1
|
Kjell J, Fischer-Sternjak J, Thompson AJ, Friess C, Sticco MJ, Salinas F, Cox J, Martinelli DC, Ninkovic J, Franze K, Schiller HB, Götz M. Defining the Adult Neural Stem Cell Niche Proteome Identifies Key Regulators of Adult Neurogenesis. Cell Stem Cell 2021; 26:277-293.e8. [PMID: 32032526 PMCID: PMC7005820 DOI: 10.1016/j.stem.2020.01.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/24/2019] [Accepted: 01/02/2020] [Indexed: 12/22/2022]
Abstract
The mammalian brain contains few niches for neural stem cells (NSCs) capable of generating new neurons, whereas other regions are primarily gliogenic. Here we leverage the spatial separation of the sub-ependymal zone NSC niche and the olfactory bulb, the region to which newly generated neurons from the sub-ependymal zone migrate and integrate, and present a comprehensive proteomic characterization of these regions in comparison to the cerebral cortex, which is not conducive to neurogenesis and integration of new neurons. We find differing compositions of regulatory extracellular matrix (ECM) components in the neurogenic niche. We further show that quiescent NSCs are the main source of their local ECM, including the multi-functional enzyme transglutaminase 2, which we show is crucial for neurogenesis. Atomic force microscopy corroborated indications from the proteomic analyses that neurogenic niches are significantly stiffer than non-neurogenic parenchyma. Together these findings provide a powerful resource for unraveling unique compositions of neurogenic niches.
Collapse
Affiliation(s)
- Jacob Kjell
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany
| | - Judith Fischer-Sternjak
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany
| | - Amelia J Thompson
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| | - Christian Friess
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany
| | - Matthew J Sticco
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Favio Salinas
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Jürgen Cox
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - David C Martinelli
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA
| | - Jovica Ninkovic
- Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany; Division of Cell Biology and Anatomy, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; SYNERGY, Excellence Cluster Systems Neurology, Ludwig-Maximilians-Universitaet, Muenchen, Germany
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| | - Herbert B Schiller
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany; Institute of Lung Biology and Disease, Member of the German Center for Lung Research, Helmholtz Zentrum Muenchen, Germany
| | - Magdalena Götz
- Division of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universitaet, Muenchen, Germany; Institute for Stem Cell Research, Helmholtz Zentrum Muenchen, Germany; SYNERGY, Excellence Cluster Systems Neurology, Ludwig-Maximilians-Universitaet, Muenchen, Germany.
| |
Collapse
|
2
|
Scalabrino G. Epidermal Growth Factor in the CNS: A Beguiling Journey from Integrated Cell Biology to Multiple Sclerosis. An Extensive Translational Overview. Cell Mol Neurobiol 2020; 42:891-916. [PMID: 33151415 PMCID: PMC8942922 DOI: 10.1007/s10571-020-00989-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
This article reviews the wealth of papers dealing with the different effects of epidermal growth factor (EGF) on oligodendrocytes, astrocytes, neurons, and neural stem cells (NSCs). EGF induces the in vitro and in vivo proliferation of NSCs, their migration, and their differentiation towards the neuroglial cell line. It interacts with extracellular matrix components. NSCs are distributed in different CNS areas, serve as a reservoir of multipotent cells, and may be increased during CNS demyelinating diseases. EGF has pleiotropic differentiative and proliferative effects on the main CNS cell types, particularly oligodendrocytes and their precursors, and astrocytes. EGF mediates the in vivo myelinotrophic effect of cobalamin on the CNS, and modulates the synthesis and levels of CNS normal prions (PrPCs), both of which are indispensable for myelinogenesis and myelin maintenance. EGF levels are significantly lower in the cerebrospinal fluid and spinal cord of patients with multiple sclerosis (MS), which probably explains remyelination failure, also because of the EGF marginal role in immunology. When repeatedly administered, EGF protects mouse spinal cord from demyelination in various experimental models of autoimmune encephalomyelitis. It would be worth further investigating the role of EGF in the pathogenesis of MS because of its multifarious effects.
Collapse
Affiliation(s)
- Giuseppe Scalabrino
- Department of Biomedical Sciences, University of Milan, Via Mangiagalli 31, 20133, Milan, Italy.
| |
Collapse
|
3
|
Lepko T, Pusch M, Müller T, Schulte D, Ehses J, Kiebler M, Hasler J, Huttner HB, Vandenbroucke RE, Vandendriessche C, Modic M, Martin‐Villalba A, Zhao S, LLorens‐Bobadilla E, Schneider A, Fischer A, Breunig CT, Stricker SH, Götz M, Ninkovic J. Choroid plexus-derived miR-204 regulates the number of quiescent neural stem cells in the adult brain. EMBO J 2019; 38:e100481. [PMID: 31304985 PMCID: PMC6717894 DOI: 10.15252/embj.2018100481] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of adult neural stem cell (NSC) number is critical for lifelong neurogenesis. Here, we identified a post-transcriptional control mechanism, centered around the microRNA 204 (miR-204), to control the maintenance of quiescent (q)NSCs. miR-204 regulates a spectrum of transcripts involved in cell cycle regulation, neuronal migration, and differentiation in qNSCs. Importantly, inhibition of miR-204 function reduced the number of qNSCs in the subependymal zone (SEZ) by inducing pre-mature activation and differentiation of NSCs without changing their neurogenic potential. Strikingly, we identified the choroid plexus of the mouse lateral ventricle as the major source of miR-204 that is released into the cerebrospinal fluid to control number of NSCs within the SEZ. Taken together, our results describe a novel mechanism to maintain adult somatic stem cells by a niche-specific miRNA repressing activation and differentiation of stem cells.
Collapse
Affiliation(s)
- Tjasa Lepko
- Institute of Stem Cell ResearchHelmholtz Center MunichNeuherbergGermany
- Graduate School of Systemic NeurosciencesLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Physiological GenomicsBiomedical CenterMedical FacultyLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
| | - Melanie Pusch
- Institute of Stem Cell ResearchHelmholtz Center MunichNeuherbergGermany
| | - Tamara Müller
- Institute of Neurology (Edinger Institute)University HospitalGoethe University FrankfurtFrankfurtGermany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute)University HospitalGoethe University FrankfurtFrankfurtGermany
| | - Janina Ehses
- Department for Cell Biology and AnatomyBiomedical CenterLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
| | - Michael Kiebler
- Department for Cell Biology and AnatomyBiomedical CenterLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
| | - Julia Hasler
- Institute of Stem Cell ResearchHelmholtz Center MunichNeuherbergGermany
| | - Hagen B Huttner
- Department of NeurologyUniversity Hospital ErlangenFriedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Ghent Gut Inflammation Group (GGIG)Ghent UniversityGhentBelgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
- Ghent Gut Inflammation Group (GGIG)Ghent UniversityGhentBelgium
| | - Miha Modic
- The Francis Crick InstituteLondonUK
- Department for Neuromuscular DiseasesUCL Queen Square Institute of NeurologyLondonUK
| | | | - Sheng Zhao
- Molecular NeurobiologyGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Anja Schneider
- Translational Dementia Research GroupGerman Center for Neurodegenerative Diseases (DZNE) BonnBonnGermany
- Department of Neurodegenerative Diseases and Geriatric PsychiatryUniversity Clinic BonnBonnGermany
| | - Andre Fischer
- Department for Epigenetics and Systems MedicineGerman Center for Neurodegenerative Diseases (DZNE) GöttingenGöttingenGermany
| | - Christopher T Breunig
- MCN Junior Research GroupMunich Center for NeurosciencesBioMedical CenterLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Epigenetic EngineeringHelmholtz Zentrum MünchenNeuherbergGermany
| | - Stefan H Stricker
- MCN Junior Research GroupMunich Center for NeurosciencesBioMedical CenterLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Epigenetic EngineeringHelmholtz Zentrum MünchenNeuherbergGermany
| | - Magdalena Götz
- Institute of Stem Cell ResearchHelmholtz Center MunichNeuherbergGermany
- Physiological GenomicsBiomedical CenterMedical FacultyLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Jovica Ninkovic
- Institute of Stem Cell ResearchHelmholtz Center MunichNeuherbergGermany
- Physiological GenomicsBiomedical CenterMedical FacultyLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Department for Cell Biology and AnatomyBiomedical CenterLudwig‐Maximilians UniversitaetPlanegg‐MartinsriedGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| |
Collapse
|
4
|
Magistri M, Khoury N, Mazza EMC, Velmeshev D, Lee JK, Bicciato S, Tsoulfas P, Faghihi MA. A comparative transcriptomic analysis of astrocytes differentiation from human neural progenitor cells. Eur J Neurosci 2016; 44:2858-2870. [PMID: 27564458 DOI: 10.1111/ejn.13382] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/25/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
Abstract
Astrocytes are a morphologically and functionally heterogeneous population of cells that play critical roles in neurodevelopment and in the regulation of central nervous system homeostasis. Studies of human astrocytes have been hampered by the lack of specific molecular markers and by the difficulties associated with purifying and culturing astrocytes from adult human brains. Human neural progenitor cells (NPCs) with self-renewal and multipotent properties represent an appealing model system to gain insight into the developmental genetics and function of human astrocytes, but a comprehensive molecular characterization that confirms the validity of this cellular system is still missing. Here we used an unbiased transcriptomic analysis to characterize in vitro culture of human NPCs and to define the gene expression programs activated during the differentiation of these cells into astrocytes using FBS or the combination of CNTF and BMP4. Our results demonstrate that in vitro cultures of human NPCs isolated during the gliogenic phase of neurodevelopment mainly consist of radial glial cells (RGCs) and glia-restricted progenitor cells. In these cells the combination of CNTF and BMP4 activates the JAK/STAT and SMAD signaling cascades, leading to the inhibition of oligodendrocytes lineage commitment and activation of astrocytes differentiation. On the other hand, FBS-derived astrocytes have properties of reactive astrocytes. Our work suggests that in vitro culture of human NPCs represents a valuable cellular system to study human disorders characterized by impairment of astrocytes development and function. Our datasets represent an important resource for researchers studying human astrocytes development and might set the basis for the discovery of novel human-specific astrocyte markers.
Collapse
Affiliation(s)
- Marco Magistri
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Nathalie Khoury
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Emilia Maria Cristina Mazza
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Dmitry Velmeshev
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Pantelis Tsoulfas
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mohammad Ali Faghihi
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| |
Collapse
|
5
|
Götz M. Glial Cells Generate Neurons—Master Control within CNS Regions: Developmental Perspectives on Neural Stem Cells. Neuroscientist 2016; 9:379-97. [PMID: 14580122 DOI: 10.1177/1073858403257138] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A common problem in neural stem cell research is the poor generation of neuronal or oligodendroglial descendants. The author takes a developmental perspective to propose solutions to this problem. After a general overview of the recent progress in developmental neurobiology, she highlights the necessity of the sequential and hierarchical specification of CNS precursors toward the generation of specific cell types, for example, neurons. In the developing as well as the adult CNS, multipotent stem cells do not directly generate neurons but give rise to precursors that are specified and restricted toward the generation of neurons. Some molecular determinants of this fate restriction have been identified during recent years and reveal that progression via this fate-restricted state is a necessary step of neurogenesis. These discoveries also demonstrate that neuronal fate specification is inseparably linked at the molecular level to regionalization of the developing CNS. These fate determinants and their specific action in distinct region-specific con-texts are essential to direct the progeny of stem cells more efficiently toward the generation of the desired cell types. Recent data are discussed that demonstrate the common identity of precursors and stem cells in the developing and adult nervous system as radial glia, astroglia, or non-myelinating glia. A novel line-age model is proposed that incorporates these new views and explains why the default pathway of stem cells is astroglia. These new insights into the cellular and molecular mechanisms of neurogenesis help to design novel approaches for reconstitutive therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Magdalena Götz
- Max-Planck Institute of Neurobiology, Planegg-Martinsried/Munich, Germany.
| |
Collapse
|
6
|
The proteome of the differentiating mesencephalic progenitor cell line CSM14.1 in vitro. BIOMED RESEARCH INTERNATIONAL 2014; 2014:351821. [PMID: 24592386 PMCID: PMC3925624 DOI: 10.1155/2014/351821] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 12/16/2013] [Indexed: 11/23/2022]
Abstract
The treatment of Parkinson's disease by transplantation of dopaminergic (DA) neurons from human embryonic mesencephalic tissue is a promising approach. However, the origin of these cells causes major problems: availability and standardization of the graft. Therefore, the generation of unlimited numbers of DA neurons from various types of stem or progenitor cells has been brought into focus. A source for DA neurons might be conditionally immortalized progenitor cells. The temperature-sensitive immortalized cell line CSM14.1 derived from the mesencephalon of an embryonic rat has been used successfully for transplantation experiments. This cell line was analyzed by unbiased stereology of cell type specific marker proteins and 2D-gel electrophoresis followed by mass spectrometry to characterize the differentially expressed proteome. Undifferentiated CSM14.1 cells only expressed the stem cell marker nestin, whereas differentiated cells expressed GFAP or NeuN and tyrosine hydroxylase. An increase of the latter cells during differentiation could be shown. By using proteomics an explanation on the protein level was found for the observed changes in cell morphology during differentiation, when CSM14.1 cells possessed the morphology of multipolar neurons. The results obtained in this study confirm the suitability of CSM14.1 cells as an in vitro model for the study of neuronal and dopaminergic differentiation in rats.
Collapse
|
7
|
Pellegrini L, Bennis Y, Guillet B, Velly L, Bruder N, Pisano P. [Cell therapy for stroke: from myth to reality]. Rev Neurol (Paris) 2012; 169:291-306. [PMID: 23246427 DOI: 10.1016/j.neurol.2012.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/13/2012] [Accepted: 08/09/2012] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Stroke is one of the leading causes of death and disability worldwide. Intravenous recombinant tissue plasminogen activator is the only available therapy for acute ischemic stroke, but its use is limited by a narrow therapeutic window and cannot stimulate endogenous repair and regeneration of damaged brain tissue. Stem cell-based approaches hold much promise as potential novel treatments to restore neurological function after stroke. STATE OF THE ART In this review, we summarize data from preclinical and clinical studies to investigate the potential application of stem cell therapies for treatment of 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. Various stem cells from multiple sources can generate neural cells that survive and form synaptic connections after transplantation in the stroke-injured brain. Stem cells also exhibit neurorevitalizing properties that may ameliorate neurological deficits through stimulation of neurogenesis, angiogenesis and inhibition of inflammation. PERSPECTIVES/CONCLUSION Performed in stroke, cell therapy would decrease brain damage and reduce functional deficits. After the damage has been done, it would still improve neurological functions by activating endogenous repair. Nevertheless, many questions raised by experimental studies particularly related to long-term safety and technical details of cell preparation and administration must be resolved before wider clinical use.
Collapse
Affiliation(s)
- L Pellegrini
- Service d'anesthésie-réanimation 1, CHU de la Timone, Assistance publique-Hôpitaux de Marseille, 264, rue Saint-Pierre, 13385 Marseille cedex 5, France.
| | | | | | | | | | | |
Collapse
|
8
|
Kozlova EN, Berens C. Guiding Differentiation of Stem Cells in Vivo by Tetracycline-Controlled Expression of Key Transcription Factors. Cell Transplant 2012; 21:2537-54. [DOI: 10.3727/096368911x637407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplantation of stem or progenitor cells is an attractive strategy for cell replacement therapy. However, poor long-term survival and insufficiently reproducible differentiation to functionally appropriate cells in vivo still present major obstacles for translation of this methodology to clinical applications. Numerous experimental studies have revealed that the expression of just a few transcription factors can be sufficient to drive stem cell differentiation toward a specific cell type, to transdifferentiate cells from one fate to another, or to dedifferentiate mature cells to pluripotent stem/progenitor cells (iPSCs). We thus propose here to apply the strategy of expressing the relevant key transcription factors to guide the differentiation of transplanted cells to the desired cell fate in vivo. To achieve this requires tools allowing us to control the expression of these genes in the transplant. Here, we describe drug-inducible systems that allow us to sequentially and timely activate gene expression from the outside, with a particular emphasis on the Tet system, which has been widely and successfully used in stem cells. These regulatory systems offer a tool for strictly limiting gene expression to the respective optimal stage after transplantation. This approach will direct the differentiation of the immature stem/progenitor cells in vivo to the desired cell type.
Collapse
Affiliation(s)
- Elena N Kozlova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
9
|
Yang Z, Qiao H, Sun Z, Li X. Effect of BDNF-plasma-collagen matrix controlled delivery system on the behavior of adult rats neural stem cells. J Biomed Mater Res A 2012; 101:599-606. [PMID: 23090850 DOI: 10.1002/jbm.a.34331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 06/24/2012] [Accepted: 06/26/2012] [Indexed: 12/14/2022]
Abstract
The neurogenesis amount in central nervous system (CNS) stimulated by the injury or diseases is so small that neural stem cells (NSCs) cannot specifically differentiate into the ideal phenotypes to repair the injured CNS. The transplanted exogenous NSCs also have such problems as poor survival and insufficient neuronal differentiation. In this study, the behavior of NSCs from the spinal cord of adult rats was compared at the neurosphere level after the respective addition of the brain-derived neurotrophic factor (BDNF) daily, the BDNF-loaded plasma-collagen matrix, the plasma-collagen matrix alone, or the defined medium alone. The results suggested that the BDNF, either in the control release form or in the soluble form, initiated NSCs proliferation and differentiation by activating receptors Trk B and p75NTR. BDNF also increased the differentiation percentage of adult NSCs into neurons and supported the long-term cell survival and growth. The BDNF was stably released by the plasma-collagen matrix for up to 21 days. The plasma-collagen matrix alone showed its biocompatibility with cells by facilitating the adhesion, survival, and differentiation of NSCs. The NSCs in the defined medium alone group showed poor survival and a very low level of neuronal differentiation and proliferation abilities than above three groups. This study suggested that the BDNF-loaded plasma-collagen matrix may provide a promising means to resolve either the poor survival and insufficient neuronal differentiation of transplanted exogenous NSCs, or stimulating the intrinsic NSCs to proliferate and differentiate into neurons so as to repair the injured adult CNS.
Collapse
Affiliation(s)
- Zhaoyang Yang
- Beijing Institute for Neuroscience, Capital Medical University, Beijing 100069, China
| | | | | | | |
Collapse
|
10
|
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: 4.2] [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
|
11
|
Rodrigues CAV, Diogo MM, da Silva CL, Cabral JMS. Microcarrier expansion of mouse embryonic stem cell-derived neural stem cells in stirred bioreactors. Biotechnol Appl Biochem 2011; 58:231-42. [PMID: 21838797 DOI: 10.1002/bab.37] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neural stem cells (NSCs) are self-renewing multipotent cells, able to differentiate into the phenotypes present in the central nervous system. Applications of NSCs may include toxicology, fundamental research, or cell therapies. The culture of floating cell clusters, called "neurospheres," is widely used for the propagation of NSC populations in vitro but shows several limitations, which may be circumvented by expansion under adherent conditions. In particular, the derivation of distinct populations of NSCs from embryonic stem cells capable of long-term culture under adherent conditions without losing differentiation potential was recently described. However, the expansion of these cells in agitated bioreactors has not been addressed until now and was the aim of this study. Selected microcarriers were tested under dynamic conditions in spinner flasks. Superior performance was observed with polystyrene beads coated with a recombinant peptide containing the Arg-Gly-Asp (RGD) motif (Pronectin F). After optimization of the culture, a 35-fold increase in cell number was achieved after 6 days. High cellular viability and multipotency were maintained throughout the culture. The study presented here may be the basis for the development of larger scale bioprocesses for expansion of these and other populations of adherent NSCs, either from mouse or human origin.
Collapse
Affiliation(s)
- Carlos A V Rodrigues
- Department of Bioengineering, Instituto Superior Técnico, Technical University of Lisbon, Lisboa, Portugal
| | | | | | | |
Collapse
|
12
|
Baizabal JM, Cano-Martínez A, Valencia C, Santa-Olalla J, Young KM, Rietze RL, Bartlett PF, Covarrubias L. Glial commitment of mesencephalic neural precursor cells expanded as neurospheres precludes their engagement in niche-dependent dopaminergic neurogenesis. Stem Cells Dev 2011; 21:1047-58. [PMID: 21615282 DOI: 10.1089/scd.2011.0241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neural precursor cells (NPCs) with high proliferative potential are commonly expanded in vitro as neurospheres. As a population, neurosphere cells show long-term self-renewal capacity and multipotentiality in vitro. These features have led to the assumption that neurosphere cells represent an expansion of the endogenous NPCs residing within the embryonic and adult brain. If this is the case, in principle, bona-fide expansion of endogenous NPCs should not significantly affect their capacity to respond to their original niche of differentiation. To address this issue, we generated primary neurospheres from the dopaminergic niche of the ventral mesencephalon and then transplanted these cells to their original niche within mesencephalic explant cultures. Primary neurosphere cells showed poor capacity to generate dopaminergic neurons in the mesencephalic niche of dopaminergic neurogenesis. Instead, most primary neurosphere cells showed glial commitment as they differentiated into astrocytes in an exclusively neurogenic niche. Subculture of primary cells demonstrated that the neurosphere assay does not amplify niche-responsive dopaminergic progenitors. Further, neurospheres cells were largely unable to acquire the endogenous positional identity within the Nkx6.1(+), Nkx2.2(+), and Pax7(+) domains of mesencephalic explants. Finally, we demonstrate that our observations are not specific for embryonic mesencephalic cells, as NPCs in the adult subventricular zone also showed an intrinsic fate switch from neuronal to glial potential upon neurosphere amplification. Our data suggest that neurosphere formation does not expand the endogenous neurogenic NPCs but rather promotes amplification of gliogenic precursors that do not respond to niche-derived signals of cellular specification and differentiation.
Collapse
Affiliation(s)
- José-Manuel Baizabal
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, México
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Rodrigues CAV, Diogo MM, da Silva CL, Cabral JMS. Hypoxia enhances proliferation of mouse embryonic stem cell-derived neural stem cells. Biotechnol Bioeng 2010; 106:260-70. [PMID: 20014442 DOI: 10.1002/bit.22648] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neural stem (NS) cells can provide a source of material with potential applications for neural drug testing, developmental studies, or novel treatments for neurodegenerative diseases. Herein, the ex vivo expansion of a model system of mouse embryonic stem (mES) cell-derived NS cells was characterized and optimized, cells being cultivated under adherent conditions. Culture was first optimized in terms of initial cell plating density and oxygen concentration, known to strongly influence brain-derived NS cells. To this end, the growth of cells cultured under hypoxic (2%, 5%, and 10% O(2)) and normoxic (20% O(2)) conditions was compared. The results showed that 2-5% oxygen, without affecting multipotency, led to fold increase values in total cell number about twice higher than observed under 20% oxygen (20-fold vs. 10-fold, respectively) this effect being more pronounced when cells were plated at low density. With an optimal cell density of 10(4) cells/cm(2), the maximum growth rates were 1.9 day(-1) under hypoxia versus 1.7 day(-1) under normoxia. Cell division kinetics analysis by flow cytometry based on PKH67 tracking showed that when cultured in hypoxia, cells are at least one divisional generation ahead compared to normoxia. In terms of cell cycle, a larger population in a quiescent G(0) phase was observed in normoxic conditions. The optimization of NS cell culture performed here represents an important step toward the generation of a large number of neural cells from a reduced initial population, envisaging the potential application of these cells in multiple settings.
Collapse
Affiliation(s)
- Carlos A V Rodrigues
- Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | | | | | | |
Collapse
|
14
|
Ebert AD, Barber AE, Heins BM, Svendsen CN. Ex vivo delivery of GDNF maintains motor function and prevents neuronal loss in a transgenic mouse model of Huntington's disease. Exp Neurol 2010; 224:155-62. [PMID: 20227407 DOI: 10.1016/j.expneurol.2010.03.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Revised: 02/17/2010] [Accepted: 03/02/2010] [Indexed: 01/24/2023]
|
15
|
Leipzig ND, Xu C, Zahir T, Shoichet MS. Functional immobilization of interferon-gamma induces neuronal differentiation of neural stem cells. J Biomed Mater Res A 2010; 93:625-33. [PMID: 19591237 DOI: 10.1002/jbm.a.32573] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Stem cell transplantation provides significant promise to regenerative strategies after injury in the central nervous system. Neural stem/progenitor cells (NSPCs) have been studied in terms of their regenerative capacity and their ability to differentiate into neurons when exposed to various soluble factors. In this study, interferon-gamma (IFN-gamma) was compared with brain-derived neurotrophic factor (BDNF) and erythropoietin and was shown to be the best single growth factor for inducing neuronal differentiation from adult rat brain-derived NSPCs. Next, IFN-gamma was surface immobilized to a methacrylamide chitosan (MAC) scaffold that was specifically designed to match the modulus of brain tissue and neuronal differentiation of NSPCs was examined in vitro by immunohistochemistry. Bioactive IFN-gamma was successfully immobilized and quantified by ELISA. Both soluble and immobilized IFN-gamma on MAC surfaces showed dose dependent neuronal differentiation with soluble saturation occurring at 100 ng/mL and the most effective immobilized IFN-gamma dose at 37.5 ng/cm(2), where significantly more neurons resulted compared with controls including soluble IFN-gamma.
Collapse
Affiliation(s)
- Nic D Leipzig
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
16
|
Yang Z, Duan H, Mo L, Qiao H, Li X. The effect of the dosage of NT-3/chitosan carriers on the proliferation and differentiation of neural stem cells. Biomaterials 2010; 31:4846-54. [PMID: 20346501 DOI: 10.1016/j.biomaterials.2010.02.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 02/06/2010] [Indexed: 12/16/2022]
Abstract
This study aimed to determine the optimal dosage range of NT-3 in the soluble form or loaded with chitosan carriers by using NT-3/chitosan carriers to support the survival and proliferation of neural stem cells (NSCs) and induce them to differentiate into desired phenotypes. NSCs were co-cultured with chitosan carriers loaded with different doses of NT-3. As the control, NSCs were cultured in the defined medium, into which were added different doses of NT-3 in the soluble form every day. The ELISA kit was used to study the NT-3 releasing kinetics, which showed that, in the initial co-culture stage from 1 h to 14 weeks, the chitosan carriers loaded with different doses of NT-3 released NT-3 stably and constantly. The 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was conducted to measure the cell viability, and the immunocytochemical methods were adopted to quantitatively analyze the phenotypes differentiating from the NSCs. Compared to the 100 ng NT-3 daily addition group (1400 ng over 14 days), the 25 ng, 50 ng and 200 ng NT-3 daily addition group showed dramatically shorter processes length and much lower differentiation percentage from NSCs into neurons. By contrast, the NT-3 (25 ng)-chitosan carriers group had not only higher cell viability, but also similar processes length and differentiation percentage from NSCs into neurons to the 100 ng NT-3 daily addition group. The method developed in this study significantly reduced the NT-3 amount required to support the survival, proliferation and differentiation of NSCs in vitro. Meanwhile, the chitosan carriers used here provided an ideal 3-dimensional scaffold for the adhesion, migration, proliferation and differentiation of NSC and the differentiated cells. Therefore, this method may open a new field for the large-scaled culture and amplification of NSCs in vitro to replace the lost neural cells, meanwhile lower the consumption of neurotrophic factors in the cell transplantation therapy of brain and spinal injury.
Collapse
Affiliation(s)
- Zhaoyang Yang
- The School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | | | | | | | | |
Collapse
|
17
|
Matos MA, Cicerone MT. Alternating current electric field effects on neural stem cell viability and differentiation. Biotechnol Prog 2010; 26:664-70. [DOI: 10.1002/btpr.389] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
18
|
Kumagai G, Okada Y, Yamane J, Nagoshi N, Kitamura K, Mukaino M, Tsuji O, Fujiyoshi K, Katoh H, Okada S, Shibata S, Matsuzaki Y, Toh S, Toyama Y, Nakamura M, Okano H. Roles of ES cell-derived gliogenic neural stem/progenitor cells in functional recovery after spinal cord injury. PLoS One 2009; 4:e7706. [PMID: 19893739 PMCID: PMC2768792 DOI: 10.1371/journal.pone.0007706] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 10/09/2009] [Indexed: 01/22/2023] Open
Abstract
Transplantation of neural stem/progenitor cells (NS/PCs) following the sub-acute phase of spinal cord injury (SCI) has been shown to promote functional recovery in rodent models. However, the types of cells most effective for treating SCI have not been clarified. Taking advantage of our recently established neurosphere-based culture system of ES cell-derived NS/PCs, in which primary neurospheres (PNS) and passaged secondary neurospheres (SNS) exhibit neurogenic and gliogenic potentials, respectively, here we examined the distinct effects of transplanting neurogenic and gliogenic NS/PCs on the functional recovery of a mouse model of SCI. ES cell-derived PNS and SNS transplanted 9 days after contusive injury at the Th10 level exhibited neurogenic and gliogenic differentiation tendencies, respectively, similar to those seen in vitro. Interestingly, transplantation of the gliogenic SNS, but not the neurogenic PNS, promoted axonal growth, remyelination, and angiogenesis, and resulted in significant locomotor functional recovery after SCI. These findings suggest that gliogenic NS/PCs are effective for promoting the recovery from SCI, and provide essential insight into the mechanisms through which cellular transplantation leads to functional improvement after SCI.
Collapse
Affiliation(s)
- Gentaro Kumagai
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yohei Okada
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Kanrinmaru Project, Keio University School of Medicine, Tokyo, Japan
| | - Junichi Yamane
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazuya Kitamura
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiko Mukaino
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Rehabilitation Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Osahiko Tsuji
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kanehiro Fujiyoshi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Katoh
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Seiji Okada
- Department of Research Super Star Program Stem Cell Unit, Graduate School of Medical Science, Kyusyu University, Fukuoka, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yumi Matsuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Toh
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshiaki Toyama
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Neumeister B, Grabosch A, Basak O, Kemler R, Taylor V. Neural progenitors of the postnatal and adult mouse forebrain retain the ability to self-replicate, form neurospheres, and undergo multipotent differentiation in vivo. Stem Cells 2009; 27:714-23. [PMID: 19096037 DOI: 10.1634/stemcells.2008-0985] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Somatic stem cells are reservoirs to replace lost cells or damaged tissue. Cells with neural stem cell (NSC) characteristics can be isolated from the postnatal mammalian brain into adulthood and expanded as neurospheres. We addressed the ability of these in vitro expanded putative NSCs to retain progenitor characteristics in vivo, in analogy to hematopoietic stem cells. When transplanted in utero, both postnatal and adult neural progenitors colonize host brains and contribute neurons and glia. In stark contrast to what has been reported when transplanted in postnatal hosts, epidermal growth factor-expanded cells also remain self-replicating and multipotent in vivo over many months and can be serially transplanted into multiple hosts. Surprisingly, embryonically transplanted NSCs remain in the neurogenic regions in adult hosts, where they express progenitor cell markers and continue to proliferate even after 6 months without tumor formation. These data indicate that spherogenic cells of the postnatal and adult mammalian brain retain their potential in vitro and in vivo throughout the life of the organism and beyond transplantation, which has important implications for cell replacement strategies.
Collapse
Affiliation(s)
- Bettina Neumeister
- Department of Molecular Embryology, Max-Planck Institute of Immunobiology, Freiburg, Germany
| | | | | | | | | |
Collapse
|
20
|
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: 154] [Impact Index Per Article: 10.3] [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
|
21
|
Wakeman DR, Hofmann MR, Redmond DE, Teng YD, Snyder EY. Long-term multilayer adherent network (MAN) expansion, maintenance, and characterization, chemical and genetic manipulation, and transplantation of human fetal forebrain neural stem cells. ACTA ACUST UNITED AC 2009; Chapter 2:Unit2D.3. [PMID: 19455542 DOI: 10.1002/9780470151808.sc02d03s9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human neural stem/precursor cells (hNSC/hNPC) have been targeted for application in a variety of research models and as prospective candidates for cell-based therapeutic modalities in central nervous system (CNS) disorders. To this end, the successful derivation, expansion, and sustained maintenance of undifferentiated hNSC/hNPC in vitro, as artificial expandable neurogenic micro-niches, promises a diversity of applications as well as future potential for a variety of experimental paradigms modeling early human neurogenesis, neuronal migration, and neurogenetic disorders, and could also serve as a platform for small-molecule drug screening in the CNS. Furthermore, hNPC transplants provide an alternative substrate for cellular regeneration and restoration of damaged tissue in neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. Human somatic neural stem/progenitor cells (NSC/NPC) have been derived from a variety of cadaveric sources and proven engraftable in a cytoarchitecturally appropriate manner into the developing and adult rodent and monkey brain while maintaining both functional and migratory capabilities in pathological models of disease. In the following unit, we describe a new procedure that we have successfully employed to maintain operationally defined human somatic NSC/NPC from developing fetal, pre-term post-natal, and adult cadaveric forebrain. Specifically, we outline the detailed methodology for in vitro expansion, long-term maintenance, manipulation, and transplantation of these multipotent precursors.
Collapse
Affiliation(s)
- Dustin R Wakeman
- University of California at San Diego, La Jolla, California, USA
| | | | | | | | | |
Collapse
|
22
|
Vergaño-Vera E, Méndez-Gómez HR, Hurtado-Chong A, Cigudosa JC, Vicario-Abejón C. Fibroblast growth factor-2 increases the expression of neurogenic genes and promotes the migration and differentiation of neurons derived from transplanted neural stem/progenitor cells. Neuroscience 2009; 162:39-54. [PMID: 19318120 DOI: 10.1016/j.neuroscience.2009.03.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 03/09/2009] [Accepted: 03/11/2009] [Indexed: 10/21/2022]
Abstract
The capacity of neural stem cells (NSC) to generate different types of neurons and glia depends on the action of intrinsic determinants and extracellular signals. Here, we isolated adult olfactory bulb stem cells (aOBSC) that express nestin, RC2 and Sox2, and that have the capacity to generate neurons possessing mature features in culture and in vivo. The differentiation of aOBSC into neurons and glia, as well as their genetic profile, was compared to that of embryonic OBSC (eOBSC) and ganglionic eminence stem cells (GESC). While these eOBSC express neurogenin (Ngn) 1 and 2, two telencephalic dorsal markers, GESC only express Ngn2. Adult OBSC express either little or no detectable Ngn1 and 2, and they produced significantly fewer neurons in culture than eOBSC. By contrast, Dlx2 transcripts (a telencephalic ventral marker) were only clearly detected in GESC. When transplanted into the early postnatal P5-P7 OB, each of the three populations gave rise to cells with a distinct pattern of neuronal migration and/or dendritic arborization. Overall, these findings suggest that cultured NSC partially maintain their regional and temporal specification. Notably, significant neuronal migration and differentiation were only observed in vivo when the NSC were briefly exposed to fibroblast growth factor-2 (FGF-2) before grafting, a treatment that enhanced the neurogenin expression. Hence, the migration and maturation of neurons derived from transplanted NSC can be promoted by upregulating neurogenic gene expression with FGF-2.
Collapse
Affiliation(s)
- E Vergaño-Vera
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda Dr. Arce 37, E-28002 Madrid, Spain
| | | | | | | | | |
Collapse
|
23
|
Behrstock S, Ebert AD, Klein S, Schmitt M, Moore JM, Svendsen CN. Lesion-induced increase in survival and migration of human neural progenitor cells releasing GDNF. Cell Transplant 2008; 17:753-62. [PMID: 19044202 DOI: 10.3727/096368908786516819] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The use of human neural progenitor cells (hNPC) has been proposed to provide neuronal replacement or astrocytes delivering growth factors for brain disorders such as Parkinson's and Huntington's disease. Success in such studies likely requires migration from the site of transplantation and integration into host tissue in the face of ongoing damage. In the current study, hNPC modified to release glial cell line-derived neurotrophic factor (hNPCGDNF) were transplanted into either intact or lesioned animals. GDNF release itself had no effect on the survival, migration, or differentiation of the cells. The most robust migration and survival was found using a direct lesion of striatum (Huntington's model) with indirect lesions of the dopamine system (Parkinson's model) or intact animals showing successively less migration and survival. No lesion affected differentiation patterns. We conclude that the type of brain injury dictates migration and integration of hNPC, which has important consequences when considering transplantation of these cells as a therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Soshana Behrstock
- The Waisman Center, University of Wisconsin Madison, Madison, WI 53705-2280, USA
| | | | | | | | | | | |
Collapse
|
24
|
Mitome M, Low HP, Lora Rodriguez KM, Kitamoto M, Kitamura T, Schwartz WJ. Neuronal differentiation of EGF-propagated neurosphere cells after engraftment to the nucleus of the solitary tract. Neurosci Lett 2008; 444:250-3. [PMID: 18761057 DOI: 10.1016/j.neulet.2008.08.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Revised: 08/15/2008] [Accepted: 08/16/2008] [Indexed: 11/30/2022]
Abstract
Neural precursor cells expanded with epidermal growth factor (EGF) exhibit multipotentiality in vitro, but they differentiate predominantly as glial phenotypes after their transplantation in vivo. Here we demonstrate that EGF-propagated precursors from the murine striatal subventricular zone can exhibit robust incorporation and neuronal differentiation within the nucleus of the solitary tract (NST) after injection into the cisterna magna of neonatal or young adult mice. About two-third of engrafted cells appeared NeuN positive in the region of the gelatinous subnucleus, a region notable for its lack of myelinated fibers. The NST may provide a useful model for understanding the physiological and metabolic regulation of postnatal neurogenesis.
Collapse
Affiliation(s)
- Masato Mitome
- Department of Pediatric Dentistry, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan.
| | | | | | | | | | | |
Collapse
|
25
|
Setting the conditions for efficient, robust and reproducible generation of functionally active neurons from adult subventricular zone-derived neural stem cells. Cell Death Differ 2008; 15:1847-56. [PMID: 19011641 DOI: 10.1038/cdd.2008.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although new culture conditions enable homogeneous and long-term propagation of radial glia-like neural stem (NS) cells in monolayer and serum-free conditions, the efficiency of the conversion of NS cells into terminally differentiated, functionally mature neurons is relatively limited and poorly characterized. We demonstrate that NS cells derived from adult mouse subventricular zone robustly develop properties of mature neurons when exposed to an optimized neuronal differentiation protocol. A high degree of cell viability was preserved. At 22 days in vitro, most cells (65%) were microtubule-associated protein 2(+) and coexpressed gamma-aminobutyric acid (GABA), GAD67, calbindin and parvalbumin. Nearly all neurons exhibited sodium, potassium and calcium currents, and 70% of them fired action potentials. These neurons expressed functional GABA(A) receptors, whereas activable kainate, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and N-methyl-D-aspartic acid receptors were present in approximately 80, 30 and 2% of cells, respectively. Antigenic and functional properties were efficiently and reliably reproduced across experiments and cell passages (up to 68). This is the first report showing a consistent and reproducible generation of large amounts of neurons from long-term passaged adult neural stem cells. Remarkably, the neuronal progeny carries a defined set of antigenic, biochemical and functional characteristics that make this system suitable for studies of NS cell biology as well as for genetic and chemical screenings.
Collapse
|
26
|
Novel and immortalization-based protocols for the generation of neural CNS stem cell lines for gene therapy approaches. Methods Mol Biol 2008. [PMID: 18369767 DOI: 10.1007/978-1-59745-133-8_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Transplantation of neural cells engineered to produce growth factors or molecules with antitumor effects have the potential of grafted cells to be used as vectors for protein delivery in animal models of diseases. In this context, neural stem cells (NSCs), since their identification, have been considered an attractive subject for therapeutic applications to the damaged brain. NSCs have been shown to include attributes important for potential successful ex vivo gene therapy approaches: they show extensive in vitro expansion and, in some cases, a particular tropism toward pathological brain areas. Clearly, the challenges for future clinical development of this approach are in the definition of the most appropriate stem cells for a given application, what genes or chemicals can be delivered, and what diseases are suitable targets. Ideally, NSC lines should be homogeneous and well characterized in terms of their in vitro stability and grafting capacity. We discuss two possible approaches to produce homogeneous and stable progenitor and NSC lines that exploit an oncogene-based immortalization, or, in the second case, a novel protocol for growth factor expansion of stem cells with radial glia-like features. Furthermore, we describe the use of retroviral particles for genetic engineering.
Collapse
|
27
|
Lu H, Li M, Song T, Qian Y, Xiao X, Chen X, Zhang P, Feng X, Parker T, Liu Y. Retrovirus delivered neurotrophin-3 promotes survival, proliferation and neuronal differentiation of human fetal neural stem cells in vitro. Brain Res Bull 2008; 77:158-64. [PMID: 19875351 DOI: 10.1016/j.brainresbull.2008.02.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/10/2008] [Accepted: 02/18/2008] [Indexed: 12/21/2022]
Abstract
Poor survival and insufficient neuronal differentiation are the main obstacles to neural stem cell (NSC) transplantation therapy. Genetic modification of NSCs with neurotrophins is considered a promising approach to overcome these difficulties. In this study, the effects on survival, proliferation and neuronal differentiation of human fetal NSCs (hfNSCs) were observed after infection by a neurotrophin-3 (NT-3) recombinant retrovirus. The hfNSCs, from 12-week human fetal brains formed neurospheres, expressed the stem cell marker nestin and differentiated into the three main cell types of the nervous system. NT-3 recombinant retrovirus (Retro-NT-3) infected hfNSCs efficiently expressed NT-3 gene for at least 8 weeks, presented an accelerated proliferation, and therefore produced an increased number of neurospheres and after differentiation in vitro, contained a higher percentage of neuronal cells. Eight weeks after infection, 37.9+/-4.2% of hfNSCs in the Retro-NT-3 infection group expressed the neuronal marker, this was significantly higher than the control and mock infection groups. NT-3 transduced hfNSCs also displayed longer protruding neurites compared with other groups. Combined these results demonstrate that NT-3 modification promote the survival/proliferation, neuronal differentiation and growth of neurites of hfNSCs in vitro. This study proposes recombinant retrovirus mediated NT-3 modification may provide a promising means to resolve the poor survival and insufficient neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Haixia Lu
- Institute of Neurobiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
OBJECTIVES The aim of this review is to provide an overview of the fundamental features of the neurosphere assay (NSA), which was initially described in 1992, and has since been used not only to detect the presence of stem cells in embryonic and adult mammalian neural tissues, but also to study their characteristics in vitro. Implicit in this review is a detailed examination of the limitations of the NSA, and how this assay is most accurately and appropriately used. Finally we will point out criteria that should be challenged to design alternative ways to overcome the limits of this assay. METHODS NSA is used to isolate putative neural stem cells (NSCs) from the central nervous system (CNS) and to demonstrate the critical stem cell attributes of proliferation, extensive self-renewal and the ability to give rise to a large number of differentiated and functional progeny. Nevertheless, the capability of neural progenitor cells to form neurospheres precludes its utilisation to accurately quantify bona fide stem cell frequency based simply on neurosphere numbers. New culture conditions are needed to be able to distinguish the activity of progenitor cells from stem cells. CONCLUSION A commonly used, and arguably misused, methodology, the NSA has provided a wealth of information on precursor activity of cells derived from the embryonic through to the aged CNS. Importantly, the NSA has contributed to the demise of the 'no new neurogenesis' dogma, and the beginning of a new era of CNS regenerative medicine. Nevertheless, the interpretations arising from the utilisation of the NSA need to take into consideration its limits, so as not to be used beyond its specificity and sensitivity.
Collapse
Affiliation(s)
- Loic P Deleyrolle
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Rodney L Rietze
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Brent A Reynolds
- 1Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
29
|
|
30
|
Muraoka K, Shingo T, Yasuhara T, Kameda M, Yuen WJ, Uozumi T, Matsui T, Miyoshi Y, Date I. Comparison of the therapeutic potential of adult and embryonic neural precursor cells in a rat model of Parkinson disease. J Neurosurg 2008; 108:149-59. [DOI: 10.3171/jns/2008/108/01/0149] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Object
The therapeutic effects of adult and embryonic neural precursor cells (NPCs) were evaluated and their therapeutic potential compared in a rat model of Parkinson disease.
Methods
Adult NPCs were obtained from the subventricular zone and embryonic NPCs were taken from the ganglionic eminence of 14-day-old embryos. Each NPC type was cultured with epidermal growth factor. The in vitro neuronal differentiation rate of adult NPCs was approximately equivalent to that of embryonic NPCs after two passages. Next, the NPCs were transfected with either green fluorescent protein or glial cell line–derived neurotrophic factor (GDNF) by adenoviral infection and transplanted into the striata in a rat model of Parkinson disease (PD) induced by unilateral intrastriatal injection of 6-hydroxydopamine. An amphetamine-induced rotation test was used to evaluate rat behavioral improvement, and immunohistochemical analysis was performed to compare grafted cell survival, differentiation, and host tissue changes.
Results
The rats with GDNF-transfected NPCs had significantly fewer amphetamine-induced rotations and less histological damage. Except for the proportion of surviving grafted cells, there were no significant differences between adult and embryonic NPCs.
Conclusions
Adult and embryonic NPCs have a comparable therapeutic potential in a rat model of PD.
Collapse
|
31
|
Malatesta P, Appolloni I, Calzolari F. Radial glia and neural stem cells. Cell Tissue Res 2007; 331:165-78. [PMID: 17846796 DOI: 10.1007/s00441-007-0481-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Accepted: 07/17/2007] [Indexed: 01/19/2023]
Abstract
During the last decade, the role of radial glia has been radically revisited. Rather than being considered a mere structural component serving to guide newborn neurons towards their final destinations, radial glia is now known to be the main source of neurons in several regions of the central nervous system, notably in the cerebral cortex. Radial glial cells differentiate from neuroepithelial progenitors at the beginning of neurogenesis and share with their ancestors the bipolar shape and the expression of some molecular markers. Radial glia, however, can be distinguished from neuroepithelial progenitors by the expression of astroglial markers. Clonal analyses showed that radial glia is a heterogeneous population, comprising both pluripotent and different lineage-restricted neural progenitors. At late-embryonic and postnatal stages, radial glial cells give rise to the neural stem cells responsible for adult neurogenesis. Embryonic pluripotent radial glia and adult neural stem cells may be clonally linked, thus representing a lineage displaying stem cell features in both the developing and mature central nervous system.
Collapse
Affiliation(s)
- Paolo Malatesta
- Dipartimento di Oncologia, Biologia e Genetica, Università degli Studi di Genova, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | | | | |
Collapse
|
32
|
Anderson L, Caldwell MA. Human neural progenitor cell transplants into the subthalamic nucleus lead to functional recovery in a rat model of Parkinson’s disease. Neurobiol Dis 2007; 27:133-40. [PMID: 17587588 DOI: 10.1016/j.nbd.2007.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Revised: 03/21/2007] [Accepted: 03/27/2007] [Indexed: 01/03/2023] Open
Abstract
Despite the success of foetal nigral transplantation for the treatment of Parkinson's disease, supply limitations of tissue means that alternative sources must be found. Transplantation of human neural progenitor cells (HNPCs) may offer a solution, however few studies have shown functional recovery in animal models of PD without cell modification. Here we show that unmodified HNPC grafted into the subthalamic nucleus (STN) show excellent survival of up to 5 months and induce significant functional recovery following amphetamine-induced rotations within 4 weeks. For the first time we also show that HNPCs, which remain in an immature nestin-positive state, produce VEGF in vivo allowing further modification of the host brain. This suggests that even in the absence of cell replacement strategies utilising immature progenitor cells could be of real therapeutic value.
Collapse
Affiliation(s)
- Lucy Anderson
- Centre for Brain Repair and Department of Clinical Neurosciences, University Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK
| | | |
Collapse
|
33
|
Honda S, Toda K, Tozuka Y, Yasuzawa S, Iwabuchi K, Tomooka Y. Migration and differentiation of neural cell lines transplanted into mouse brains. Neurosci Res 2007; 59:124-35. [PMID: 17651850 DOI: 10.1016/j.neures.2007.06.1467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 05/25/2007] [Accepted: 06/11/2007] [Indexed: 11/29/2022]
Abstract
In the past few years, the plasticity of the regional specification of the CNS has been widely debated on the results from in utero transplantation. Two different results are reported with this transplantation method. One is that the distribution of transplanted cells is dependent on the donor origin, and the other is that the distribution is independent on the donor cell origin. The present study attempted to examine closely the plasticity of the regional specification by in utero transplantation method with clonal neural cell lines, 2Y-3t and 2Y-5o2b. These lines were established from a cerebellum of an adult p53-deficient mouse. Our results showed that transplanted cells migrated into various regions of the CNS and supported the independent distribution. Moreover, different distribution patterns of transplanted cells were observed between host sexes. Labeled cells were localized around the ventricle of neonatal host brains, where they were undifferentiated. In 2-3 weeks after birth, labeled cells were found in the brain parenchyma and some of them took neuronal morphology. In the rostral migratory stream (RMS), cells with unipolar or bipolar morphology were still undifferentiated. In other regions, labeled cells were often associated with blood vessels; the soma were on the surface of vessels, extending processes or neurites into surrounding brain parenchyma. Time-lapse imaging demonstrated that they were migrating with blood vessels.
Collapse
Affiliation(s)
- Shinya Honda
- Department of Biological Science and Technology, and Tissue Engineering Research Center, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Rice HE, Hsu EW, Sheng H, Evenson DA, Freemerman AJ, Safford KM, Provenzale JM, Warner DS, Johnson GA. Superparamagnetic iron oxide labeling and transplantation of adipose-derived stem cells in middle cerebral artery occlusion-injured mice. AJR Am J Roentgenol 2007; 188:1101-8. [PMID: 17377054 DOI: 10.2214/ajr.06.0663] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Adipose-derived stem cells are an alternative stem cell source for CNS therapies. The goals of the current study were to label adipose-derived stem cells with superparamagnetic iron oxide (SPIO) particles, to use MRI to guide the transplantation of adipose-derived stem cells in middle cerebral artery occlusion (MCAO)-injured mice, and to localize donor adipose-derived stem cells in the injured brain using MRI. We hypothesized that we would successfully label adipose-derived stem cells and image them with MRI. MATERIALS AND METHODS Adipose-derived stem cells harvested from mice inbred for green fluorescent protein were labeled with SPIO ferumoxide particles through the use of poly-L-lysine. Adipose-derived stem cell viability, iron staining, and proliferation were measured after SPIO labeling, and the sensitivity of MRI in the detection of SPIO-labeled adipose-derived stem cells was assessed ex vivo. Adult mice (n = 12) were subjected to unilateral MCAO. Two weeks later, in vivo 7-T MRI was performed to guide stereotactic transplantation of SPIO-labeled adipose-derived stem cells into brain tissue adjacent to the infarct. After 24 hours, the mice were sacrificed for high-resolution ex vivo 7-T or 9.4-T MRI and histologic study. RESULTS Adipose-derived stem cells were efficiently labeled with SPIO particles without loss of cell viability or proliferation. Using MRI, we guided precise transplantation of adipose-derived stem cells. MR images of mice given injections of SPIO-labeled adipose-derived stem cells had hypointense regions that correlated with the histologic findings in donor cells. CONCLUSION MRI proved useful in transplantation of adipose-derived stem cells in vivo. This imaging technique may be useful for studies of CNS stem cell therapies.
Collapse
Affiliation(s)
- Henry E Rice
- Department of Surgery, Duke University Medical Center, Box 3815, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Corti S, Nizzardo M, Nardini M, Donadoni C, Locatelli F, Papadimitriou D, Salani S, Del Bo R, Ghezzi S, Strazzer S, Bresolin N, Comi GP. Isolation and characterization of murine neural stem/progenitor cells based on Prominin-1 expression. Exp Neurol 2007; 205:547-62. [PMID: 17466977 DOI: 10.1016/j.expneurol.2007.03.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2006] [Revised: 02/28/2007] [Accepted: 03/15/2007] [Indexed: 12/18/2022]
Abstract
The identification of strategies for the isolation of neural stem cells (NSCs) has important implications for the understanding of their biology and the development of therapeutic applications. It has been previously described that human neural stem and progenitor cells (NSPCs) can be isolated from the central nervous system (CNS) using antibodies to prominin (CD133) and fluorescence-activated cell sorting (FACS). Although this antigen displayed an identical membrane topology in several human and murine tissues there was uncertainty as to the relationship between human and mouse prominin because of the low level of amino acid identity. Here we show that prominin expression can be used to identify and isolate also murine NSPCs from the developing or adult brain. Prominin is co-expressed with known neural stem markers like SOX 1-2, Musashi and Nestin. Moreover, neurosphere-forming cells with multipotency and self-renewal capacity reside within the prominin-positive fraction. Transplantation experiments show that CD133-positive cells give rise to neurons and glial cells in vivo, and that many neurons display appropriate phenotypic characteristics of the recipient tissues. The demonstration that CD133 is a stem cell antigen for murine NSPCs as it is for human NSPCs is useful for the investigation of mammal neurogenesis and development of preclinical tests of NSPCs transplantation in mouse analogues of human diseases.
Collapse
Affiliation(s)
- Stefania Corti
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Foundation, Ospedale Maggiore, Policlinico Mangiagalli and Regina Elena, Padiglione Ponti, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Canola K, Angénieux B, Tekaya M, Quiambao A, Naash MI, Munier FL, Schorderet DF, Arsenijevic Y. Retinal stem cells transplanted into models of late stages of retinitis pigmentosa preferentially adopt a glial or a retinal ganglion cell fate. Invest Ophthalmol Vis Sci 2007; 48:446-54. [PMID: 17197566 PMCID: PMC2823590 DOI: 10.1167/iovs.06-0190] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To characterize the potential of newborn retinal stem cells (RSCs) isolated from the radial glia population to integrate the retina, this study was conducted to investigate the fate of in vitro expanded RSCs transplanted into retinas devoid of photoreceptors (adult rd1 and old VPP mice and rhodopsin-mutated transgenic mice) or partially degenerated retina (adult VPP mice) retinas. METHODS Populations of RSCs and progenitor cells were isolated either from DBA2J newborn mice and labeled with the red lipophilic fluorescent dye (PKH26) or from GFP (green fluorescent protein) transgenic mice. After expansion in EGF+FGF2 (epidermal growth factor+fibroblast growth factor), cells were transplanted intravitreally or subretinally into the eyes of adult wild-type, transgenic mice undergoing slow (VPP strain) or rapid (rd1 strain) retinal degeneration. RESULTS Only limited migration and differentiation of the cells were observed in normal mice injected subretinally or in VPP and rd1 mice injected intravitreally. After subretinal injection in old VPP mice, transplanted cells massively migrated into the ganglion cell layer and, at 1 and 4 weeks after injection, harbored neuronal and glial markers expressed locally, such as beta-tubulin-III, NeuN, Brn3b, or glial fibrillary acidic protein (GFAP), with a marked preference for the glial phenotype. In adult VPP retinas, the grafted cells behaved similarly. Few grafted cells stayed in the degenerating outer nuclear layer (ONL). These cells were, in rare cases, positive for rhodopsin or recoverin, markers specific for photoreceptors and some bipolar cells. CONCLUSIONS These results show that the grafted cells preferentially integrate into the GCL and IPL and express ganglion cell or glial markers, thus exhibiting migratory and differentiation preferences when injected subretinally. It also appears that the retina, whether partially degenerated or already degenerated, does not provide signals to induce massive differentiation of RSCs into photoreceptors. This observation suggests that a predifferentiation of RSCs into photoreceptors before transplantation may be necessary to obtain graft integration in the ONL.
Collapse
Affiliation(s)
- Kriss Canola
- Unit of Gene Therapy and Stem Cell Biology, Jules Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Brigitte Angénieux
- Unit of Gene Therapy and Stem Cell Biology, Jules Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Meriem Tekaya
- Unit of Gene Therapy and Stem Cell Biology, Jules Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alexander Quiambao
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Muna I. Naash
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Francis L. Munier
- Unit of Clinical Oculogenetics, Jules Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | | | - Yvan Arsenijevic
- Unit of Gene Therapy and Stem Cell Biology, Jules Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
37
|
Baudet ML, Martin B, Hassanali Z, Parker E, Sanders EJ, Harvey S. Expression, translation, and localization of a novel, small growth hormone variant. Endocrinology 2007; 148:103-15. [PMID: 17008400 DOI: 10.1210/en.2006-1070] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A novel transcript of the GH gene has been identified in ocular tissues of chick embryos. It is, however, unknown whether this transcript (small chicken GH, scGH) is translated. This possibility was therefore assessed. The expression of scGH mRNA was confirmed by RT-PCR, using primers that amplified a 426-bp cDNA of its coding sequence. This cDNA was inserted into an expression plasmid to transfect HEK 293 cells, and its translation was shown by specific scGH immunoreactivity in extracts of these cells. This immunoreactivity was directed against the unique N terminus of scGH and was associated with a protein of 16 kDa, comparable with its predicted size. Most of the immunoreactivity detected was, however, associated with a 31-kDa moiety, suggesting scGH is normally dimerized. Neither protein was, however, present in media of the transfected HEK cells, consistent with scGH's lack of a signal sequence. Similar moieties of 16 and 31 kDa were also found in proteins extracted from ocular tissues (neural retina, pigmented epithelium, lens, cornea, choroid) of embryos, although they were not consistently present in vitreous humor. Specific scGH immunoreactivity was also detected in these tissues by immunocytochemistry but not in axons in the optic fiber layer or the optic nerve head, which were immunoreactive for full-length GH. In summary, we have established that scGH expression and translation occurs in ocular tissues of chick embryos, in which its localization in the neural retina and the optic nerve head is distinct from that of the full-length protein.
Collapse
Affiliation(s)
- M-L Baudet
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | | | | | | | | | | |
Collapse
|
38
|
Johann V, Schiefer J, Sass C, Mey J, Brook G, Krüttgen A, Schlangen C, Bernreuther C, Schachner M, Dihné M, Kosinski CM. Time of transplantation and cell preparation determine neural stem cell survival in a mouse model of Huntington’s disease. Exp Brain Res 2006; 177:458-70. [PMID: 17013619 DOI: 10.1007/s00221-006-0689-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Accepted: 08/24/2006] [Indexed: 10/24/2022]
Abstract
Cell replacement therapies for neurodegenerative diseases, using multipotent neural stem cells (NSCs), require above all, a good survival of the graft. In this study, we unilaterally injected quinolinic acid (QA) into the striatum of adult mice and transplanted syngeneic NSCs of enhanced green fluorescent protein-transgenic mice into the lesioned striatum. The injection of QA leads to an excitotoxic lesion with selective cell death of the medium sized spiny neurons, the same cells that are affected in Huntington's disease. In order to investigate the best timing of transplantation for the survival of donor cells, we transplanted the stem cells at 2, 7 and 14 days after injury. In addition, the influence of graft preparation prior to transplantation, i.e., intact neurospheres versus dissociated cell suspension on graft survival was investigated. By far the best survival was found with the combination of early transplantation (i.e., 2 days after QA-lesion) with the use of neurospheres instead of dissociated cell suspension. This might be due to the different states of host's astrocytic and microglia activation which we found to be moderate at 2, but pronounced at 7 and 14 days after QA-lesion. We also investigated brain derived neurotrophic factor (BDNF)-expression in the striatum after QA-lesion and found no significant change in BDNF protein-level. We conclude that already the method of graft preparation of NSCs for transplantation, as well as the timing of the transplantation procedure strongly affects the survival of the donor cells when grafted into the QA-lesioned striatum of adult mice.
Collapse
Affiliation(s)
- Verena Johann
- Department of Neurology, University Hospital RWTH, Pauwelsstr. 30, 52074 Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yang Y, Ren W, Chen F. Knockdown of Stat3 in C17.2 neural stem cells facilitates the generation of neurons: a possibility of transplantation with a low level of oncogene. Neuroreport 2006; 17:235-8. [PMID: 16462589 DOI: 10.1097/01.wnr.0000199470.29357.8f] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
This study investigates the role of a low level of Stat3 in the C17.2 neural stem cells, which are popular stem cell candidates for transplantation research. The results reveal that C17.2 neural stem cells will undergo increased differentiation into neurons without generating glia after knockdown of Stat3 expression via an interfering RNA expression plasmid. As constitutively activated Stat3 is considered to be an oncogene, this study raises the possibility of stem cell transplantation with a low level of Stat3 to reduce the oncogenesis and facilitate the generation of neurons.
Collapse
Affiliation(s)
- Yang Yang
- Laboratory of Neural Molecular Biology, School of Life Sciences, Shanghai University, Shanghai, PR China
| | | | | |
Collapse
|
40
|
Conti L, Reitano E, Cattaneo E. Neural stem cell systems: diversities and properties after transplantation in animal models of diseases. Brain Pathol 2006; 16:143-54. [PMID: 16768755 PMCID: PMC8095762 DOI: 10.1111/j.1750-3639.2006.00009.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Currently available effective treatments of the diseased or damaged central nervous system (CNS) are restricted to a limited pharmacological relief of symptoms or those given to avoid further damage. Therefore the search is on for treatments that can restore function in the CNS. During recent years replacement of damaged neurons by cell transplantation is being enthusiastically explored as a potential treatment for many neurodegenerative diseases, stroke and traumatic brain injury. Several references in both scientific journals and popular newspapers concerning different types of cultured stem cells, potentially exploitable to treat pathological conditions of the brain, raise important questions pertinent to the fundamental and realistic differences between grafts of primary neural cells and the transplantation of in vitro expanded neural stem cells (NSCs). Our aim is to review the available information on the grafting of different NSC types into the adult rodent brain, focusing on critical aspects for the development of clinical therapies to replace damaged neurons.
Collapse
Affiliation(s)
- Luciano Conti
- Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Erika Reitano
- Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| | - Elena Cattaneo
- Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milano, Milano, Italy
| |
Collapse
|
41
|
Barraud P, Thompson L, Kirik D, Björklund A, Parmar M. Isolation and characterization of neural precursor cells from theSox1-GFP reporter mouse. Eur J Neurosci 2005; 22:1555-69. [PMID: 16197496 DOI: 10.1111/j.1460-9568.2005.04352.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have made use of a reporter mouse line in which enhanced green fluorescence protein (GFP) is inserted into the Sox1 locus. We show that the GFP reporter is coexpressed with the Sox1 protein as well as with other known markers for neural stem and progenitor cells, and can be used to identify and isolate these cells by fluorescence-activated cell sorting (FACS) from the developing or adult brain and from neurosphere cultures. All neurosphere-forming cells with the capacity for multipotency and self-renewal reside in the Sox1-GFP-expressing population. Thus, the Sox1-GFP reporter system is highly useful for identification, isolation and characterization of neural stem and progenitor cells, as well as for the validation of alternative means for isolating neural stem and progenitor cells. Further, transplantation experiments show that Sox1-GFP cells isolated from the foetal brain give rise to neurons and glia in vivo, and that many of the neurons display phenotypic characteristics appropriate for the developing brain region from which the Sox1-GFP precursors were derived. On the other hand, Sox1-GFP cells isolated from the adult subventricular zone or expanded neurosphere cultures gave rise almost exclusively to glial cells following transplantation. Thus, not all Sox1-GFP cells possess the same capacity for neuronal differentiation in vivo.
Collapse
Affiliation(s)
- Perrine Barraud
- Wallenberg Neuroscience Center, BMC A11, Section of Neurobiology, and Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund, University, SE-221 84 Lund, Sweden
| | | | | | | | | |
Collapse
|
42
|
Corti S, Locatelli F, Papadimitriou D, Donadoni C, Del Bo R, Fortunato F, Strazzer S, Salani S, Bresolin N, Comi GP. Multipotentiality, homing properties, and pyramidal neurogenesis of CNS‐derived LeX(ssea‐1)
+
/CXCR4
+
stem cells. FASEB J 2005; 19:1860-2. [PMID: 16150803 DOI: 10.1096/fj.05-4170fje] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Achieving efficient distribution of neural stem cells throughout the central nervous system (CNS) and robust generation of specific neurons is a major challenge for the development of cell-mediated therapy for neurodegenerative diseases. We isolated a primitive neural stem cell subset, double positive for LeX(Le) and CXCR4(CX) antigens that possesses CNS homing potential and extensive neuronal repopulating capacity. Le+CX+ cells are multipotential and can generate neurons as well as myogenic and endothelial cells. In vivo Le+CX+ cells displayed widespread incorporation and differentiated into cortical and hippocampal pyramidal neurons. Since intravenous delivery could be a less invasive route of transplantation, we investigated whether Le+CX+ cells could migrate across endothelial monolayers. Intracerebral coadministration of SDF enabled migration of intravenously injected Le+CX+ cells into the CNS and a small, yet significant, number of donor cells differentiated into neurons. The isolation of a specific neural stem cell population could offer major advantages to neuronal replacement strategies.
Collapse
Affiliation(s)
- Stefania Corti
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Foundation, Ospedale Maggiore Policlinico, Mangiagalli and Regina Elena, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Mawatari Y, Fukushima M, Inoue T, Setoguchi T, Taga T, Tanihara H. Preferential differentiation of neural progenitor cells into the glial lineage through gp130 signaling in N-methyl-d-aspartate-treated retinas. Brain Res 2005; 1055:7-14. [PMID: 16098488 DOI: 10.1016/j.brainres.2005.06.047] [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] [Received: 02/02/2005] [Revised: 05/31/2005] [Accepted: 06/05/2005] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to investigate the differentiation of neural progenitor cells (NPCs) following retinal transplantation in N-methyl-D-aspartate (NMDA)-treated eyes. NMDA was injected into the vitreous cavity of adult rat eyes. NPCs were prepared from telencephalic neuroepithelium of enhanced green fluorescence protein (EGFP) transgenic mice on embryonic day 14.5. A cell suspension was injected into the vitreous cavity in experimental eyes. Immunohistochemistry was conducted at 1, 2 or 4 weeks after transplantation of NPCs in an effort to determine the survival and differentiation of transplanted NPCs. Similar experiments were conducted using glycoprotein (gp)130-null (-/-) mice. Examination of retinal sections revealed that transplanted NPCs could survive for at least 4 weeks in NMDA-treated retinas. Immunohistochemical studies for specific cell-type markers revealed that, among the transplanted NPCs at 2 weeks after transplantation, the mean percentage (+/-standard deviation) of glial fibrillary acidic protein (GFAP)-positive (glial) cells was 63.5 +/- 7.4%, demonstrating the differentiation of transplanted NPCs with a preference for the glial lineage. Furthermore, the mean percentage of betaIII-tubulin-positive (mature neuronal) cells was 18.8 +/- 4.5%. Following transplantation of NPCs isolated from gp130-/- mice into NMDA-treated retinas, the mean percentage of GFAP-positive cells (17.6 +/- 7.0%), was significantly lower than that in NPCs isolated from wild-type mice (59.1 +/- 6.0%, P = 0.04, Mann-Whitney U test). Preferential differentiation of NPCs into the glial lineage is induced through gp130 signaling in NMDA-treated eyes.
Collapse
Affiliation(s)
- Yuki Mawatari
- Department of Ophthalmology and Visual Science, Kumamoto University Graduate School of Medical Sciences, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, Biella G, Sun Y, Sanzone S, Ying QL, Cattaneo E, Smith A. Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol 2005; 3:e283. [PMID: 16086633 PMCID: PMC1184591 DOI: 10.1371/journal.pbio.0030283] [Citation(s) in RCA: 673] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2004] [Accepted: 06/14/2005] [Indexed: 12/11/2022] Open
Abstract
Pluripotent mouse embryonic stem (ES) cells multiply in simple monoculture by symmetrical divisions. In vivo, however, stem cells are generally thought to depend on specialised cellular microenvironments and to undergo predominantly asymmetric divisions. Ex vivo expansion of pure populations of tissue stem cells has proven elusive. Neural progenitor cells are propagated in combination with differentiating progeny in floating clusters called neurospheres. The proportion of stem cells in neurospheres is low, however, and they cannot be directly observed or interrogated. Here we demonstrate that the complex neurosphere environment is dispensable for stem cell maintenance, and that the combination of fibroblast growth factor 2 (FGF-2) and epidermal growth factor (EGF) is sufficient for derivation and continuous expansion by symmetrical division of pure cultures of neural stem (NS) cells. NS cells were derived first from mouse ES cells. Neural lineage induction was followed by growth factor addition in basal culture media. In the presence of only EGF and FGF-2, resulting NS cells proliferate continuously, are diploid, and clonogenic. After prolonged expansion, they remain able to differentiate efficiently into neurons and astrocytes in vitro and upon transplantation into the adult brain. Colonies generated from single NS cells all produce neurons upon growth factor withdrawal. NS cells uniformly express morphological, cell biological, and molecular features of radial glia, developmental precursors of neurons and glia. Consistent with this profile, adherent NS cell lines can readily be established from foetal mouse brain. Similar NS cells can be generated from human ES cells and human foetal brain. The extrinsic factors EGF plus FGF-2 are sufficient to sustain pure symmetrical self-renewing divisions of NS cells. The resultant cultures constitute the first known example of tissue-specific stem cells that can be propagated without accompanying differentiation. These homogenous cultures will enable delineation of molecular mechanisms that define a tissue-specific stem cell and allow direct comparison with pluripotent ES cells.
Collapse
Affiliation(s)
- Luciano Conti
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Steven M Pollard
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Thorsten Gorba
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Erika Reitano
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Mauro Toselli
- 3Institute of Physiological and Pharmacological Sciences, University of Pavia, Pavia, Italy
| | - Gerardo Biella
- 3Institute of Physiological and Pharmacological Sciences, University of Pavia, Pavia, Italy
| | - Yirui Sun
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Sveva Sanzone
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Qi-Long Ying
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Elena Cattaneo
- 2Department of Pharmacological Sciences and Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Austin Smith
- 1Institute for Stem Cell Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
45
|
Sánchez-Pernaute R, Studer L, Ferrari D, Perrier A, Lee H, Viñuela A, Isacson O. Long-term survival of dopamine neurons derived from parthenogenetic primate embryonic stem cells (cyno-1) after transplantation. Stem Cells 2005; 23:914-22. [PMID: 15941857 PMCID: PMC2654596 DOI: 10.1634/stemcells.2004-0172] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dopamine (DA) neurons can be derived from human and primate embryonic stem (ES) cells in vitro. An ES cell-based replacement therapy for patients with Parkinson's disease requires that in vitro-generated neurons maintain their phenotype in vivo. Other critical issues relate to their proliferative capacity and risk of tumor formation, and the capability of migration and integration in the adult mammalian brain. Neural induction was achieved by coculture of primate parthenogenetic ES cells (Cyno-1) with stromal cells, followed by sequential exposure to midbrain patterning and differentiation factors to favor DA phenotypic specification. Differentiated ES cells were treated with mitomycin C and transplanted into adult immunosuppressed rodents and into a primate (allograft) with out immunosuppression. A small percentage of DA neurons survived in both rodent and primate hosts for the entire term of the study (4 and 7 months, respectively). Other neuronal and glial populations derived from Cyno-1 ES cells showed, in vivo, phenotypic characteristics and growth and migration patterns similar to fetal primate transplants, and a majority of cells (>80%) expressed the forebrain transcription factor brain factor 1. No teratoma formation was observed. In this study, we demonstrate long-term survival of DA neurons obtained in vitro from primate ES cells. Optimization of differentiation, cell selection, and cell transfer is required for functional studies of ES-derived DA neurons for future therapeutic applications.
Collapse
Affiliation(s)
- Rosario Sánchez-Pernaute
- McLean Hospital/Harvard University Udall Parkinson's Disease Research Center of Excellence and Neuroregeneration Laboratories, McLean Hospital, 115 Mill St., Belmont, Massachusetts 02478, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Nagato M, Heike T, Kato T, Yamanaka Y, Yoshimoto M, Shimazaki T, Okano H, Nakahata T. Prospective characterization of neural stem cells by flow cytometry analysis using a combination of surface markers. J Neurosci Res 2005; 80:456-66. [PMID: 15795928 DOI: 10.1002/jnr.20442] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neural stem cells (NSCs) with self-renewal and multilineage differentiation properties can potentially repair degenerating or damaged neural tissue. Here, we have enriched NSCs from neurospheres, which make up a heterogeneous population, by fluorescence-activated cell sorting (FACS) with antibodies against syndecan-1, Notch-1, and integrin-beta1, which were chosen as candidates for hematopoietic cell-or somatic stem cell-markers. Antigen-positive cells readily initiated neurosphere formation, but cells lacking these markers did so less readily. Doubly positive cells expressing both syndecan-1 and Notch-1 underwent neurosphere formation more efficiently than did singly positive cells. The progeny of sorted cells could differentiate into neurons and glial cells both in vitro and in vivo. These antibodies were also useful for isolating cells from the murine embryonic day 14.5 brain that efficiently formed neurospheres. In contrast, there was no distinct difference in neurosphere formation efficiency between Hoechst 33342-stained side population cells and main population cells, although the former are known to have a stem cell phenotype in various tissues. These results indicate the usefulness of syndecan-1, Notch-1, and integrin-beta1 as NSC markers.
Collapse
Affiliation(s)
- Masako Nagato
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Klein S, Svendsen CN. Stem cells in the injured spinal cord: reducing the pain and increasing the gain. Nat Neurosci 2005; 8:259-60. [PMID: 15746908 DOI: 10.1038/nn0305-259] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Gréco B, Low HP, Johnson EC, Salmonsen RA, Gallant J, Jones SN, Ross AH, Recht LD. Differentiation prevents assessment of neural stem cell pluripotency after blastocyst injection. ACTA ACUST UNITED AC 2005; 22:600-8. [PMID: 15277705 DOI: 10.1634/stemcells.22-4-600] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Earlier studies reported that neural stem (NS) cells injected into blastocysts appeared to be pluripotent, differentiating into cells of all three germ layers. In this study, we followed in vitro green fluorescent protein (GFP)-labeled NS and embryonic stem (ES) cells injected into blastocysts. Forty-eight hours after injection, significantly fewer blastocysts contained GFP-NS cells than GFP-ES cells. By 96 hours, very few GFP-NS cells remained in blastocysts compared with ES cells. Moreover, 48 hours after injection, GFP-NS cells in blastocysts extended long cellular processes, ceased expressing the NS cell marker nestin, and instead expressed the astrocytic marker glial fibrillary acidic protein. GFP-ES cells in blastocysts remained morphologically undifferentiated, continuing to express the pluripotent marker stage-specific embryonic antigen-1. Selecting cells from the NS cell population that preferentially formed neurospheres for injection into blastocysts resulted in identical results. Consistent with this in vitro behavior, none of almost 80 mice resulting from NS cell-injected blastocysts replaced into recipient mothers were chimeric. These results strongly support the idea that NS cells cannot participate in chimera formation because of their rapid differentiation into glia-like cells. Thus, these results raise doubts concerning the pluripotency properties of NS cells.
Collapse
Affiliation(s)
- Béatrice Gréco
- Department of Neurology, University of Massachusetts Medical School, Worcester, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Jin K, Sun Y, Xie L, Mao XO, Childs J, Peel A, Logvinova A, Banwait S, Greenberg DA. Comparison of ischemia-directed migration of neural precursor cells after intrastriatal, intraventricular, or intravenous transplantation in the rat. Neurobiol Dis 2005; 18:366-74. [PMID: 15686965 DOI: 10.1016/j.nbd.2004.10.010] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2004] [Revised: 08/04/2004] [Accepted: 10/13/2004] [Indexed: 01/24/2023] Open
Abstract
Cell replacement therapy may have the potential to promote brain repair and recovery after stroke. To compare how focal cerebral ischemia affects the entry, migration, and phenotypic features of neural precursor cells transplanted by different routes, we administered neuronal precursors from embryonic cerebral cortex of green fluorescent protein (GFP)-expressing transgenic mice to rats that had undergone middle cerebral artery occlusion (MCAO) by the intrastriatal, intraventricular, and intravenous routes. MCAO increased the entry of GFP-immunoreactive cells, most of which expressed neuroepithelial (nestin) or neuronal (doublecortin) markers, from the ventricles and bloodstream into the brain, and enhanced their migration when delivered by any of these routes. Transplanted neural precursors migrated into the ischemic striatum and cerebral cortex. Thus, transplantation of neural precursors by a variety of routes can deliver cells with the potential to replace injured neurons to ischemic brain regions.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945-0638, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Armstrong RJE, Jain M, Barker RA. Stem cell transplantation as an approach to brain repair. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.11.10.1563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|