51
|
Labeed FH, Lu J, Mulhall HJ, Marchenko SA, Hoettges KF, Estrada LC, Lee AP, Hughes MP, Flanagan LA. Biophysical characteristics reveal neural stem cell differentiation potential. PLoS One 2011; 6:e25458. [PMID: 21980464 PMCID: PMC3184132 DOI: 10.1371/journal.pone.0025458] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Accepted: 09/05/2011] [Indexed: 12/15/2022] Open
Abstract
Background Distinguishing human neural stem/progenitor cell (huNSPC) populations that will predominantly generate neurons from those that produce glia is currently hampered by a lack of sufficient cell type-specific surface markers predictive of fate potential. This limits investigation of lineage-biased progenitors and their potential use as therapeutic agents. A live-cell biophysical and label-free measure of fate potential would solve this problem by obviating the need for specific cell surface markers. Methodology/Principal Findings We used dielectrophoresis (DEP) to analyze the biophysical, specifically electrophysiological, properties of cortical human and mouse NSPCs that vary in differentiation potential. Our data demonstrate that the electrophysiological property membrane capacitance inversely correlates with the neurogenic potential of NSPCs. Furthermore, as huNSPCs are continually passaged they decrease neuron generation and increase membrane capacitance, confirming that this parameter dynamically predicts and negatively correlates with neurogenic potential. In contrast, differences in membrane conductance between NSPCs do not consistently correlate with the ability of the cells to generate neurons. DEP crossover frequency, which is a quantitative measure of cell behavior in DEP, directly correlates with neuron generation of NSPCs, indicating a potential mechanism to separate stem cells biased to particular differentiated cell fates. Conclusions/Significance We show here that whole cell membrane capacitance, but not membrane conductance, reflects and predicts the neurogenic potential of human and mouse NSPCs. Stem cell biophysical characteristics therefore provide a completely novel and quantitative measure of stem cell fate potential and a label-free means to identify neuron- or glial-biased progenitors.
Collapse
Affiliation(s)
- Fatima H. Labeed
- Centre for Biomedical Engineering, University of Surrey, Guildford, United Kingdom
| | - Jente Lu
- Department of Neurology and Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Hayley J. Mulhall
- Centre for Biomedical Engineering, University of Surrey, Guildford, United Kingdom
| | - Steve A. Marchenko
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, United States of America
| | - Kai F. Hoettges
- Centre for Biomedical Engineering, University of Surrey, Guildford, United Kingdom
| | - Laura C. Estrada
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
- Laboratory for Fluorescence Dynamics, University of California Irvine, Irvine, California, United States of America
| | - Abraham P. Lee
- Department of Biomedical Engineering, University of California Irvine, Irvine, California, United States of America
| | - Michael P. Hughes
- Centre for Biomedical Engineering, University of Surrey, Guildford, United Kingdom
| | - Lisa A. Flanagan
- Department of Neurology and Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
52
|
Abstract
The developing nervous system derives from neuroepithelial progenitor cells that divide to generate all of the mature neuronal types. For the proper complement of cell types to form, the progenitors must produce postmitotic cells, yet also replenish the progenitor pool. Progenitor divisions can be classified into three general types: symmetric proliferative (producing two progenitors), asymmetric neurogenic (producing one progenitor and one postmitotic cell), and symmetric neurogenic (producing two postmitotic cells). The appropriate ratios for these modes of cell division require intrinsic polarity, which is one of the characteristics that define neuroepithelial progenitor cells. The type of division an individual progenitor undergoes can be influenced by cellular features, or behaviors, which are heterogeneous within the population of progenitors. Here we review three key cellular parameters, asymmetric inheritance, cell cycle kinetics, and interkinetic nuclear migration, and the possible mechanisms for how these features influence progenitor fates.
Collapse
Affiliation(s)
- Minde I Willardsen
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
53
|
Hwang HJ, Rulifson E. Serial specification of diverse neuroblast identities from a neurogenic placode by Notch and Egfr signaling. Development 2011; 138:2883-93. [PMID: 21653613 PMCID: PMC3119302 DOI: 10.1242/dev.055681] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We used the brain insulin-producing cell (IPC) lineage and its identified neuroblast (IPC NB) as a model to understand a novel example of serial specification of NB identities in the Drosophila dorsomedial protocerebral neuroectoderm. The IPC NB was specified from a small, molecularly identified group of cells comprising an invaginated epithelial placode. By progressive delamination of cells, the placode generated a series of NB identities, including the single IPC NB, a number of other canonical Type I NBs, and a single Type II NB that generates large lineages by transient amplification of neural progenitor cells. Loss of Notch function caused all cells of the placode to form as supernumerary IPC NBs, indicating that the placode is initially a fate equivalence group for the IPC NB fate. Loss of Egfr function caused all placodal cells to apoptose, except for the IPC NB, indicating a requirement of Egfr signaling for specification of alternative NB identities. Indeed, both derepressed Egfr activity in yan mutants and ectopic EGF activity produced supernumerary Type II NBs from the placode. Loss of both Notch and Egfr function caused all placode cells to become IPC NBs and survive, indicating that commitment to NB fate nullified the requirement of Egfr activity for placode cell survival. We discuss the surprising parallels between the serial specification of neural fates from this neurogenic placode and the fly retina.
Collapse
Affiliation(s)
- Helen J Hwang
- Biomedical Sciences Graduate Program, University of California-San Francisco, CA 94143, USA
| | | |
Collapse
|
54
|
Costa MR, Ortega F, Brill MS, Beckervordersandforth R, Petrone C, Schroeder T, Götz M, Berninger B. Continuous live imaging of adult neural stem cell division and lineage progression in vitro. Development 2011; 138:1057-68. [PMID: 21343361 DOI: 10.1242/dev.061663] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Little is known about the intrinsic specification of adult neural stem cells (NSCs) and to what extent they depend on their local niche. To observe adult NSC division and lineage progression independent of their niche, we isolated cells from the adult mouse subependymal zone (SEZ) and cultured them at low density without growth factors. We demonstrate here that SEZ cells in this culture system are primarily neurogenic and that adult NSCs progress through stereotypic lineage trees consisting of asymmetric stem cell divisions, symmetric transit-amplifying divisions and final symmetric neurogenic divisions. Stem cells, identified by their astro/radial glial identity and their slow-dividing nature, were observed to generate asymmetrically and fast-dividing cells that maintained an astro/radial glia identity. These, in turn, gave rise to symmetrically and fast-dividing cells that lost glial hallmarks, but had not yet acquired neuronal features. The number of amplifying divisions was limited to a maximum of five in this system. Moreover, we found that cell growth correlated with the number of subsequent divisions of SEZ cells, with slow-dividing astro/radial glia exhibiting the most substantial growth prior to division. The fact that in the absence both of exogenously supplied growth factors and of signals provided by the local niche neurogenic lineage progression takes place in such stereotypic fashion, suggests that lineage progression is, to a significant degree, cell intrinsic or pre-programmed at the beginning of the lineage.
Collapse
Affiliation(s)
- Marcos R Costa
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, 59072-970 Natal, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
55
|
Khodosevich K, Watanabe Y, Monyer H. EphA4 preserves postnatal and adult neural stem cells in an undifferentiated state in vivo. J Cell Sci 2011; 124:1268-79. [DOI: 10.1242/jcs.076059] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In the postnatal brain, new neurons continue to be generated in two neurogenic areas, the subventricular zone of the lateral ventricles (SVZ) and the subgranular zone of the hippocampus. There is evidence that ephrins and their Eph receptors belong to a signaling network that regulates neurogenesis. On the basis of previous data, we have identified Eph receptor A4 (EphA4) as a potential regulator of neurogenesis. We showed by immunohistochemistry that in adult neurogenic niches EphA4 is expressed only by neural stem cells (NSCs). Using in vitro and in vivo assays, we demonstrated that EphA4 expression maintains NSCs in an undifferentiated state. Specifically, in neurosphere cultures Epha4 knockdown resulted in a decrease of NSC proliferation and premature differentiation. In postnatal and adult brain, Epha4 knockdown caused a decrease in NSCs in the SVZ, eventually resulting in a reduced number of postnatally generated neuroblasts. Both in vitro and in vivo effects were rescued by co-infection with a modified EphA4 that was resistant to Epha4 shRNA.
Collapse
Affiliation(s)
- Konstantin Khodosevich
- Department of Clinical Neurobiology, Heidelberg University Medical Center, 69120 Heidelberg, Germany
- Department of Clinical Neurobiology/A230, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Yasuhito Watanabe
- Department of Clinical Neurobiology, Heidelberg University Medical Center, 69120 Heidelberg, Germany
- Department of Clinical Neurobiology/A230, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology, Heidelberg University Medical Center, 69120 Heidelberg, Germany
- Department of Clinical Neurobiology/A230, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
56
|
Sun Y, Meijer DH, Alberta JA, Mehta S, Kane MF, Tien AC, Fu H, Petryniak MA, Potter GB, Liu Z, Powers JF, Runquist IS, Rowitch DH, Stiles CD. Phosphorylation state of Olig2 regulates proliferation of neural progenitors. Neuron 2011; 69:906-17. [PMID: 21382551 PMCID: PMC3065213 DOI: 10.1016/j.neuron.2011.02.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2010] [Indexed: 10/18/2022]
Abstract
The bHLH transcription factors that regulate early development of the central nervous system can generally be classified as either antineural or proneural. Initial expression of antineural factors prevents cell cycle exit and thereby expands the pool of neural progenitors. Subsequent (and typically transient) expression of proneural factors promotes cell cycle exit, subtype specification, and differentiation. Against this backdrop, the bHLH transcription factor Olig2 in the oligodendrocyte lineage is unorthodox, showing antineural functions in multipotent CNS progenitor cells but also sustained expression and proneural functions in the formation of oligodendrocytes. We show here that the proliferative function of Olig2 is controlled by developmentally regulated phosphorylation of a conserved triple serine motif within the amino-terminal domain. In the phosphorylated state, Olig2 maintains antineural (i.e., promitotic) functions that are reflected in human glioma cells and in a genetically defined murine model of primary glioma.
Collapse
Affiliation(s)
- Yu Sun
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - Dimphna H. Meijer
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - John A. Alberta
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - Shwetal Mehta
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - Michael F. Kane
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - An-Chi Tien
- Departments of Pediatrics and Neurological Surgery; Howard Hughes Medical Institute, UCSF, 513 Parnassus Avenue, San Francisco CA 94143
| | - Hui Fu
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - Magdalena A. Petryniak
- Departments of Pediatrics and Neurological Surgery; Howard Hughes Medical Institute, UCSF, 513 Parnassus Avenue, San Francisco CA 94143
| | - Gregory B. Potter
- Departments of Pediatrics and Neurological Surgery; Howard Hughes Medical Institute, UCSF, 513 Parnassus Avenue, San Francisco CA 94143
| | - Zijing Liu
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | | | - I. Sophie Runquist
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| | - David H. Rowitch
- Departments of Pediatrics and Neurological Surgery; Howard Hughes Medical Institute, UCSF, 513 Parnassus Avenue, San Francisco CA 94143
| | - Charles D. Stiles
- Department of Cancer Biology, Harvard Medical School and Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115
| |
Collapse
|
57
|
Huang SL, Shi W, Jiao Q, He XJ. Change of Neural Stem Cells in the Choroid Plexuses of Developing Rat. Int J Neurosci 2011; 121:310-5. [DOI: 10.3109/00207454.2011.556282] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
58
|
Sonic hedgehog and notch signaling can cooperate to regulate neurogenic divisions of neocortical progenitors. PLoS One 2011; 6:e14680. [PMID: 21379383 PMCID: PMC3040755 DOI: 10.1371/journal.pone.0014680] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 01/03/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hedgehog (Hh) signaling is crucial for the generation and maintenance of both embryonic and adult stem cells, thereby regulating development and tissue homeostasis. In the developing neocortex, Sonic Hedgehog (Shh) regulates neural progenitor cell proliferation. During neurogenesis, radial glial cells of the ventricular zone (VZ) are the predominant neocortical progenitors that generate neurons through both symmetric and asymmetric divisions. Despite its importance, relatively little is known of the molecular pathways that control the switch from symmetric proliferative to differentiative/neurogenic divisions in neural progenitors. PRINCIPAL FINDINGS Here, we report that conditional inactivation of Patched1, a negative regulator of the Shh pathway, in Nestin positive neural progenitors of the neocortex leads to lamination defects due to improper corticogenesis and an increase in the number of symmetric proliferative divisions of the radial glial cells. Hedgehog-activated VZ progenitor cells demonstrated a concomitant upregulation of Hes1 and Blbp, downstream targets of Notch signaling. The Notch signaling pathway plays a pivotal role in the maintenance of stem/progenitor cells and the regulation of glial versus neuronal identity. To study the effect of Notch signaling on Hh-activated neural progenitors, we inactivated both Patched1 and Rbpj, a transcriptional mediator of Notch signaling, in Nestin positive cells of the neocortex. CONCLUSIONS Our data indicate that by mid neurogenesis (embryonic day 14.5), attenuation of Notch signaling reverses the effect of Patched1 deletion on neurogenesis by restoring the balance between symmetric proliferative and neurogenic divisions. Hence, our results demonstrate that correct corticogenesis is an outcome of the interplay between the Hh and Notch signaling pathways.
Collapse
|
59
|
Gomes FLAF, Zhang G, Carbonell F, Correa JA, Harris WA, Simons BD, Cayouette M. Reconstruction of rat retinal progenitor cell lineages in vitro reveals a surprising degree of stochasticity in cell fate decisions. Development 2010; 138:227-35. [PMID: 21148186 DOI: 10.1242/dev.059683] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In vivo cell lineage-tracing studies in the vertebrate retina have revealed that the sizes and cellular compositions of retinal clones are highly variable. It has been challenging to ascertain whether this variability reflects distinct but reproducible lineages among many different retinal progenitor cells (RPCs) or is the product of stochastic fate decisions operating within a population of more equivalent RPCs. To begin to distinguish these possibilities, we developed a method for long-term videomicroscopy to follow the lineages of rat perinatal RPCs cultured at clonal density. In such cultures, cell-cell interactions between two different clones are eliminated and the extracellular environment is kept constant, allowing us to study the cell-intrinsic potential of a given RPC. Quantitative analysis of the reconstructed lineages showed that the mode of division of RPCs is strikingly consistent with a simple stochastic pattern of behavior in which the decision to multiply or differentiate is set by fixed probabilities. The variability seen in the composition and order of cell type genesis within clones is well described by assuming that each of the four different retinal cell types generated at this stage is chosen stochastically by differentiating neurons, with relative probabilities of each type set by their abundance in the mature retina. Although a few of the many possible combinations of cell types within clones occur at frequencies that are incompatible with a fully stochastic model, our results support the notion that stochasticity has a major role during retinal development and therefore possibly in other parts of the central nervous system.
Collapse
Affiliation(s)
- Francisco L A F Gomes
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | | | | | | | | | | | | |
Collapse
|
60
|
Bilitou A, Ohnuma SI. The role of cell cycle in retinal development: cyclin-dependent kinase inhibitors co-ordinate cell-cycle inhibition, cell-fate determination and differentiation in the developing retina. Dev Dyn 2010; 239:727-36. [PMID: 20108332 DOI: 10.1002/dvdy.22223] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The mature retina is formed through multi-step developmental processes, including eye field specification, optic vesicle evagination, and cell-fate determination. Co-ordination of these developmental events with cell-proliferative activity is essential to achieve formation of proper retinal structure and function. In particular, the molecular and cellular dynamics of the final cell cycle significantly influence the identity that a cell acquires, since cell fate is largely determined at the final cell cycle for the production of postmitotic cells. This review summarizes our current understanding of the cellular mechanisms that underlie the co-ordination of cell-cycle and cell-fate determination, and also describes a molecular role of cyclin-dependent kinase inhibitors (CDKIs) as co-ordinators of cell-cycle arrest, cell-fate determination and differentiation.
Collapse
Affiliation(s)
- Aikaterini Bilitou
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | |
Collapse
|
61
|
|
62
|
Phoenix TN, Temple S. Spred1, a negative regulator of Ras-MAPK-ERK, is enriched in CNS germinal zones, dampens NSC proliferation, and maintains ventricular zone structure. Genes Dev 2010; 24:45-56. [PMID: 20047999 DOI: 10.1101/gad.1839510] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neural stem cells (NSCs) have great potential for self-renewal, which must be tightly regulated to generate appropriate cell numbers during development and to prevent tumor formation. The Ras-MAPK-ERK pathway affects mitogen-stimulated proliferation, and negative regulators are likely to be important for keeping self-renewal in check. Sprouty-related protein with an EVH1 domain (Spred1) is a recently discovered negative Ras-MAPK-ERK regulator linked to a neurofibromatosis 1 (NF-1)-like human syndrome; however, its role in CNS development has not been explored. We show that Spred1 is highly enriched in CNS germinal zones during neurogenesis. Spred1 knockdown increases NSC self-renewal and progenitor proliferation cell-autonomously, and overexpression causes premature differentiation. Surprisingly, Spred1 knockdown in vivo in the embryonic mouse forebrain frequently resulted in periventricular heterotopia, developmental abnormalities often associated with mutations in genes in the vesicular trafficking pathway that cause disruption of germinal zones and impair cell migration. In cortical progenitor cells, Spred1 localizes within distinct vesicles, indicating a potential role in transport. Spred1 knockdown gradually leads to disruption of the apical ventricular zone and loss of radial glia alignment. This impairs late neuronal migration, resulting in the formation of periventricular masses. Thus, Spred1 is critical for normal cortical development, as it modulates progenitor self-renewal/proliferation and helps maintain the integrity and organization of germinal zones.
Collapse
Affiliation(s)
- Timothy N Phoenix
- New York Neural Stem Cell Institute, Rensselaer, New York 12144, USA
| | | |
Collapse
|
63
|
Sahara S, O’Leary DDM. Fgf10 regulates transition period of cortical stem cell differentiation to radial glia controlling generation of neurons and basal progenitors. Neuron 2009; 63:48-62. [PMID: 19607792 PMCID: PMC2746711 DOI: 10.1016/j.neuron.2009.06.006] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Revised: 05/13/2009] [Accepted: 06/09/2009] [Indexed: 11/21/2022]
Abstract
Radial glia (RG), the progenitors of cortical neurons and basal progenitors (BPs), differentiate from neuroepithelial cells (NCs) with stem cell properties. We show that the morphogen Fgf10 is transiently expressed by NCs coincident with the transition period of NC differentiation into RG. Targeted deletion of Fgf10 delays RG differentiation, whereas overexpression has opposing effects. Delayed RG differentiation in Fgf10 mutants occurs selectively in rostral cortex, paralleled by an extended period of symmetric NC divisions increasing progenitor number, coupled with delayed and initially diminished production of neurons and BPs. RG eventually differentiate in excess number and overproduce neurons and BPs rostrally resulting in tangential expansion of frontal areas and increased laminar thickness. Thus, transient Fgf10 expression regulates timely differentiation of RG, and through this function, determines both length of the early progenitor expansion phase and onset of neurogenesis and ultimately the number of progenitors and neurons fated to specific cortical areas.
Collapse
Affiliation(s)
- Setsuko Sahara
- Molecular Neurobiology Laboratory, The Salk Institute, 10010 N. Torrey Pines Rd., La Jolla, CA 92037
| | - Dennis D. M. O’Leary
- Molecular Neurobiology Laboratory, The Salk Institute, 10010 N. Torrey Pines Rd., La Jolla, CA 92037
| |
Collapse
|
64
|
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: 12] [Impact Index Per Article: 0.8] [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
|
65
|
Nakagomi T, Taguchi A, Fujimori Y, Saino O, Nakano-Doi A, Kubo S, Gotoh A, Soma T, Yoshikawa H, Nishizaki T, Nakagomi N, Stern DM, Matsuyama T. Isolation and characterization of neural stem/progenitor cells from post-stroke cerebral cortex in mice. Eur J Neurosci 2009; 29:1842-52. [PMID: 19473237 DOI: 10.1111/j.1460-9568.2009.06732.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The CNS has the potential to marshal strong reparative mechanisms, including activation of endogenous neurogenesis, after a brain injury such as stroke. However, the response of neural stem/progenitor cells to stroke is poorly understood. Recently, neural stem/progenitor cells have been identified in the cerebral cortex, as well as previously recognized regions such as the subventricular or subgranular zones of the hippocampus, suggesting that a contribution of cortex-derived neural stem/progenitor cells may repair ischemic lesions of the cerebral cortex. In the present study, using a highly reproducible murine model of cortical infarction, we have found nestin-positive cells in the post-stroke cerebral cortex, but not in the non-ischemic cortex. Cells obtained from the ischemic core of the post-stroke cerebral cortex formed neurosphere-like cell clusters expressing nestin; such cells had the capacity for self-renewal and differentiated into electrophysiologically functional neurons, astrocytes and myelin-producing oligodendrocytes. Nestin-positive cells from the stroke-affected cortex migrated into the peri-infarct area and differentiated into glial cells in vivo. Although we could not detect differentiation of nestin-positive cells into neurons in vivo, our current observations indicate that endogenous neural stem/progenitors with the potential to become neurons can develop within post-stroke cerebral cortex.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Hyogo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Costa MR, Bucholz O, Schroeder T, Götz M. Late Origin of Glia-Restricted Progenitors in the Developing Mouse Cerebral Cortex. Cereb Cortex 2009; 19 Suppl 1:i135-43. [DOI: 10.1093/cercor/bhp046] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
67
|
Schwamborn JC, Berezikov E, Knoblich JA. The TRIM-NHL protein TRIM32 activates microRNAs and prevents self-renewal in mouse neural progenitors. Cell 2009; 136:913-25. [PMID: 19269368 PMCID: PMC2988196 DOI: 10.1016/j.cell.2008.12.024] [Citation(s) in RCA: 333] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 08/22/2008] [Accepted: 12/10/2008] [Indexed: 02/07/2023]
Abstract
In the mouse neocortex, neural progenitor cells generate both differentiating neurons and daughter cells that maintain progenitor fate. Here, we show that the TRIM-NHL protein TRIM32 regulates protein degradation and microRNA activity to control the balance between those two daughter cell types. In both horizontally and vertically dividing progenitors, TRIM32 becomes polarized in mitosis and is concentrated in one of the two daughter cells. TRIM32 overexpression induces neuronal differentiation while inhibition of TRIM32 causes both daughter cells to retain progenitor cell fate. TRIM32 ubiquitinates and degrades the transcription factor c-Myc but also binds Argonaute-1 and thereby increases the activity of specific microRNAs. We show that Let-7 is one of the TRIM32 targets and is required and sufficient for neuronal differentiation. TRIM32 is the mouse ortholog of Drosophila Brat and Mei-P26 and might be part of a protein family that regulates the balance between differentiation and proliferation in stem cell lineages.
Collapse
Affiliation(s)
- Jens C. Schwamborn
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Dr Bohr Gasse 3, 1030 Vienna, Austria
| | - Eugene Berezikov
- Hubrecht Institute, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Juergen A. Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Dr Bohr Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
68
|
Kelly TK, Karsten SL, Geschwind DH, Kornblum HI. Cell lineage and regional identity of cultured spinal cord neural stem cells and comparison to brain-derived neural stem cells. PLoS One 2009; 4:e4213. [PMID: 19148290 PMCID: PMC2615219 DOI: 10.1371/journal.pone.0004213] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 12/10/2008] [Indexed: 01/25/2023] Open
Abstract
Neural stem cells (NSCs) can be isolated from different regions of the central nervous system. There has been controversy whether regional differences amongst stem and progenitor cells are cell intrinsic and whether these differences are maintained during expansion in culture. The identification of inherent regional differences has important implications for the use of these cells in neural repair. Here, we compared NSCs derived from the spinal cord and embryonic cortex. We found that while cultured cortical and spinal cord derived NSCs respond similarly to mitogens and are equally neuronogenic, they retain and maintain through multiple passages gene expression patterns indicative of the region from which they were isolated (e.g Emx2 and HoxD10). Further microarray analysis identified 229 genes that were differentially expressed between cortical and spinal cord derived neurospheres, including many Hox genes, Nuclear receptors, Irx3, Pace4, Lhx2, Emx2 and Ntrk2. NSCs in the cortex express LeX. However, in the embryonic spinal cord there are two lineally related populations of NSCs: one that expresses LeX and one that does not. The LeX negative population contains few markers of regional identity but is able to generate LeX expressing NSCs that express markers of regional identity. LeX positive cells do not give rise to LeX-negative NSCs. These results demonstrate that while both embryonic cortical and spinal cord NSCs have similar self-renewal properties and multipotency, they retain aspects of regional identity, even when passaged long-term in vitro. Furthermore, there is a population of a LeX negative NSC that is present in neurospheres derived from the embryonic spinal cord but not the cortex.
Collapse
Affiliation(s)
- Theresa K Kelly
- The Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | | | |
Collapse
|
69
|
|
70
|
Slater JL, Landman KA, Hughes BD, Shen Q, Temple S. Cell lineage tree models of neurogenesis. J Theor Biol 2008; 256:164-79. [PMID: 18977364 DOI: 10.1016/j.jtbi.2008.09.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 09/10/2008] [Accepted: 09/24/2008] [Indexed: 01/06/2023]
Abstract
The production of neurons to form the mammalian cortex, known as embryonic cortical neurogenesis, is a complex developmental process. Insight into the process of cell division during neurogenesis is provided by murine cortical cell lineage trees, recorded through experimental observation. Recurring patterns within cell lineage trees may be indicative of predetermined cell behaviour. The application of mathematical modelling to this process requires careful consideration and identification of the key features to be incorporated into the model. A biologically plausible stochastic model of evolution of cell lineage trees is developed, based on the most important known features of neurogenesis. Tractable means of measuring lineage tree shape are discussed. Symmetry is identified as a significant feature of shape and is measured using Colless's Index of Imbalance. Distributions of tree size and imbalance for large tree sizes are computed and results compared to experimental data. Several refinements to the model are investigated, when the cell division probabilities are weighted according to cell generation. Two models involving generation-dependent cell division probabilities produce imbalance distributions which are the most consistent with the available experimental results. The results indicate that a stochastic cell division mechanism is a plausible basis of mammalian neurogenesis.
Collapse
Affiliation(s)
- Jennifer L Slater
- Department of Mathematics and Statistics, University of Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
71
|
Howard BM, Zhicheng Mo, Filipovic R, Moore AR, Antic SD, Zecevic N. Radial glia cells in the developing human brain. Neuroscientist 2008; 14:459-73. [PMID: 18467668 PMCID: PMC2702478 DOI: 10.1177/1073858407313512] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human radial glia (RG) share many of the features described in rodents, but also have a number of characteristics unique to the human brain. Results obtained from different mammalian species including human and non-human primates reveal differences in the involvement of RG in neurogenesis and oligodendrogenesis and in the timing of the initial expression of typical RG immunomarkers. A common problem in studying the human brain is that experimental procedures using modern molecular and genetic methods, such as in vivo transduction with retroviruses or creation of knockout or transgenic mutants, are not possible. Nevertheless, abundant and valuable information about the development of the human brain has been revealed using postmortem human material. Additionally, a combination and spectrum of in vitro techniques are used to gain knowledge about normal developmental processes in the human brain, including better understanding of RG as progenitor cells. Molecular and functional characterization of multipotent progenitors, such as RG, is important for future cell replacement therapies in neurological and psychiatric disorders, which are often resistant to conventional treatments. The protracted time of development and larger size of the human brain could provide insight into processes that may go unnoticed in the much smaller rodent cortex, which develops over a much shorter period. With that in mind, we summarize results on the role of RG in the human fetal brain.
Collapse
|
72
|
Fasano CA, Dimos JT, Ivanova NB, Lowry N, Lemischka IR, Temple S. shRNA knockdown of Bmi-1 reveals a critical role for p21-Rb pathway in NSC self-renewal during development. Cell Stem Cell 2008; 1:87-99. [PMID: 18371338 DOI: 10.1016/j.stem.2007.04.001] [Citation(s) in RCA: 248] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 01/25/2007] [Accepted: 04/02/2007] [Indexed: 12/31/2022]
Abstract
Knockout studies have shown that the polycomb gene Bmi-1 is important for postnatal, but not embryonic, neural stem cell (NSC) self-renewal and have identified the cell-cycle inhibitors p16/p19 as molecular targets. Here, using lentiviral-delivered shRNAs in vitro and in vivo, we determined that Bmi-1 is also important for NSC self-renewal in the embryo. We found that neural progenitors depend increasingly on Bmi-1 for proliferation as development proceeds from embryonic through adult stages. Acute shRNA-mediated Bmi-1 reduction causes defects in embryonic and adult NSC proliferation and self-renewal that, unexpectedly, are mediated by a different cell-cycle inhibitor, p21. Gene array analyses revealed developmental differences in Bmi-1-controlled expression of genes in the p21-Rb cell cycle regulatory pathway. Our data therefore implicate p21 as an important Bmi-1 target in NSCs, potentially with stage-related differences. Understanding stage-related mechanisms underlying NSC self-renewal has important implications for development of stem cell-based therapies.
Collapse
Affiliation(s)
- Christopher A Fasano
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | | | | | | | | | | |
Collapse
|
73
|
Wai MSM, Shi C, Kwong WH, Zhang L, Lam WP, Yew DT. Development of the human insular cortex: differentiation, proliferation, cell death, and appearance of 5HT-2A receptors. Histochem Cell Biol 2008; 130:1199-204. [DOI: 10.1007/s00418-008-0497-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2008] [Indexed: 11/29/2022]
|
74
|
Corbin JG, Gaiano N, Juliano SL, Poluch S, Stancik E, Haydar TF. Regulation of neural progenitor cell development in the nervous system. J Neurochem 2008; 106:2272-87. [PMID: 18819190 PMCID: PMC2640107 DOI: 10.1111/j.1471-4159.2008.05522.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The mammalian telencephalon, which comprises the cerebral cortex, olfactory bulb, hippocampus, basal ganglia, and amygdala, is the most complex and intricate region of the CNS. It is the seat of all higher brain functions including the storage and retrieval of memories, the integration and processing of sensory and motor information, and the regulation of emotion and drive states. In higher mammals such as humans, the telencephalon also governs our creative impulses, ability to make rational decisions, and plan for the future. Despite its massive complexity, exciting work from a number of groups has begun to unravel the developmental mechanisms for the generation of the diverse neural cell types that form the circuitry of the mature telencephalon. Here, we review our current understanding of four aspects of neural development. We first begin by providing a general overview of the broad developmental mechanisms underlying the generation of neuronal and glial cell diversity in the telencephalon during embryonic development. We then focus on development of the cerebral cortex, the most complex and evolved region of the brain. We review the current state of understanding of progenitor cell diversity within the cortical ventricular zone and then describe how lateral signaling via the Notch-Delta pathway generates specific aspects of neural cell diversity in cortical progenitor pools. Finally, we review the signaling mechanisms required for development, and response to injury, of a specialized group of cortical stem cells, the radial glia, which act both as precursors and as migratory scaffolds for newly generated neurons.
Collapse
Affiliation(s)
- Joshua G. Corbin
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| | - Nicholas Gaiano
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | - Sylvie Poluch
- Program in Neuroscience, USUHS, Bethesda, Maryland 20814
| | - Elizabeth Stancik
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| | - Tarik F. Haydar
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| |
Collapse
|
75
|
Nekrasova T, Jobes ML, Ting JH, Wagner GC, Minden A. Targeted disruption of the Pak5 and Pak6 genes in mice leads to deficits in learning and locomotion. Dev Biol 2008; 322:95-108. [PMID: 18675265 DOI: 10.1016/j.ydbio.2008.07.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 06/12/2008] [Accepted: 07/07/2008] [Indexed: 11/26/2022]
Abstract
PAK6 is a member of the group B family of PAK serine/threonine kinases, and is highly expressed in the brain. The group B PAKs, including PAK4, PAK5, and PAK6, were first identified as effector proteins for the Rho GTPase Cdc42. They have important roles in filopodia formation, the extension of neurons, and cell survival. Pak4 knockout mice die in utero, and the embryos have several abnormalities, including a defect in the development of motor neurons. In contrast, Pak5 knockout mice do not have any noticeable abnormalities. So far nothing is known about the biological function of Pak6. To address this, we have deleted the Pak6 gene in mice. Since Pak6 and Pak5 are both expressed in the brain, we also generated Pak5/Pak6 double knockout mice. These mice were viable and fertile, but had several locomotor and behavioral deficits. Our results indicate that Pak5 and Pak6 together are not required for viability, but are required for a normal level of locomotion and activity as well as for learning and memory. This is consistent with a role for the group B PAKs in the nervous system.
Collapse
Affiliation(s)
- Tanya Nekrasova
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | | | | | | | | |
Collapse
|
76
|
Nemeth MJ, Bodine DM. Regulation of hematopoiesis and the hematopoietic stem cell niche by Wnt signaling pathways. Cell Res 2008; 17:746-58. [PMID: 17768401 DOI: 10.1038/cr.2007.69] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare population of cells that are responsible for life-long generation of blood cells of all lineages. In order to maintain their numbers, HSCs must establish a balance between the opposing cell fates of self-renewal (in which the ability to function as HSCs is retained) and initiation of hematopoietic differentiation. Multiple signaling pathways have been implicated in the regulation of HSC cell fate. One such set of pathways are those activated by the Wnt family of ligands. Wnt signaling pathways play a crucial role during embryogenesis and deregulation of these pathways has been implicated in the formation of solid tumors. Wnt signaling also plays a role in the regulation of stem cells from multiple tissues, such as embryonic, epidermal, and intestinal stem cells. However, the function of Wnt signaling in HSC biology is still controversial. In this review, we will discuss the basic characteristics of the adult HSC and its regulatory microenvironment, the "niche", focusing on the regulation of the HSC and its niche by the Wnt signaling pathways.
Collapse
Affiliation(s)
- Michael J Nemeth
- Hematopoiesis Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda, MD 20892-4442, USA.
| | | |
Collapse
|
77
|
|
78
|
Lowry N, Goderie SK, Adamo M, Lederman P, Charniga C, Gill J, Silver J, Temple S. Multipotent embryonic spinal cord stem cells expanded by endothelial factors and Shh/RA promote functional recovery after spinal cord injury. Exp Neurol 2008; 209:510-22. [DOI: 10.1016/j.expneurol.2007.09.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 09/22/2007] [Indexed: 01/27/2023]
|
79
|
Pinto L, Mader MT, Irmler M, Gentilini M, Santoni F, Drechsel D, Blum R, Stahl R, Bulfone A, Malatesta P, Beckers J, Götz M. Prospective isolation of functionally distinct radial glial subtypes--lineage and transcriptome analysis. Mol Cell Neurosci 2008; 38:15-42. [PMID: 18372191 DOI: 10.1016/j.mcn.2008.01.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 01/07/2008] [Indexed: 12/18/2022] Open
Abstract
Since the discovery of radial glia as the source of neurons, their heterogeneity in regard to neurogenesis has been described by clonal and time-lapse analysis in vitro. However, the molecular determinants specifying neurogenic radial glia differently from radial glia that mostly self-renew remain ill-defined. Here, we isolated two radial glial subsets that co-exist at mid-neurogenesis in the developing cerebral cortex and their immediate progeny. While one subset generates neurons directly, the other is largely non-neurogenic but also gives rise to Tbr2-positive basal precursors, thereby contributing indirectly to neurogenesis. Isolation of these distinct radial glia subtypes allowed determining interesting differences in their transcriptome. These transcriptomes were also strikingly different from the transcriptome of radial glia isolated at the end of neurogenesis. This analysis therefore identifies, for the first time, the lineage origin of basal progenitors and the molecular differences of this lineage in comparison to directly neurogenic and gliogenic radial glia.
Collapse
Affiliation(s)
- Luisa Pinto
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Stem Cell Research, Ingolstädter Landstr. 1, 85764 Neuherberg/Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Ashton RS, Peltier J, Fasano CA, O'Neill A, Leonard J, Temple S, Schaffer DV, Kane RS. High-throughput screening of gene function in stem cells using clonal microarrays. Stem Cells 2007; 25:2928-35. [PMID: 17673524 DOI: 10.1634/stemcells.2007-0468] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We describe a microarray-based approach for the high-throughput screening of gene function in stem cells and demonstrate the potential of this method by growing and isolating clonal populations of both adult and embryonic neural stem cells. Clonal microarrays are constructed by seeding a population of cells at clonal density on micropatterned surfaces generated using soft lithographic microfabrication techniques. Clones of interest can be isolated after assaying in parallel for various cellular processes and functions, including proliferation, signal transduction, and differentiation. We demonstrate the compatibility of the technique with both gain- and loss-of-function studies using cell populations infected with cDNA libraries or DNA constructs that induce RNA interference. The infection of cells with a library prior to seeding and the compact but isolated growth of clonal cell populations will facilitate the screening of large libraries in a wide variety of mammalian cells, including those that are difficult to transfect by conventional methods.
Collapse
Affiliation(s)
- Randolph S Ashton
- The Howard P. Isermann Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, USA
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Bani-Yaghoub M, Kubu CJ, Cowling R, Rochira J, Nikopoulos GN, Bellum S, Verdi JM. A switch in numb isoforms is a critical step in cortical development. Dev Dyn 2007; 236:696-705. [PMID: 17253625 DOI: 10.1002/dvdy.21072] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Loss of numb function suggests that numb maintains progenitors in an undifferentiated state. Herein, we demonstrate that numb1 and numb3 are expressed in undifferentiated cortical progenitors, whereas numb2 and numb4 become prominent throughout differentiation. To further assess the role of different numb isoforms in cortical neural development, we first created a Numb-null state with antisense morpholino, followed by the re-expression of specific numb isoforms. The re-expression of numb1 or numb3 resulted in a significant reduction of neural differentiation, correlating with an expansion of the cortical progenitor pool. In contrast, the expression of numb2 or numb4 resulted in a reduction of proliferating progenitors and a corresponding increase in mammalian achete-scute homologue (MASH1) expression, concurrent with the appearance of the microtubule[corrected]-associated [corrected] protein-2-positive neurons. Of interest, the effect of numb isoforms on neural differentiation could not be directly related to Notch, because classic canonical Notch signaling assays failed to uncover any differences in the four isoforms to inhibit the Notch downstream events. This finding suggests that numb may have other signaling properties during neuronal differentiation in addition to augmenting notch signal strength.
Collapse
Affiliation(s)
- Mahmud Bani-Yaghoub
- Laboratory of Neural Stem Cell Biology, Robarts Research Institute, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
82
|
Spella M, Britz O, Kotantaki P, Lygerou Z, Nishitani H, Ramsay RG, Flordellis C, Guillemot F, Mantamadiotis T, Taraviras S. Licensing regulators Geminin and Cdt1 identify progenitor cells of the mouse CNS in a specific phase of the cell cycle. Neuroscience 2007; 147:373-87. [PMID: 17533120 DOI: 10.1016/j.neuroscience.2007.03.050] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 03/27/2007] [Accepted: 03/31/2007] [Indexed: 01/16/2023]
Abstract
Nervous system formation integrates control of cellular proliferation and differentiation and is mediated by multipotent neural progenitor cells that become progressively restricted in their developmental potential before they give rise to differentiated neurons and glial cells. Evidence from different experimental systems suggests that Geminin is a candidate molecule linking proliferation and differentiation during nervous system development. We show here that Geminin and its binding partner Cdt1 are expressed abundantly by neural progenitor cells during early mouse neurogenesis. Their expression levels decline at late developmental stages and become undetectable upon differentiation. Geminin and Cdt1 expressing cells also express Sox2 while no overlap is detected with cells expressing markers of a differentiated neuronal phenotype. A fraction of radial glial cells expressing RC2 and Pax6 are also immunoreactive for Geminin and Cdt1. The majority of the Geminin and Cdt1 expressing cell populations appears to be distinct from fate-restricted precursor cells expressing Mash1 or Neurogenin2. Bromo-deoxy-uridine (BrdU) incorporation experiments reveal a cell cycle specific expression in neural progenitor cells, with Geminin being present from S to M phase, while Cdt1 expression characterizes progenitor cells in G1 phase. Furthermore, in vitro differentiation of adult neurosphere cultures shows downregulation of Geminin/Cdt1 in the differentiated state, in line with our data showing that Geminin is present in neural progenitor cells of the CNS during mouse embryogenesis and adulthood and becomes downregulated upon cell fate specification and differentiation. This suggests a role for Geminin in the formation and maintenance of the neural progenitor cells.
Collapse
Affiliation(s)
- M Spella
- Department of Pharmacology, School of Medicine, University of Patras, 26500 Rio, Patras, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Zhou Y, Atkins JB, Rompani SB, Bancescu DL, Petersen PH, Tang H, Zou K, Stewart SB, Zhong W. The Mammalian Golgi Regulates Numb Signaling in Asymmetric Cell Division by Releasing ACBD3 during Mitosis. Cell 2007; 129:163-78. [PMID: 17418793 DOI: 10.1016/j.cell.2007.02.037] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2006] [Revised: 11/27/2006] [Accepted: 02/16/2007] [Indexed: 01/04/2023]
Abstract
Mammalian neural progenitor cells divide asymmetrically to self-renew and produce a neuron by segregating cytosolic Numb proteins primarily to one daughter cell. Numb signaling specifies progenitor over neuronal fates but, paradoxically, also promotes neuronal differentiation. Here we report that ACBD3 is a Numb partner in cell-fate specification. ACBD3 and Numb proteins interact through an essential Numb domain, and the respective loss- and gain-of-function mutant mice share phenotypic similarities. Interestingly, ACBD3 associates with the Golgi apparatus in neurons and interphase progenitor cells but becomes cytosolic after Golgi fragmentation during mitosis, when Numb activity is needed to distinguish the two daughter cells. Accordingly, cytosolic ACBD3 can act synergistically with Numb to specify cell fates, and its continuing presence during the progenitor cell cycle inhibits neuron production. We propose that Golgi fragmentation and reconstitution during cell cycle differentially regulate Numb signaling through changes in ACBD3 subcellular distribution and represent a mechanism for coupling cell-fate specification and cell-cycle progression.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Ramunas J, Illman M, Kam A, Farn K, Kelly L, Morshead CM, Jervis EJ. True monolayer cell culture in a confined 3D microenvironment enables lineage informatics. Cytometry A 2007; 69:1202-11. [PMID: 17066473 DOI: 10.1002/cyto.a.20341] [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] [Indexed: 01/10/2023]
Abstract
BACKGROUND There is a need for methods to (1) track cells continuously to generate lineage trees; (2) culture cells in in vivo-like microenvironments; and (3) measure many biological parameters simultaneously and noninvasively. Herein, we present a novel imaging culture chamber that facilitates "lineage informatics," a lineage-centric approach to cytomics. METHODS We cultured cells in a confined monolayer using a novel "gap chamber" that produces images with confocal-like qualities using standard DIC microscopy. Lineage and other cytometric data were semiautomatically extracted from image sets of neural stem and progenitor cells and analyzed using lineage informatics. RESULTS Cells imaged in the chamber every 3 min could be tracked for at least 6 generations allowing for the construction of extensive lineage trees with multiparameter data sets at hundreds of time points for each cell. The lineage informatics approach reveals relationships between lineage, phenotype, and microenvironment. Mass transfer characteristics and 3D geometry make the chamber more in vivo-like than traditional culture systems. CONCLUSIONS The gap chamber allows cells to be cultured, imaged, and tracked in true monolayers permitting detailed informatics analysis of cell lineage, phenotype, and fate determinants. The chamber is biomimetic and straightforward to build and use, and should find many applications in long-term cell imaging.
Collapse
Affiliation(s)
- John Ramunas
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | | | | | | | | | | |
Collapse
|
85
|
Nicolis SK. Cancer stem cells and "stemness" genes in neuro-oncology. Neurobiol Dis 2007; 25:217-29. [PMID: 17141509 DOI: 10.1016/j.nbd.2006.08.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Accepted: 08/27/2006] [Indexed: 11/29/2022] Open
Abstract
The main properties of stem cells include long-term self-renewal and the capacity to give rise to one or more types of differentiated progeny. Recently, much evidence was provided that leukemia and tumor maintenance and growth are sustained by a small proportion of cells exhibiting stem cell properties. In neural tumors, stem cells have been detected in glioblastoma, medulloblastoma and ependymoma. These observations imply that normal stem cells could be the origin of cancer stem cells; alternatively, a more differentiated progeny may revert to a "stem-like" status, and give rise to cancer stem cells. In adult brain residual stem cells are located in the hippocampus, the subventricular zone and possibly the cerebellum. However, evidence for the ability of more differentiated progeny (astroglia, oligodendroglia) to convert into "stem cells" in vitro has also been provided, thus greatly expanding the potential target of oncogenic mutations. In the framework of the cancer stem cell hypothesis, genes originally identified as important for normal neural stem cells may be essential to support cancer stem cells as well. Stem cell genes act in several ways: they stimulate stem cell self-replication, inhibit differentiation, control excessive replication that might lead to "exhaustion" of the stem cell pool. Mutations in man and mouse, in spontaneous or experimental brain tumors, often target stem cell genes or genes lying in their functional pathway, the main examples being the Sonic hedgehog and the Wnt pathways. Interestingly, several stem cell genes are often overexpressed in brain tumors, even if they are not mutated. This suggests that these genes may be important for the generation of cancer stem cells from more differentiated precursors, or for cancer stem cell maintenance. Cancer stem cells partially differentiate in vivo, and in vitro they also give rise to seemingly normal differentiated progeny, like normal stem cells: thus, their main defect, leading to cancer, may lie in the unbalance between self-replication and terminal differentiation of this minority cell population. Knowledge of extrinsic diffusible factors affecting the activity of stem cell genes may help identifying tools for inducing cancer stem cell differentiation, which might be of use in therapy.
Collapse
Affiliation(s)
- Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano Bicocca, piazza della Scienza 2, 20126 Milano, Italy.
| |
Collapse
|
86
|
Miyata T. Asymmetric cell division during brain morphogenesis. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2007; 45:121-42. [PMID: 17585499 DOI: 10.1007/978-3-540-69161-7_6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The division patterns of neural progenitor cells in developing vertebrate brains have traditionally been classified into three types: (i) "symmetric" divisions producing two progenitor cells (P/P division), (ii) "symmetric" divisions producing two neurons (N/N division), and (iii) "asymmetric" divisions producing one progenitor cell and one neuron (P/N division). Many studies examining the mechanism(s) regulating P/N divisions have focused on mitotic cleavage orientation and the possible uneven distribution of cell-fate determining molecules such as Numb. Although these two factors may intrinsically determine daughter cell fate arising from M-phase progenitor cells, no unified explanations have yet to be put forth incorporating all available data. In this review, I will discuss recent advances in techniques allowing the more detailed monitoring of daughter cell behavior in a heterogeneously pseudostratified neuroepithelium that demonstrate previously unrecognized asymmetries in P/P divisions. Careful observations of daughter cell behavior suggest that, immediately after their birth at the apical surface of the neuroepithelium, generated cells may not yet be fate committed but rather integrate extrinsic and intrinsic signals during GI phase before continuing down a developmental pathway.
Collapse
Affiliation(s)
- Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya, Aichi 466-8550, Japan.
| |
Collapse
|
87
|
Lathia JD, Rao MS, Mattson MP, ffrench-Constant C. The microenvironment of the embryonic neural stem cell: Lessons from adult niches? Dev Dyn 2007; 236:3267-82. [DOI: 10.1002/dvdy.21319] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
88
|
Abstract
Currently there is an intense effort being made to elucidate the factors that control stem and progenitor cell fate. Developments in our understanding of the FGF/FGFR pathway and its role as an effector of stem cell pluripotency have heightened expectations that a therapeutic use for stem cells will move from a possibility to a probability. Mounting evidence is revealing the molecular mechanisms by which fibroblast growth factor (FGF) signaling, together with a large number of other growth and adhesive factors, is controlled by the extracellular sugar, heparan sulfate (HS). What has resulted is a novel means of augmenting and thus regulating the growth factor control of stem and progenitor cell fate. Here, we review the numerous bioactivities of HS, and the development of strategies to implement HS-induced control of cell fate decisions.
Collapse
Affiliation(s)
- Simon M Cool
- Laboratory of Stem Cells and Tissue Repair, Institute of Molecular and Cell Biology, Proteos, 61 Biopolis Drive, Singapore 138673.
| | | |
Collapse
|
89
|
Bouhon IA, Joannides A, Kato H, Chandran S, Allen ND. Embryonic stem cell-derived neural progenitors display temporal restriction to neural patterning. Stem Cells 2006; 24:1908-13. [PMID: 16627686 DOI: 10.1634/stemcells.2006-0031] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Neural stem cells have considerable therapeutic potential because of their ability to generate defined neuronal cell types for use in drug screening studies or cell-based therapies for neurodegenerative diseases. In this study, we differentiate mouse embryonic stem cells to neural progenitors with an initial forebrain identity in a defined system that enables systematic manipulation to generate more caudal fates, including motoneurons. We demonstrate that the ability to pattern embryonic stem cell-derived neural progenitors is temporally restricted and show that the loss of responsiveness to morphogenetic cues correlates with constitutive expression of the basic helix-loop-helix transcription factors Olig2 and Mash1, epidermal growth factor receptor, and vimentin and parallels the onset of gliogenesis. We provide evidence for two temporal classes of embryonic stem cell-derived putative radial glia that coincide with a transition from neurogenesis to gliogenesis and a concomitant loss of regional identity.
Collapse
Affiliation(s)
- Isabelle A Bouhon
- Neurobiology Programme, The Babraham Institute, Babraham, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
90
|
Branching stochastic processes with immigration in analysis of renewing cell populations. Math Biosci 2006; 203:37-63. [PMID: 16904129 DOI: 10.1016/j.mbs.2006.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 02/22/2006] [Accepted: 06/06/2006] [Indexed: 10/24/2022]
Abstract
This paper considers the utility of a new class of stochastic branching processes with non-homogeneous immigration in modeling complex renewing cell systems. Such systems typically include the population of stem cells that provides an inexhaustible supply of cells necessary for maintaining the cellular composition of a tissue. A stem cell may be induced to transform (differentiate) into a progenitor cell. Progenitor cells retain the ability to proliferate and their function is believed to provide a quick proliferative response to an increased demand for cells in the population. There may be several sub-types of progenitor cells. Terminally differentiated cells do not divide under normal conditions; they are responsible for maintaining tissue-specific functions. Recent advancements in experimental techniques offer considerable scope for quantitative studies of in vivo cell kinetics based on stochastic modeling of renewing cell populations. However, no ready-made theory is currently available to take full advantage of these advancements. This paper introduces such a theory with a special focus on its feasibility in biological applications.
Collapse
|
91
|
|
92
|
Shen Q, Wang Y, Dimos JT, Fasano CA, Phoenix TN, Lemischka IR, Ivanova NB, Stifani S, Morrisey EE, Temple S. The timing of cortical neurogenesis is encoded within lineages of individual progenitor cells. Nat Neurosci 2006; 9:743-51. [PMID: 16680166 DOI: 10.1038/nn1694] [Citation(s) in RCA: 461] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 04/04/2006] [Indexed: 01/03/2023]
Abstract
In the developing cerebral cortex, neurons are born on a predictable schedule. Here we show in mice that the essential timing mechanism is programmed within individual progenitor cells, and its expression depends solely on cell-intrinsic and environmental factors generated within the clonal lineage. Multipotent progenitor cells undergo repeated asymmetric divisions, sequentially generating neurons in their normal in vivo order: first preplate cells, including Cajal-Retzius neurons, then deep and finally superficial cortical plate neurons. As each cortical layer arises, stem cells and neuroblasts become restricted from generating earlier-born neuron types. Growth as neurospheres or in co-culture with younger cells did not restore their plasticity. Using short-hairpin RNA (shRNA) to reduce Foxg1 expression reset the timing of mid- but not late-gestation progenitors, allowing them to remake preplate neurons and then cortical-plate neurons. Our data demonstrate that neural stem cells change neuropotency during development and have a window of plasticity when restrictions can be reversed.
Collapse
Affiliation(s)
- Qin Shen
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York 12208, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Bani-Yaghoub M, Tremblay RG, Lei JX, Zhang D, Zurakowski B, Sandhu JK, Smith B, Ribecco-Lutkiewicz M, Kennedy J, Walker PR, Sikorska M. Role of Sox2 in the development of the mouse neocortex. Dev Biol 2006; 295:52-66. [PMID: 16631155 DOI: 10.1016/j.ydbio.2006.03.007] [Citation(s) in RCA: 220] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 03/03/2006] [Accepted: 03/08/2006] [Indexed: 01/24/2023]
Abstract
The mammalian neocortex is established from neural stem and progenitor cells that utilize specific transcriptional and environmental factors to create functional neurons and astrocytes. Here, we examined the mechanism of Sox2 action during neocortical neurogenesis and gliogenesis. We established a robust Sox2 expression in neural stem and progenitor cells within the ventricular zone, which persisted until the cells exited the cell cycle. Overexpression of constitutively active Sox2 in neural progenitors resulted in upregulation of Notch1, recombination signal-sequence binding protein-J (RBP-J) and hairy enhancer of split 5 (Hes5) transcripts and the Sox2 high mobility group (HMG) domain seemed sufficient to confer these effects. While Sox2 overexpression permitted the differentiation of progenitors into astroglia, it inhibited neurogenesis, unless the Notch pathway was blocked. Moreover, neuronal precursors engaged a serine protease(s) to eliminate the overexpressed Sox2 protein and relieve the repression of neurogenesis. Glial precursors and differentiated astrocytes, on the other hand, maintained Sox2 expression until they reached a quiescent state. Sox2 expression was re-activated by signals that triggered astrocytic proliferation (i.e., injury, mitogenic and gliogenic factors). Taken together, Sox2 appears to act upstream of the Notch signaling pathway to maintain the cell proliferative potential and to ensure the generation of sufficient cell numbers and phenotypes in the developing neocortex.
Collapse
Affiliation(s)
- Mahmud Bani-Yaghoub
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council of Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON, Canada K1A 0R6.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Petersen PH, Tang H, Zou K, Zhong W. The Enigma of the Numb-Notch Relationship during Mammalian Embryogenesis. Dev Neurosci 2006; 28:156-68. [PMID: 16508312 DOI: 10.1159/000090761] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Accepted: 10/17/2005] [Indexed: 01/01/2023] Open
Abstract
Asymmetric cell division is an attractive means to diversify cell fates during development and for stem cells to balance self-renewal and differentiation. In Drosophila, two signaling pathways, one mediated by Numb and the other by Notch, play essential but antagonistic roles in enabling the two daughters to adopt different fates after a wide variety of asymmetric cell divisions. However, recent studies show that mutating mammalian Numb homologues, m-Numb and Numblike (Numbl or Nbl), and eliminating Notch signaling in the developing nervous system both lead to premature depletion of neural stem/progenitor cells in mice. These findings raise an interesting question as to whether and how the antagonistic roles of Numb and Notch signaling are conserved in vertebrates. Here we provide evidence that loss of m-Numb and Numbl outside the embryonic nervous system also causes phenotypes similar to those exhibited by mice with defective Notch signaling. We further show that very little Numb protein is necessary for embryogenesis and that the presence of different m-Numb isoforms is unlikely to provide a molecular basis for differential regulation of Notch signaling in mammals, as these isoforms are functionally indistinguishable in cell fate specification. We discuss possible mechanisms by which the antagonistic roles of Numb and Notch are evolutionarily conserved to meet the needs of stem cell maintenance during mammalian neurogenesis.
Collapse
Affiliation(s)
- Petur H Petersen
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
95
|
Capela A, Temple S. LeX is expressed by principle progenitor cells in the embryonic nervous system, is secreted into their environment and binds Wnt-1. Dev Biol 2006; 291:300-13. [PMID: 16458284 DOI: 10.1016/j.ydbio.2005.12.030] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 12/12/2005] [Accepted: 12/14/2005] [Indexed: 11/22/2022]
Abstract
LeX/SSEA1/CD15 is an extracellular matrix-associated carbohydrate expressed by ES cells and by adult neural and bone marrow stem cells. It is important for cell adhesion, compaction and FGF2 responses of early embryonic stem cells; however, its function at later stages is not clear. We now show that LeX is expressed by primary mouse neural progenitor cells, including neural stem cells, neuroblasts and glioblasts, but not by their more differentiated products. LeX distinguishes highly proliferative cells even in the primitive neuroepithelium, demonstrating heterogeneity in cell potential before radial glia arise. At later stages, LeX expressing progenitors are frequently radial in morphology. Surface LeX expression can be used to enrich neural stem and progenitor cells from different CNS regions throughout development by FACS. We found that LeX expression is particularly strong in neural regions with prolonged neurogenesis, e.g., the olfactory epithelium, hippocampus, basal forebrain and cerebellum. These regions also express high levels of the growth factors FGF8 and/or Wnt-1. We show here that LeX-containing molecules in the developing nervous system bind Wnt-1. Our findings suggest that LeX, which is present on the surface of principle neural progenitors and secreted into their extracellular niche, may bind and present growth factors important for their proliferation and self-renewal.
Collapse
Affiliation(s)
- Alexandra Capela
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, NY 12208, USA
| | | |
Collapse
|
96
|
Abstract
During the development of the mammalian central nervous system, neural stem cells and their derivative progenitor cells generate neurons by asymmetric and symmetric divisions. The proliferation versus differentiation of these cells and the type of division are closely linked to their epithelial characteristics, notably, their apical-basal polarity and cell-cycle length. Here, we discuss how these features change during development from neuroepithelial to radial glial cells, and how this transition affects cell fate and neurogenesis.
Collapse
Affiliation(s)
- Magdalena Götz
- Institute for Stem Cell Research, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg/Munich, Germany.
| | | |
Collapse
|
97
|
Mitrusková B, Orendácová J, Raceková E. Fluoro Jade-B detection of dying cells in the SVZ and RMS of adult rats after bilateral olfactory bulbectomy. Cell Mol Neurobiol 2005; 25:1255-64. [PMID: 16388336 PMCID: PMC11529220 DOI: 10.1007/s10571-005-8502-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Accepted: 12/23/2004] [Indexed: 11/30/2022]
Abstract
A novel fluorochrome, Fluoro-Jade B, was used to detect dying precursor cells in the subventricular zone (SVZ) and rostral migratory stream (RMS) of adult rats after bilateral olfactory bulbectomy and in control intact rats. The animals in experimental group were left to survive 3 days and from 3 till 16 months after surgical procedure. 1. In the control animals, Fluoro-Jade B positive cells were visible in the SVZ and within the whole extent of the RMS. The number of Fluoro-Jade B positive cells increased in the elbow in comparison to the rest parts of the RMS. 2. In the experimental animals surviving either 3 days or from 3 till 16 months after bilateral olfactory bulbectomy, Fluoro-Jade B positive cells displayed the similar pattern of distribution as in the control animals. However, some quantitative differences in the labeled cells number along the rostral migratory pathway appeared. 3. The average number of degenerating cells within the control SVZ and RMS was 26.24+/- 0.686. In bulbectomized animals, regardless of survival time, an insignificant increase of Fluoro-Jade B positive cells number occurred. We can conclude that dying of precursor cells is a physiological process running within the SVZ/RMS in both control and experimental animals. Moreover, this physiological process is not influenced by survival period after bilateral olfactory bulbectomy. Our results demonstrate Fluoro-Jade B as a useful marker of dying cells.
Collapse
Affiliation(s)
- Barbora Mitrusková
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic.
| | | | | |
Collapse
|
98
|
Riaz SS, Bradford HF. Factors involved in the determination of the neurotransmitter phenotype of developing neurons of the CNS: Applications in cell replacement treatment for Parkinson's disease. Prog Neurobiol 2005; 76:257-78. [PMID: 16256257 DOI: 10.1016/j.pneurobio.2005.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Revised: 06/07/2005] [Accepted: 08/04/2005] [Indexed: 02/08/2023]
Abstract
The developmental stages involved in the conversion of stem cells to fully functional neurons of specific neurotransmitter phenotype are complex and not fully understood. Over the past decade many studies have been published that demonstrate that in vitro manipulation of the epigenetic environment of the stem cells allows experimental control of final neuronal phenotypic choice. This review presents the evidence for the involvement of a number of endogenous neurobiochemicals, which have been reported to potently influence DAergic (and other neurotransmitter) phenotype expression in vitro. They act at different stages on the pathway to neurotransmitter phenotype determination, and in different ways. Many are better known for their involvement in other aspects of development, and in other biochemical roles. Their proper place, and precise roles, in neurotransmitter phenotype determination in vivo will no doubt be determined in the future. Meanwhile, considerable medical benefits are offered from producing large, long-term, viable cryostores of self-regenerating multipotential neural precursor cells (i.e., brain stem cells), which can be used for cell replacement therapies in the treatment of degenerative brain diseases, such as Parkinson's disease.
Collapse
Affiliation(s)
- S S Riaz
- Department of Biological Sciences, Imperial College of Science, Technology and Medicine, Biochemistry Building, South Kensington Campus, Imperial College Road, SW7 2AZ London, UK
| | | |
Collapse
|
99
|
Abramova N, Charniga C, Goderie SK, Temple S. Stage-specific changes in gene expression in acutely isolated mouse CNS progenitor cells. Dev Biol 2005; 283:269-81. [PMID: 15890332 DOI: 10.1016/j.ydbio.2005.03.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2004] [Revised: 03/04/2005] [Accepted: 03/31/2005] [Indexed: 01/06/2023]
Abstract
Neural progenitor cells can be derived from a variety of developmental stages when they are preferentially proliferating, undergoing neurogenesis or undergoing gliogenesis. We used FACS sorting and the LeX surface marker to enrich neural progenitor cells from different embryonic stages and adult and compared their gene expression profiles using Affymetrix Microarrays. Our results show that, while there are common genes expressed in the progenitor cell population from all stages, there are also significant differences in gene expression patterns that correlate with stage-related behaviors. These data indicate that progenitor cells change during development and that adult and embryonic neural progenitor cells are intrinsically different.
Collapse
Affiliation(s)
- Natalia Abramova
- Center for Neuropharmacology and Neuroscience, Albany Medical College, NY 12208, USA.
| | | | | | | |
Collapse
|
100
|
Sun Y, Goderie SK, Temple S. Asymmetric distribution of EGFR receptor during mitosis generates diverse CNS progenitor cells. Neuron 2005; 45:873-86. [PMID: 15797549 DOI: 10.1016/j.neuron.2005.01.045] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 12/23/2004] [Accepted: 01/27/2005] [Indexed: 11/25/2022]
Abstract
It has been debated whether asymmetric distribution of cell surface receptors during mitosis could generate asymmetric cell divisions by yielding daughters with different environmental responsiveness and, thus, different fates. We have found that in mouse embryonic forebrain ventricular and subventricular zones, the EGFR can distribute asymmetrically during mitosis in vivo and in vitro. This occurs during divisions yielding two Nestin+ progenitor cells, via an actin-dependent mechanism. The resulting sibling progenitor cells respond differently to EGFR ligand in terms of migration and proliferation. Moreover, they express different phenotypic markers: the EGFRhigh daughter usually has radial glial/astrocytic markers, while its EGFRlow sister lacks them, indicating fate divergence. Lineage trees of cultured cortical glioblasts reveal repeated EGFR asymmetric distribution, and asymmetric divisions underlie formation of oligodendrocytes and astrocytes in clones. These data suggest that asymmetric EGFR distribution contributes to forebrain development by creating progenitors with different proliferative, migratory, and differentiation responses to ligand.
Collapse
Affiliation(s)
- Yu Sun
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York 12208, USA
| | | | | |
Collapse
|