151
|
Srikanth P, Young-Pearse TL. Stem cells on the brain: modeling neurodevelopmental and neurodegenerative diseases using human induced pluripotent stem cells. J Neurogenet 2014; 28:5-29. [PMID: 24628482 PMCID: PMC4285381 DOI: 10.3109/01677063.2014.881358] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seven years have passed since the initial report of the generation of induced pluripotent stem cells (iPSCs) from adult human somatic cells, and in the intervening time the field of neuroscience has developed numerous disease models using this technology. Here, we review progress in the field and describe both the advantages and potential pitfalls of modeling neurodegenerative and neurodevelopmental diseases using this technology. We include tables with information on neural differentiation protocols and studies that developed human iPSC lines to model neurological diseases. We also discuss how one can: investigate effects of genetic mutations with iPSCs, examine cell fate-specific phenotypes, best determine the specificity of a phenotype, and bring in vivo relevance to this in vitro technique.
Collapse
Affiliation(s)
- Priya Srikanth
- Center for Neurologic Diseases, Brigham and Women's Hospital , Boston, Massachusetts , USA
| | | |
Collapse
|
152
|
Tabar V, Studer L. Pluripotent stem cells in regenerative medicine: challenges and recent progress. Nat Rev Genet 2014; 15:82-92. [PMID: 24434846 PMCID: PMC4539940 DOI: 10.1038/nrg3563] [Citation(s) in RCA: 324] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
After years of incremental progress, several recent studies have succeeded in deriving disease-relevant cell types from human pluripotent stem cell (hPSC) sources. The prospect of an unlimited cell source, combined with promising preclinical data, indicates that hPSC technology may be on the verge of clinical translation. In this Review, we discuss recent progress in directed differentiation, some of the new technologies that have facilitated the success of hPSC therapies and the remaining hurdles on the road towards developing hPSC-based cell therapies.
Collapse
Affiliation(s)
- Viviane Tabar
- Center for Stem Cell Biology and Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York 10065, USA
| | - Lorenz Studer
- Center for Stem Cell Biology and Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York 10065, USA
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York 10065, USA
| |
Collapse
|
153
|
Li Y, Liu M, Yan Y, Yang ST. Neural differentiation from pluripotent stem cells: The role of natural and synthetic extracellular matrix. World J Stem Cells 2014; 6:11-23. [PMID: 24567784 PMCID: PMC3927010 DOI: 10.4252/wjsc.v6.i1.11] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/23/2013] [Accepted: 11/02/2013] [Indexed: 02/06/2023] Open
Abstract
Neural cells differentiated from pluripotent stem cells (PSCs), including both embryonic stem cells and induced pluripotent stem cells, provide a powerful tool for drug screening, disease modeling and regenerative medicine. High-purity oligodendrocyte progenitor cells (OPCs) and neural progenitor cells (NPCs) have been derived from PSCs recently due to the advancements in understanding the developmental signaling pathways. Extracellular matrices (ECM) have been shown to play important roles in regulating the survival, proliferation, and differentiation of neural cells. To improve the function and maturation of the derived neural cells from PSCs, understanding the effects of ECM over the course of neural differentiation of PSCs is critical. During neural differentiation of PSCs, the cells are sensitive to the properties of natural or synthetic ECMs, including biochemical composition, biomechanical properties, and structural/topographical features. This review summarizes recent advances in neural differentiation of human PSCs into OPCs and NPCs, focusing on the role of ECM in modulating the composition and function of the differentiated cells. Especially, the importance of using three-dimensional ECM scaffolds to simulate the in vivo microenvironment for neural differentiation of PSCs is highlighted. Future perspectives including the immediate applications of PSC-derived neural cells in drug screening and disease modeling are also discussed.
Collapse
Affiliation(s)
- Yan Li
- Yan Li, Yuanwei Yan, Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, United States
| | - Meimei Liu
- Yan Li, Yuanwei Yan, Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, United States
| | - Yuanwei Yan
- Yan Li, Yuanwei Yan, Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, United States
| | - Shang-Tian Yang
- Yan Li, Yuanwei Yan, Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, United States
| |
Collapse
|
154
|
Sinha S, Iyer D, Granata A. Embryonic origins of human vascular smooth muscle cells: implications for in vitro modeling and clinical application. Cell Mol Life Sci 2014; 71:2271-88. [PMID: 24442477 PMCID: PMC4031394 DOI: 10.1007/s00018-013-1554-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/03/2013] [Accepted: 12/30/2013] [Indexed: 01/06/2023]
Abstract
Vascular smooth muscle cells (SMCs) arise from multiple origins during development, raising the possibility that differences in embryological origins between SMCs could contribute to site-specific localization of vascular diseases. In this review, we first examine the developmental pathways and embryological origins of vascular SMCs and then discuss in vitro strategies for deriving SMCs from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We then review in detail the potential for vascular disease modeling using iPSC-derived SMCs and consider the pathological implications of heterogeneous embryonic origins. Finally, we touch upon the role of human ESC-derived SMCs in therapeutic revascularization and the challenges remaining before regenerative medicine using ESC- or iPSC-derived cells comes of age.
Collapse
Affiliation(s)
- Sanjay Sinha
- Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK,
| | | | | |
Collapse
|
155
|
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
|
156
|
Tam RY, Fuehrmann T, Mitrousis N, Shoichet MS. Regenerative therapies for central nervous system diseases: a biomaterials approach. Neuropsychopharmacology 2014; 39:169-88. [PMID: 24002187 PMCID: PMC3857664 DOI: 10.1038/npp.2013.237] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 02/07/2023]
Abstract
The central nervous system (CNS) has a limited capacity to spontaneously regenerate following traumatic injury or disease, requiring innovative strategies to promote tissue and functional repair. Tissue regeneration strategies, such as cell and/or drug delivery, have demonstrated promising results in experimental animal models, but have been difficult to translate clinically. The efficacy of cell therapy, which involves stem cell transplantation into the CNS to replace damaged tissue, has been limited due to low cell survival and integration upon transplantation, while delivery of therapeutic molecules to the CNS using conventional methods, such as oral and intravenous administration, have been limited by diffusion across the blood-brain/spinal cord-barrier. The use of biomaterials to promote graft survival and integration as well as localized and sustained delivery of biologics to CNS injury sites is actively being pursued. This review will highlight recent advances using biomaterials as cell- and drug-delivery vehicles for CNS repair.
Collapse
Affiliation(s)
- Roger Y Tam
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Tobias Fuehrmann
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Nikolaos Mitrousis
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada,Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada,Department of Chemistry, University of Toronto, Toronto, ON, Canada,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Centre for Cellular and Biomolecular Research, 160 College Street, Room 514, Toronto, ON, Canada, Tel: +416 978 1460, Fax: +416 978 4317, E-mail:
| |
Collapse
|
157
|
The role of mesenchymal stromal cells in spinal cord injury, regenerative medicine and possible clinical applications. Biochimie 2013; 95:2257-70. [DOI: 10.1016/j.biochi.2013.08.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 08/05/2013] [Indexed: 12/13/2022]
|
158
|
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol 2013; 114:25-57. [PMID: 24269804 DOI: 10.1016/j.pneurobio.2013.11.002] [Citation(s) in RCA: 531] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that affects thousands of individuals each year. Over the past decades an enormous progress has been made in our understanding of the molecular and cellular events generated by SCI, providing insights into crucial mechanisms that contribute to tissue damage and regenerative failure of injured neurons. Current treatment options for SCI include the use of high dose methylprednisolone, surgical interventions to stabilize and decompress the spinal cord, and rehabilitative care. Nonetheless, SCI is still a harmful condition for which there is yet no cure. Cellular, molecular, rehabilitative training and combinatorial therapies have shown promising results in animal models. Nevertheless, work remains to be done to ascertain whether any of these therapies can safely improve patient's condition after human SCI. This review provides an extensive overview of SCI research, as well as its clinical component. It starts covering areas from physiology and anatomy of the spinal cord, neuropathology of the SCI, current clinical options, neuronal plasticity after SCI, animal models and techniques to assess recovery, focusing the subsequent discussion on a variety of promising neuroprotective, cell-based and combinatorial therapeutic approaches that have recently moved, or are close, to clinical testing.
Collapse
Affiliation(s)
- Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
159
|
Stacpoole S, Spitzer S, Bilican B, Compston A, Karadottir R, Chandran S, Franklin R. High yields of oligodendrocyte lineage cells from human embryonic stem cells at physiological oxygen tensions for evaluation of translational biology. Stem Cell Reports 2013; 1:437-50. [PMID: 24286031 PMCID: PMC3841262 DOI: 10.1016/j.stemcr.2013.09.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 02/09/2023] Open
Abstract
We have established and efficient system to specify NG2/PDGF-Rα/OLIG2+ oligodendrocyte precursor cells (OPCs) from human embryonic stem cells (hESCs) at low, physiological (3%) oxygen levels. This was achieved via both forebrain and spinal cord origins, with up to 98% of cells expressing NG2. Developmental insights reveal a critical role for fibroblast growth factor 2 (FGF-2) in OLIG2 induction via ventral forebrain pathways. The OPCs mature in vitro to express O4 (46%) and subsequently become galactocerebroside (GALC), O1, and myelin basic protein-positive (MBP+) multibranching oligodendrocytes. These were cultured alongside hESC-derived neurons. The electrophysiological properties of human OPCs are similar to those of rat OPCs, with large voltage-gated sodium currents and the ability to fire action potentials. Exposure to a selective retinoid X receptor agonist increased the proportion of O4+ oligodendrocytes that express MBP from 5% to 30%. Thus, we have established a developmentally engineered system to investigate the biological properties of human OPCs and test the effects of putative remyelinating agents prior to clinical application.
Collapse
Affiliation(s)
- Sybil R.L. Stacpoole
- Department of Clinical Neurosciences, Cambridge University, Cambridge CB2 0PY, UK
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | - Sonia Spitzer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | - Bilada Bilican
- Centre for Neuroregeneration, Edinburgh University, Edinburgh EH16 4SB, UK
| | - Alastair Compston
- Department of Clinical Neurosciences, Cambridge University, Cambridge CB2 0PY, UK
| | - Ragnhildur Karadottir
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| | | | - Robin J.M. Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, Cambridge University, Cambridge CB3 0ES, UK
| |
Collapse
|
160
|
Alsanie WF, Niclis JC, Petratos S. Human embryonic stem cell-derived oligodendrocytes: protocols and perspectives. Stem Cells Dev 2013; 22:2459-76. [PMID: 23621561 PMCID: PMC3760471 DOI: 10.1089/scd.2012.0520] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 04/26/2013] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocytes play a fundamental supportive role in the mammalian central nervous system (CNS) as the myelinating-glial cells. Disruption of fast axonal transport mechanisms can occur as a consequence of mature oligodendrocyte loss following spinal cord injury, stroke, or due to neuroinflammatory conditions, such as multiple sclerosis. As a result of the limited remyelination ability in the CNS after injury or disease, human embryonic stem cells (hESCs) may prove to be a promising option for the generation and replacement of mature oligodendrocytes. Moreover, hESC-derived oligodendrocytes may be experimentally utilized to unravel fundamental questions of oligodendrocyte development, along with their therapeutic potential through growth factor support of axons and neurons. However, an intensive characterization and examination of hESC-derived oligodendrocytes prior to preclinical or clinical trials is required to facilitate greater success in their integration following cellular replacement therapy (CRT). Currently, the protocols utilized to derive oligodendrocytes from hESCs consist of significant variations in culture style, time-length of differentiation, and the provision of growth factors in culture. Further, these differing protocols also report disparate patterns in the expression of oligodendroglial markers by these derived oligodendrocytes, throughout their differentiation in culture. We have comprehensively reviewed the published protocols describing the derivation of oligodendrocytes from hESCs and the studies that examine their efficacy to remyelinate, along with the fundamental issues of their safety as a viable CRT. Additionally, this review will highlight particular issues of concern and suggestions for troubleshooting to provide investigators critical information for the future improvement of establishing in vitro hESC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Walaa F Alsanie
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia.
| | | | | |
Collapse
|
161
|
Asmani MN, Ai J, Amoabediny G, Noroozi A, Azami M, Ebrahimi-Barough S, Navaei-Nigjeh M, Ai A, Jafarabadi M. Three-dimensional culture of differentiated endometrial stromal cells to oligodendrocyte progenitor cells (OPCs) in fibrin hydrogel. Cell Biol Int 2013; 37:1340-9. [PMID: 24038753 DOI: 10.1002/cbin.10171] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 07/22/2012] [Indexed: 01/08/2023]
Abstract
Neural tissue engineering is one of the most promising strategies for treatment of nerve tissue injuries. Three-dimensional (3D) environment mimics in vivo conditions for cells. 3D distribution and growth of the cells within the scaffold are both important for neural tissue engineering. In this study, endometrial stromal cell-derived oligodendrocyte progenitor cells (EnSC-derived OPCs) were cultured in fibrin gel and cell differentiation and viability were evaluated after 8 days of post-culture. The structural and mechanical characteristics of fibrin gel-like scaffold were examined with rheological analysis. EnSCs were isolated from donor tissue and were induced to OPCs with growth factors (FGF2/EGF/PDGF-AA) for 12 days, then were cultured in fibrin gel with Triiodothyronine (T3) medium for another 8 days. The viability of cells was analyzed using MTT assay for a period of 8 days culturing in a fibrin matrix. Structure of fibrin matrix and cell morphology was analyzed with SEM. TEM, immunostaining and quantitative RT-PCR was performed for OPCs markers after cell culturing in fibrin matrix. Cell viability is enhanced in fibrin matrix after 8 days. SEM and TEM show that cells are in good integration with nano-fibers. Moreover, immunohistochemistry and quantitative RT-PCR of OPCs differentiation markers showed that Olig2, Sox10, PDGFRa, CNP, and A2B5 are expressed after 8 days culturing within fibrin matrix. Fibrin can provide a suitable 3-D scaffold for EnSCs differentiated cells for the regeneration of CNS.
Collapse
Affiliation(s)
- Mohammad Nabi Asmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for New Technologies in Life Science Engineering, University of Tehran, Tehran, Iran; Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Review of transplantation of neural stem/progenitor cells for spinal cord injury. Int J Dev Neurosci 2013; 31:701-13. [DOI: 10.1016/j.ijdevneu.2013.07.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 07/02/2013] [Accepted: 07/26/2013] [Indexed: 11/17/2022] Open
|
163
|
Bang WS, Kim KT, Cho DC, Kim HJ, Sung JK. Valproic Acid increases expression of neuronal stem/progenitor cell in spinal cord injury. J Korean Neurosurg Soc 2013; 54:8-13. [PMID: 24044073 PMCID: PMC3772294 DOI: 10.3340/jkns.2013.54.1.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/22/2013] [Accepted: 07/17/2013] [Indexed: 02/06/2023] Open
Abstract
Objective This study investigates the effect of valproic acid (VPA) on expression of neural stem/progenitor cells (NSPCs) in a rat spinal cord injury (SCI) model. Methods Adult male rats (n=24) were randomly and blindly allocated into three groups. Laminectomy at T9 was performed in all three groups. In group 1 (sham), only laminectomy was performed. In group 2 (SCI-VPA), the animals received a dose of 200 mg/kg of VPA. In group 3 (SCI-saline), animals received 1.0 mL of the saline vehicle solution. A modified aneurysm clip with a closing force of 30 grams was applied extradurally around the spinal cord at T9, and then rapidly released with cord compression persisting for 2 minutes. The rats were sacrificed and the spinal cord were collected one week after SCI. Immunohistochemistry (IHC) and western blotting sample were obtained from 5 mm rostral region to the lesion and prepared. We analyzed the nestin immunoreactivity from the white matter of ventral cord and the ependyma of central canal. Nestin and SOX2 were used for markers for NSPCs and analyzed by IHC and western blotting, respectively. Results Nestin and SOX2 were expressed significantly in the SCI groups but not in the sham group. Comparing SCI groups, nestin and SOX2 expression were much stronger in SCI-VPA group than in SCI-saline group. Conclusion Nestin and SOX2 as markers for NSPCs showed increased expression in SCI-VPA group in comparison with SCI-saline group. This result suggests VPA increases expression of spinal NSPCs in SCI.
Collapse
Affiliation(s)
- Woo-Seok Bang
- Department of Neurosurgery, Kyungpook National University Hospital, Daegu, Korea
| | | | | | | | | |
Collapse
|
164
|
Awe JP, Lee PC, Ramathal C, Vega-Crespo A, Durruthy-Durruthy J, Cooper A, Karumbayaram S, Lowry WE, Clark AT, Zack JA, Sebastiano V, Kohn DB, Pyle AD, Martin MG, Lipshutz GS, Phelps PE, Pera RAR, Byrne JA. Generation and characterization of transgene-free human induced pluripotent stem cells and conversion to putative clinical-grade status. Stem Cell Res Ther 2013; 4:87. [PMID: 23890092 PMCID: PMC3854769 DOI: 10.1186/scrt246] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/17/2013] [Indexed: 02/06/2023] Open
Abstract
Introduction The reprogramming of a patient’s somatic cells back into induced pluripotent stem cells (iPSCs) holds significant promise for future autologous cellular therapeutics. The continued presence of potentially oncogenic transgenic elements following reprogramming, however, represents a safety concern that should be addressed prior to clinical applications. The polycistronic stem cell cassette (STEMCCA), an excisable lentiviral reprogramming vector, provides, in our hands, the most consistent reprogramming approach that addresses this safety concern. Nevertheless, most viral integrations occur in genes, and exactly how the integration, epigenetic reprogramming, and excision of the STEMCCA reprogramming vector influences those genes and whether these cells still have clinical potential are not yet known. Methods In this study, we used both microarray and sensitive real-time PCR to investigate gene expression changes following both intron-based reprogramming and excision of the STEMCCA cassette during the generation of human iPSCs from adult human dermal fibroblasts. Integration site analysis was conducted using nonrestrictive linear amplification PCR. Transgene-free iPSCs were fully characterized via immunocytochemistry, karyotyping and teratoma formation, and current protocols were implemented for guided differentiation. We also utilized current good manufacturing practice guidelines and manufacturing facilities for conversion of our iPSCs into putative clinical grade conditions. Results We found that a STEMCCA-derived iPSC line that contains a single integration, found to be located in an intronic location in an actively transcribed gene, PRPF39, displays significantly increased expression when compared with post-excised stem cells. STEMCCA excision via Cre recombinase returned basal expression levels of PRPF39. These cells were also shown to have proper splicing patterns and PRPF39 gene sequences. We also fully characterized the post-excision iPSCs, differentiated them into multiple clinically relevant cell types (including oligodendrocytes, hepatocytes, and cardiomyocytes), and converted them to putative clinical-grade conditions using the same approach previously approved by the US Food and Drug Administration for the conversion of human embryonic stem cells from research-grade to clinical-grade status. Conclusion For the first time, these studies provide a proof-of-principle for the generation of fully characterized transgene-free human iPSCs and, in light of the limited availability of current good manufacturing practice cellular manufacturing facilities, highlight an attractive potential mechanism for converting research-grade cell lines into putatively clinical-grade biologics for personalized cellular therapeutics.
Collapse
|
165
|
Tao F, Li Q, Liu S, Wu H, Skinner J, Hurtado A, Belegu V, Furmanski O, Yang Y, McDonald JW, Johns RA. Role of neuregulin-1/ErbB signaling in stem cell therapy for spinal cord injury-induced chronic neuropathic pain. Stem Cells 2013; 31:83-91. [PMID: 23097328 DOI: 10.1002/stem.1258] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 10/01/2012] [Indexed: 12/12/2022]
Abstract
Chronic neuropathic pain is a common and debilitating consequence of spinal cord injury (SCI). In a rat contusion injury model, we observed that chronic neuropathic pain is present on day 7 after SCI and persists for the entire 56-day observation period. However, currently available pain therapies are inadequate for SCI-induced neuropathic pain. In this study, we show that spinal transplantation of mouse embryonic stem cell-derived oligodendrocyte progenitor cells (OPCs) enhances remyelination in the injured spinal cord and reduces SCI-induced chronic neuropathic pain. Moreover, we found that SCI reduces the protein level of neuregulin-1 and ErbB4 in the injured spinal cord and that OPC transplantation enhances the spinal expression of both proteins after SCI. Finally, intrathecal injection of neuregulin-1 small interfering RNA, but not the control nontarget RNA, diminishes OPC transplantation-produced remyelination and reverses the antinociceptive effect of OPC transplantation. Our findings suggest that the transplantation of embryonic stem cell-derived OPCs is an appropriate therapeutic intervention for treatment of SCI-induced chronic neuropathic pain, and that neuregulin-1/ErbB signaling plays an important role in central remyelination under pathological conditions and contributes to the alleviation of such pain.
Collapse
Affiliation(s)
- Feng Tao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Sykova E, Forostyak S. Stem cells in regenerative medicine. Laser Ther 2013; 22:87-92. [PMID: 24155553 DOI: 10.3136/islsm.22.87] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/03/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND A number of cardiovascular, neurological, musculoskeletal and other diseases have a limited capacity for repair and only a modest progress has been made in treatment of brain diseases. The discovery of stem cells has opened new possibilities for the treatment of these maladies, and cell therapy now stands at the cutting-edge of modern regenerative medicine and tissue engineering. Experimental data and the first clinical trials employing stem cells have shown their broad therapeutic potential and have brought hope to patients suffering from devastating pathologies of different organs and systems. AIMS Here, we briefly review the main achievements and trends in cell-based therapy, with an emphasis on the main types of stem cells: embryonic, mesenchymal stromal and induced pluripotent cells. DISCUSSION Many questions regarding the application of stem cells remain unanswered, particularly tumorigenicity, immune rejection and danger of gene manipulation. Currently, only MSC seems to be safe and might be considered to be a leading candidate for human application to treat pathologies that affect the cardiovascular, neurological and musculoskeletal systems.
Collapse
Affiliation(s)
- Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Videnska 1083, Prague, 14220, Czech Republic ; Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, Prague, 15006, Czech Republic
| | | |
Collapse
|
167
|
Abstract
BACKGROUND A number of cardiovascular, neurological, musculoskeletal and other diseases have a limited capacity for repair and only a modest progress has been made in treatment of brain diseases. The discovery of stem cells has opened new possibilities for the treatment of these maladies, and cell therapy now stands at the cutting-edge of modern regenerative medicine and tissue engineering. Experimental data and the first clinical trials employing stem cells have shown their broad therapeutic potential and have brought hope to patients suffering from devastating pathologies of different organs and systems. AIMS Here, we briefly review the main achievements and trends in cell-based therapy, with an emphasis on the main types of stem cells: embryonic, mesenchymal stromal and induced pluripotent cells. DISCUSSION Many questions regarding the application of stem cells remain unanswered, particularly tumorigenicity, immune rejection and danger of gene manipulation. Currently, only MSC seems to be safe and might be considered to be a leading candidate for human application to treat pathologies that affect the cardiovascular, neurological and musculoskeletal systems.
Collapse
Affiliation(s)
- Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Science of the Czech Republic, Videnska 1083, Prague, 14220, Czech Republic ; Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, Prague, 15006, Czech Republic
| | | |
Collapse
|
168
|
Pol SU, Lang JK, O'Bara MA, Cimato TR, McCallion AS, Sim FJ. Sox10-MCS5 enhancer dynamically tracks human oligodendrocyte progenitor fate. Exp Neurol 2013; 247:694-702. [PMID: 23507034 DOI: 10.1016/j.expneurol.2013.03.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/05/2013] [Accepted: 03/08/2013] [Indexed: 10/27/2022]
Abstract
In this study, we sought to establish a novel method to prospectively and dynamically identify live human oligodendrocyte precursor cells (OPCs) and oligodendrocyte lineage cells from brain dissociates and pluripotent stem cell culture. We selected a highly conserved enhancer element of the Sox10 gene, known as MCS5, which directs reporter expression to oligodendrocyte lineage cells in mouse and zebrafish. We demonstrate that lentiviral Sox10-MCS5 induced expression of GFP at high levels in a subpopulation of human CD140a/PDGFαR-sorted OPCs as well as their immature oligodendrocyte progeny. Furthermore, we show that almost all Sox10-MCS5:GFP(high) cells expressed OPC antigen CD140a and human OPCs expressing SOX10, OLIG2, and PDGFRA mRNAs could be prospectively identified using GFP based fluorescence activated cells sorting alone. Additionally, we established a human induced pluripotent cell (iPSC) line transduced with the Sox10-MCS5:GFP reporter using a Rex-Neo cassette. Similar to human primary cells, GFP expression was restricted to embryoid bodies containing both oligodendrocyte progenitor and oligodendrocyte cells and co-localized with NG2 and O4-positive cells respectively. As such, we have developed a novel reporter system that can track oligodendrocyte commitment in human cells, establishing a valuable tool to improve our understanding and efficiency of human oligodendrocyte derivation.
Collapse
Affiliation(s)
- Suyog U Pol
- Department of Pharmacology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|
169
|
Programming of human endometrial-derived stromal cells (EnSCs) into pre-oligodendrocyte cells by overexpression of miR-219. Neurosci Lett 2013; 537:65-70. [DOI: 10.1016/j.neulet.2013.01.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/15/2013] [Accepted: 01/17/2013] [Indexed: 01/01/2023]
|
170
|
Li Y, Gautam A, Yang J, Qiu L, Melkoumian Z, Weber J, Telukuntla L, Srivastava R, Whiteley EM, Brandenberger R. Differentiation of oligodendrocyte progenitor cells from human embryonic stem cells on vitronectin-derived synthetic peptide acrylate surface. Stem Cells Dev 2013; 22:1497-505. [PMID: 23249362 DOI: 10.1089/scd.2012.0508] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human embryonic stem cell (hESC)-derived oligodendrocyte progenitor cells (OPCs) are being studied for cell replacement therapies, including the treatment of acute spinal cord injury. Current methods of differentiating OPCs from hESCs require complex, animal-derived biological extracellular matrices (ECMs). Defined, low-cost, robust, and scalable culture methods will need to be developed for the widespread deployment and commercialization of hESC-derived cell therapies. Here we describe a defined culture system that uses a vitronectin-derived synthetic peptide acrylate surface (VN-PAS; commercially available as Corning(®) Synthemax(®) surface) in combination with a defined culture medium for hESC growth and differentiation to OPCs. We show that synthetic VN-PAS supports OPC attachment and differentiation, and that hESCs grown on VN-PAS are able to differentiate into OPCs on VN-PAS. Compared to OPCs derived from hESCs grown on ECM of animal origin, higher levels of NG2, a chondroitin sulfate proteoglycan expressed by OPCs, were observed in OPCs differentiated from H1 hESCs grown on VN-PAS, while the expression levels of Nestin and PDGFRα were comparable. In summary, this study demonstrates that synthetic VN-PAS can replace complex, animal-origin ECM to support OPC differentiation from hESCs.
Collapse
Affiliation(s)
- Yan Li
- Geron Corporation , Menlo Park, California, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Ebert AD, Shelley BC, Hurley AM, Onorati M, Castiglioni V, Patitucci TN, Svendsen SP, Mattis VB, McGivern JV, Schwab AJ, Sareen D, Kim HW, Cattaneo E, Svendsen CN. EZ spheres: a stable and expandable culture system for the generation of pre-rosette multipotent stem cells from human ESCs and iPSCs. Stem Cell Res 2013; 10:417-427. [PMID: 23474892 DOI: 10.1016/j.scr.2013.01.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 01/19/2013] [Accepted: 01/28/2013] [Indexed: 02/01/2023] Open
Abstract
We have developed a simple method to generate and expand multipotent, self-renewing pre-rosette neural stem cells from both human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs) without utilizing embryoid body formation, manual selection techniques, or complex combinations of small molecules. Human ESC and iPSC colonies were lifted and placed in a neural stem cell medium containing high concentrations of EGF and FGF-2. Cell aggregates (termed EZ spheres) could be expanded for long periods using a chopping method that maintained cell-cell contact. Early passage EZ spheres rapidly down-regulated OCT4 and up-regulated SOX2 and nestin expression. They retained the potential to form neural rosettes and consistently differentiated into a range of central and peripheral neural lineages. Thus, they represent a very early neural stem cell with greater differentiation flexibility than other previously described methods. As such, they will be useful for the rapidly expanding field of neurological development and disease modeling, high-content screening, and regenerative therapies based on pluripotent stem cell technology.
Collapse
Affiliation(s)
- Allison D Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226
| | - Brandon C Shelley
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Amanda M Hurley
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Marco Onorati
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Via Viotti 3/5, 20133 Milano, Italy
| | - Valentina Castiglioni
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Via Viotti 3/5, 20133 Milano, Italy
| | - Teresa N Patitucci
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226
| | - Soshana P Svendsen
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Virginia B Mattis
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Jered V McGivern
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226
| | - Andrew J Schwab
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226
| | - Dhruv Sareen
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Ho Won Kim
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Elena Cattaneo
- Department of Biosciences and Centre for Stem Cell Research, Università degli Studi di Milano, Via Viotti 3/5, 20133 Milano, Italy
| | - Clive N Svendsen
- Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| |
Collapse
|
172
|
Ebrahimi-Barough S, Kouchesfahani HM, Ai J, Massumi M. Differentiation of human endometrial stromal cells into oligodendrocyte progenitor cells (OPCs). J Mol Neurosci 2013; 51:265-73. [PMID: 23338937 DOI: 10.1007/s12031-013-9957-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/09/2013] [Indexed: 01/21/2023]
Abstract
Oligodendrocytes are myelinating cells in the central nervous system that form the myelin sheath of axons to support rapid nerve conduction. Human endometrial stromal cells (EnSCs) are the abundant and easy available source for cell replacement therapy. In the present study, the EnSCs were coaxed to oligodendrocyte progenitor programming by induction of neuronal condition media, including bFGF, epidermal growth factor, and platelet-derived growth factor (PDGF)-AA signaling molecules as well as triiodothyronine. Differentiated cells were analyzed for expression of oligodendrocytic markers by quantitative reverse transcription PCR and immunocytochemistry. The results showed the expression of oligodendrocyte lineage markers such as nestin, PDGF receptor alpha (PDGFRα), Sox10, and Olig2 in the level of mRNAs. The expression of nestin and PDGFRα increased after 8 days posttreatment. Interestingly, the expression of nestin and PDGFRα genes at the levels of mRNA and proteins decreased 24 days after induction. The expression of A2B5, O4, and Olig2 proteins in EnSCs was confirmed using immunocytochemistry. The results confirmed that EnSCs could response to the signaling molecules which routinely applied for oligodendrocyte differentiation. Here for the first time, we demonstrated that EnSCs could be programmed into oligodendrocyte progenitor cells and may convince to consider these cells as suitable source for cell therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Somayeh Ebrahimi-Barough
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University (TMU), Tehran, Iran,
| | | | | | | |
Collapse
|
173
|
Therapeutic strategies for the treatment of spinal muscular atrophy. Future Med Chem 2013; 4:1733-50. [PMID: 22924510 DOI: 10.4155/fmc.12.107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an inherited neurodegenerative disease that results in progressive dysfunction of motor neurons of the anterior horn of the spinal cord. SMA is caused by the loss of full-length protein expression from the survival of motor neuron 1 (SMN1) gene. The disease has a unique genetic profile as it is autosomal recessive for the loss of SMN1, but a nearly identical homolog, SMN2, acts as a disease modifier whose expression is inversely correlated to clinical severity. Targeted therapeutic approaches primarily focus on increasing the levels of full-length SMN protein, through either gene replacement or regulation of SMN2 expression. There is currently no US FDA approved treatment for SMA. This is an exciting time as multiple efforts from academic and industrial laboratories are reaching the preclinical and clinical testing stages.
Collapse
|
174
|
Neural lineage development in the rhesus monkey with embryonic stem cells. Transl Neurosci 2013. [DOI: 10.2478/s13380-013-0135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThere are three controversial and undetermined models of neurogenesis and gliogenesis from neuroepithelial cells in the early neural tube; the first in which neurons and glia were proposed to originate from a single homogenous population, the second from two separate pools of committed glial and neuronal progenitors, or, lastly, from transit radial glial (RG). Issues concerning embryonic neural lineage development in primates are not well understood due to restrictions imposed by ethics and material sources. In this study, early neural lineage development was investigated in vitro with rhesus monkey embryonic stem cells (rESC) by means of immunofluorescence with lineage specific markers. It was revealed that neural differentiation likely progresses in a sequential lineage restriction pathway from neuroepithelial stem/progenitor cells to neurons and glia via RG and intermediate precursors: neuronal precursors and glial progenitors. In conclusion, our results suggest that the early neural lineage development of rESC in vitro supported the model in which neuroepithelial cells develop into RG capable of generating both neurons and glia. This work should facilitate understanding of the mechanism of development of the nervous system in primates.
Collapse
|
175
|
Kim SU. Regenerative Medicine in the Central Nervous System: Stem Cell-Based Cell- and Gene-Therapy. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
176
|
Gongora M, Peressutti C, Machado S, Teixeira S, Velasques B, Ribeiro P. Progress and prospects in neurorehabilitation: clinical applications of stem cells and brain–computer interface for spinal cord lesions. Neurol Sci 2012; 34:427-33. [DOI: 10.1007/s10072-012-1232-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/18/2012] [Indexed: 12/19/2022]
|
177
|
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating neurodegenerative disorder of the brain and spinal cord that causes significant disability in young adults. Although the precise aetiopathogenesis of MS remains unresolved, its pathological hallmarks include inflammation, demyelination, axonal injury (acute and chronic), astrogliosis and variable remyelination. Despite major recent advances in therapeutics for the early stage of the disease there are currently no disease modifying treatments for the progressive stage of disease, whose pathological substrate is axonal degeneration. This represents the great and unmet clinical need in MS. Against this background, human stem cells offer promise both to improve understanding of disease mechanism(s) through in-vitro modeling as well as potentially direct use to supplement and promote remyelination, an endogenous reparative process where entire myelin sheaths are restored to demyelinated axons. Conceptually, stem cells can act directly to myelinate axons or indirectly through different mechanisms to promote endogenous repair; importantly these two mechanisms of action are not mutually exclusive. We propose that discovery of novel methods to invoke or enhance remyelination in MS may be the most effective therapeutic strategy to limit axonal damage and instigate restoration of structure and function in this debilitating condition. Human stem cell derived neurons and glia, including patient specific cells derived through reprogramming, provide an unprecedented experimental system to model MS “in a dish” as well as enable high-throughput drug discovery. Finally, we speculate upon the potential role for stem cell based therapies in MS.
Collapse
|
178
|
Cao HQ, Dong ED. An update on spinal cord injury research. Neurosci Bull 2012; 29:94-102. [PMID: 23124646 DOI: 10.1007/s12264-012-1277-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/26/2012] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) can have a range of debilitating effects and permanently alter the capabilities and quality of life of survivors. The first specialized centers of care for SCI were established in 1944 and since then an increasing amount of research has been carried out in this area. Despite this, the present treatment and care levels for SCI are not comparable to those in other areas of medicine. In the clinic, the aim of SCI treatment is primarily to limit secondary damage by reducing compression in trauma spots and stabilizing the spinal column. Currently, no effective strategy for functional recovery is offered. In this review, we focus on research progress on the molecular mechanisms underlying SCI, and assess the treatment outcomes of SCI in animal models, i.e., neurotrophins and stem cells are discussed as pre-clinical therapies in animal models. We also assess the resources available and national research projects carried out on SCI in China in recent years, as well as making recommendations for the future allocation of funds in this area.
Collapse
Affiliation(s)
- He-Qi Cao
- Division of Neurological Disorders and Mental Health, Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China.
| | | |
Collapse
|
179
|
Mothe AJ, Tator CH. Advances in stem cell therapy for spinal cord injury. J Clin Invest 2012; 122:3824-34. [PMID: 23114605 DOI: 10.1172/jci64124] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition producing great personal and societal costs and for which there is no effective treatment. Stem cell transplantation is a promising therapeutic strategy, though much preclinical and clinical research work remains. Here, we briefly describe SCI epidemiology, pathophysiology, and experimental and clinical stem cell strategies. Research in stem cell biology and cell reprogramming is rapidly advancing, with the hope of moving stem cell therapy closer to helping people with SCI. We examine issues important for clinical translation and provide a commentary on recent developments, including termination of the first human embryonic stem cell transplantation trial in human SCI.
Collapse
Affiliation(s)
- Andrea J Mothe
- Toronto Western Research Institute and Krembil Neuroscience Centre, Toronto Western Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
180
|
Pêgo AP, Kubinova S, Cizkova D, Vanicky I, Mar FM, Sousa MM, Sykova E. Regenerative medicine for the treatment of spinal cord injury: more than just promises? J Cell Mol Med 2012; 16:2564-82. [PMID: 22805417 PMCID: PMC4118226 DOI: 10.1111/j.1582-4934.2012.01603.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 07/09/2012] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury triggers a complex set of events that lead to tissue healing without the restoration of normal function due to the poor regenerative capacity of the spinal cord. Nevertheless, current knowledge about the intrinsic regenerative ability of central nervous system axons, when in a supportive environment, has made the prospect of treating spinal cord injury a reality. Among the range of strategies under investigation, cell-based therapies offer the most promising results, due to the multifactorial roles that these cells can fulfil. However, the best cell source is still a matter of debate, as are clinical issues that include the optimal cell dose as well as the timing and route of administration. In this context, the role of biomaterials is gaining importance. These can not only act as vehicles for the administered cells but also, in the case of chronic lesions, can be used to fill the permanent cyst, thus creating a more favourable and conducive environment for axonal regeneration in addition to serving as local delivery systems of therapeutic agents to improve the regenerative milieu. Some of the candidate molecules for the future are discussed in view of the knowledge derived from studying the mechanisms that facilitate the intrinsic regenerative capacity of central nervous system neurons. The future challenge for the multidisciplinary teams working in the field is to translate the knowledge acquired in basic research into effective combinatorial therapies to be applied in the clinic.
Collapse
Affiliation(s)
- Ana Paula Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
181
|
Goldman SA, Nedergaard M, Windrem MS. Glial progenitor cell-based treatment and modeling of neurological disease. Science 2012; 338:491-5. [PMID: 23112326 PMCID: PMC3548656 DOI: 10.1126/science.1218071] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The diseases of myelin are among the most prevalent and disabling conditions in neurology. These diseases include both the vascular and inflammatory demyelinating disorders of adulthood, as well as the childhood leukodystrophies and cerebral palsy. These fundamentally glial disorders may be amenable to treatment by glial progenitor cells (GPCs), which give rise to astroglia and myelin-producing oligodendrocytes. Given the development of new methods for generating and isolating human GPCs, the myelin disorders may now be compelling targets for cell-based therapy. In addition, the efficient engraftment and expansion of human GPCs in murine hosts has led to the development of human glial chimeric mouse brains, which provides new opportunities for studying the species-specific roles of human glia in cognition, as well as in disease pathogenesis.
Collapse
Affiliation(s)
- Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
182
|
Oldershaw RA. Cell sources for the regeneration of articular cartilage: the past, the horizon and the future. Int J Exp Pathol 2012; 93:389-400. [PMID: 23075006 DOI: 10.1111/j.1365-2613.2012.00837.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 06/15/2012] [Indexed: 11/29/2022] Open
Abstract
Avascular, aneural articular cartilage has a low capacity for self-repair and as a consequence is highly susceptible to degradative diseases such as osteoarthritis. Thus the development of cell-based therapies that repair focal defects in otherwise healthy articular cartilage is an important research target, aiming both to delay the onset of degradative diseases and to decrease the need for joint replacement surgery. This review will discuss the cell sources which are currently being investigated for the generation of chondrogenic cells. Autologous chondrocyte implantation using chondrocytes expanded ex vivo was the first chondrogenic cellular therapy to be used clinically. However, limitations in expansion potential have led to the investigation of adult mesenchymal stem cells as an alternative cell source and these therapies are beginning to enter clinical trials. The chondrogenic potential of human embryonic stem cells will also be discussed as a developmentally relevant cell source, which has the potential to generate chondrocytes with phenotype closer to that of articular cartilage. The clinical application of these chondrogenic cells is much further away as protocols and tissue engineering strategies require additional optimization. The efficacy of these cell types in the regeneration of articular cartilage tissue that is capable of withstanding biomechanical loading will be evaluated according to the developing regulatory framework to determine the most appropriate cellular therapy for adoption across an expanding patient population.
Collapse
Affiliation(s)
- Rachel A Oldershaw
- North East England Stem Cell Institute (NESCI), Institute of Cellular Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK.
| |
Collapse
|
183
|
Abstract
Stem cells are the seeds of tissue repair and regeneration and a promising source for novel therapies. However, apart from hematopoietic stem cell (HSC) transplantation, essentially all other stem cell treatments remain experimental. High hopes have inspired numerous clinical trials, but it has been difficult to obtain unequivocal evidence for robust clinical benefit. In recent years, unproven therapies have been widely practiced outside the standard clinical trial network, threatening the cause of legitimate clinical investigation. Numerous challenges and technical barriers must be overcome before novel stem cell therapies can achieve meaningful clinical impact.
Collapse
|
184
|
Jaerve A, Schira J, Müller HW. Concise review: the potential of stromal cell-derived factor 1 and its receptors to promote stem cell functions in spinal cord repair. Stem Cells Transl Med 2012. [PMID: 23197665 DOI: 10.5966/sctm.2012-0068] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transplanted stem cells provide beneficial effects on regeneration/recovery after spinal cord injury (SCI) by the release of growth-promoting factors, increased tissue preservation, and provision of a permissive environment for axon regeneration. A rise in chemokine stromal cell-derived factor 1 (SDF-1/CXCL12) expression levels in central nervous system (CNS) injury sites has been shown to play a central role in recruiting transplanted stem cells. Although technically more challenging, it has been shown that after SCI few endogenous stem cells are recruited via SDF-1/CXCR4 signaling. Evidence is accumulating that increasing SDF-1 levels at the injury site (e.g., by exogenous application or transfection methods) further enhances stem cell recruitment. Moreover, SDF-1 might, in addition to migration, also influence survival, proliferation, differentiation, and cytokine secretion of stem cells. Here, we discuss the experimental data available on the role of SDF-1 in stem and progenitor cell biology following CNS injury and suggest strategies for how manipulation of the SDF-1 system could facilitate stem cell-based therapeutic approaches in SCI. In addition, we discuss challenges such as how to circumvent off-target effects in order to facilitate the transfer of SDF-1 to the clinic.
Collapse
Affiliation(s)
- Anne Jaerve
- Molecular Neurobiology Laboratory, Department of Neurology, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany
| | | | | |
Collapse
|
185
|
Volarevic V, Erceg S, Bhattacharya SS, Stojkovic P, Horner P, Stojkovic M. Stem cell-based therapy for spinal cord injury. Cell Transplant 2012; 22:1309-23. [PMID: 23043847 DOI: 10.3727/096368912x657260] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells (SCs) represent a new therapeutic approach for spinal cord injury (SCI) by enabling improved sensory and motor functions in animal models. The main goal of SC-based therapy for SCI is the replacement of neurons and glial cells that undergo cell death soon after injury. Stem cells are able to promote remyelination via oligodendroglia cell replacement to produce trophic factors enhancing neurite outgrowth, axonal elongation, and fiber density and to activate resident or transplanted progenitor cells across the lesion cavity. While several SC transplantation strategies have shown promising yet partial efficacy, mechanistic proof is generally lacking and is arguably the largest impediment toward faster progress and clinical application. The main challenge ahead is to spur on cooperation between clinicians, researchers, and patients in order to define and optimize the mechanisms of SC function and to establish the ideal source/s of SCs that produce efficient and also safe therapeutic approaches.
Collapse
Affiliation(s)
- Vladislav Volarevic
- Center for Molecular Medicine and Stem Cell Research, Medical Faculty, University of Kragujevac, Serbia
| | | | | | | | | | | |
Collapse
|
186
|
Cellular programming and reprogramming: sculpting cell fate for the production of dopamine neurons for cell therapy. Stem Cells Int 2012; 2012:412040. [PMID: 22988464 PMCID: PMC3441013 DOI: 10.1155/2012/412040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 07/05/2012] [Indexed: 12/14/2022] Open
Abstract
Pluripotent stem cells are regarded as a promising cell source to obtain human dopamine neurons in sufficient amounts and purity for cell replacement therapy. Importantly, the success of clinical applications depends on our ability to steer pluripotent stem cells towards the right neuronal identity. In Parkinson disease, the loss of dopamine neurons is more pronounced in the ventrolateral population that projects to the sensorimotor striatum. Because synapses are highly specific, only neurons with this precise identity will contribute, upon transplantation, to the synaptic reconstruction of the dorsal striatum. Thus, understanding the developmental cell program of the mesostriatal dopamine neurons is critical for the identification of the extrinsic signals and cell-intrinsic factors that instruct and, ultimately, determine cell identity. Here, we review how extrinsic signals and transcription factors act together during development to shape midbrain cell fates. Further, we discuss how these same factors can be applied in vitro to induce, select, and reprogram cells to the mesostriatal dopamine fate.
Collapse
|
187
|
Lukovic D, Moreno Manzano V, Stojkovic M, Bhattacharya SS, Erceg S. Concise Review: Human Pluripotent Stem Cells in the Treatment of Spinal Cord Injury. Stem Cells 2012; 30:1787-92. [DOI: 10.1002/stem.1159] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
188
|
Abstract
Clonal cell culture is crucial for experimental protocols that require growth or selection of pure populations of cells. High-density derivation of neural progenitors from human embryonic stem cells (hESCs) can lead to incomplete differentiation, and transplantation of resulting heterogeneous cell mixtures can cause proliferation of tumorigenic clusters in vivo. We have identified the neural precursor that resides among normal hESC colonies as a TRA-1-60(-)/SSEA4(-)/SOX1(+) cell and developed a method that allows for the clonal expansion of these FACS-selected progenitors to neural stem cells (NSCs) in serum-free conditions. Single TRA-1-60(-)/SSEA4(-)/SOX1(+) cells grown in serum-free media give rise to multipotent NSCs with an efficiency of 0.7%. The fate of the TRA-1-60(-)/SSEA4(-)/SOX1(+) neural precursor becomes specified in maintenance conditions by inhibition of BMP signaling. This clonal culture method can be scaled up to produce NSCs for differentiation and use in cell therapies.
Collapse
|
189
|
Chen C, Daugherty D, Jiang P, Deng W. Oligodendrocyte progenitor cells derived from mouse embryonic stem cells give rise to type-1 and type-2 astrocytes in vitro. Neurosci Lett 2012; 523:180-5. [PMID: 22781495 DOI: 10.1016/j.neulet.2012.06.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 11/18/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) in primary culture can give rise to mature oligodendrocytes and type-2, but not type-1 astrocytes depending on the culture conditions. The OPCs thus are called oligodendrocyte-type-2 astrocyte (O2-A) progenitor cells. Mouse embryonic stem cells (mESCs) have been efficiently differentiated into OPCs; however, the fate plasticity of mESC-derived OPCs is not well characterized. In the present study, using GFP-Olig2 mESC line, we showed that the Olig2(+)/GFP(+)/A2B5(+)/NG2(+) OPCs derived from GFP-Olig2 mESCs can mature into oligodendrocytes when co-cultured with mESC-derived neurons. Interestingly, when induced to astrocytic differentiation by bone morphogenetic protein-4, these mESC-derived OPCs can not only generate type-2 astrocytes, but also type-1 astrocytes. These results challenge the dogma that OPCs in culture can only generate type-2, but not type-1 astrocytes, and support the in vivo finding that during perinatal development, OPCs can give rise to a subset of type-1 astrocytes.
Collapse
Affiliation(s)
- Chen Chen
- Department of Neurology, Institute of Neurology, Tianjin General Hospital of Tianjin Medical University, Tianjin, China
| | | | | | | |
Collapse
|
190
|
Tirotta E, Kirby LA, Hatch MN, Lane TE. IFN-γ-induced apoptosis of human embryonic stem cell derived oligodendrocyte progenitor cells is restricted by CXCR2 signaling. Stem Cell Res 2012; 9:208-17. [PMID: 22885102 DOI: 10.1016/j.scr.2012.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/18/2012] [Accepted: 06/20/2012] [Indexed: 12/01/2022] Open
Abstract
Engraftment of human embryonic stem cell (hESC)-derived OPCs in animal models of demyelination results in remyelination and clinical recovery, supporting the feasibility of cell replacement therapies in promoting repair of damaged neural tissue. A critical gap in our understanding of the mechanisms associated with repair revolves around the effects of the local microenvironment on transplanted cell survival. We have determined that treatment of human ESC-derived OPCs with the pleiotropic cytokine IFN-γ promotes apoptosis that is associated with mitochondrial cytochrome c released into the cytosol with subsequent caspase 3 activation. IFN-γ-induced apoptosis is mediated, in part, by secretion of the CXC chemokine ligand 10 (CXCL10) from IFN-γ-treated cells. Signaling through the chemokine receptor CXCR2 by the ligand CXCL1 functions in a tonic manner by muting apoptosis and this is associated with reduced levels of cytosolic cytochrome c and impaired cleavage of caspase 3. These findings support a role for both IFN-γ and CXCL10 in contributing to neuropathology by promoting OPC apoptosis. In addition, these data suggest that hOPCs used for therapeutic treatment for human neurologic disease/damage are susceptible to death through exposure to local inflammatory cytokines present within the inflammatory milieu.
Collapse
Affiliation(s)
- Emanuele Tirotta
- Department of Molecular Biology and Biochemistry, Multiple Sclerosis Research Center, Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697-3900, USA
| | | | | | | |
Collapse
|
191
|
Neman J, de Vellis J. A method for deriving homogenous population of oligodendrocytes from mouse embryonic stem cells. Dev Neurobiol 2012; 72:777-88. [DOI: 10.1002/dneu.22008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
192
|
Bernstein HS, Hyun WC. Strategies for enrichment and selection of stem cell-derived tissue precursors. Stem Cell Res Ther 2012; 3:17. [PMID: 22575029 PMCID: PMC3392764 DOI: 10.1186/scrt108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Human embryonic stem cells have the capacity for self-renewal and pluripotency and thus are a primary candidate for tissue engineering and regenerative therapies. These cells also provide an opportunity to study the development of human tissues ex vivo. To date, numerous human embryonic stem cell lines have been derived and characterized. In this review, we will detail the strategies used to direct tissue-specific differentiation of embryonic stem cells. We also will discuss how these strategies have produced new sources of tissue-specific progenitor cells. Finally, we will describe the next generation of methods being developed to identify and select stem cell-derived tissue precursors for experimental study and clinical use.
Collapse
Affiliation(s)
- Harold S Bernstein
- Department of Pediatrics (Cardiology), University of California San Francisco, San Francisco, CA 94143-1346, USA.
| | | |
Collapse
|
193
|
Nazor KL, Altun G, Lynch C, Tran H, Harness JV, Slavin I, Garitaonandia I, Müller FJ, Wang YC, Boscolo FS, Fakunle E, Dumevska B, Lee S, Park HS, Olee T, D’Lima DD, Semechkin R, Parast MM, Galat V, Laslett AL, Schmidt U, Keirstead HS, Loring JF, Laurent LC. Recurrent variations in DNA methylation in human pluripotent stem cells and their differentiated derivatives. Cell Stem Cell 2012; 10:620-34. [PMID: 22560082 PMCID: PMC3348513 DOI: 10.1016/j.stem.2012.02.013] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 01/18/2012] [Accepted: 02/06/2012] [Indexed: 11/23/2022]
Abstract
Human pluripotent stem cells (hPSCs) are potential sources of cells for modeling disease and development, drug discovery, and regenerative medicine. However, it is important to identify factors that may impact the utility of hPSCs for these applications. In an unbiased analysis of 205 hPSC and 130 somatic samples, we identified hPSC-specific epigenetic and transcriptional aberrations in genes subject to X chromosome inactivation (XCI) and genomic imprinting, which were not corrected during directed differentiation. We also found that specific tissue types were distinguished by unique patterns of DNA hypomethylation, which were recapitulated by DNA demethylation during in vitro directed differentiation. Our results suggest that verification of baseline epigenetic status is critical for hPSC-based disease models in which the observed phenotype depends on proper XCI or imprinting and that tissue-specific DNA methylation patterns can be accurately modeled during directed differentiation of hPSCs, even in the presence of variations in XCI or imprinting.
Collapse
Affiliation(s)
- Kristopher L. Nazor
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Gulsah Altun
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Candace Lynch
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Ha Tran
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Julie V. Harness
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California 92697, USA
| | - Ileana Slavin
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Ibon Garitaonandia
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Franz-Josef Müller
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- Center for Psychiatry, ZIP-Kiel, University Hospital Schleswig Holstein, Niemannsweg 147, D-24105 Kiel, Germany
| | - Yu-Chieh Wang
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Francesca S. Boscolo
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Eyitayo Fakunle
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Biljana Dumevska
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Division of Materials Science & Engineering Clayton, Victoria3168, Australia
| | - Sunray Lee
- Laboratory of Stem Cell Niche, CEFO Co. Inc, 46-21 Susong-dong, Jongno-gu, Seoul 110-140, South Korea
| | - Hyun Sook Park
- University of California, San Diego, Department of Reproductive Medicine, 200 West Arbor Drive, San Diego, California 92035, USA
| | - Tsaiwei Olee
- Shiley Center for Orthopaedic Research & Education, Scripps Clinic, La Jolla, California 92037, USA
| | - Darryl D. D’Lima
- Shiley Center for Orthopaedic Research & Education, Scripps Clinic, La Jolla, California 92037, USA
| | - Ruslan Semechkin
- International Stem Cell Corporation, Carlsbad, California 92008, USA
| | - Mana M. Parast
- University of California, San Diego, Department of Pathology, 200 West Arbor Drive, San Diego, California 92035, USA
| | - Vasiliy Galat
- Developmental Biology Program, iPS and Human Stem Cell Core Facility, Children’s Memorial Research Center, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Andrew L. Laslett
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Division of Materials Science & Engineering Clayton, Victoria3168, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3168, Australia
| | - Uli Schmidt
- Stem Cell Laboratory, Genea, Sydney, New South Wales 2000, Australia
| | - Hans S. Keirstead
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy and Neurobiology, School of Medicine, University of California at Irvine, Irvine, California 92697, USA
| | - Jeanne F. Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Louise C. Laurent
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
- University of California, San Diego, Department of Reproductive Medicine, 200 West Arbor Drive, San Diego, California 92035, USA
| |
Collapse
|
194
|
Garbossa D, Boido M, Fontanella M, Fronda C, Ducati A, Vercelli A. Recent therapeutic strategies for spinal cord injury treatment: possible role of stem cells. Neurosurg Rev 2012; 35:293-311; discussion 311. [PMID: 22539011 DOI: 10.1007/s10143-012-0385-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 09/27/2011] [Accepted: 11/20/2011] [Indexed: 01/01/2023]
Abstract
Spinal cord injury (SCI) often results in significant dysfunction and disability. A series of treatments have been proposed to prevent and overcome the formation of the glial scar and inhibitory factors to axon regrowth. In the last decade, cell therapy has emerged as a new tool for several diseases of the nervous system. Stem cells act as minipumps providing trophic and immunomodulatory factors to enhance axonal growth, to modulate the environment, and to reduce neuroinflammation. This capability can be boosted by genetical manipulation to deliver trophic molecules. Different types of stem cells have been tested, according to their properties and the therapeutic aims. They differ from each other for origin, developmental stage, stage of differentiation, and fate lineage. Related to this, stem cells differentiating into neurons could be used for cell replacement, even though the feasibility that stem cells after transplantation in the adult lesioned spinal cord can differentiate into neurons, integrate within neural circuits, and emit axons reaching the muscle is quite remote. The timing of cell therapy has been variable, and may be summarized in the acute and chronic phases of disease, when stem cells interact with a completely different environment. Even though further experimental studies are needed to elucidate the mechanisms of action, the therapeutic, and the side effects of cell therapy, several clinical protocols have been tested or are under trial. Here, we report the state-of-the-art of cell therapy in SCI, in terms of feasibility, outcome, and side effects.
Collapse
Affiliation(s)
- D Garbossa
- Department of Neurosurgery, S. Giovanni Battista Hospital, University of Torino, Via Cherasco 15, 10126, Torino, Italy.
| | | | | | | | | | | |
Collapse
|
195
|
Domingo A, Al-Yahya AA, Asiri Y, Eng JJ, Lam T. A systematic review of the effects of pharmacological agents on walking function in people with spinal cord injury. J Neurotrauma 2012; 29:865-79. [PMID: 22142289 PMCID: PMC4496059 DOI: 10.1089/neu.2011.2052] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Studies of spinalized animals indicate that some pharmacological agents may act on receptors in the spinal cord, helping to produce coordinated locomotor movement. Other drugs may help to ameliorate the neuropathological changes resulting from spinal cord injury (SCI), such as spasticity or demyelination, to improve walking. The purpose of this study was to systematically review the effects of pharmacological agents on gait in people with SCI. A keyword literature search of articles that evaluated the effects of drugs on walking after SCI was performed using the databases MEDLINE/PubMed, CINAHL, EMBASE, PsycINFO, and hand searching. Two reviewers independently evaluated each study, using the Physiotherapy Evidence Database (PEDro) tool for randomized clinical trials (RCTs), and the modified Downs & Black scale for all other studies. Results were tabulated and levels of evidence were assigned. Eleven studies met the inclusion criteria. One RCT provided Level 1 evidence that GM-1 ganglioside in combination with physical therapy improved motor scores, walking velocity, and distance better than placebo and physical therapy in persons with incomplete SCI. Multiple studies (levels of evidence 1-5) showed that clonidine and cyproheptadine may improve locomotor function and walking speed in severely impaired individuals with incomplete SCI. Gains in walking speed associated with GM-1, cyproheptadine, and clonidine are low compared to those seen with locomotor training. There was also Level 1 evidence that 4-aminopyridine and L-dopa were no better than placebo in helping to improve gait. Two Level 5 studies showed that baclofen had little to no effect on improving walking in persons with incomplete SCI. There is limited evidence that pharmacological agents tested so far would facilitate the recovery of walking after SCI. More studies are needed to better understand the effects of drugs combined with gait training on walking outcomes in people with SCI.
Collapse
Affiliation(s)
- Antoinette Domingo
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | |
Collapse
|
196
|
Olig1 function is required for remyelination potential of transplanted neural progenitor cells in a model of viral-induced demyelination. Exp Neurol 2012; 235:380-7. [PMID: 22449475 DOI: 10.1016/j.expneurol.2012.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 03/04/2012] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) resulting in cumulative neurologic deficits associated with progressive myelin loss. We have previously shown that transplantation of neural progenitor cells (NPCs) into mice persistently infected with the JHM strain of mouse hepatitis virus (JHMV) results in enhanced differentiation into oligodendrocyte progenitor cells (OPCs) that is associated with remyelination and axonal sparing. The current study examines the contributions of the transcription factor Olig1 on NPC differentiation and remyelination. Under defined conditions, NPCs preferentially differentiate into oligodendroglia whereas NPCs isolated from Olig1-deficient (Olig1-/-) mice exhibit enhanced differentiation into astrocytes. Transplantation of Olig1-/- and Olig1+/+ NPCs into JHMV-infected mice resulted in similar cell survival, proliferation, and selective migration to areas of demyelination. However, only recipients of wild type NPCs exhibited extensive remyelination compared to mice receiving Olig1-/- NPCs. In vivo characterization of NPCs revealed that Olig1+/+ NPCs preferentially differentiated into NG2-positive OPCs and formed processes expressing myelin basic protein that encircled axons. In contrast, the majority of transplanted Olig1-/- NPCs differentiated into GFAP-positive cells consistent with the astrocyte lineage. These results indicate that exogenous NPCs contribute to improved clinical and histological outcome and this is associated with remyelination by this donor population. Further, these findings reveal that Olig1function is required for the remyelination potential of NPCs after transplant, through specification and/or maintenance of oligodendroglial identity.
Collapse
|
197
|
Neural stem cells for spinal cord repair. Cell Tissue Res 2012; 349:349-62. [DOI: 10.1007/s00441-012-1363-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 02/02/2012] [Indexed: 12/20/2022]
|
198
|
Cromer Berman SM, Kshitiz, Wang CJ, Orukari I, Levchenko A, Bulte JWM, Walczak P. Cell motility of neural stem cells is reduced after SPIO-labeling, which is mitigated after exocytosis. Magn Reson Med 2012; 69:255-62. [PMID: 22374813 DOI: 10.1002/mrm.24216] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/12/2011] [Accepted: 01/23/2012] [Indexed: 11/05/2022]
Abstract
MRI is used for tracking of superparamagnetic iron oxide (SPIO)-labeled neural stem cells. Studies have shown that long-term MR tracking of rapidly dividing cells underestimates their migration distance. Time-lapse microscopy of random cellular motility and cell division was performed to evaluate the effects of SPIO-labeling on neural stem cell migration. Labeled cells divided symmetrically and exhibited no changes in cell viability, proliferation, or apoptosis. However, SPIO-labeling resulted in decreased motility of neural stem cells as compared with unlabeled controls. When SPIO-labeled neural stem cells and human induced pluripotent stem cells were transplanted into mouse brain, rapid exocytosis of SPIO by live cells was observed as early as 48 h postengraftment, with SPIO-depleted cells showing the farthest migration distance. As label dilution is negligible at this early time point, we conclude that MRI underestimation of cell migration can also occur as a result of reduced cell motility, which appears to be mitigated following SPIO exocytosis.
Collapse
Affiliation(s)
- Stacey M Cromer Berman
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
199
|
Benchoua A, Onteniente B. Intracerebral transplantation for neurological disorders. Lessons from developmental, experimental, and clinical studies. Front Cell Neurosci 2012; 6:2. [PMID: 22319470 PMCID: PMC3267364 DOI: 10.3389/fncel.2012.00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/09/2012] [Indexed: 01/24/2023] Open
Abstract
The use of human pluripotent stem cells (PSCs) for cell therapy faces a number of challenges that are progressively answered by results from clinical trials and experimental research. Among these is the control of differentiation before transplantation and the prediction of cell fate after administration into the human brain, two aspects that condition both the safety and efficacy of the approach. For neurological disorders, this includes two steps: firstly, the identification of the optimal maturation stage for transplantation along the continuum that transforms PSCs into fully differentiated neural cell types, together with the derivation of robust protocols for large-scale production of biological products, and, secondly, the understanding of the effects of environmental cues and their possible interference with transplanted cells commitment. This review will firstly summarize our knowledge on developmental processes that have been applied to achieve robust in vitro differentiation of PSCs into neural progenitors. In a second part, we summarize results from experimental and clinical transplantation studies that help understanding the dialogue that establishes between transplanted cells and their host brain.
Collapse
|
200
|
Davis H, Guo X, Lambert S, Stancescu M, Hickman JJ. Small Molecule Induction of Human Umbilical Stem Cells into MBP-positive Oligodendrocytes in a Defined Three-Dimensional Environment. ACS Chem Neurosci 2012; 3:31-39. [PMID: 22582139 DOI: 10.1021/cn200082q] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Stem cells from umbilical cord would be a favorable alternative to embryonic stem cells for therapeutic applications. In this study, human multipotent progenitor cells (MLPCs) from umbilical cord were differentiated into oligodendrocytes by exposure to a range of microenvironmental chemical and physical cues. Chemical cues were represented by a novel defined differentiation medium containing the neurotransmitter norepinephrine (NE). In traditional 2 dimensional (2D) conditions, the MLPCs differentiated into oligodendrocyte precursors, but did not progress further. However, in a 3 dimensional (3D) environment, the MLPCs differentiated into committed oligodendrocytes that expressed MBP. The apparent method of interaction of NE in stimulating the differentiation process was identified to occur through the adenergic pathway while all prior differentiation methods have used other routes. This novel method of obtaining functional human oligodendrocytes from MLPCs would eliminate many of the difficulties associated with their differentiation from embryonic stem cells.
Collapse
Affiliation(s)
- Hedvika Davis
- Hybrid Systems Laboratory, NanoScience
Technology Center, University of Central Florida, Orlando, Florida 32826, United States
- College of Medicine, Biomedical
Science Program, University of Central Florida, Orlando, Florida 32826, United States
| | - Xiufang Guo
- Hybrid Systems Laboratory, NanoScience
Technology Center, University of Central Florida, Orlando, Florida 32826, United States
| | - Stephen Lambert
- Hybrid Systems Laboratory, NanoScience
Technology Center, University of Central Florida, Orlando, Florida 32826, United States
- College of Medicine, Biomedical
Science Program, University of Central Florida, Orlando, Florida 32826, United States
| | - Maria Stancescu
- Hybrid Systems Laboratory, NanoScience
Technology Center, University of Central Florida, Orlando, Florida 32826, United States
| | - James J. Hickman
- Hybrid Systems Laboratory, NanoScience
Technology Center, University of Central Florida, Orlando, Florida 32826, United States
- College of Medicine, Biomedical
Science Program, University of Central Florida, Orlando, Florida 32826, United States
| |
Collapse
|