1
|
Kano M, Suga H, Arima H. Induction of Functional Hypothalamus and Pituitary Tissues From Pluripotent Stem Cells for Regenerative Medicine. J Endocr Soc 2020; 5:bvaa188. [PMID: 33604493 PMCID: PMC7880040 DOI: 10.1210/jendso/bvaa188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Indexed: 12/22/2022] Open
Abstract
The hypothalamus and pituitary have been identified to play essential roles in maintaining homeostasis. Various diseases can disrupt the functions of these systems, which can often result in serious lifelong symptoms. The current treatment for hypopituitarism involves hormone replacement therapy. However, exogenous drug administration cannot mimic the physiological changes that are a result of hormone requirements. Therefore, patients are at a high risk of severe hormone deficiency, including adrenal crisis. Pluripotent stem cells (PSCs) self-proliferate and differentiate into all types of cells. The generation of endocrine tissues from PSCs has been considered as another new treatment for hypopituitarism. Our colleagues established a 3-dimensional (3D) culture method for embryonic stem cells (ESCs). In this culture, the ESC-derived aggregates exhibit self-organization and spontaneous formation of highly ordered patterning. Recent results have shown that strict removal of exogenous patterning factors during early differentiation efficiently induces rostral hypothalamic progenitors from mouse ESCs. These hypothalamic progenitors generate vasopressinergic neurons, which release neuropeptides upon exogenous stimulation. Subsequently, we reported adenohypophysis tissue self-formation in 3D cultures of mouse ESCs. The ESCs were found to differentiate into both nonneural oral ectoderm and hypothalamic neuroectoderm in adjacent layers. Interactions between the 2 tissues appear to be critically important for in vitro induction of a Rathke’s pouch-like developing embryo. Various endocrine cells were differentiated from nonneural ectoderm. The induced corticotrophs efficiently secreted adrenocorticotropic hormone when engrafted in vivo, which rescued hypopituitary hosts. For future regenerative medicine, generation of hypothalamic and pituitary tissues from human PSCs is necessary. We and other groups succeeded in establishing a differentiation method with the use of human PSCs. Researchers could use these methods for models of human diseases to elucidate disease pathology or screen potential therapeutics.
Collapse
Affiliation(s)
- Mayuko Kano
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| |
Collapse
|
2
|
Protective Mechanism and Treatment of Neurogenesis in Cerebral Ischemia. Neurochem Res 2020; 45:2258-2277. [PMID: 32794152 DOI: 10.1007/s11064-020-03092-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/18/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the fifth leading cause of death worldwide and is a main cause of disability in adults. Neither currently marketed drugs nor commonly used treatments can promote nerve repair and neurogenesis after stroke, and the repair of neurons damaged by ischemia has become a research focus. This article reviews several possible mechanisms of stroke and neurogenesis and introduces novel neurogenic agents (fibroblast growth factors, brain-derived neurotrophic factor, purine nucleosides, resveratrol, S-nitrosoglutathione, osteopontin, etc.) as well as other treatments that have shown neuroprotective or neurogenesis-promoting effects.
Collapse
|
3
|
Bagheri-Mohammadi S, Karimian M, Alani B, Verdi J, Tehrani RM, Noureddini M. Stem cell-based therapy for Parkinson's disease with a focus on human endometrium-derived mesenchymal stem cells. J Cell Physiol 2018; 234:1326-1335. [PMID: 30146713 DOI: 10.1002/jcp.27182] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) as an increasing clinical syndrome is a multifunctional impairment with systemic involvement. At present, therapeutic approaches such as l-3,4-dihydroxy-phenylalanine replacement therapy, dopaminergic agonist administration, and neurosurgical treatment intend to relieve PD symptoms which are palliative and incompetent in counteracting PD progression. These mentioned therapies have not been able to replace the lost cells and they could not effectively slow down the relentless neurodegenerative process. Till now, there is a lack of eligible treatment for PD, and stem cells therapy recently has been considered for PD treatment. In this review, we demonstrate how human stem cell technology especially human endometrium-derived stem cells have made advancement as a therapeutic source for PD compared with other treatments.
Collapse
Affiliation(s)
- Saeid Bagheri-Mohammadi
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Physiology Research Centre, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Behrang Alani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Rana Moradian Tehrani
- Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahdi Noureddini
- Physiology Research Centre, Kashan University of Medical Sciences, Kashan, Iran.,Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
4
|
Suga H. Application of pluripotent stem cells for treatment of human neuroendocrine disorders. Cell Tissue Res 2018; 375:267-278. [PMID: 30078102 DOI: 10.1007/s00441-018-2880-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
Abstract
The neuroendocrine system is composed of many types of functional cells. Matured cells are generally irreversible to progenitor cells and it is difficult to obtain enough from our body. Therefore, studying specific subtypes of human neuroendocrine cells in vitro has not been feasible. One of the solutions is pluripotent stem cells, such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells. These are unlimited sources and, in theory, are able to give rise to all cell types of our body. Therefore, we can use them for regenerative medicine, developmental basic research and disease modeling. Based on this idea, differentiation methods have been studied for years. Recent studies have successfully induced hypothalamic-like progenitors from mouse and human ES/iPS cells. The induced hypothalamic-like progenitors generated hypothalamic neurons, for instance, vasopressin neurons. Induction to adenohypophysis was also reported in the manner of self-formation by three-dimensional floating cultures. Rathke's pouch-like structures, i.e., pituitary anlage, were self-organized in accordance with pituitary development in embryo. Pituitary hormone-producing cells were subsequently differentiated. The induced corticotrophs secreted adrenocorticotropic hormone in response to corticotropin-releasing hormone. When engrafted in vivo, these cells rescued systemic glucocorticoid levels in hypopituitary mice. These culture methods were characterized by replication of stepwise embryonic differentiation. It is based on the idea of mimicking the molecular environment of embryogenesis. Thanks to these improvements, these days, we can generate hormone-secreting neuroendocrine cells from pluripotent stem cells. The next problems that need to be solved are improving differentiation efficiency even further and structuring networks.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
5
|
Abstract
The hypothalamic-pituitary system is essential for maintaining life and controlling systemic homeostasis. The functional disorder makes patients suffer from various symptoms all their lives. Pluripotent stem cells, such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, differentiate into neuroectodermal progenitors when cultured as floating aggregates under serum-free conditions. Recent results have shown that strict removal of exogenous patterning factors during the early differentiation period induces rostral hypothalamic-like progenitors from mouse ES cells. The use of growth factor-free, chemically defined medium was critical for this induction. The ES cell-derived hypothalamic-like progenitors generated rostral-dorsal hypothalamic neurons, in particular magnocellular vasopressinergic neurons. We subsequently reported self-formation of adenohypophysis in three-dimensional floating cultures of mouse ES cells. The ES cell aggregates were stimulated to differentiate into both non-neural head ectoderm and hypothalamic neuroectoderm in adjacent layers. Self-organization of Rathke's pouch-like structures occurred at the interface of the two epithelia in vitro. Various pituitary endocrine cells including corticotrophs and somatotrophs were subsequently produced from the Rathke's pouch-like structures. The induced corticotrophs efficiently secreted ACTH in response to CRH. Furthermore, when engrafted in vivo, these cells rescued systemic glucocorticoid levels in hypopituitary mice. Our latest study aimed to prepare hypothalamic and pituitary tissues from human pluripotent stem cells. We succeeded in establishing the differentiation method using human ES/iPS cells. The culture method is characterized by replication of stepwise embryonic differentiation. Therefore, these methods could potentially be used as developmental and disease models, as well as for future regenerative medicine.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Hospital, Nagoya 466-8550, Japan
| |
Collapse
|
6
|
Katsukawa M, Nakajima Y, Fukumoto A, Doi D, Takahashi J. Fail-Safe Therapy by Gamma-Ray Irradiation Against Tumor Formation by Human-Induced Pluripotent Stem Cell-Derived Neural Progenitors. Stem Cells Dev 2016; 25:815-25. [DOI: 10.1089/scd.2015.0394] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mitsuko Katsukawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Signal Transductions, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yusuke Nakajima
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Akiko Fukumoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Clinical Neuroscience, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
7
|
Abstract
In Parkinson’s disease (PD), dopamine neurons in the substantia nigra are degenerated and lost. Cell therapy for PD replaces the lost dopamine neurons by transplanting donor dopamine neural progenitor cells. Cell therapy for PD has been performed in the clinic since the 1980s and uses donor cells from the mesencephalon of aborted embryos. Regenerative medicine for PD using induced pluripotent stem (iPS) cell technology is drawing attention, because it offers a limitless and more advantageous source of donor cells than aborted embryos.
Collapse
Affiliation(s)
- Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University
| | | |
Collapse
|
8
|
Li XY, Bao XJ, Wang RZ. Potential of neural stem cell-based therapies for Alzheimer's disease. J Neurosci Res 2015; 93:1313-24. [PMID: 25601591 DOI: 10.1002/jnr.23555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/23/2014] [Accepted: 12/15/2014] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), known to be a leading cause of dementia that causes heavy social and financial burdens worldwide, is characterized by progressive loss of neurons and synaptic connectivity after depositions of amyloid-β (Aβ) protein. Current therapies for AD patients can only alleviate symptoms but cannot deter the neural degeneration, thus providing no long-term recovery. Neural stem cells (NSCs), capable of self-renewal and of differentiation into functional neurons and glia, have been shown to repair damaged networks and reverse memory and learning deficits in animal studies, providing new hope for curing AD patients by cell transplantation. Under AD pathology, the microenvironment also undergoes great alterations that affect the propagation of NSCs and subsequent therapeutic efficiency, calling for measures to improve the hostile environment for cell transplantation. This article reviews the therapeutic potential of both endogenous and exogenous NSCs in the treatment of AD and the challenges to application of stem cells in AD treatment, particularly those from the microenvironmental alterations, in the hope of providing more information for future research in exploiting stem cell-based therapies for AD. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xue-Yuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Xin-Jie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Ren-Zhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
9
|
Suga H. Differentiation of pluripotent stem cells into hypothalamic and pituitary cells. Neuroendocrinology 2015; 101:18-24. [PMID: 25428763 DOI: 10.1159/000369821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/05/2014] [Indexed: 11/19/2022]
Abstract
The hypothalamic-pituitary system is essential to maintain life and control systemic homeostasis, but it is negatively affected by various diseases, leading to serious symptoms. Embryonic stem (ES) cells differentiate into neuroectodermal progenitors when cultured as floating aggregates under serum-free conditions. Recently, our colleagues have shown that strict removal of exogenous patterning factors during early differentiation steps induced efficient generation of rostral hypothalamic-like progenitors from mouse ES cell-derived neuroectodermal cells. The use of growth factor-free chemically defined medium was critical for this induction. The ES cell-derived hypothalamic-like progenitors generated rostral-dorsal hypothalamic neurons, especially magnocellular vasopressinergic neurons that release the hormone upon stimulation. Subsequently, we reported efficient self-formation of 3-dimensional adenohypophysis tissues in aggregate cultures of mouse ES cells. The ES cells were stimulated to differentiate into nonneural head ectoderm and hypothalamic neuroectoderm in adjacent layers within the aggregate and then treated with hedgehog. Self-organization of Rathke's pouch-like structures occurred at the interface of the two epithelia, as observed in vivo, and various endocrine cells including corticotrophs and somatotrophs were subsequently produced. The corticotrophs efficiently secreted adrenocorticotropic hormone in response to corticotropin-releasing hormone. Furthermore, when engrafted in vivo, these cells rescued the systemic glucocorticoid level in hypopituitary mice. Our present research aims are to prepare hypothalamic and pituitary tissues from human induced pluripotent stem cells and establish effective transplantation techniques with clinical applications. To replicate the complex and precise control of the hypothalamic-pituitary system, regenerative medicine using pluripotent cells may be a hopeful option.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
10
|
Guloglu MO, Larsen A, Brundin P. Adipocytes derived from PA6 cells reliably promote the differentiation of dopaminergic neurons from human embryonic stem cells. J Neurosci Res 2014; 92:564-73. [PMID: 24482287 DOI: 10.1002/jnr.23355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 11/26/2013] [Accepted: 11/30/2013] [Indexed: 11/09/2022]
Abstract
The PA6 stromal cell line comprises a heterogeneous population of cells that can induce both mouse and human embryonic stem cells to differentiate into dopaminergic neurons. This ability of PA6 cells has been termed stromal cell-derived inducing activity (SDIA). The level of SDIA has been found to vary considerably between and within batches of PA6 cells. Not only are the molecular mechanisms that underlie SDIA unknown but also the cell type(s) within the heterogeneous PA6 cultures that underlie SDIA remain poorly defined. In this study, we reveal that adipocytes, which are present within the heterogeneous PA6 cell population, robustly release the factors mediating SDIA. Furthermore, we report that the coculture of human embryonic stem cells with PA6-derived adipocytes reliably induces their differentiation into midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- M O Guloglu
- Neuronal Survival Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden
| | | | | |
Collapse
|
11
|
Yang T, Liu LY, Ma YY, Zhang W. Notch signaling-mediated neural lineage selection facilitates intrastriatal transplantation therapy for ischemic stroke by promoting endogenous regeneration in the hippocampus. Cell Transplant 2013; 23:221-38. [PMID: 23295012 DOI: 10.3727/096368912x661355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Acquisition of highly efficient neural differentiation based on understanding of initial lineage commitment of human embryonic stem (hES) cells remains a challenge. This study describes a simple three-stage protocol to induce hES cells into neural lineage cells using a 2-week coculture with murine bone marrow stromal cell (BMSC) PA6 followed by a 2-week propagation culture in PA6-conditioned medium and an additional 2-week selection culture in chemically defined neurobasal medium. This protocol generated a relatively high yield of neural lineage cells without mesodermal and endodermal lineage cell contamination. Notably, we demonstrated that PA6 coculture can significantly enhance the expression level of Notch signaling components and promote neural lineage entry of hES cell derivatives. Manipulation of Notch signaling can boost or suppress neural differentiation of hES cell derivatives, suggesting that Notch signaling may underlie the PA6-mediated neural induction. In vivo studies demonstrated that derived neural cells could improve the cognitive function of ischemic stroke rats. Intrastriatal human neural cell grafts were noted to migrate to damaged cerebral regions, enhance basic fibroblast growth factor production in the hippocampus, and restore the pyramidal neuron density and morphology in the hippocampal CA1 region, although only a small number of human donor cells were present in the hippocampus, suggesting that donor cells can boost hippocampal reconstruction by promoting the endogenous regeneration process. These findings demonstrate a pivotal role for Notch in hES cell fate determination and that manipulation of Notch signaling is therefore likely to be a key factor in taking command of hES cell lineage choice. This study suggested the potential of utilizing PA6 coculture to imitate the embryonic niche for hES cell neural induction via Notch signaling and a high application potential of BMSC-involved protocol, which can yield a whole lineage of human neural cells to promote endogenous regeneration in the hippocampus upon transplantation for potential therapy of ischemic stroke.
Collapse
Affiliation(s)
- Tao Yang
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, P. R. China
| | | | | | | |
Collapse
|
12
|
Moon J, Lee HS, Kang JM, Park J, Leung A, Hong S, Chung S, Kim KS. Stem cell grafting improves both motor and cognitive impairments in a genetic model of Parkinson's disease, the aphakia (ak) mouse. Cell Transplant 2012; 22:1263-79. [PMID: 23031199 DOI: 10.3727/096368912x657242] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell-based cell replacement of lost midbrain dopamine (mDA) neurons is a potential therapy for Parkinson's disease (PD). Toward this goal, it is critical to optimize various aspects of cell transplantation and to assess functional recovery through behavioral tests in validated animal model(s) of PD. At present, cell transplantation studies are being done almost exclusively in neurotoxin-based animal models, because few genetic models of PD exhibit robust mDA neuronal loss. Here we used a genetic model of PD, the aphakia mouse, which demonstrates selective degeneration of mDA neurons in the substantia nigra. We systematically investigated the functional effects of transplanting embryonic stem cell-derived cells at different stages of in vitro differentiation: embryoid body (EB), neural progenitor (NP), and neuronal differentiated (ND) stages. We found that transplantation of NP cells yielded the best outcomes for both survival and behavioral improvement, while transplantation of EB and ND cells resulted in high teratoma-like tumor formation and poor survival, respectively. In behavioral paradigms specific to basal ganglia, the NP cells group prominently improved motor behavioral defects 1 and 2 months posttransplantation. Furthermore, we found that NP cell transplantation also improved cognitive impairments of aphakia mice, as examined by the passive avoidance task. Importantly, these graft-induced functional improvements well correlated with survival of tyrosine hydroxylase-positive DA neurons. Taken together, we propose that the aphakia mouse can serve as a novel and useful platform for cell transplantation studies to assess both neurological and cognitive improvements and that NP stage cells represent an optimal stage for transplantation.
Collapse
Affiliation(s)
- Jisook Moon
- Molecular Neurobiology Laboratory, McLean Hospital/Harvard Medical School, Belmont, MA 02178, USA.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
The synergistic effect of beta-boswellic acid and Nurr1 overexpression on dopaminergic programming of antioxidant glutathione peroxidase-1-expressing murine embryonic stem cells. Neuroscience 2012; 222:404-16. [DOI: 10.1016/j.neuroscience.2012.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 07/05/2012] [Accepted: 07/05/2012] [Indexed: 11/22/2022]
|
14
|
Ogura A, Morizane A, Nakajima Y, Miyamoto S, Takahashi J. γ-secretase inhibitors prevent overgrowth of transplanted neural progenitors derived from human-induced pluripotent stem cells. Stem Cells Dev 2012; 22:374-82. [PMID: 23020188 DOI: 10.1089/scd.2012.0198] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although transplanted pluripotent stem cell-derived neurons can contribute to functional recovery in animal models of Parkinson's disease, the risk of tumor formation hinders clinical applications of this approach. Removing undifferentiated cells from the donor population is critical to reduce tumorigenesis. Moreover, immature neural progenitors in transplants can proliferate unpredictably, resulting in neural overgrowth and long-term risks of compressing the surrounding host tissue. Because Notch signaling plays a role in maintaining the multipotency and proliferative capacity of neural progenitors, we used γ-secretase inhibitors (GSIs) to dampen Notch signaling in human-induced pluripotent stem cell-derived neural progenitors before transplantation and examined the effects on the growth of proliferative grafts. We observed a marked reduction in the percentage of dividing cells and increased neuronal maturation in GSI-treated samples in vitro. Next, grafts were transplanted into the striata of nonobese diabetic/severe combined immune deficiency mice. Histological analyses performed 8 weeks after the operation showed that grafts pretreated with GSIs--N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester or compound E--were significantly smaller than control samples. Immunohistologic analyses revealed that briefly treating the donor population with GSIs not only reduced the graft volume, but also altered the composition of the graft; control grafts showed neural overgrowth with numerous PAX6+ and Ki67+ neural rosettes, whereas GSI-treated samples developed into mature neuronal grafts containing primarily Tubβ3+ cells. These results suggest that pretreating potentially proliferative progenitors with GSIs may improve the safety of cell replacement therapies using pluripotent stem cells.
Collapse
Affiliation(s)
- Aya Ogura
- Department of Biological Repair, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | |
Collapse
|
15
|
Shin E, Palmer MJ, Li M, Fricker RA. GABAergic neurons from mouse embryonic stem cells possess functional properties of striatal neurons in vitro, and develop into striatal neurons in vivo in a mouse model of Huntington's disease. Stem Cell Rev Rep 2012; 8:513-31. [PMID: 21720791 DOI: 10.1007/s12015-011-9290-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disease where GABAergic medium spiny neurons (MSNs) in the striatum degenerate. Embryonic stem cell-derived neural transplantation may provide an appropriate therapy for HD. Here we aimed to develop a suitable protocol to obtain a high percentage of functional GABAergic neurons from mouse embryonic stem cells (mESCs), and then tested their differentiation potential in vivo. The monolayer method was compared with the embryoid body and five stage method for its efficiency in generating GABAergic neurons from mESCs. All three methods yielded a similar percentage of GABAergic neurons from mESCs. Monolayer method-derived GABAergic neurons expressed the MSN marker dopamine- and cyclic AMP-regulated phosphoprotein (DARPP32). The pluripotent stem cell population could be eliminated in vitro by treating cells with puromycin and retinoic acid. Using patch-clamp recordings, the functional properties of GABAergic neurons derived from mESCs were compared to GABAergic neurons derived from primary lateral ganglionic eminence. Both types of neurons showed active membrane properties (voltage-gated Na(+) and K(+) currents, Na(+)-dependent action potentials, and spontaneous postsynaptic currents) and possessed functional glutamatergic receptors and transporters. mESC-derived neural progenitors were transplanted into a mouse model of HD. Grafted cells differentiated to mature neurons expressing glutamate decarboxylase, dopamine type 1 receptors, and DARPP32. Also, neural precursors and dividing populations were found in the grafts. In summary, mESCs are able to differentiate efficiently into functional GABAergic neurons using defined in vitro conditions, and these survive and differentiate following grafting to a mouse model of HD.
Collapse
Affiliation(s)
- Eunju Shin
- Institute for Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK
| | | | | | | |
Collapse
|
16
|
Sasselli V, Micci MA, Kahrig KM, Pasricha PJ. Evaluation of ES-derived neural progenitors as a potential source for cell replacement therapy in the gut. BMC Gastroenterol 2012; 12:81. [PMID: 22735038 PMCID: PMC3412704 DOI: 10.1186/1471-230x-12-81] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 06/26/2012] [Indexed: 01/28/2023] Open
Abstract
Background Stem cell-based therapy has recently been explored for the treatment of disorders of the enteric nervous system (ENS). Pluripotent embryonic stem (ES) cells represent an attractive cell source; however, little or no information is currently available on how ES cells will respond to the gut environment. In this study, we investigated the ability of ES cells to respond to environmental cues derived from the ENS and related tissues, both in vitro and in vivo. Methods Neurospheres were generated from mouse ES cells (ES-NS) and co-cultured with organotypic preparations of gut tissue consisting of the longitudinal muscle layers with the adherent myenteric plexus (LM-MP). Results LM-MP co-culture led to a significant increase in the expression of pan-neuronal markers (βIII-tubulin, PGP 9.5) as well as more specialized markers (peripherin, nNOS) in ES-NS, both at the transcriptional and protein level. The increased expression was not associated with increased proliferation, thus confirming a true neurogenic effect. LM-MP preparations exerted also a myogenic effect on ES-NS, although to a lesser extent. After transplantation in vivo into the mouse pylorus, grafted ES-NS failed to acquire a distinct phenotype al least 1 week following transplantation. Conclusions This is the first study reporting that the gut explants can induce neuronal differentiation of ES cells in vitro and induce the expression of nNOS, a key molecule in gastrointestinal motility regulation. The inability of ES-NS to adopt a neuronal phenotype after transplantation in the gastrointestinal tract is suggestive of the presence of local inhibitory influences that prevent ES-NS differentiation in vivo.
Collapse
Affiliation(s)
- Valentina Sasselli
- Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | |
Collapse
|
17
|
Denham M, Parish CL, Leaw B, Wright J, Reid CA, Petrou S, Dottori M, Thompson LH. Neurons derived from human embryonic stem cells extend long-distance axonal projections through growth along host white matter tracts after intra-cerebral transplantation. Front Cell Neurosci 2012; 6:11. [PMID: 22470319 PMCID: PMC3311135 DOI: 10.3389/fncel.2012.00011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 03/01/2012] [Indexed: 12/17/2022] Open
Abstract
Human pluripotent stem cells have the capacity for directed differentiation into a wide variety of neuronal subtypes that may be useful for brain repair. While a substantial body of research has lead to a detailed understanding of the ability of neurons in fetal tissue grafts to structurally and functionally integrate after intra-cerebral transplantation, we are only just beginning to understand the in vivo properties of neurons derived from human pluripotent stem cells. Here we have utilized the human embryonic stem (ES) cell line Envy, which constitutively expresses green fluorescent protein (GFP), in order to study the in vivo properties of neurons derived from human ES cells. Rapid and efficient neural induction, followed by differentiation as neurospheres resulted in a GFP+ neural precursor population with traits of neuroepithelial and dorsal forebrain identity. Ten weeks after transplantation into neonatal rats, GFP+ fiber patterns revealed extensive axonal growth in the host brain, particularly along host white matter tracts, although innervation of adjacent nuclei was limited. The grafts were composed of a mix of neural cell types including differentiated neurons and glia, but also dividing neural progenitors and migrating neuroblasts, indicating an incomplete state of maturation at 10 weeks. This was reflected in patch-clamp recordings showing stereotypical properties appropriate for mature functional neurons, including the ability to generate action potentials, as well profiles consistent for more immature neurons. These findings illustrate the intrinsic capacity for neurons derived from human ES cells to integrate at a structural and functional level following transplantation.
Collapse
Affiliation(s)
- Mark Denham
- Centre for Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ordonez MP, Roberts EA, Kidwell CU, Yuan SH, Plaisted WC, Goldstein LSB. Disruption and therapeutic rescue of autophagy in a human neuronal model of Niemann Pick type C1. Hum Mol Genet 2012; 21:2651-62. [PMID: 22437840 DOI: 10.1093/hmg/dds090] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An unresolved issue about many neurodegenerative diseases is why neurons are particularly sensitive to defects in ubiquitous cellular processes. One example is Niemann Pick type C1, caused by defects in cholesterol trafficking in all cells, but where neurons are preferentially damaged. Understanding this selective failure is limited by the difficulty in obtaining live human neurons from affected patients. To solve this problem, we generated neurons with decreased function of NPC1 from human embryonic stem cells and used them to test the hypothesis that defective cholesterol handling leads to enhanced pathological phenotypes in neurons. We found that human NPC1 neurons have strong spontaneous activation of autophagy, and, contrary to previous reports in patient fibroblasts, a block of autophagic progression leading to defective mitochondrial clearance. Mitochondrial fragmentation is an exceptionally severe phenotype in NPC1 neurons compared with fibroblasts, causing abnormal accumulation of mitochondrial proteins. Contrary to expectation, these abnormal phenotypes were rescued by treatment with the autophagy inhibitor 3-methyladenine and by treatment with the potential therapeutic cyclodextrin, which mobilizes cholesterol from the lysosomal compartment. Our findings suggest that neurons are especially sensitive to lysosomal cholesterol accumulation because of autophagy disruption and accumulation of fragmented mitochondria, thus defining a new route to effective drug development for NPC1 disease.
Collapse
Affiliation(s)
- M Paulina Ordonez
- Department of Cellular and Molecular Medicine, Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
19
|
Azari H, Osborne GW, Yasuda T, Golmohammadi MG, Rahman M, Deleyrolle LP, Esfandiari E, Adams DJ, Scheffler B, Steindler DA, Reynolds BA. Purification of immature neuronal cells from neural stem cell progeny. PLoS One 2011; 6:e20941. [PMID: 21687800 PMCID: PMC3109004 DOI: 10.1371/journal.pone.0020941] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 05/16/2011] [Indexed: 01/01/2023] Open
Abstract
Large-scale proliferation and multi-lineage differentiation capabilities make neural stem cells (NSCs) a promising renewable source of cells for therapeutic applications. However, the practical application for neuronal cell replacement is limited by heterogeneity of NSC progeny, relatively low yield of neurons, predominance of astrocytes, poor survival of donor cells following transplantation and the potential for uncontrolled proliferation of precursor cells. To address these impediments, we have developed a method for the generation of highly enriched immature neurons from murine NSC progeny. Adaptation of the standard differentiation procedure in concert with flow cytometry selection, using scattered light and positive fluorescent light selection based on cell surface antibody binding, provided a near pure (97%) immature neuron population. Using the purified neurons, we screened a panel of growth factors and found that bone morphogenetic protein-4 (BMP-4) demonstrated a strong survival effect on the cells in vitro, and enhanced their functional maturity. This effect was maintained following transplantation into the adult mouse striatum where we observed a 2-fold increase in the survival of the implanted cells and a 3-fold increase in NeuN expression. Additionally, based on the neural-colony forming cell assay (N-CFCA), we noted a 64 fold reduction of the bona fide NSC frequency in neuronal cell population and that implanted donor cells showed no signs of excessive or uncontrolled proliferation. The ability to provide defined neural cell populations from renewable sources such as NSC may find application for cell replacement therapies in the central nervous system.
Collapse
Affiliation(s)
- Hassan Azari
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Laboratory for Stem Cell Research, Department of Anatomical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, McKnight Brain Institute, The University of Florida, Gainesville, Florida, United States of America
| | - Geoffrey W. Osborne
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Takahiro Yasuda
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia
| | - Mohammad G. Golmohammadi
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Department of Anatomical Sciences, Ardebil University of Medical Sciences, Ardebil, Iran
| | - Maryam Rahman
- Department of Neurosurgery, McKnight Brain Institute, The University of Florida, Gainesville, Florida, United States of America
| | - Loic P. Deleyrolle
- Department of Neurosurgery, McKnight Brain Institute, The University of Florida, Gainesville, Florida, United States of America
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - David J. Adams
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Health Innovations Research Institute, RMIT University, Bundoora, Victoria, Australia
| | - Bjorn Scheffler
- Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Dennis A. Steindler
- Department of Neurosurgery, McKnight Brain Institute, The University of Florida, Gainesville, Florida, United States of America
| | - Brent A. Reynolds
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- Department of Neurosurgery, McKnight Brain Institute, The University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
20
|
Morizane A, Darsalia V, Guloglu MO, Hjalt T, Carta M, Li JY, Brundin P. A simple method for large-scale generation of dopamine neurons from human embryonic stem cells. J Neurosci Res 2011; 88:3467-78. [PMID: 20981866 DOI: 10.1002/jnr.22515] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dopamine (DA) neurons derived from human embryonic stem cells (hESCs) are potentially valuable in drug screening and as a possible source of donor tissue for transplantation in Parkinson's disease. However, existing culture protocols that promote the differentiation of DA neurons from hESCs are complex, involving multiple steps and having unreliable results between cultures. Here we report a simple and highly reproducible culture protocol that induces expandable DA neuron progenitors from hESCs in attached cultures. We found that the hESC-derived neuronal progenitors retain their full capacity to generate DA neurons after repeated passaging in the presence of basic fibroblast growth factor (bFGF) and medium conditioned with PA6 stromal cells. Using immunocytochemistry and RT-PCR, we found that the differentiated DA neurons exhibit a midbrain phenotype and express, e.g., Aldh1a, Ptx3, Nurr1, and Lmx1a. Using HPLC, we monitored their production of DA. We then demonstrated that the expanded progenitors are possible to cryopreserve without loosing the dopaminergic phenotype. With our protocol, we obtained large and homogeneous populations of dopaminergic progenitors and neurons. We conclude that our protocol can be used to generate human DA neurons suitable for the study of disease mechanisms, toxicology, drug screening, and intracerebral transplantation.
Collapse
Affiliation(s)
- Asuka Morizane
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
21
|
Anisimov SV. Cell-based therapeutic approaches for Parkinson's disease: progress and perspectives. Rev Neurosci 2010; 20:347-81. [PMID: 20397620 DOI: 10.1515/revneuro.2009.20.5-6.347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Motor dysfunctions in Parkinson's disease are believed to be primarily due to the degeneration of dopaminergic neurons located in the substantia nigra pars compacta. Because a single-type cell population is depleted, Parkinson's disease is considered a primary target for cell replacement-based therapeutic strategies. Extensive studies have confirmed transplantation of donor neurons could be beneficial, yet identifying an alternative cell source is clearly essential. Human embryonic stem cells (hESCs) have been proposed as a renewable source of dopaminergic neurons for transplantation in Parkinson's disease; other potential sources could include neural stem cells (hNSCs) and adult mesenchymal stem cells (hMSCs). However, numerous difficulties avert practical application of stem cell-based therapeutic approaches for the treatment of Parkinson's disease. Among the latter, ethical, safety (including xeno- and tumor formation-associated risks) and technical issues stand out. This review aims to provide a balanced and updated outlook on various issues associated with stem cells in regard to their potential in the treatment of Parkinson's disease. Essential features of the individual stem cell subtypes, principles of available differentiation protocols, transplantation, and safety issues are discussed extensively.
Collapse
Affiliation(s)
- Sergey V Anisimov
- Department of Intracellular Signalling and Transport, Institute of Cytology, Russian Academy of Sciences and Research, Saint-Petersburg, Russia.
| |
Collapse
|
22
|
Murine embryonic stem cell-derived pyramidal neurons integrate into the cerebral cortex and appropriately project axons to subcortical targets. J Neurosci 2010; 30:894-904. [PMID: 20089898 DOI: 10.1523/jneurosci.4318-09.2010] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Although embryonic stem (ES) cells have been induced to differentiate into diverse neuronal cell types, the production of cortical projection neurons with the correct morphology and axonal connectivity has not been demonstrated. Here, we show that in vitro patterning is critical for generating neural precursor cells (ES-NPCs) competent to form cortical pyramidal neurons. During the first week of neural induction, these ES-NPCs begin to express genes that are specific for forebrain progenitors; an additional week of differentiation produces mature neurons with many features of cortical pyramidal neurons. After transplantation into the murine cerebral cortex, these specified ES-NPCs manifest the correct dendritic and axonal connectivity for their areal location. ES-NPCs transplanted into the deep layers of the motor cortex differentiate into layer 5 pyramidal neurons and extend axons to distant subcortical targets such as the pons and as far caudal as the pyramidal decussation and descending spinal tract and, importantly, do not extend axons to inappropriate targets such as the superior colliculus (SC). ES-NPCs transplanted into the visual cortex extend axons to the dorsal aspect of the SC and pons but avoid ventral SC and the pyramidal tract, whereas cells transplanted deep into the somatosensory cortex project axons to the ventral SC, avoiding the dorsal SC. Thus, these data establish that ES-derived cortical projection neurons can integrate into anatomically relevant circuits.
Collapse
|
23
|
Meyer AK, Maisel M, Hermann A, Stirl K, Storch A. Restorative approaches in Parkinson's Disease: Which cell type wins the race? J Neurol Sci 2010; 289:93-103. [DOI: 10.1016/j.jns.2009.08.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Tremblay RG, Sikorska M, Sandhu JK, Lanthier P, Ribecco-Lutkiewicz M, Bani-Yaghoub M. Differentiation of mouse Neuro 2A cells into dopamine neurons. J Neurosci Methods 2009; 186:60-7. [PMID: 19903493 DOI: 10.1016/j.jneumeth.2009.11.004] [Citation(s) in RCA: 212] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Revised: 10/06/2009] [Accepted: 11/04/2009] [Indexed: 01/19/2023]
Abstract
Neuro 2A (N2a) is a mouse neural crest-derived cell line that has been extensively used to study neuronal differentiation, axonal growth and signaling pathways. A convenient characteristic of these cells is their ability to differentiate into neurons within a few days. However, most differentiation methods reported for N2a cells do not provide information about the neuronal types obtained after each treatment. In this study, we evaluated the generation of N2a dopamine neurons following treatment with a number of factors known to induce neuronal differentiation. Our results showed that N2a cells express Nurr-related factor 1 (Nurr1) and produce low levels of tyrosine hydroxylase (TH) and dopamine. Both TH and dopamine levels were significantly enhanced in the presence of dibutyryl cyclic adenosine monophosphate (dbcAMP), as evidenced by Western blot, immunocytochemistry and high performance liquid chromatography (HPLC). In contrast to dbcAMP, other factors such as transforming growth factor beta1 (TGF beta 1), bone morphogenetic protein 4 (BMP4), glial cell-derived neurotrophic factor (GDNF) and retinoic acid (RA) did not increase TH expression. Further investigation confirmed that the effect of dbcAMP on production of TH-positive neurons was mediated through cyclic AMP (cAMP) responsive element binding protein (CREB) and it was antagonized by RA. Thus, although various treatments can be used to generate N2a neurons, only dbcAMP significantly enhanced the formation of dopamine neurons. Taken together, this study provided a simple and reliable method to generate dopamine neurons for rapid and efficient physiological and pharmacological assays.
Collapse
Affiliation(s)
- Roger G Tremblay
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, Ottawa, ON, K1A 0R6, Canada
| | | | | | | | | | | |
Collapse
|
25
|
Farin A, Liu CY, Langmoen IA, Apuzzo ML. BIOLOGICAL RESTORATION OF CENTRAL NERVOUS SYSTEM ARCHITECTURE AND FUNCTION. Neurosurgery 2009; 65:831-59; discussion 859. [DOI: 10.1227/01.neu.0000351721.81175.0b] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
26
|
Yang T, Tsang KS, Poon WS, Ng HK. Neurotrophism of bone marrow stromal cells to embryonic stem cells: noncontact induction and transplantation to a mouse ischemic stroke model. Cell Transplant 2009; 18:391-404. [PMID: 19622227 DOI: 10.3727/096368909788809767] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Embryonic stem (ES) cell-derived cell products may serve as a source of cells for regenerative medicine. Currently available technologies for the induction of ES cells into neural lineage cells require extended culturing in vitro and complex procedural manipulations, with variable yields of heterogeneous cells, which have hindered the prospective use of cell derivatives for treatment of ischemic stroke. We established a simple and efficient method to derive mouse ES cells into neural lineage cells using an 8-day coculture with the bone marrow stromal cells MS5, followed by a 6-day propagation culture and a 4-day selection culture. The protocol generated a relatively high yield of neural lineage cells without any mesodermal and endodermal lineage commitment. In in vivo study, these derived cells could improve the cognitive function of ischemic stroke mice. Three weeks after transplantation, migration of implanted cells to lesioned areas was noted. It was also evident of a normalization of pyramidal neuron density and morphology in hippocampal CA1 region. One (1/17) episode of teratoma development was noted. Data suggested that MS5 cells may exert a neurotrophic effect to enhance neural differentiation of ES cells and MS5-induced ES cell-derived cells appeared to be applicable to cell therapy for ischemic stroke.
Collapse
Affiliation(s)
- Tao Yang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong; Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | | | | | | |
Collapse
|
27
|
Burns TC, Verfaillie CM, Low WC. Stem cells for ischemic brain injury: a critical review. J Comp Neurol 2009; 515:125-44. [PMID: 19399885 DOI: 10.1002/cne.22038] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
No effective therapy is currently available to promote recovery following ischemic stroke. Stem cells have been proposed as a potential source of new cells to replace those lost due to central nervous system injury, as well as a source of trophic molecules to minimize damage and promote recovery. We undertook a detailed review of data from recent basic science and preclinical studies to investigate the potential application of endogenous and exogenous stem cell therapies for treatment of cerebral ischemia. To date, spontaneous endogenous neurogenesis has been observed in response to ischemic injury, and can be enhanced via infusion of appropriate cytokines. Exogenous stem cells from multiple sources can generate neural cells that survive and form synaptic connections after transplantation in the stroke-injured brain. Stem cells from multiple sources cells also exhibit neuroprotective properties that may ameliorate stroke deficits. In many cases, functional benefits observed are likely independent of neural differentiation, although the exact mechanisms remain poorly understood. Future studies of neuroregeneration will require the demonstration of function in endogenously born neurons following focal ischemia. Further, methods are currently lacking to demonstrate definitively the therapeutic effect of newly introduced neural cells. Increased plasticity following stroke may facilitate the functional integration of new neurons, but the loss of appropriate guidance cues and supporting architecture in the infarct cavity will likely impede the restoration of lost circuitry. Thus careful investigation of the mechanisms underlying trophic benefits will be essential. Evidence to date suggests that continued development of stem cell therapies may ultimately lead to viable treatment options for ischemic brain injury.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
28
|
Efficient production of mesencephalic dopamine neurons by Lmx1a expression in embryonic stem cells. Proc Natl Acad Sci U S A 2009; 106:7613-8. [PMID: 19383789 DOI: 10.1073/pnas.0902396106] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Signaling factors involved in CNS development have been used to control the differentiation of embryonic stem cells (ESCs) into mesencephalic dopamine (mesDA) neurons, but tend to generate a limited yield of desired cell type. Here we show that forced expression of Lmx1a, a transcription factor functioning as a determinant of mesDA neurons during embryogenesis, effectively can promote the generation of mesDA neurons from mouse and human ESCs. Under permissive culture conditions, 75%-95% of mouse ESC-derived neurons express molecular and physiological properties characteristic of bona fide mesDA neurons. Similar to primary mesDA neurons, these cells integrate and innervate the striatum of 6-hydroxy dopamine lesioned neonatal rats. Thus, the enriched generation of functional mesDA neurons by forced expression of Lmx1a may be of future importance in cell replacement therapy of Parkinson disease.
Collapse
|
29
|
Teramura T, Onodera Y, Murakami H, Ito S, Mihara T, Takehara T, Kato H, Mitani T, Anzai M, Matsumoto K, Saeki K, Fukuda K, Sagawa N, Osoi Y. Mouse androgenetic embryonic stem cells differentiated to multiple cell lineages in three embryonic germ layers in vitro. J Reprod Dev 2009; 55:283-92. [PMID: 19305126 DOI: 10.1262/jrd.20146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The embryos of some rodents and primates can precede early development without the process of fertilization; however, they cease to develop after implantation because of restricted expressions of imprinting genes. Asexually developed embryos are classified into parthenote/gynogenote and androgenote by their genomic origins. Embryonic stem cells (ESCs) derived from asexual origins have also been reported. To date, ESCs derived from parthenogenetic embryos (PgESCs) have been established in some species, including humans, and the possibility to be alternative sources for autologous cell transplantation in regenerative medicine has been proposed. However, some developmental characteristics, which might be important for therapeutic applications, such as multiple differentiation capacity and transplantability of the ESCs of androgenetic origin (AgESCs) are uncertain. Here, we induced differentiation of mouse AgESCs and observed derivation of neural cells, cardiomyocytes and hepatocytes in vitro. Following differentiated embryoid body (EB) transplantation in various mouse strains including the strain of origin, we found that the EBs could engraft in theoretically MHC-matched strains. Our results indicate that AgESCs possess at least two important characteristics, multiple differentiation properties in vitro and transplantability after differentiation, and suggest that they can also serve as a source of histocompatible tissues for transplantation.
Collapse
Affiliation(s)
- Takeshi Teramura
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Mie University.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kuai XL, Gagliardi C, Flaat M, Bunnell BA. Differentiation of nonhuman primate embryonic stem cells along neural lineages. Differentiation 2009; 77:229-238. [PMID: 19272521 PMCID: PMC2749555 DOI: 10.1016/j.diff.2008.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 09/30/2008] [Accepted: 10/06/2008] [Indexed: 11/22/2022]
Abstract
The differentiation of embryonic stem cells (ESCs) into neurons and glial cells represents a promising cell-based therapy for neurodegenerative diseases. Because the rhesus macaque is physiologically and phylogenetically similar to humans, it is a clinically relevant animal model for ESC research. In this study, the pluripotency and neural differentiation potential of a rhesus monkey ESC line (ORMES6) was investigated. ORMES6 was derived from an in vitro produced blastocyst, which is the same way human ESCs have been derived. ORMES6 stably expressed the embryonic transcription factors POU5F1 (Oct4), Sox2 and NANOG. Stage-specific embryonic antigen 4 (SSEA 4) and the glycoproteins TRA-1-60 and TRA-1-81 were also expressed. The embryoid bodies (EBs) formed from ORMES6 ESCs spontaneously gave rise to cells of three germ layers. After exposure to basic fibroblast growth factor (bFGF) for 14-16 days, columnar rosette cells formed in the EB outgrowths. Sox2, microtubule-associated protein (MAP2), beta-tublinIII and glial fibrillary acidic protein (GFAP) genes and Nestin, FoxD3, Pax6 and beta-tublinIII antigens were expressed in the rosette cells. Oct4 and NANOG expression were remarkably down-regulated in these cells. After removal of bFGF from the medium, the rosette cells differentiated along neural lineages. The differentiated cells expressed MAP2, beta-tublinIII, Neuro D and GFAP genes. Most differentiated cells expressed early neuron-specific antigen beta-tublinIII (73+/-4.7%) and some expressed intermediate neuron antigen MAP2 (18+/-7.2%). However, some differentiated cells expressed the glial cell antigens A2B5 (7.17%+/-1.2%), GFAP (4.93+/-1.9%), S100 (7+/-3.5%) and O4 (0.27+/-0.2%). The rosette cells were transplanted into the striatum of immune-deficient NIHIII mice. The cells persisted for approximately 2 weeks and expressed Ki67, NeuN, MAP2 and GFAP. These results demonstrate that the rhesus monkey ESC line ORMES6 retains the pluripotent characteristics of ESCs and can be efficiently induced to differentiate along neural lineages.
Collapse
Affiliation(s)
- Xiao Ling Kuai
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, USA
| | - Christine Gagliardi
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, USA
- Department of Pharmacology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, USA
| | - Mette Flaat
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, USA
- Department of Biochemistry, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, USA
| | - Bruce A. Bunnell
- Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, USA
- Department of Pharmacology, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, USA
- Center for Gene Therapy, Tulane University Health Sciences Center, Tulane University, New Orleans, LA, USA
| |
Collapse
|
31
|
Streckfuss-Bömeke K, Vlasov A, Hülsmann S, Yin D, Nayernia K, Engel W, Hasenfuss G, Guan K. Generation of functional neurons and glia from multipotent adult mouse germ-line stem cells. Stem Cell Res 2009; 2:139-54. [DOI: 10.1016/j.scr.2008.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Revised: 09/10/2008] [Accepted: 09/14/2008] [Indexed: 11/24/2022] Open
|
32
|
Protocols for generating ES cell-derived dopamine neurons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 651:101-11. [PMID: 19731555 DOI: 10.1007/978-1-4419-0322-8_10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Shintani A, Nakao N, Kakishita K, Itakura T. Generation of dopamine neurons from embryonic stem cells in the presence of the neuralizing activity of bone marrow stromal cells derived from adult mice. J Neurosci Res 2008; 86:2829-38. [DOI: 10.1002/jnr.21748] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Bartosh T, Wang Z, Rosales AA, Dimitrijevich SD, Roque RS. 3D-model of adult cardiac stem cells promotes cardiac differentiation and resistance to oxidative stress. J Cell Biochem 2008; 105:612-23. [DOI: 10.1002/jcb.21862] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
35
|
Ideguchi M, Shinoyama M, Gomi M, Hayashi H, Hashimoto N, Takahashi J. Immune or inflammatory response by the host brain suppresses neuronal differentiation of transplanted ES cell-derived neural precursor cells. J Neurosci Res 2008; 86:1936-43. [PMID: 18335525 DOI: 10.1002/jnr.21652] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Embryonic stem (ES) cells are a promising donor source for transplantation therapy, but several problems must be solved before they can be clinically useful. One of these is the host immune reaction to allogeneic grafts. In this article, we examine the effect of the host immune reaction on survival and differentiation of grafted ES cell-derived neural precursor cells (NPCs). We induced NPCs from mouse ES cells by stromal cell-derived inducing activity and then transplanted them into mouse brains with or without administering the immunosuppressant cyclosporine A (CsA). Two and 8 weeks following transplantation, the accumulation of host-derived microglia/macrophages and lymphocytes was observed around the graft. This effect was reduced by CsA treatment, although no significant difference in graft volume was observed. These data suggest that an immune response occurs in allografts of ES cell-derived NPCs. Intriguingly, however, the ratio of neurons to astrocytes in the graft was higher in immunosuppressed mice. Because inflammatory or immune cells produce various cytokines, we examined the effect of IL-1beta, IL-6, IFN-gamma, and TNF-alpha on the differentiation of NPCs in vitro. Only IL-6 promoted glial cell fate, and this effect could be reversed by the addition of an IL-6 neutralizing antibody. These results suggest that allogeneic ES cell-derived NPCs can cause an immune response by the host brain, but it is not strong enough to reject the graft. More important, activated microglia and lymphocytes can suppress neuronal differentiation of grafted NPCs in vivo by producing cytokines such as IL-6.
Collapse
Affiliation(s)
- Makoto Ideguchi
- Department of Neurosurgery, Clinical Neuroscience, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Pomp O, Brokhman I, Ziegler L, Almog M, Korngreen A, Tavian M, Goldstein RS. PA6-induced human embryonic stem cell-derived neurospheres: a new source of human peripheral sensory neurons and neural crest cells. Brain Res 2008; 1230:50-60. [PMID: 18671952 DOI: 10.1016/j.brainres.2008.07.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Revised: 07/01/2008] [Accepted: 07/04/2008] [Indexed: 11/19/2022]
Abstract
Human embryonic stem cells (hESC) have been directed to differentiate into CNS cells with clinical importance. However, for study of development and regeneration of the human PNS, and peripheral neuropathies, it would be useful to have a source of human PNS derivatives. We have demonstrated that peripheral sensory neuron-like cells (PSN) can also be derived from hESC via neural crest-like (NC) intermediates, and from neural progenitors induced from hESC using noggin. Here we report the generation of higher purity PSN from passagable neurospheres (NSP) induced by murine PA6 stromal cells. hESC were cultured with PA6, and colonies that developed a specific morphology were cut from the plates. Culture of these colonies under non-adhesive conditions yielded NSPs. Several NC marker genes were expressed in the NSP, and these were also detected in 3-5week gestation human embryos containing migrating NC. These NSPs passaged for 2-8weeks and re-plated on PA6 gave rise to many Brn3a+/peripherin+ cells, characteristic of early sensory-like neurons. Re-culturing PA6-induced NSP cells with PA6 resulted in about 25% of the human cells in the co-cultures differentiating to PSN after 1week, compared to only about 10% PSN obtained after 3 weeks when noggin-induced NSP were used. Two month adherent cultures of PA6-induced NSP cells contained neurons expressing several PSN neuropeptides, and voltage-dependent currents and action potentials were obtained from a molecularly identified PSN. hESC-derived PA6-induced NSP cells are therefore an excellent potential source of human PSN for study of differentiation and modeling of PNS disease.
Collapse
Affiliation(s)
- Oz Pomp
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900 Ramat-Gan, Israel
| | | | | | | | | | | | | |
Collapse
|
37
|
Emerging restorative treatments for Parkinson's disease. Prog Neurobiol 2008; 85:407-32. [PMID: 18586376 DOI: 10.1016/j.pneurobio.2008.05.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 04/03/2008] [Accepted: 05/06/2008] [Indexed: 01/18/2023]
Abstract
Several exciting approaches for restorative therapy in Parkinson's disease have emerged over the past two decades. This review initially describes experimental and clinical data regarding growth factor administration. We focus on glial cell line-derived neurotrophic factor (GDNF), particularly its role in neuroprotection and in regeneration in Parkinson's disease. Thereafter, we discuss the challenges currently facing cell transplantation in Parkinson's disease and briefly consider the possibility to continue testing intrastriatal transplantation of fetal dopaminergic progenitors clinically. We also give a more detailed overview of the developmental biology of dopaminergic neurons and the potential of certain stem cells, i.e. neural and embryonic stem cells, to differentiate into dopaminergic neurons. Finally, we discuss adult neurogenesis as a potential tool for restoring lost dopamine neurons in patients suffering from Parkinson's disease.
Collapse
|
38
|
Koyanagi M, Takahashi J, Arakawa Y, Doi D, Fukuda H, Hayashi H, Narumiya S, Hashimoto N. Inhibition of the Rho/ROCK pathway reduces apoptosis during transplantation of embryonic stem cell-derived neural precursors. J Neurosci Res 2008; 86:270-80. [PMID: 17828770 DOI: 10.1002/jnr.21502] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Rho-GTPase has been implicated in the apoptosis of many cell types, including neurons, but the mechanism by which it acts is not fully understood. Here, we investigate the roles of Rho and ROCK in apoptosis during transplantation of embryonic stem cell-derived neural precursor cells. We find that dissociation of neural precursors activates Rho and induces apoptosis. Treatment with the Rho inhibitor C3 exoenzyme and/or the ROCK inhibitor Y-27632 decreases the amount of dissociation-induced apoptosis (anoikis) by 20-30%. Membrane blebbing, which is an early morphological sign of apoptosis; cleavage of caspase-3; and release of cytochrome c from the mitochondria are also reduced by ROCK inhibition. These results suggest that dissociation of neural precursor cells elicits an intrinsic pathway of cell death that is at least partially mediated through the Rho/ROCK pathway. Moreover, in an animal transplantation model, inhibition of Rho and/or ROCK suppresses acute apoptosis of grafted cells. After transplantation, tumor necrosis factor-alpha and pro-nerve growth factor are strongly expressed around the graft. ROCK inhibition also suppresses apoptosis enhanced by these inflammatory cytokines. Taken together, these results indicate that inhibition of Rho/ROCK signaling may improve survival of grafted cells in cell replacement therapy.
Collapse
Affiliation(s)
- Masaomi Koyanagi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hayashi H, Morizane A, Koyanagi M, Ono Y, Sasai Y, Hashimoto N, Takahashi J. Meningeal cells induce dopaminergic neurons from embryonic stem cells. Eur J Neurosci 2008; 27:261-8. [PMID: 18215228 DOI: 10.1111/j.1460-9568.2008.06027.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neural induction of midbrain dopaminergic (DA) neurons from embryonic stem (ES) cells can be achieved by culturing them on a bone marrow-derived stromal cell line, PA6, which possesses stromal cell-derived inducing activity (SDIA). The mechanism of SDIA is unknown, but clinical application of ES cell transplantation requires the use of defined factors for DA neuron induction. Here, we demonstrate that meningeal cells harvested from the developing dura can induce DA neuron differentiation from mouse and human ES cells, as assessed by midbrain DA marker expression and secretion of DA in response to potassium stimuli. Intriguingly, the inductive strength of meningeal cells depends on their developmental stage, with those harvested from embryonic day 18 embryos showing the highest activity. Among six soluble factors known to be involved in DA neuron differentiation, only Wnt-5a and transforming growth factor-beta3 were expressed by both meningeal and PA6 cells, and the expression of Wnt-5a correlated with the DA neuron induction activity of these cells. Furthermore, the induction of DA neuron differentiation by PA6 cell-conditioned medium was reversed by addition of a Wnt-5a neutralizing antibody, whereas recombinant Wnt-5a promoted DA neuron induction when cells were cultured on Matrigel. These results indicate that meningeal cells can be used as feeders to induce DA neurons from ES cells, and that Wnt-5a plays an important role in DA neuron induction by SDIA.
Collapse
Affiliation(s)
- Hideki Hayashi
- Department of Biological Repair, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Anwar MR, Andreasen CM, Lippert SK, Zimmer J, Martinez-Serrano A, Meyer M. Dopaminergic differentiation of human neural stem cells mediated by co-cultured rat striatal brain slices. J Neurochem 2008; 105:460-70. [DOI: 10.1111/j.1471-4159.2007.05164.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Gong J, Sagiv O, Cai H, Tsang SH, Del Priore LV. Effects of extracellular matrix and neighboring cells on induction of human embryonic stem cells into retinal or retinal pigment epithelial progenitors. Exp Eye Res 2008; 86:957-65. [PMID: 18472095 DOI: 10.1016/j.exer.2008.03.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 01/31/2008] [Accepted: 03/18/2008] [Indexed: 11/17/2022]
Abstract
To determine the effects of extracellular matrix and neighboring cells on the differentiation of human embryonic stem cells (hESC) into progenitors of retinal cells and/or retinal pigment epithelium (RPE). HESC were cultured on mouse PA6 stromal cells for approximately 2weeks to obtain neural progenitors. To induce photoreceptor marker expression, the neural progenitors were cultured on a confluent monolayer of ARPE19 or on laminin-coated dishes. To induce RPE markers, the neural progenitors were seeded onto human Bruch's membrane or Matrigel. Cells were examined morphologically and stained with different RPE or neural progenitor markers. Microarray techniques were used to compare the gene expression profiles of hESC cultured on mouse fibroblasts or neural progenitors on PA6 cells to the transcriptome of the adult neural retina and RPE. HESC cultured on PA6 cells expressed neural progenitor markers beta-tubulin III, PAX6, neural filament, GFAP and vimentin. Culturing these neural progenitors on confluent ARPE19 monolayer induced expression of the photoreceptor progenitor cell marker CRX; culturing neural progenitors on laminin substrates induced a neuronal phenotype with neurite formation. Neural progenitors expressed the RPE marker ZO-1 after culturing on Matrigel-coated dishes and the RPE marker Bestrophin after culturing on human Bruch's membrane explants. Hierarchical clustering analysis of samples suggested that when cultured on PA6 stromal cells hESC exhibited genetic characteristics towards differentiating into neural retina. Microarray analysis showed that after culturing on PA6 cells, stem cells expressed 117 new genes; among these there were 22 genes present in neural retina or RPE cells. The functions of these genes were highly related to cell proliferation, nervous system development and cell adhesion. HESC can be induced to differentiate into neural progenitors after culturing on PA6 cells. These neural progenitors can express RPE markers when cultured on Bruch's membrane or Matrigel, or photoreceptor markers when cultured on confluent ARPE19 or laminin. Additional studies are required to assess the function of hESC induced to express retinal or RPE markers prior to successful intraocular transplantation into animal models of retinal degeneration.
Collapse
Affiliation(s)
- Jie Gong
- Department of Ophthalmology, Harkness Eye Institute, Columbia University, 635 West 165th Street, New York, USA
| | | | | | | | | |
Collapse
|
42
|
Derive and conquer: sourcing and differentiating stem cells for therapeutic applications. Nat Rev Drug Discov 2008; 7:131-42. [PMID: 18079756 DOI: 10.1038/nrd2403] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although great progress has been made in the isolation and culture of stem cells, the future of stem-cell-based therapies and their productive use in drug discovery and regenerative medicine depends on two key factors: finding reliable sources of multipotent and pluripotent cells and the ability to control their differentiation to generate desired derivatives. It is essential for clinical applications to establish reliable sources of pathogen-free human embryonic stem cells (ESCs) and develop suitable differentiation techniques. Here, we address some of the problems associated with the sourcing of human ESCs and discuss the current status of stem-cell differentiation technology.
Collapse
|
43
|
Hall VJ, Li JY, Brundin P. Restorative cell therapy for Parkinson's disease: A quest for the perfect cell. Semin Cell Dev Biol 2007; 18:859-69. [DOI: 10.1016/j.semcdb.2007.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Accepted: 09/05/2007] [Indexed: 12/09/2022]
|
44
|
Abstract
Certain regions of the adult brain have the ability for partial self-repair after injury through production of new neurons via activation of neural stem/progenitor cells (NSCs). Nonetheless, there is no evidence yet for pervasive spontaneous replacement of dead neurons by newly formed neurons leading to functional recovery in the injured brain. Consequently, there is enormous interest for stimulating endogenous NSCs in the brain to produce new neurons or for grafting of NSCs isolated and expanded from different brain regions or embryonic stem cells into the injured brain. Temporal lobe epilepsy (TLE), characterized by hyperexcitability in the hippocampus and spontaneous seizures, is a possible clinical target for stem cell-based therapies. This is because these approaches have the potential to curb epileptogenesis and prevent chronic epilepsy development and learning and memory dysfunction after hippocampal damage related to status epilepticus or head injury. Grafting of NSCs may also be useful for restraining seizures during chronic epilepsy. The aim of this review is to evaluate current knowledge and outlook pertaining to stem cell-based therapies for TLE. The first section discusses the behavior of endogenous hippocampal NSCs in human TLE and animal models of TLE and evaluates the role of hippocampal neurogenesis in the pathophysiology and treatment of TLE. The second segment considers the prospects for preventing or suppressing seizures in TLE using exogenously applied stem cells. The final part analyzes problems that remain to be resolved before initiating clinical application of stem cell-based therapies for TLE. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ashok K Shetty
- Department of Surgery (Neurosurgery), Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
45
|
Itoh T, Satou T, Nishida S, Hashimoto S, Ito H. Cultured Rat Astrocytes Give Rise to Neural Stem Cells. Neurochem Res 2006; 31:1381-7. [PMID: 17053966 DOI: 10.1007/s11064-006-9186-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Accepted: 09/26/2006] [Indexed: 11/29/2022]
Abstract
Previously, we reported the occurrence of neural stem cells (NSCs) around an area of damage after rat traumatic brain injury (TBI), but it was unclear if this was due to blastgenesis in astrocytes, or to NSCs migrating from the subventricular zone (SVZ). In this study, NSCs were isolated and cultured from cultured type 1 astrocytes taken from newborn rat cortex in which the subventricular zone and hippocampus had been discarded. All cultured type 1 astrocytes showed glial fibrillary acidic protein (GFAP) immunopositivity. Nestin immunopositive spheres were isolated from type 1 astrocytes and cultured in the presence of bFGF and EGF in the medium. Neurospheres differentiated into Tuj1-, GFAP- and A2B5-positive cells after 4 days of culture without bFGF and EGF. These results indicate that isolated neurospheres from brain cortex astrocytes can differentiate into neurons and glia and might contribute to neurogenesis and neuroplasticity.
Collapse
Affiliation(s)
- Tatsuki Itoh
- Department of Pathology, Kinki University School of Medicine, Osaka, Japan.
| | | | | | | | | |
Collapse
|