characterized neural stem cell, mouse embryonic fibroblast, dendritic cell, bone marrow mononuclear cell, and splenocyte populations, all isolated or cultured from C57BL/6-eGFP transgenic mice, below the capsula externa (CE) of healthy C57BL/6 mice and below the inflamed/demyelinated CE of cuprizone-treated C57BL/6 mice. Two weeks postgrafting, an extensive quantitative multicolor histological analysis was performed in order (i) to quantify cell graft localization, migration, survival, and toxicity and (ii) to characterize endogenous CNS immune responses against the different cell grafts. Obtained results indicate dependence on the cell type grafted: (i) a different degree of cell graft migration, survival, and toxicity and (ii) a different organization of the endogenous immune response. Based on these observations, we warrant that further research should be undertaken to understand-and eventually control-cell graft-induced tissue damage and activation of the brain's innate immune system. The latter will be inevitable before cell grafting in the CNS can be performed safely and successfully in clinical settings.
Collapse
Affiliation(s)
- Jelle Praet
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
De Vocht N, Bergwerf I, Vanhoutte G, Daans J, De Visscher G, Chatterjee S, Pauwels P, Berneman Z, Ponsaerts P, Van der Linden A. Labeling of Luciferase/eGFP-expressing bone marrow-derived stromal cells with fluorescent micron-sized iron oxide particles improves quantitative and qualitative multimodal imaging of cellular grafts in vivo. Mol Imaging Biol 2012; 13:1133-45. [PMID: 21246293 DOI: 10.1007/s11307-011-0469-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Development of multimodal imaging strategies is currently of utmost importance for the validation of preclinical stem cell therapy studies. PROCEDURES We performed a combined labeling strategy for bone marrow-derived stromal cells (BMSC) based on genetic modification with the reporter genes Luciferase and eGFP (BMSC-Luc/eGFP) and physical labeling with blue fluorescent micron-sized iron oxide particles (MPIO) in order to unambiguously identify BMSC localization, survival, and differentiation following engraftment in the central nervous system of mice by in vivo bioluminescence (BLI) and magnetic resonance imaging and postmortem histological analysis. RESULTS Using this combination, a significant increase of in vivo BLI signal was observed for MPIO-labeled BMSC-Luc/eGFP. Moreover, MPIO labeling of BMSC-Luc/eGFP allows for the improved identification of implanted cells within host tissue during histological observation. CONCLUSIONS This study describes an optimized labeling strategy for multimodal stem cell imaging resulting in improved quantitative and qualitative detection of cellular grafts.
Collapse
|
99
|
Leung L. Cellular therapies for treating pain associated with spinal cord injury. J Transl Med 2012; 10:37. [PMID: 22394650 PMCID: PMC3320547 DOI: 10.1186/1479-5876-10-37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 03/06/2012] [Indexed: 12/21/2022] Open
Abstract
Spinal cord injury leads to immense disability and loss of quality of life in human with no satisfactory clinical cure. Cell-based or cell-related therapies have emerged as promising therapeutic potentials both in regeneration of spinal cord and mitigation of neuropathic pain due to spinal cord injury. This article reviews the various options and their latest developments with an update on their therapeutic potentials and clinical trialing.
Collapse
Affiliation(s)
- Lawrence Leung
- Centre of Neurosciences Study, Queen's University, 18 Stuart Street, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
100
|
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
|