101
|
Human stem cell-derived spinal cord astrocytes with defined mature or reactive phenotypes. Cell Rep 2013; 4:1035-1048. [PMID: 23994478 DOI: 10.1016/j.celrep.2013.06.021] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 05/15/2013] [Accepted: 06/18/2013] [Indexed: 12/23/2022] Open
Abstract
Differentiation of astrocytes from human stem cells has significant potential for analysis of their role in normal brain function and disease, but existing protocols generate only immature astrocytes. Using early neuralization, we generated spinal cord astrocytes from mouse or human embryonic or induced pluripotent stem cells with high efficiency. Remarkably, short exposure to fibroblast growth factor 1 (FGF1) or FGF2 was sufficient to direct these astrocytes selectively toward a mature quiescent phenotype, as judged by both marker expression and functional analysis. In contrast, tumor necrosis factor alpha and interleukin-1β, but not FGFs, induced multiple elements of a reactive inflammatory phenotype but did not affect maturation. These phenotypically defined, scalable populations of spinal cord astrocytes will be important both for studying normal astrocyte function and for modeling human pathological processes in vitro.
Collapse
|
102
|
Molofsky AV, Glasgow SM, Chaboub LS, Tsai HH, Murnen AT, Kelley KW, Fancy SPJ, Yuen TJ, Madireddy L, Baranzini S, Deneen B, Rowitch DH, Oldham MC. Expression profiling of Aldh1l1-precursors in the developing spinal cord reveals glial lineage-specific genes and direct Sox9-Nfe2l1 interactions. Glia 2013; 61:1518-32. [PMID: 23840004 DOI: 10.1002/glia.22538] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/10/2013] [Accepted: 05/15/2013] [Indexed: 01/26/2023]
Abstract
Developmental regulation of gliogenesis in the mammalian CNS is incompletely understood, in part due to a limited repertoire of lineage-specific genes. We used Aldh1l1-GFP as a marker for gliogenic radial glia and later-stage precursors of developing astrocytes and performed gene expression profiling of these cells. We then used this dataset to identify candidate transcription factors that may serve as glial markers or regulators of glial fate. Our analysis generated a database of developmental stage-related markers of Aldh1l1+ cells between murine embryonic day 13.5-18.5. Using these data we identify the bZIP transcription factor Nfe2l1 and demonstrate that it promotes glial fate under direct Sox9 regulatory control. Thus, this dataset represents a resource for identifying novel regulators of glial development.
Collapse
Affiliation(s)
- Anna V Molofsky
- Department of Pediatrics, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Prion replication elicits cytopathic changes in differentiated neurosphere cultures. J Virol 2013; 87:8745-55. [PMID: 23740992 DOI: 10.1128/jvi.00572-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The molecular mechanisms of prion-induced cytotoxicity remain largely obscure. Currently, only a few cell culture models have exhibited the cytopathic changes associated with prion infection. In this study, we introduced a cell culture model based on differentiated neurosphere cultures isolated from the brains of neonatal prion protein (PrP)-null mice and transgenic mice expressing murine PrP (dNP0 and dNP20 cultures). Upon exposure to mouse Chandler prions, dNP20 cultures supported the de novo formation of abnormal PrP and the resulting infectivity, as assessed by bioassays. Furthermore, this culture was susceptible to various prion strains, including mouse-adapted scrapie, bovine spongiform encephalopathy, and Gerstmann-Sträussler-Scheinker syndrome prions. Importantly, a subset of the cells in the infected culture that was mainly composed of astrocyte lineage cells consistently displayed late-occurring, progressive signs of cytotoxicity as evidenced by morphological alterations, decreased cell viability, and increased lactate dehydrogenase release. These signs of cytotoxicity were not observed in infected dNP0 cultures, suggesting the requirement of endogenous PrP expression for prion-induced cytotoxicity. Degenerated cells positive for glial fibrillary acidic protein accumulated abnormal PrP and exhibited features of apoptotic death as assessed by active caspase-3 and terminal deoxynucleotidyltransferase nick-end staining. Furthermore, caspase inhibition provided partial protection from prion-mediated cell death. These results suggest that differentiated neurosphere cultures can provide an in vitro bioassay for mouse prions and permit the study of the molecular basis for prion-induced cytotoxicity at the cellular level.
Collapse
|
104
|
Repair of the injured spinal cord by transplantation of neural stem cells in a hyaluronan-based hydrogel. Biomaterials 2013; 34:3775-83. [DOI: 10.1016/j.biomaterials.2013.02.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 02/01/2013] [Indexed: 12/29/2022]
|
105
|
Bugiani M, Postma N, Polder E, Dieleman N, Scheffer PG, Sim FJ, van der Knaap MS, Boor I. Hyaluronan accumulation and arrested oligodendrocyte progenitor maturation in vanishing white matter disease. ACTA ACUST UNITED AC 2013; 136:209-22. [PMID: 23365098 DOI: 10.1093/brain/aws320] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Vanishing white matter disease is a genetic leukoencephalopathy caused by mutations in eukaryotic translation initiation factor 2B. Patients experience a slowly progressive neurological deterioration with episodes of rapid clinical worsening triggered by stress. The disease may occur at any age and leads to early death. Characteristic neuropathological findings include cystic degeneration of the white matter with feeble, if any, reactive gliosis, dysmorphic astrocytes and paucity of myelin despite an increase in oligodendrocytic density. These features have been linked to a maturation defect of astrocytes and oligodendrocytes. However, the nature of the link between glial immaturity and the observed neuropathological features is unclear. We hypothesized that the defects in maturation and function of astrocytes and oligodendrocytes are related. Brain tissue of seven patients with genetically proven vanishing white matter disease was investigated using immunohistochemistry, western blotting, quantitative polymerase chain reaction and size exclusion chromatography. The results were compared with those obtained from normal brain tissue of age-matched controls, from chronic demyelinated multiple sclerosis lesions and from other genetic and acquired white matter disorders. We found that the white matter of patients with vanishing white matter disease is enriched in CD44-expressing astrocyte precursor cells and accumulates the glycosaminoglycan hyaluronan. Hyaluronan is a major component of the extracellular matrix, and CD44 is a hyaluronan receptor. We found that a high molecular weight form of hyaluronan is overabundant, especially in the most severely affected areas. Comparison between the more severely affected frontal white matter and the relatively spared cerebellum confirms that high molecular weight hyaluronan accumulation is more pronounced in the frontal white matter than in the cerebellum. High molecular weight hyaluronan is known to inhibit astrocyte and oligodendrocyte precursor maturation and can explain the arrested glial progenitor maturation observed in vanishing white matter disease. In conclusion, high molecular weight species of hyaluronan accumulate in the white matter of patients with vanishing white matter disease, and by inhibiting glial maturation and proper function, they may be a major determinant of the white matter pathology and lack of repair.
Collapse
Affiliation(s)
- Marianna Bugiani
- Department of Paediatrics/Child Neurology, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Majumder A, Dhara SK, Swetenburg R, Mithani M, Cao K, Medrzycki M, Fan Y, Stice SL. Inhibition of DNA methyltransferases and histone deacetylases induces astrocytic differentiation of neural progenitors. Stem Cell Res 2013; 11:574-86. [PMID: 23644509 DOI: 10.1016/j.scr.2013.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/06/2013] [Accepted: 03/25/2013] [Indexed: 11/25/2022] Open
Abstract
Understanding how to specify rapid differentiation of human neural progenitor towards enriched non-transformed human astrocyte progenitors will provide a critical cell source to further our understanding of how astrocytes play a pivotal role in neural function and development. Human neural progenitors derived from pluripotent embryonic stem cells and propagated in adherent serum-free cultures provide a fate restricted renewable source for quick production of neural cells; however, such cells are highly refractive to astrocytogenesis and show a strong neurogenic bias, similar to neural progenitors from the early embryonic central nervous system (CNS). We found that several astrocytic genes are hypermethylated in such progenitors potentially preventing generation of astrocytes and leading to the proneuronal fate of these progenitors. However, epigenetic modification by Azacytidine (Aza-C) and Trichostatin A (TSA), with concomitant signaling from BMP2 and LIF in neural progenitor cultures shifts this bias, leading to expression of astrocytic markers as early as 5days of differentiation, with near complete suppression of neuronal differentiation. The resultant cells express major astrocytic markers, are amenable to co-culture with neurons, can be propagated as astrocyte progenitors and are cryopreservable. Although previous reports have generated astrocytes from pluripotent cells, the differentiation required extensive culture or selection based on cell surface antigens. The development of a label free and rapid differentiation process will expedite future derivation of astrocytes from various sources pluripotent cells including, but not limited to, human astrocytes associated with various neurological diseases.
Collapse
Affiliation(s)
- Anirban Majumder
- Regenerative Bioscience Center, University of Georgia, 425 River Rd, Athens, GA 30602, USA
| | | | | | | | | | | | | | | |
Collapse
|
107
|
Deboux C, Ladraa S, Cazaubon S, Ghribi-Mallah S, Weiss N, Chaverot N, Couraud PO, Evercooren ABV. Overexpression of CD44 in neural precursor cells improves trans-endothelial migration and facilitates their invasion of perivascular tissues in vivo. PLoS One 2013; 8:e57430. [PMID: 23468987 PMCID: PMC3585392 DOI: 10.1371/journal.pone.0057430] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/22/2013] [Indexed: 02/02/2023] Open
Abstract
Neural precursor (NPC) based therapies are used to restore neurons or oligodendrocytes and/or provide neuroprotection in a large variety of neurological diseases. In multiple sclerosis models, intravenously (i.v) -delivered NPCs reduced clinical signs via immunomodulation. We demonstrated recently that NPCs were able to cross cerebral endothelial cells in vitro and that the multifunctional signalling molecule, CD44 involved in trans-endothelial migration of lymphocytes to sites of inflammation, plays a crucial role in extravasation of syngeneic NPCs. In view of the role of CD44 in NPCs trans-endothelial migration in vitro, we questioned presently the benefit of CD44 overexpression by NPCs in vitro and in vivo, in EAE mice. We show that overexpression of CD44 by NPCs enhanced over 2 folds their trans-endothelial migration in vitro, without impinging on the proliferation or differentiation potential of the transduced cells. Moreover, CD44 overexpression by NPCs improved significantly their elongation, spreading and number of filopodia over the extracellular matrix protein laminin in vitro. We then tested the effect of CD44 overexpression after i.v. delivery in the tail vein of EAE mice. CD44 overexpression was functional invivo as it accelerated trans-endothelial migration and facilitated invasion of HA expressing perivascular sites. These in vitro and in vivo data suggest that CD44 may be crucial not only for NPC crossing the endothelial layer but also for facilitating invasion of extravascular tissues.
Collapse
Affiliation(s)
- Cyrille Deboux
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Sophia Ladraa
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Sylvie Cazaubon
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Siham Ghribi-Mallah
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Nicolas Weiss
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nathalie Chaverot
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pierre Olivier Couraud
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Baron-Van Evercooren
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
- Assitance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Fédération de Neurologie
- * E-mail:
| |
Collapse
|
108
|
Human neural stem/progenitor cells derived from embryonic stem cells and fetal nervous system present differences in immunogenicity and immunomodulatory potentials in vitro. Stem Cell Res 2013; 10:325-37. [PMID: 23416350 DOI: 10.1016/j.scr.2013.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/18/2012] [Accepted: 01/02/2013] [Indexed: 12/29/2022] Open
Abstract
To develop cell therapies for damaged nervous tissue with human neural stem/progenitor cells (hNPCs), the risk of an immune response and graft rejection must be considered. There are conflicting results and lack of knowledge concerning the immunocompetence of hNPCs of different origin. Here, we studied the immunogenicity and immunomodulatory potentials of hNPCs cultured under equivalent conditions after derivation from human embryonic stem cells (hESC-NPCs) or human fetal spinal cord tissue (hfNPCs). The expression patterns of human leukocyte antigen, co-stimulatory and adhesion molecules in hESC-NPCs and hfNPCs were relatively similar and mostly not affected by inflammatory cytokines. Unstimulated hfNPCs secreted more transforming growth factor-β1 (TGF-β1) and β2 but similar level of interleukin (IL)-10 compared to hESC-NPCs. In contrast to hfNPCs, hESC-NPCs displayed 4-6 fold increases in TGF-β1, TGF-β2 and IL-10 under inflammatory conditions. Both hNPCs reduced the alloreaction between allogeneic peripheral blood mononuclear cells (PBMCs) and up-regulated CD4(+)CD25(+)forkhead box P3 (FOXP3)(+) T cells. However, hESC-NPCs but not hfNPCs dose-dependently triggered PBMC proliferation, which at least partly may be due to TGF-β signaling. To conclude, hESC-NPCs and hfNPCs displayed similarities but also significant differences in their immunocompetence and interaction with allogeneic PBMCs, differences may be crucial for the outcome of cell therapy.
Collapse
|
109
|
Naruse M, Shibasaki K, Yokoyama S, Kurachi M, Ishizaki Y. Dynamic changes of CD44 expression from progenitors to subpopulations of astrocytes and neurons in developing cerebellum. PLoS One 2013; 8:e53109. [PMID: 23308146 PMCID: PMC3537769 DOI: 10.1371/journal.pone.0053109] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/26/2012] [Indexed: 12/19/2022] Open
Abstract
We previously reported that CD44-positive cells were candidates for astrocyte precursor cells in the developing cerebellum, because cells expressing high levels of CD44 selected by fluorescence-activated cell sorting (FACS) gave rise only to astrocytes in vitro. However, whether CD44 is a specific cell marker for cerebellar astrocyte precursor cells in vivo is unknown. In this study, we used immunohistochemistry, in situ hybridization, and FACS to analyze the spatial and temporal expression of CD44 and characterize the CD44-positive cells in the mouse cerebellum during development. CD44 expression was observed not only in astrocyte precursor cells but also in neural stem cells and oligodendrocyte precursor cells (OPCs) at early postnatal stages. CD44 expression in OPCs was shut off during oligodendrocyte differentiation. Interestingly, during development, CD44 expression was limited specifically to Bergmann glia and fibrous astrocytes among three types of astrocytes in cerebellum, and expression in astrocytes was shut off during postnatal development. CD44 expression was also detected in developing Purkinje and granule neurons but was limited to granule neurons in the adult cerebellum. Thus, at early developmental stages of the cerebellum, CD44 was widely expressed in several types of precursor cells, and over the course of development, the expression of CD44 became restricted to granule neurons in the adult.
Collapse
Affiliation(s)
- Masae Naruse
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koji Shibasaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
- * E-mail: (KS); (YS)
| | - Shuichi Yokoyama
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
- * E-mail: (KS); (YS)
| |
Collapse
|
110
|
Abstract
Glioma is a heterogeneous disease process with differential histology and treatment response. It was previously thought that the histological features of glial tumors indicated their cell of origin. However, the discovery of continuous neuro-gliogenesis in the normal adult brain and the identification of brain tumor stem cells within glioma have led to the hypothesis that these brain tumors originate from multipotent neural stem or progenitor cells, which primarily divide asymmetrically during the postnatal period. Asymmetric cell division allows these cell types to concurrently self-renew whilst also producing cells for the differentiation pathway. It has recently been shown that increased symmetrical cell division, favoring the self-renewal pathway, leads to oligodendroglioma formation from oligodendrocyte progenitor cells. In contrast, there is some evidence that asymmetric cell division maintenance in tumor stem-like cells within astrocytoma may lead to acquisition of treatment resistance. Therefore cell division mode in normal brain stem and progenitor cells may play a role in setting tumorigenic potential and the type of tumor formed. Moreover, heterogeneous tumor cell populations and their respective cell division mode may confer differential sensitivity to therapy. This review aims to shed light on the controllers of cell division mode which may be therapeutically targeted to prevent glioma formation and improve treatment response.
Collapse
|
111
|
Piao JH, Wang Y, Duncan ID. CD44 is required for the migration of transplanted oligodendrocyte progenitor cells to focal inflammatory demyelinating lesions in the spinal cord. Glia 2012; 61:361-7. [DOI: 10.1002/glia.22438] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 09/25/2012] [Indexed: 11/06/2022]
|
112
|
A critical cell-intrinsic role for serum response factor in glial specification in the CNS. J Neurosci 2012; 32:8012-23. [PMID: 22674276 DOI: 10.1523/jneurosci.5633-11.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Astrocytes and oligodendrocytes play crucial roles in nearly every facet of nervous system development and function, including neuronal migration, synaptogenesis, synaptic plasticity, and myelination. Previous studies have widely characterized the signaling pathways important for astrocyte differentiation and unveiled a number of transcription factors that guide oligodendrocyte differentiation in the CNS. However, the identities of the transcription factors critical for astrocyte specification in the brain remain unknown. Here we show that deletion of the stimulus-dependent transcription factor, serum response factor (SRF), in neural precursor cells (NPCs) (Srf-Nestin-cKO) results in nearly 60% loss in astrocytes and 50% loss in oligodendrocyte precursors at birth. Cultured SRF-deficient NPCs exhibited normal growth rate and capacity to self-renew. However, SRF-deficient NPCs generated fewer astrocytes and oligodendrocytes in response to several lineage-specific differentiation factors. These deficits in glial differentiation were rescued by ectopic expression of wild-type SRF in SRF-deficient NPCs. Interestingly, ectopic expression of a constitutively active SRF (SRF-VP16) in NPCs augmented astrocyte differentiation in the presence of pro-astrocytic factors. However, SRF-VP16 expression in NPCs had an inhibitory effect on oligodendrocyte differentiation. In contrast, mice carrying conditional deletion of SRF in developing forebrain neurons (Srf-NEX-cKO) did not exhibit any deficits in astrocytes in the brain. Together, our observations suggest that SRF plays a critical cell-autonomous role in NPCs to regulate astrocyte and oligodendrocyte specification in vivo and in vitro.
Collapse
|
113
|
Katz AM, Amankulor NM, Pitter K, Helmy K, Squatrito M, Holland EC. Astrocyte-specific expression patterns associated with the PDGF-induced glioma microenvironment. PLoS One 2012; 7:e32453. [PMID: 22393407 PMCID: PMC3290579 DOI: 10.1371/journal.pone.0032453] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 01/31/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The tumor microenvironment contains normal, non-neoplastic cells that may contribute to tumor growth and maintenance. Within PDGF-driven murine gliomas, tumor-associated astrocytes (TAAs) are a large component of the tumor microenvironment. The function of non-neoplastic astrocytes in the glioma microenvironment has not been fully elucidated; moreover, the differences between these astrocytes and normal astrocytes are unknown. We therefore sought to identify genes and pathways that are increased in TAAs relative to normal astrocytes and also to determine whether expression of these genes correlates with glioma behavior. METHODOLOGY/PRINCIPAL FINDINGS We compared the gene expression profiles of TAAs to normal astrocytes and found the Antigen Presentation Pathway to be significantly increased in TAAs. We then identified a gene signature for glioblastoma (GBM) TAAs and validated the expression of some of those genes within the tumor. We also show that TAAs are derived from the non-tumor, stromal environment, in contrast to the Olig2+ tumor cells that constitute the neoplastic elements in our model. Finally, we validate this GBM TAA signature in patients and show that a TAA-derived gene signature predicts survival specifically in the human proneural subtype of glioma. CONCLUSIONS/SIGNIFICANCE Our data identifies unique gene expression patterns between populations of TAAs and suggests potential roles for stromal astrocytes within the glioma microenvironment. We show that certain stromal astrocytes in the tumor microenvironment express a GBM-specific gene signature and that the majority of these stromal astrocyte genes can predict survival in the human disease.
Collapse
Affiliation(s)
- Amanda M. Katz
- Biochemistry, Cell, and Molecular Biology Program, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Nduka M. Amankulor
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Departments of Neurosurgery, Neurology and Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ken Pitter
- Biochemistry, Cell, and Molecular Biology Program, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Karim Helmy
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Massimo Squatrito
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Eric C. Holland
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Brain Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Departments of Neurosurgery, Neurology and Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
114
|
Haas C, Neuhuber B, Yamagami T, Rao M, Fischer I. Phenotypic analysis of astrocytes derived from glial restricted precursors and their impact on axon regeneration. Exp Neurol 2012; 233:717-32. [PMID: 22101004 PMCID: PMC3272137 DOI: 10.1016/j.expneurol.2011.11.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/12/2011] [Accepted: 11/01/2011] [Indexed: 12/16/2022]
Abstract
Although astrocytes are involved in the production of an inhibitory glial scar following injury, they are also capable of providing neuroprotection and supporting axonal growth. There is growing appreciation for a diverse and dynamic population of astrocytes, specified by a variety of glial precursors, whose function is regulated regionally and temporally. Consequently, the therapeutic application of glial precursors and astrocytes by effective transplantation protocols requires a better understanding of their phenotypic and functional properties and effective protocols for their preparation. We present a systematic analysis of astrocyte differentiation using multiple preparations of glial-restricted precursors (GRP), evaluating their morphological and phenotypic properties following treatment with fetal bovine serum (FBS), bone morphogenetic protein 4 (BMP-4), or ciliary neurotrophic factor (CNTF) in comparison to controls treated with basic fibroblast growth factor (bFGF), which maintains undifferentiated GRP. We found that treatments with FBS or BMP-4 generated similar profiles of highly differentiated astrocytes that were A2B5-/GFAP+. Treatment with FBS generated the most mature astrocytes, with a distinct and near-homogeneous morphology of fibroblast-like flat cells, whereas BMP-4 derived astrocytes had a stellate, but heterogeneous morphology. Treatment with CNTF induced differentiation of GRP to an intermediate state of GFAP+cells that maintained immature markers and had relatively long processes. Furthermore, astrocytes generated by BMP-4 or CNTF showed considerable experimental plasticity, and their morphology and phenotypes could be reversed with complementary treatments along a wide range of mature-immature states. Importantly, when GRP or GRP treated with BMP-4 or CNTF were transplanted acutely into a dorsal column lesion of the spinal cord, cells from all 3 groups survived and generated permissive astrocytes that supported axon growth and regeneration of host sensory axons into, but not out of the lesion. Our study underscores the dynamic nature of astrocytes prepared from GRP and their permissive properties, and suggest that future therapeutic applications in restoring connectivity following CNS injury are likely to require a combination of treatments.
Collapse
Affiliation(s)
| | | | - Takaya Yamagami
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, Life Technologies, Frederick, MD
| | | | - Itzhak Fischer
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, Life Technologies, Frederick, MD
| |
Collapse
|
115
|
Abstract
Cells with certain attributes of very immature astroglial cells and their radial precursors can act as stem and/or progenitor cells during developmental and persistent neurogenesis. Neural stem/progenitor cells both express and are affected by a variety of developmentally regulated macromolecules and growth factors, and such signaling or recognition molecules are being uncovered through extensive genomic and proteomic studies, as well as tested using in vitro/in vivo cell growth bioassays. Glycosylated molecules are appreciated as distinct signaling molecules during morphogenesis in a variety of tissues and organs, with glycoconjugates (glycoproteins, glycolipids, and glycosaminoglycans) serving as mediators for the interactions of cells with each other and their substrates, to confer growth and differentiation cues to precursor cells in search of identity. Neurogenic astrocytes and associated glycoconjugates, especially extracellular matrix molecules, are discussed in the context of neurogenesis and stem/progenitor cell growth, fate choice, and differentiation.
Collapse
Affiliation(s)
- Dennis A Steindler
- Department of Neuroscience, The Evelyn F. and William L. McKnight Brain Institute, The University of Florida, Gainesville, FL, USA.
| |
Collapse
|
116
|
Taverna E, Haffner C, Pepperkok R, Huttner WB. A new approach to manipulate the fate of single neural stem cells in tissue. Nat Neurosci 2011; 15:329-37. [PMID: 22179113 DOI: 10.1038/nn.3008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 11/14/2011] [Indexed: 12/19/2022]
Abstract
A challenge in the field of neural stem cell biology is the mechanistic dissection of single stem cell behavior in tissue. Although such behavior can be tracked by sophisticated imaging techniques, current methods of genetic manipulation do not allow researchers to change the level of a defined gene product on a truly acute time scale and are limited to very few genes at a time. To overcome these limitations, we established microinjection of neuroepithelial/radial glial cells (apical progenitors) in organotypic slice culture of embryonic mouse brain. Microinjected apical progenitors showed cell cycle parameters that were indistinguishable to apical progenitors in utero, underwent self-renewing divisions and generated neurons. Microinjection of single genes, recombinant proteins or complex mixtures of RNA was found to elicit acute and defined changes in apical progenitor behavior and progeny fate. Thus, apical progenitor microinjection provides a new approach to acutely manipulating single neural stem and progenitor cells in tissue.
Collapse
Affiliation(s)
- Elena Taverna
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | | |
Collapse
|
117
|
Directed differentiation of functional astroglial subtypes from human pluripotent stem cells. Nat Protoc 2011; 6:1710-7. [PMID: 22011653 DOI: 10.1038/nprot.2011.405] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Regionally and functionally diverse types of astrocytes exist throughout the central nervous system and participate in nearly every aspect of normal and abnormal neural function. Therefore, human astrocyte subtypes are useful tools for understanding brain function, modulating disease processes and promoting neural regeneration. Here we describe a protocol for directed differentiation and maintenance of functional astroglia from human pluripotent stem cells in a chemically defined system. Human stem cells are first differentiated into neuroepithelial cells with or without exogenous patterning molecules (days 0-21). Regular dissociation of the neuroepithelial clusters in suspension, and in the presence of mitogens, permits generation of astroglial subtypes over a long-term expansion (days 21-90). Finally, the astroglial progenitors are either amplified for an extended time or differentiated into functional astrocytes on removal of mitogens and the addition of ciliary neurotrophic factor (days >90). This method generates robust populations of functionally diversified astrocytes with high efficiency.
Collapse
|
118
|
Astrocytes carrying the superoxide dismutase 1 (SOD1G93A) mutation induce wild-type motor neuron degeneration in vivo. Proc Natl Acad Sci U S A 2011; 108:17803-8. [PMID: 21969586 DOI: 10.1073/pnas.1103141108] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recent studies highlight astrocytes as key drivers of motor neuron (MN) degeneration and disease propagation in mutant human superoxide dismutase 1 (mSOD1)-mediated amyotrophic lateral sclerosis. However, in vivo analysis of specific astrocytic influence in amyotrophic lateral sclerosis has proven difficult because mSOD1 is ubiquitously expressed throughout the CNS of rodent models studied. Here, we transplanted SOD1(G93A) glial-restricted precursor cells--glial progenitors capable of differentiating into astrocytes--into the cervical spinal cord of WT rats to reveal how mutant astrocytes influence WT MNs and other cells types (microglia and astrocytes) in an in vivo setting. Transplanted SOD1(G93A) glial-restricted precursor cells survived and differentiated efficiently into astrocytes. Graft-derived SOD1(G93A) astrocytes induced host MN ubiquitination and death, forelimb motor and respiratory dysfunction, reactive astrocytosis, and reduced GLT-1 transporter expression in WT animals. The SOD1(G93A) astrocyte-induced MN death seemed in part mediated by host microglial activation. These findings show that mSOD1 astrocytes alone can induce WT MN death and associated pathological changes in vivo.
Collapse
|
119
|
Macarthur CC, Xue H, Van Hoof D, Lieu PT, Dudas M, Fontes A, Swistowski A, Touboul T, Seerke R, Laurent LC, Loring JF, German MS, Zeng X, Rao MS, Lakshmipathy U, Chesnut JD, Liu Y. Chromatin insulator elements block transgene silencing in engineered human embryonic stem cell lines at a defined chromosome 13 locus. Stem Cells Dev 2011; 21:191-205. [PMID: 21699412 DOI: 10.1089/scd.2011.0163] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lineage reporters of human embryonic stem cell (hESC) lines are useful for differentiation studies and drug screening. Previously, we created reporter lines driven by an elongation factor 1 alpha (EF1α) promoter at a chromosome 13q32.3 locus in the hESC line WA09 and an abnormal hESC line BG01V in a site-specific manner. Expression of reporters in these lines was maintained in long-term culture at undifferentiated state. However, when these cells were differentiated into specific lineages, reduction in reporter expression was observed, indicating transgene silencing. To develop an efficient and reliable genetic engineering strategy in hESCs, we used chromatin insulator elements to flank single-copy transgenes and integrated the combined expression constructs via PhiC31/R4 integrase-mediated recombination technology to the chromosome 13 locus precisely. Two copies of cHS4 double-insulator sequences were placed adjacent to both 5' and 3' of the promoter reporter constructs. The green fluorescent protein (GFP) gene was driven by EF1α or CMV early enhancer/chicken β actin (CAG) promoter. In the engineered hESC lines, for both insulated CAG-GFP and EF1α-GFP, constitutive expression at the chromosome 13 locus was maintained during prolonged culture and in directed differentiation assays toward diverse types of neurons, pancreatic endoderm, and mesodermal progeny. In particular, described here is the first normal hESC fluorescent reporter line that robustly expresses GFP in both the undifferentiated state and throughout dopaminergic lineage differentiation. The dual strategy of utilizing insulator sequences and integration at the constitutive chromosome 13 locus ensures appropriate transgene expression. This is a valuable tool for lineage development study, gain- and loss-of-function experiments, and human disease modeling using hESCs.
Collapse
Affiliation(s)
- Chad C Macarthur
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Cai N, Kurachi M, Shibasaki K, Okano-Uchida T, Ishizaki Y. CD44-Positive Cells Are Candidates for Astrocyte Precursor Cells in Developing Mouse Cerebellum. THE CEREBELLUM 2011; 11:181-93. [DOI: 10.1007/s12311-011-0294-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
121
|
Krencik R, Weick JP, Liu Y, Zhang Z, Zhang SC. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat Biotechnol 2011; 29:528-34. [PMID: 21602806 PMCID: PMC3111840 DOI: 10.1038/nbt.1877] [Citation(s) in RCA: 314] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 04/20/2011] [Indexed: 12/23/2022]
Abstract
Human pluripotent stem cells (hPSCs) have been differentiated efficiently to neuronal cell types. However, directed differentiation of hPSCs to astrocytes and astroglial subtypes remains elusive. In this study, hPSCs were directed to nearly uniform populations of immature astrocytes (>90% S100β(+) and GFAP(+)) in large quantities. The immature human astrocytes exhibit similar gene expression patterns as primary astrocytes, display functional properties such as glutamate uptake and promotion of synaptogenesis, and become mature astrocytes by forming connections with blood vessels after transplantation into the mouse brain. Furthermore, hPSC-derived neuroepithelia, patterned to rostral-caudal and dorsal-ventral identities with the same morphogens used for neuronal subtype specification, generate immature astrocytes that express distinct homeodomain transcription factors and display phenotypic differences of different astroglial subtypes. These human astroglial progenitors and immature astrocytes will be useful for studying astrocytes in brain development and function, understanding the roles of astrocytes in disease processes and developing novel treatments for neurological disorders.
Collapse
Affiliation(s)
- Robert Krencik
- Neuroscience Training Program, Fudan University Shanghai Medical School, Shanghai, China
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
| | - Jason P. Weick
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
| | - Yan Liu
- Department of Human Anatomy and Histology, Fudan University Shanghai Medical School, Shanghai, China
| | - Zhijian Zhang
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
| | - Su-Chun Zhang
- Neuroscience Training Program, Fudan University Shanghai Medical School, Shanghai, China
- Department of Human Anatomy and Histology, Fudan University Shanghai Medical School, Shanghai, China
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave., Madison, Wisconsin 53705
| |
Collapse
|
122
|
Yuan SH, Martin J, Elia J, Flippin J, Paramban RI, Hefferan MP, Vidal JG, Mu Y, Killian RL, Israel MA, Emre N, Marsala S, Marsala M, Gage FH, Goldstein LSB, Carson CT. Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS One 2011; 6:e17540. [PMID: 21407814 PMCID: PMC3047583 DOI: 10.1371/journal.pone.0017540] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 02/08/2011] [Indexed: 12/22/2022] Open
Abstract
Background Neural induction of human pluripotent stem cells often yields heterogeneous cell populations that can hamper quantitative and comparative analyses. There is a need for improved differentiation and enrichment procedures that generate highly pure populations of neural stem cells (NSC), glia and neurons. One way to address this problem is to identify cell-surface signatures that enable the isolation of these cell types from heterogeneous cell populations by fluorescence activated cell sorting (FACS). Methodology/Principal Findings We performed an unbiased FACS- and image-based immunophenotyping analysis using 190 antibodies to cell surface markers on naïve human embryonic stem cells (hESC) and cell derivatives from neural differentiation cultures. From this analysis we identified prospective cell surface signatures for the isolation of NSC, glia and neurons. We isolated a population of NSC that was CD184+/CD271−/CD44−/CD24+ from neural induction cultures of hESC and human induced pluripotent stem cells (hiPSC). Sorted NSC could be propagated for many passages and could differentiate to mixed cultures of neurons and glia in vitro and in vivo. A population of neurons that was CD184−/CD44−/CD15LOW/CD24+ and a population of glia that was CD184+/CD44+ were subsequently purified from cultures of differentiating NSC. Purified neurons were viable, expressed mature and subtype-specific neuronal markers, and could fire action potentials. Purified glia were mitotic and could mature to GFAP-expressing astrocytes in vitro and in vivo. Conclusions/Significance These findings illustrate the utility of immunophenotyping screens for the identification of cell surface signatures of neural cells derived from human pluripotent stem cells. These signatures can be used for isolating highly pure populations of viable NSC, glia and neurons by FACS. The methods described here will enable downstream studies that require consistent and defined neural cell populations.
Collapse
Affiliation(s)
- Shauna H. Yuan
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jody Martin
- BD Biosciences, La Jolla, California, United States of America
| | - Jeanne Elia
- BD Biosciences, La Jolla, California, United States of America
| | - Jessica Flippin
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | | - Mike P. Hefferan
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Jason G. Vidal
- BD Biosciences, La Jolla, California, United States of America
| | - Yangling Mu
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Rhiannon L. Killian
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Mason A. Israel
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, California, United States of America
| | - Nil Emre
- BD Biosciences, La Jolla, California, United States of America
| | - Silvia Marsala
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - Martin Marsala
- Anesthesiology Research Laboratory, Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- Institute of Neurobiology, Slovak Academy of Sciences, Košice, Slovakia
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Lawrence S. B. Goldstein
- Howard Hughes Medical Institute and Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | | |
Collapse
|
123
|
Thirant C, Bessette B, Varlet P, Puget S, Cadusseau J, Dos Reis Tavares S, Studler JM, Silvestre DC, Susini A, Villa C, Miquel C, Bogeas A, Surena AL, Dias-Morais A, Léonard N, Pflumio F, Bièche I, Boussin FD, Sainte-Rose C, Grill J, Daumas-Duport C, Chneiweiss H, Junier MP. Clinical relevance of tumor cells with stem-like properties in pediatric brain tumors. PLoS One 2011; 6:e16375. [PMID: 21297991 PMCID: PMC3030582 DOI: 10.1371/journal.pone.0016375] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 12/19/2010] [Indexed: 11/19/2022] Open
Abstract
Background Primitive brain tumors are the leading cause of cancer-related death in children. Tumor cells with stem-like properties (TSCs), thought to account for tumorigenesis and therapeutic resistance, have been isolated from high-grade gliomas in adults. Whether TSCs are a common component of pediatric brain tumors and are of clinical relevance remains to be determined. Methodology/Principal Findings Tumor cells with self-renewal properties were isolated with cell biology techniques from a majority of 55 pediatric brain tumors samples, regardless of their histopathologies and grades of malignancy (57% of embryonal tumors, 57% of low-grade gliomas and neuro-glial tumors, 70% of ependymomas, 91% of high-grade gliomas). Most high-grade glioma-derived oncospheres (10/12) sustained long-term self-renewal akin to neural stem cells (>7 self-renewals), whereas cells with limited renewing abilities akin to neural progenitors dominated in all other tumors. Regardless of tumor entities, the young age group was associated with self-renewal properties akin to neural stem cells (P = 0.05, chi-square test). Survival analysis of the cohort showed an association between isolation of cells with long-term self-renewal abilities and a higher patient mortality rate (P = 0.013, log-rank test). Sampling of low- and high-grade glioma cultures showed that self-renewing cells forming oncospheres shared a molecular profile comprising embryonic and neural stem cell markers. Further characterization performed on subsets of high-grade gliomas and one low-grade glioma culture showed combination of this profile with mesenchymal markers, the radio-chemoresistance of the cells and the formation of aggressive tumors after intracerebral grafting. Conclusions/Significance In brain tumors affecting adult patients, TSCs have been isolated only from high-grade gliomas. In contrast, our data show that tumor cells with stem cell-like or progenitor-like properties can be isolated from a wide range of histological sub-types and grades of pediatric brain tumors. They suggest that cellular mechanisms fueling tumor development differ between adult and pediatric brain tumors.
Collapse
Affiliation(s)
- Cécile Thirant
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Barbara Bessette
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Pascale Varlet
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
| | - Stéphanie Puget
- Pediatric Neurosurgical Department. Hospital Necker, University Paris Descartes, Paris, France
- CNRS UMR 8203, Vectorology and Anticancer Therapeutics, Gustave Roussy Cancer Institute, Villejuif, France
| | | | | | - Jeanne-Marie Studler
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Collège de France, Paris, France
| | - David Carlos Silvestre
- Laboratoire de Radiopathologie UMR 967, CEA-INSERM-Université Paris VII, Fontenay-aux-Roses, France
| | - Aurélie Susini
- Laboratoire d'Oncogénétique - INSERM U735, Institut Curie/Hôpital René Huguenin, St-Cloud, France
| | - Chiara Villa
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
| | - Catherine Miquel
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
| | - Alexandra Bogeas
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Anne-Laure Surena
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Amélia Dias-Morais
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Nadine Léonard
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
| | - Françoise Pflumio
- Laboratoire des Cellules Souches Hématopoïétiques et Leucémiques, UMR U967, CEA-INSERM-Université Paris VII, Fontenay-aux-Roses, France
| | - Ivan Bièche
- Laboratoire d'Oncogénétique - INSERM U735, Institut Curie/Hôpital René Huguenin, St-Cloud, France
| | - François D. Boussin
- Laboratoire de Radiopathologie UMR 967, CEA-INSERM-Université Paris VII, Fontenay-aux-Roses, France
| | - Christian Sainte-Rose
- Pediatric Neurosurgical Department. Hospital Necker, University Paris Descartes, Paris, France
| | - Jacques Grill
- CNRS UMR 8203, Vectorology and Anticancer Therapeutics, Gustave Roussy Cancer Institute, Villejuif, France
| | - Catherine Daumas-Duport
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
| | - Hervé Chneiweiss
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
| | - Marie-Pierre Junier
- Inserm, UMR894, Team Glial Plasticity, University Paris Descartes, Paris, France
- Department of Neuropathology, Hospital Sainte-Anne, Paris, France
- * E-mail:
| |
Collapse
|
124
|
Shinoe T, Kuribayashi H, Saya H, Seiki M, Aburatani H, Watanabe S. Identification of CD44 as a cell surface marker for Müller glia precursor cells. J Neurochem 2010; 115:1633-42. [PMID: 20969572 DOI: 10.1111/j.1471-4159.2010.07072.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the retina, both neurons and glia differentiate from a common progenitor population. CD44 cell surface antigen is a hyaluronic acid receptor expressed on mature Müller glial cells. We found that in the developing mouse retina, expression of CD44 was transiently observed at or around birth in a subpopulation of c-kit-positive retinal progenitor cells. During in vitro culture, purified CD44/c-kit-positive retinal progenitor cells exclusively differentiated into Müller glial cells and not into neurons, suggesting that CD44 marks a subpopulation of retinal progenitor cells that are fated to become glia. Over-expression of CD44 inhibited the extension of processes by Müller glial cells and neurons. Notch signaling is known to be involved in the specification of retinal progenitors into a glial fate. Activation of Notch signaling increased the number of CD44-positive cells, and treatment with the Notch signal inhibitor, DAPT, at early, but not later, stages of retinal development abolished both CD44-positive cells and Müller glial cells. Together, CD44 was identified as an early cell surface marker of the Müller glia lineage, and Notch signalling was involved in commitment of retinal progenitor cells to CD44 positive Müller glial precursor cells.
Collapse
Affiliation(s)
- Toru Shinoe
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
125
|
Fan X, Liu S, Su F, Pan Q, Lin T. Effective enrichment of prostate cancer stem cells from spheres in a suspension culture system. Urol Oncol 2010; 30:314-8. [PMID: 20843707 DOI: 10.1016/j.urolonc.2010.03.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 03/15/2010] [Accepted: 03/26/2010] [Indexed: 01/24/2023]
Abstract
BACKGROUND Stem-like prostate cancer cells are also called prostate cancer stem cells (PrCSCs). These rare cells are supposed to be highly tumorigenic and to be involved in maintenance of tumor homeostasis and mediation of tumor metastasis. Methods for sorting PrCSCs are mainly based on sorting cells with the marker (CD133(+)/CD44(+)) or side population cells. However, CD133(+)/CD44(+) cells or side population cells are very rare or even undetectable. The scarcity of approaches for isolation and purification of PrCSCs is the main obstacle to studying PrCSCs. METHODS In the present study, suspension culture was used for enrichment of PrCSCs. And PrCSCs were verified by side population technology, drug sensitivity assays, and the molecular marker analysis of prostate cancer stem cell. RESULTS PC3 cells survived and formed spheres in nonadherent suspension culture. The percentage of CD44(+)/CD133(+) cells was 18-fold higher in the nonadherent sphere-forming cell population than in the adherent PC3 cell population (13.94% vs. 0.77%, respectively). This side population was increased to 3.1% in the nonadherent population but undetectable in adherent population. Resistance to cisplatin was higher in the nonadherent cells than adherent cells. CONCLUSION Suspension culture can be used to enrich for PrCSCs. This approach will aid prostate stem cell biology research and facilitate identification of novel therapeutic agents for prostate cancer.
Collapse
Affiliation(s)
- Xinlan Fan
- Center of Medical Research, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | |
Collapse
|
126
|
Seidlits SK, Khaing ZZ, Petersen RR, Nickels JD, Vanscoy JE, Shear JB, Schmidt CE. The effects of hyaluronic acid hydrogels with tunable mechanical properties on neural progenitor cell differentiation. Biomaterials 2010; 31:3930-40. [PMID: 20171731 DOI: 10.1016/j.biomaterials.2010.01.125] [Citation(s) in RCA: 373] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 01/20/2010] [Indexed: 11/19/2022]
Abstract
We report the ability to direct the differentiation pathway of neural progenitor cells (NPCs) within hydrogels having tunable mechanical properties. By modifying the polymeric sugar hyaluronic acid (HA), a major extracellular matrix component in the fetal mammalian brain, with varying numbers of photocrosslinkable methacrylate groups, hydrogels could be prepared with bulk compressive moduli spanning the threefold range measured for neonatal brain and adult spinal cord. Ventral midbrain-derived NPCs were photoencapsulated into HA hydrogels and remained viable after encapsulation. After three weeks, the majority of NPCs cultured in hydrogels with mechanical properties comparable to those of neonatal brain had differentiated into neurons (ss-III tubulin-positive), many of which had extended long, branched processes, indicative of a relatively mature phenotype. In contrast, NPCs within stiffer hydrogels, with mechanical properties comparable to those of adult brain, had differentiated into mostly astrocytes (glial fibrillary acidic protein (GFAP)-positive). Primary spinal astrocytes cultured in the hydrogel variants for two weeks acquired a spread and elongated morphology only in the stiffest hydrogels evaluated, with mechanical properties similar to adult tissue. Results demonstrate that the mechanical properties of these scaffolds can assert a defining influence on the differentiation of ventral midbrain-derived NPCs, which have strong clinical relevance because of their ability to mature into dopaminergic neurons of the substantia nigra, cells that idiopathically degenerate in individuals suffering from Parkinson's disease.
Collapse
Affiliation(s)
- Stephanie K Seidlits
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | | | | | | | | |
Collapse
|
127
|
Pusch A, Boeckenhoff A, Glaser T, Kaminski T, Kirfel G, Hans M, Steinfarz B, Swandulla D, Kubitscheck U, Gieselmann V, Brüstle O, Kappler J. CD44 and hyaluronan promote invasive growth of B35 neuroblastoma cells into the brain. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:261-74. [DOI: 10.1016/j.bbamcr.2009.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Revised: 11/10/2009] [Accepted: 12/16/2009] [Indexed: 11/29/2022]
|
128
|
Liu Y, Thyagarajan B, Lakshmipathy U, Xue H, Lieu P, Fontes A, MacArthur CC, Scheyhing K, Rao MS, Chesnut JD. Generation of Platform Human Embryonic Stem Cell Lines That Allow Efficient Targeting at a Predetermined Genomic Location. Stem Cells Dev 2009; 18:1459-72. [DOI: 10.1089/scd.2009.0047] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ying Liu
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Bhaskar Thyagarajan
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Uma Lakshmipathy
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Haipeng Xue
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Pauline Lieu
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Andrew Fontes
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Chad C. MacArthur
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Kelly Scheyhing
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Mahendra S. Rao
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| | - Jonathan D. Chesnut
- Primary and Stem Cell Systems, Life Technologies Corporation, Carlsbad, California
| |
Collapse
|
129
|
Xue H, Wu S, Papadeas ST, Spusta S, Swistowska AM, MacArthur CC, Mattson MP, Maragakis NJ, Capecchi MR, Rao MS, Zeng X, Liu Y. A targeted neuroglial reporter line generated by homologous recombination in human embryonic stem cells. Stem Cells 2009; 27:1836-46. [PMID: 19544414 PMCID: PMC2741170 DOI: 10.1002/stem.129] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this study, we targeted Olig2, a basic helix-loop-helix transcription factor that plays an important role in motoneuron and oligodendrocyte development, in human embryonic stem cell (hESC) line BG01 by homologous recombination. One allele of Olig2 locus was replaced by a green fluorescent protein (GFP) cassette with a targeting efficiency of 5.7%. Targeted clone R-Olig2 (like the other clones) retained pluripotency, typical hESC morphology, and a normal parental karyotype 46,XY. Most importantly, GFP expression recapitulated endogenous Olig2 expression when R-Olig2 was induced by sonic hedgehog and retinoic acid, and GFP-positive cells could be purified by fluorescence-activated cell sorting. Consistent with previous reports on rodents, early GFP-expressing cells appeared biased to a neuronal fate, whereas late GFP-expressing cells appeared biased to an oligodendrocytic fate. This was corroborated by myoblast coculture, transplantation into the rat spinal cords, and whole genome expression profiling. The present work reports an hESC reporter line generated by homologous recombination targeting a neural lineage-specific gene, which can be differentiated and sorted to obtain pure neural progenitor populations.
Collapse
Affiliation(s)
- Haipeng Xue
- Primary and Stem Cell Systems, Life Technologies Corporation, 5781 Van Allen Way, Carlsbad, California 92008, USA
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Suite 100, Room 05C12, 251 Bayview Blvd. Baltimore, Maryland 21224, USA
| | - Sen Wu
- Department of Human Genetics, University of Utah, 15 N 2030 E, Rm. 5440, Salt Lake City, Utah 84112, USA
- Howard Hughes Medical Institute, University of Utah, 15 N 2030 E, Rm. 5440, Salt Lake City, Utah 84112, USA
| | - Sophia T. Papadeas
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Meyer 6-119, Baltimore, Maryland 21287, USA
| | - Steve Spusta
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, California 94945, USA
| | | | - Chad C. MacArthur
- Primary and Stem Cell Systems, Life Technologies Corporation, 5781 Van Allen Way, Carlsbad, California 92008, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Suite 100, Room 05C12, 251 Bayview Blvd. Baltimore, Maryland 21224, USA
| | - Nicholas J. Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Meyer 6-119, Baltimore, Maryland 21287, USA
| | - Mario R. Capecchi
- Department of Human Genetics, University of Utah, 15 N 2030 E, Rm. 5440, Salt Lake City, Utah 84112, USA
- Howard Hughes Medical Institute, University of Utah, 15 N 2030 E, Rm. 5440, Salt Lake City, Utah 84112, USA
| | - Mahendra S. Rao
- Primary and Stem Cell Systems, Life Technologies Corporation, 5781 Van Allen Way, Carlsbad, California 92008, USA
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Suite 100, Room 05C12, 251 Bayview Blvd. Baltimore, Maryland 21224, USA
| | - Xianmin Zeng
- Buck Institute for Age Research, 8001 Redwood Blvd, Novato, California 94945, USA
| | - Ying Liu
- Primary and Stem Cell Systems, Life Technologies Corporation, 5781 Van Allen Way, Carlsbad, California 92008, USA
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Suite 100, Room 05C12, 251 Bayview Blvd. Baltimore, Maryland 21224, USA
| |
Collapse
|
130
|
Wilczynska KM, Singh SK, Adams B, Bryan L, Rao RR, Valerie K, Wright S, Griswold-Prenner I, Kordula T. Nuclear factor I isoforms regulate gene expression during the differentiation of human neural progenitors to astrocytes. Stem Cells 2009; 27:1173-81. [PMID: 19418463 DOI: 10.1002/stem.35] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Even though astrocytes are critical for both normal brain functions and the development and progression of neuropathological states, including neuroinflammation associated with neurodegenerative diseases, the mechanisms controlling gene expression during astrocyte differentiation are poorly understood. Thus far, several signaling pathways were shown to regulate astrocyte differentiation, including JAK-STAT, bone morphogenic protein-2/Smads, and Notch. More recently, a family of nuclear factor-1 (NFI-A, -B, -C, and -X) was implicated in the regulation of vertebral neocortex development, with NFI-A and -B controlling the onset of gliogenesis. Here, we developed an in vitro model of differentiation of stem cells towards neural progenitors (NP) and subsequently astrocytes. The transition from stem cells to progenitors was accompanied by an expected change in the expression profile of markers, including Sox-2, Musashi-1, and Oct4. Subsequently, generated astrocytes were characterized by proper morphology, increased glutamate uptake, and marker gene expression. We used this in vitro differentiation model to study the expression and functions of NFIs. Interestingly, stem cells expressed only background levels of NFIs, while differentiation to NP activated the expression of NFI-A. More importantly, NFI-X expression was induced during the later stages of differentiation towards astrocytes. In addition, NFI-X and -C were required for the expression of glial fibrillary acidic protein and secreted protein acidic and rich in cystein-like protein 1, which are the markers of astrocytes at the later stages of differentiation. We conclude that an expression program of NFIs is executed during the differentiation of astrocytes, with NFI-X and -C controlling the expression of astrocytic markers at late stages of differentiation.
Collapse
Affiliation(s)
- Katarzyna M Wilczynska
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Abstract
Gliogenesis in the mammalian CNS continues after birth, with astrocytes being generated well into the first two postnatal weeks. In this study, we have isolated an A2B5(+) astrocyte precursor (APC) from the postnatal rat forebrain, which is capable of differentiating into mature astrocytes in serum-free medium without further trophic support. Exposure to basic fibroblast growth factor (bFGF) selectively induces the APCs to proliferate, forming clusters of vimentin(+) cells, which, within 2 weeks, differentiate into GFAP(+) astrocytes. While bFGF functions as a potent mitogen, neither is it necessary to induce or maintain astrocyte differentiation, nor is it capable of maintaining the precursors in an immature, proliferative state. APCs exit the cell cycle and differentiate, even in the continued presence of fibroblast growth factor alone or in combination with other mitogenic factors such as platelet-derived growth factor. Under the culture conditions used, it was not possible to cause the astrocytes to re-enter cell cycle. After transplantation into the neonatal forebrain, APCs differentiated exclusively into astrocytes, regardless of brain region. Initially distributed widely within the forebrain, the precursors are most greatly concentrated within the subventricular zone (SVZ) and subcortical white matter, where they are maintained throughout postnatal development. APCs can be isolated from the SVZ and white matter of animals as late as 4 weeks after birth.
Collapse
Affiliation(s)
- Grace Lin
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | | |
Collapse
|
132
|
Shin S, Vemuri M. Culture and Differentiation of Human Neural Stem Cells. SPRINGER PROTOCOLS HANDBOOKS 2009. [DOI: 10.1007/978-1-60761-292-6_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
133
|
Harris MA, Yang H, Low BE, Mukherje J, Guha A, Bronson RT, Shultz LD, Israel MA, Yun K. Cancer stem cells are enriched in the side population cells in a mouse model of glioma. Cancer Res 2008; 68:10051-9. [PMID: 19074870 PMCID: PMC2841432 DOI: 10.1158/0008-5472.can-08-0786] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The recent identification of cancer stem cells (CSCs) in multiple human cancers provides a new inroad to understanding tumorigenesis at the cellular level. CSCs are defined by their characteristics of self-renewal, multipotentiality, and tumor initiation upon transplantation. By testing for these defining characteristics, we provide evidence for the existence of CSCs in a transgenic mouse model of glioma, S100beta-verbB;Trp53. In this glioma model, CSCs are enriched in the side population (SP) cells. These SP cells have enhanced tumor-initiating capacity, self-renewal, and multipotentiality compared with non-SP cells from the same tumors. Furthermore, gene expression analysis comparing fluorescence-activated cell sorting-sorted cancer SP cells to non-SP cancer cells and normal neural SP cells identified 45 candidate genes that are differentially expressed in glioma stem cells. We validated the expression of two genes from this list (S100a4 and S100a6) in primary mouse gliomas and human glioma samples. Analyses of xenografted human glioblastoma multiforme cell lines and primary human glioma tissues show that S100A4 and S100A6 are expressed in a small subset of cancer cells and that their abundance is positively correlated to tumor grade. In conclusion, this study shows that CSCs exist in a mouse glioma model, suggesting that this model can be used to study the molecular and cellular characteristics of CSCs in vivo and to further test the CSC hypothesis.
Collapse
Affiliation(s)
- Molly A. Harris
- The Jackson Laboratory, Bar Harbor, ME, 04609
- The Graduate School of Biomedical Sciences, University of Maine, Orono, ME 04469
| | - Hyuna Yang
- The Jackson Laboratory, Bar Harbor, ME, 04609
| | | | - Joydeep Mukherje
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Abhijit Guha
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | - Mark A. Israel
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756
| | - Kyuson Yun
- The Jackson Laboratory, Bar Harbor, ME, 04609
| |
Collapse
|
134
|
Canoll P, Goldman JE. The interface between glial progenitors and gliomas. Acta Neuropathol 2008; 116:465-77. [PMID: 18784926 PMCID: PMC2759726 DOI: 10.1007/s00401-008-0432-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Revised: 08/29/2008] [Accepted: 08/29/2008] [Indexed: 01/28/2023]
Abstract
The mammalian brain and spinal cord contain heterogeneous populations of cycling, immature cells. These include cells with stem cell-like properties as well as progenitors in various stages of early glial differentiation. This latter population is distributed widely throughout gray and white matter and numerically represents an extremely large cell pool. In this review, we discuss the possibility that the glial progenitors that populate the adult CNS are one source of gliomas. Indeed, the marker phenotypes, morphologies, and migratory properties of cells in gliomas strongly resemble glial progenitors in many ways. We review briefly some salient features of normal glial development and then examine the similarities and differences between normal progenitors and cells in gliomas, focusing on the phenotypic plasticity of glial progenitors and the responses to growth factors in promoting proliferation and migration of normal and glioma cells, and discussing known mutational changes in gliomas in the context of how these might affect the proliferative and migratory behaviors of progenitors. Finally, we will discuss the "cancer stem cell" hypothesis in light of the possibility that glial progenitors can generate gliomas.
Collapse
Affiliation(s)
- Peter Canoll
- Department of Pathology, Division of Neuropathology, Columbia University, 630 W. 168th St., New York, NY 10032, USA
| | - James E. Goldman
- Department of Pathology, Division of Neuropathology, Columbia University, 630 W. 168th St., New York, NY 10032, USA
| |
Collapse
|
135
|
Campanelli JT, Sandrock RW, Wheatley W, Xue H, Zheng J, Liang F, Chesnut JD, Zhan M, Rao MS, Liu Y. Expression profiling of human glial precursors. BMC DEVELOPMENTAL BIOLOGY 2008; 8:102. [PMID: 18947415 PMCID: PMC2579429 DOI: 10.1186/1471-213x-8-102] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 10/23/2008] [Indexed: 11/10/2022]
Abstract
BACKGROUND We have generated gene expression databases for human glial precursors, neuronal precursors, astrocyte precursors and neural stem cells and focused on comparing the profile of glial precursors with that of other populations. RESULTS A total of 14 samples were analyzed. Each population, previously distinguished from each other by immunocytochemical analysis of cell surface markers, expressed genes related to their key differentiation pathways. For the glial precursor cell population, we identified 458 genes that were uniquely expressed. Expression of a subset of these individual genes was validated by RT-PCR. We also report genes encoding cell surface markers that may be useful for identification and purification of human glial precursor populations. CONCLUSION We provide gene expression profile for human glial precursors. Our data suggest several signaling pathways that are important for proliferation and differentiation of human glial precursors. Such information may be utilized to further purify glial precursor populations, optimize media formulation, or study the effects of glial differentiation.
Collapse
Affiliation(s)
- James T Campanelli
- Q Therapeutics, Inc. 615 Arapeen Dr., Ste. 102, Salt Lake City, UT 84108, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Pollard SM, Wallbank R, Tomlinson S, Grotewold L, Smith A. Fibroblast growth factor induces a neural stem cell phenotype in foetal forebrain progenitors and during embryonic stem cell differentiation. Mol Cell Neurosci 2008; 38:393-403. [PMID: 18504136 DOI: 10.1016/j.mcn.2008.03.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 03/23/2008] [Accepted: 03/28/2008] [Indexed: 01/06/2023] Open
Abstract
Neural stem (NS) cell lines may be derived via differentiation of pluripotent embryonic stem (ES) cells or from foetal forebrain. However, because NS cells arise in vitro from heterogeneous populations their immediate cellular origin remains unclear. We used microarray-based expression profiling to identify a set of markers expressed by mouse NS cells but not ES cells. One differentially expressed gene encodes the cell surface protein, CD44. CD44 expression is activated by FGF-2 in a subset of cells in both differentiating ES cells and foetal forebrain cultures. Following isolation by flow cytometry the CD44+ population was found to be highly enriched for NS cell founders. We found that other NS cell marker genes are also induced by FGF in culture, including: Adam12, Cadherin20, Cx3cl1, EGFR, Frizzled9, Kitl, Olig1, Olig2 and Vav3. We speculate that the self-renewing NS cell state may be generated in vitro following transcriptional resetting induced by FGF.
Collapse
Affiliation(s)
- Steven M Pollard
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QR, UK.
| | | | | | | | | |
Collapse
|
137
|
Li H, Chang YW, Mohan K, Su HW, Ricupero CL, Baridi A, Hart RP, Grumet M. Activated Notch1 maintains the phenotype of radial glial cells and promotes their adhesion to laminin by upregulating nidogen. Glia 2008; 56:646-58. [PMID: 18286610 PMCID: PMC2712347 DOI: 10.1002/glia.20643] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Radial glia are neural stem cells that exist only transiently during central nervous system (CNS) development, where they serve as scaffolds for neuronal migration. Their instability makes them difficult to study, and therefore we have isolated stabilized radial glial clones from E14.5 cortical progenitors (e.g., L2.3) after expression of v-myc. Activated Notch1 intracellular region (actNotch1) promotes radial glia in the embryonic mouse forebrain (Gaiano et al., (2000), and when it was introduced into E14.5 cortical progenitors or radial glial clone L2.3, the cells exhibited enhanced radial morphology and increased expression of the radial glial marker BLBP. A representative clone of L2.3 cells expressing actNotch1 called NL2.3-4 migrated more extensively than L2.3 cells in culture and in white matter of the adult rat spinal cord. Microarray and RT-PCR comparisons of mRNAs expressed in these closely related clones showed extensive similarities, but differed significantly for certain mRNAs including several cell adhesion molecules. Cell adhesion assays demonstrated significantly enhanced adhesion to laminin of NL2.3-4 by comparison to L2.3 cells. The laminin binding protein nidogen was the most highly induced adhesion molecule in NL2.3-4, and immunological analyses indicated that radial glia synthesize and secrete nidogen. Adhesion of NL2.3-4 cells to laminin was inhibited by anti-nidogen antibodies and required the nidogen binding region in laminin, indicating that nidogen promotes cell adhesion to laminin. The combined results indicate that persistent expression of activated Notch1 maintains the phenotype of radial glial cells, inhibits their differentiation, and promotes their adhesion and migration on a laminin/nidogen complex.
Collapse
Affiliation(s)
- Hedong Li
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Yu-Wen Chang
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Kriti Mohan
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Hui-Wen Su
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Christopher L. Ricupero
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Ajoeb Baridi
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
- Department of Pharmacology & Anatomy, Rudolf Magnus Institute of Neuroscience, Utrecht University, Utrecht, The Netherlands
| | - Ronald P. Hart
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| | - Martin Grumet
- Department of Cell Biology and Neuroscience, and W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd, Piscataway, NJ 08854-8082
| |
Collapse
|
138
|
Glial progenitor-like phenotype in low-grade glioma and enhanced CD133-expression and neuronal lineage differentiation potential in high-grade glioma. PLoS One 2008; 3:e1936. [PMID: 18398462 PMCID: PMC2277459 DOI: 10.1371/journal.pone.0001936] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 02/18/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND While neurosphere- as well as xenograft tumor-initiating cells have been identified in gliomas, the resemblance between glioma cells and neural stem/progenitor cells as well as the prognostic value of stem/progenitor cell marker expression in glioma are poorly clarified. METHODOLOGY/PRINCIPAL FINDINGS Viable glioma cells were characterized for surface marker expression along the glial genesis hierarchy. Six low-grade and 17 high-grade glioma specimens were flow-cytometrically analyzed for markers characteristics of stem cells (CD133); glial progenitors (PDGFRalpha, A2B5, O4, and CD44); and late oligodendrocyte progenitors (O1). In parallel, the expression of glial fibrillary acidic protein (GFAP), synaptophysin and neuron-specific enolase (NSE) was immunohistochemically analyzed in fixed tissue specimens. Irrespective of the grade and morphological diagnosis of gliomas, glioma cells concomitantly expressed PDGFRalpha, A2B5, O4, CD44 and GFAP. In contrast, O1 was weakly expressed in all low-grade and the majority of high-grade glioma specimens analyzed. Co-expression of neuronal markers was observed in all high-grade, but not low-grade, glioma specimens analyzed. The rare CD133 expressing cells in low-grade glioma specimens typically co-expressed vessel endothelial marker CD31. In contrast, distinct CD133 expression profiles in up to 90% of CD45-negative glioma cells were observed in 12 of the 17 high-grade glioma specimens and the majority of these CD133 expressing cells were CD31 negative. The CD133 expression correlates inversely with length of patient survival. Surprisingly, cytogenetic analysis showed that gliomas contained normal and abnormal cell karyotypes with hitherto indistinguishable phenotype. CONCLUSIONS/SIGNIFICANCE This study constitutes an important step towards clarification of lineage commitment and differentiation blockage of glioma cells. Our data suggest that glioma cells may resemble expansion of glial lineage progenitor cells with compromised differentiation capacity downstream of A2B5 and O4 expression. The concurrent expression of neuronal markers demonstrates that high-grade glioma cells are endowed with multi-lineage differentiation potential in vivo. Importantly, enhanced CD133 expression marks a poor prognosis in gliomas.
Collapse
|
139
|
Segovia KN, McClure M, Moravec M, Luo NL, Wan Y, Gong X, Riddle A, Craig A, Struve J, Sherman LS, Back SA. Arrested oligodendrocyte lineage maturation in chronic perinatal white matter injury. Ann Neurol 2008; 63:520-30. [PMID: 18393269 PMCID: PMC3140464 DOI: 10.1002/ana.21359] [Citation(s) in RCA: 264] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Abnormal myelination is a major pathological sequela of chronic periventricular white matter injury in survivors of premature birth. We tested the hypothesis that myelination failure in chronic hypoxia-ischemia-induced periventricular white matter injury is related to persistent depletion of the oligodendrocyte (OL) precursor pool required to generate mature myelinating OLs. METHODS A neonatal rat model of hypoxia-ischemia was used where acute degeneration of late OL progenitors (preOLs) occurs via a mostly caspase-independent mechanism. The fate of OL lineage cells in chronic cerebral lesions was defined with OL lineage-specific markers. RESULTS Acute caspase-3-independent preOL degeneration from hypoxia-ischemia was significantly augmented by delayed preOL death that was caspase-3-dependent. Degeneration of preOLs was offset by a robust regenerative response that resulted in a several-fold expansion in the pool of surviving preOLs in chronic lesions. However, these preOLs displayed persistent maturation arrest with failure to differentiate and generate myelin. When preOL-rich chronic lesions sustained recurrent hypoxia-ischemia at a time in development when white matter is normally resistant to injury, an approximately 10-fold increase in caspase-dependent preOL degeneration occurred relative to lesions caused by a single episode of hypoxia-ischemia. INTERPRETATION The mechanism of myelination failure in chronic white matter lesions is related to a combination of delayed preOL degeneration and preOL maturation arrest. The persistence of a susceptible population of preOLs renders chronic white matter lesions markedly more vulnerable to recurrent hypoxia-ischemia. These data suggest that preOL maturation arrest may predispose to more severe white matter injury in preterm survivors that sustain recurrent hypoxia-ischemia.
Collapse
Affiliation(s)
- Kristen N. Segovia
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Melissa McClure
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Matthew Moravec
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Ning Ling Luo
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Ying Wan
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Xi Gong
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Art Riddle
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Andrew Craig
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
| | - Jaime Struve
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Larry S. Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Stephen A. Back
- Department of Pediatrics, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| |
Collapse
|
140
|
Abstract
Isolation and characterization of neural stem cells and lineage-specific progenitors provide important information for central nervous system development study and regenerative medicine. We describe methods for dissection of rodent embryonic spinal cords by enzymatic separation, and isolation and enrichment (or purification) of neuronal and glial precursors at different developing stages by fluorescence-activated cell sorting.
Collapse
|
141
|
Witusik M, Gresner SM, Hulas-Bigoszewska K, Krynska B, Azizi SA, Liberski PP, Brown P, Rieske P. Neuronal and astrocytic cells, obtained after differentiation of human neural GFAP-positive progenitors, present heterogeneous expression of PrPc. Brain Res 2007; 1186:65-73. [PMID: 17996224 DOI: 10.1016/j.brainres.2007.10.039] [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: 06/04/2007] [Revised: 09/28/2007] [Accepted: 10/06/2007] [Indexed: 11/19/2022]
Abstract
PrP(c) is a cellular isoform of the prion protein with an unknown normal function. One of the putative physiological roles of this protein is its involvement in cell differentiation. Recently, in vitro and in vivo studies showed that GFAP-positive cells have characteristics of stem/progenitor cells that generate neurons and glia. We used an in vitro model of human neurogenesis from GFAP-positive progenitor cells to study the expression of PrP(c) during neural differentiation. Semi-quantitative multiplex-PCR assay and Western blot analysis revealed a significant increase of PRNP expression level in differentiated cells compared to undifferentiated cell population. As determined by immunocytochemistry followed by a quantitative image analysis, the PrP(c) level increased significantly in neuronal cells and did not increase significantly in glial cells. Of note, glial and neuronal cells showed a very large heterogeneity of PrP(c) expression. Our results provide the basis for studying the role of PrP(c) in cell differentiation and neurogenesis from human GFAP-positive progenitor cells.
Collapse
Affiliation(s)
- Monika Witusik
- Department of Molecular Pathology and Neuropathology, Chair of Oncology, Medical University of Lodz, 8/10 Czechoslowacka str., Lodz, Poland
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Cassiani-Ingoni R, Muraro PA, Magnus T, Reichert-Scrivner S, Schmidt J, Huh J, Quandt JA, Bratincsak A, Shahar T, Eusebi F, Sherman LS, Mattson MP, Martin R, Rao MS. Disease progression after bone marrow transplantation in a model of multiple sclerosis is associated with chronic microglial and glial progenitor response. J Neuropathol Exp Neurol 2007; 66:637-49. [PMID: 17620989 DOI: 10.1097/nen.0b013e318093f3ef] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS), the most common nontraumatic cause of neurologic disability in young adults in economically developed countries, is characterized by inflammation, gliosis, demyelination, and neuronal degeneration in the CNS. Bone marrow transplantation (BMT) can suppress inflammatory disease in a majority of patients with MS but retards clinical progression only in patients treated in the early stages of the disease. Here, we applied BMT in a mouse model of neuroinflammation, experimental autoimmune encephalomyelitis (EAE), and investigated the kinetics of reconstitution of the immune system in the periphery and in the CNS using bone marrow cells isolated from syngeneic donors constitutively expressing green fluorescent protein. This approach allowed us to dissect the contribution of donor cells to the turnover of resident microglia and to the pathogenesis of observed disease relapses after BMT. BMT effectively blocked or delayed EAE development when mice were treated early in the course of the disease but was without effect in mice with chronic disease. We found that there is minimal overall replacement of host microglia with donor cells in the CNS and that newly transplanted cells do not appear to contribute to disease progression. In contrast, EAE relapses are accompanied by the robust activation of endogenous microglial and macroglial cells, which further involves the maturation of endogenous Olig2 glial progenitor cells into reactive astrocytes through the cytoplasmic translocation of Olig2 and the expression of CD44 on the cellular membrane. The observed maturation of large numbers of reactive astrocytes from glial progenitors and the chronic activation of host microglial cells have relevance for our understanding of the resident glial response to inflammatory injury in the CNS. Our data indicate that reactivation of a local inflammatory process after BMT is sustained predominantly by endogenous microglia/macrophages.
Collapse
Affiliation(s)
- Riccardo Cassiani-Ingoni
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Magnus T, Coksaygan T, Korn T, Xue H, Arumugam TV, Mughal MR, Eckley DM, Tang SC, Detolla L, Rao MS, Cassiani-Ingoni R, Mattson MP. Evidence that nucleocytoplasmic Olig2 translocation mediates brain-injury-induced differentiation of glial precursors to astrocytes. J Neurosci Res 2007; 85:2126-37. [PMID: 17510983 DOI: 10.1002/jnr.21368] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The mechanisms by which neural and glial progenitor cells in the adult brain respond to tissue injury are unknown. We studied the responses of these cells to stab wound injury in rats and in two transgenic mouse models in which Y/GFP is driven either by Sox2 (a neural stem cell marker) or by Talpha-1 (which marks newly born neurons). The response of neural progenitors was low in all nonneurogenic regions, and no neurogenesis occurred at the injury site. Glial progenitors expressing Olig2 and NG2 showed the greatest response. The appearance of these progenitors preceded the appearance of reactive astrocytes. Surprisingly, we found evidence of the translocation of the transcription factor Olig2 into cytoplasm in the first week after injury, a mechanism that is known to mediate the differentiation of astrocytes during brain development. Translocation of Olig2, down-regulation of NG2, and increased glial fibrillary acidic protein expression were recapitulated in vitro after exposure of glial progenitors to serum components or bone morphogentic protein by up-regulation of Notch-1. The glial differentiation and Olig2 translocation could be blocked by inhibition of Notch-1 with the gamma-secretase inhibitor DAPT. Together, these data indicate that the prompt maturation of numerous Olig2(+) glial progenitors to astrocytes underlies the repair process after a traumatic injury. In contrast, neural stem cells and neuronal progenitor cells appear to play only a minor role in the injured adult CNS.
Collapse
Affiliation(s)
- Tim Magnus
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Carmen J, Magnus T, Cassiani-Ingoni R, Sherman L, Rao MS, Mattson MP. Revisiting the astrocyte–oligodendrocyte relationship in the adult CNS. Prog Neurobiol 2007; 82:151-62. [PMID: 17448587 DOI: 10.1016/j.pneurobio.2007.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 01/26/2007] [Accepted: 03/02/2007] [Indexed: 01/31/2023]
Abstract
The lineages of both astrocytes and oligodendrocytes have been popular areas of research in the last decade. The source of these cells in the mature CNS is relevant to the study of the cellular response to CNS injury. A significant amount of evidence exists to suggest that resident precursor cells proliferate and differentiate into mature glial cells that facilitate tissue repair and recovery. Additionally, the re-entry of mature astrocytes into the cell cycle can also contribute to the pool of new astrocytes that are observed following CNS injury. In order to better understand the glial response to injury in the adult CNS we must revisit the astrocyte-oligodendrocyte relationship. Specifically, we argue that there is a common glial precursor cell from which astrocytes and oligodendrocytes differentiate and that the microenvironment surrounding the injury determines the fate of the stimulated precursor cell. Ideally, better understanding the origin of new glial cells in the injured CNS will facilitate the development of therapeutics targeted to alter the glial response in a beneficial way.
Collapse
Affiliation(s)
- Jessica Carmen
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD 21224, USA.
| | | | | | | | | | | |
Collapse
|
145
|
Gauthier AS, Furstoss O, Araki T, Chan R, Neel BG, Kaplan DR, Miller FD. Control of CNS cell-fate decisions by SHP-2 and its dysregulation in Noonan syndrome. Neuron 2007; 54:245-62. [PMID: 17442246 PMCID: PMC1900070 DOI: 10.1016/j.neuron.2007.03.027] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 02/15/2007] [Accepted: 03/16/2007] [Indexed: 02/08/2023]
Abstract
Within the developing mammalian CNS, growth factors direct multipotent precursors to generate neurons versus glia, a process that if perturbed might lead to neural dysfunction. In this regard, genetic mutations resulting in constitutive activation of the protein tyrosine phosphatase SHP-2 cause Noonan Syndrome (NS), which is associated with learning disabilities and mental retardation. Here, we demonstrate that genetic knockdown of SHP-2 in cultured cortical precursors or in the embryonic cortex inhibited basal neurogenesis and caused enhanced and precocious astrocyte formation. Conversely, expression of an NS SHP-2 mutant promoted neurogenesis and inhibited astrogenesis. Neural cell-fate decisions were similarly perturbed in a mouse knockin model that phenocopies human NS. Thus, SHP-2 instructs precursors to make neurons and not astrocytes during the neurogenic period, and perturbations in the relative ratios of these two cell types upon constitutive SHP-2 activation may contribute to the cognitive impairments in NS patients.
Collapse
Affiliation(s)
- Andrée S Gauthier
- Developmental Biology Program, Hospital for Sick Children, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| | | | | | | | | | | | | |
Collapse
|
146
|
Lim JC, Wolpaw AJ, Caldwell MA, Hladky SB, Barrand MA. Neural precursor cell influences on blood-brain barrier characteristics in rat brain endothelial cells. Brain Res 2007; 1159:67-76. [PMID: 17583679 DOI: 10.1016/j.brainres.2007.05.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 04/26/2007] [Accepted: 05/01/2007] [Indexed: 11/15/2022]
Abstract
This study explores the effects of neural precursor cells (NPCs) on barrier characteristics in brain vasculature. Primary rat brain endothelial cells were exposed to conditioned medium from NPCs isolated from day 14 embryonic rat brains and maintained as free-floating undifferentiated neurospheres. Such exposure increased brain endothelial transcript levels of the mdr1a but not mdr1b gene encoding P-glycoprotein (Pgp) and reduced proliferation but did not alter transendothelial resistance (TER). These effects were compared to those seen following co-culture with differentiating NPCs or with primary astrocytes. NPCs, if grown adherent, differentiate into glial and neuronal cells as assessed by immunocytochemical and mRNA analysis. Brain endothelial cells when co-cultured with these cells also showed reduced proliferation and enhanced mdr1a expression, but in addition increased TER. Similar increases were observed in co-culture with astrocytes. These results suggest that undifferentiated NPCs produce factors that influence Pgp expression whereas their progeny also affect tight junction integrity.
Collapse
Affiliation(s)
- Joseph C Lim
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | | | | | | | | |
Collapse
|
147
|
Shin S, Sun Y, Liu Y, Khaner H, Svant S, Cai J, Xu QX, Davidson BP, Stice SL, Smith AK, Goldman SA, Reubinoff BE, Zhan M, Rao MS, Chesnut JD. Whole Genome Analysis of Human Neural Stem Cells Derived from Embryonic Stem Cells and Stem and Progenitor Cells Isolated from Fetal Tissue. Stem Cells 2007; 25:1298-306. [PMID: 17272497 DOI: 10.1634/stemcells.2006-0660] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Multipotent neural stem cells (NSC) have been derived from human embryonic stem cells (hESC) as well as isolated from fetal tissues. However, there have been few exclusive markers of NSC identified to date, and the differences between NSC from various sources are poorly understood. Although cells isolated from these two sources share many important characteristics, it is not clear how closely they are related in terms of gene expression. Here, we compare the gene expression profiles of 11 lines of NSC derived from hESC (ES_NSC), four lines of NSC isolated from fetus (F_NSC), and two lines of restricted progenitors in order to characterize these cell populations and identify differences between NSC derived from these two sources. We showed that ES_NSC were clustered together with high transcriptional similarities but were distinguished from F_NSC, oligodendrocyte precursor cells, and astrocyte precursor cells. There were 17 genes expressed in both ES_NSC and F_NSC whose expression was not identified in restricted neural progenitors. Furthermore, the major differences between ES_NSC and F_NSC were mostly observed in genes related to the key neural differentiation pathways. Here, we show that comparison of global gene expression profiles of ES_NSC, F_NSC, and restricted neural progenitor cells makes it possible to identify some of the common characteristics of NSC and differences between similar stem cell populations derived from hESCs or isolated from fetal tissue. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Soojung Shin
- Stem Cells and Regenerative Medicine, Invitrogen, Carlsbad, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Fukuda S, Abematsu M, Mori H, Yanagisawa M, Kagawa T, Nakashima K, Yoshimura A, Taga T. Potentiation of astrogliogenesis by STAT3-mediated activation of bone morphogenetic protein-Smad signaling in neural stem cells. Mol Cell Biol 2007; 27:4931-7. [PMID: 17452461 PMCID: PMC1951480 DOI: 10.1128/mcb.02435-06] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Astrocytes play important roles in brain development and injury response. Transcription factors STAT3 and Smad1, activated by leukemia inhibitory factor (LIF) and bone morphogenetic protein 2 (BMP2), respectively, form a complex with the coactivator p300 to synergistically induce astrocytes from neuroepithelial cells (NECs) (K. Nakashima, M. Yanagisawa, H. Arakawa, N. Kimura, T. Hisatsune, M. Kawabata, K. Miyazono, and T. Taga, Science 284:479-482, 1999). However, the mechanisms that govern astrogliogenesis during the determination of the fate of neural stem cells remain elusive. Here we found that LIF induces expression of BMP2 via STAT3 activation and leads to the consequent activation of Smad1 to efficiently promote astrogliogenic differentiation of NECs. The BMP antagonist Noggin abrogated LIF-induced Smad1 activation and astrogliogenesis by inhibiting BMPs produced by NECs. NECs deficient in suppressor of cytokine signaling 3 (SOCS3), a negative regulator of STAT3, readily differentiated into astrocytes upon activation by LIF not only due to sustained activation of STAT3 but also because of the consequent activation of Smad1. Our study suggests a novel LIF-triggered positive regulatory loop that enhances astrogliogenesis.
Collapse
Affiliation(s)
- Shinji Fukuda
- Department of Cell Fate Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Identification of two novel glial-restricted cell populations in the embryonic telencephalon arising from unique origins. BMC DEVELOPMENTAL BIOLOGY 2007; 7:33. [PMID: 17439658 PMCID: PMC1858687 DOI: 10.1186/1471-213x-7-33] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2006] [Accepted: 04/17/2007] [Indexed: 01/12/2023]
Abstract
Background Considerably less attention has been given to understanding the cellular components of gliogenesis in the telencephalon when compared to neuronogenesis, despite the necessity of normal glial cell formation for neurological function. Early proposals of exclusive ventral oligodendrocyte precursor cell (OPC) generation have been challenged recently with studies revealing the potential of the dorsal telencephalon to also generate oligodendrocytes. The identification of OPCs generated from multiple regions of the developing telencephalon, together with the need of the embryonic telencephalon to provide precursor cells for oligodendrocytes as well as astrocytes in ventral and dorsal areas, raises questions concerning the identity of the precursor cell populations capable of generating macroglial subtypes during multiple developmental windows and in differing locations. Results We have identified progenitor populations in the ventral and dorsal telencephalon restricted to the generation of astrocytes and oligodendrocytes. We further demonstrate that the dorsal glial progenitor cells can be generated de novo from the dorsal telencephalon and we demonstrate their capacity for in vivo production of both myelin-forming oligodendrocytes and astrocytes upon transplantation. Conclusion Based on our results we offer a unifying model of telencephalic gliogenesis, with the generation of both oligodendrocytes and astrocytes from spatially separate, but functionally similar, glial restricted populations at different developmental times in the dorsal and ventral CNS.
Collapse
|
150
|
Sergent-Tanguy S, Véziers J, Bonnamain V, Boudin H, Neveu I, Naveilhan P. Cell surface antigens on rat neural progenitors and characterization of the CD3 (+)/CD3 (-) cell populations. Differentiation 2007; 74:530-41. [PMID: 17177850 DOI: 10.1111/j.1432-0436.2006.00098.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
While the hematopoietic lineage has been extensively studied using cluster of differentiation (CD) antibodies, very few data are available on the extracellular epitopes expressed by rat neural progenitors (rNPC) and their derivatives. In the present study, we used flow cytometry to screen 47 cell surface antigens, initially known as immune markers. The quantitative analyses were performed on rat neurospheres and compared with primary cultures of astroglial cells or cerebellar neurons. Several antigens such as CD80 or CD86 were clearly undetectable while others, like CD26 or CD161, showed a weak expression. Interestingly, 10% and 15% of the cells were immunopositive for CD172a and CD200, two immunoglobulin superfamily members preferentially expressed by glial or neuronal cells, respectively. Over 40% of the cells were immunopositive for CD3, CD71, or MHCI. The biological significance of the latter markers in rNPC remains to be determined but analyses of the CD3(-)/CD3(+) populations isolated by magnetic cell separation revealed differences in their cell fate. Indeed, CD3(+) cells did not establish neurospheres and differentiated mostly into GFAP(+) cells while CD3(-) cells were able to generate neurospheres upon mitogen treatment and gave rise to GFAP(+), A2B5(+), Tuj-1(+), and RIP(+) cells under differentiating conditions. In contrast, CD71(-)/CD71(+) cells did not show any significant difference in their proliferating and differentiating potentials. Finally, it is worth noting that an subpopulation of cells in rat neurospheres exhibit an immunoreactivity against anti-CD25 (IL2 receptor) and anti-CD62L (L-selectin) antibodies. The results reveal particular surface antigen profiles, giving new perspectives on the properties of rat brain-derived cells.
Collapse
|