551
|
Sierra A, Encinas JM, Maletic-Savatic M. Adult human neurogenesis: from microscopy to magnetic resonance imaging. Front Neurosci 2011; 5:47. [PMID: 21519376 PMCID: PMC3075882 DOI: 10.3389/fnins.2011.00047] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 03/23/2011] [Indexed: 01/18/2023] Open
Abstract
Neural stem cells reside in well-defined areas of the adult human brain and are capable of generating new neurons throughout the life span. In rodents, it is well established that the new born neurons are involved in olfaction as well as in certain forms of memory and learning. In humans, the functional relevance of adult human neurogenesis is being investigated, in particular its implication in the etiopathology of a variety of brain disorders. Adult neurogenesis in the human brain was discovered by utilizing methodologies directly imported from the rodent research, such as immunohistological detection of proliferation and cell-type specific biomarkers in postmortem or biopsy tissue. However, in the vast majority of cases, these methods do not support longitudinal studies; thus, the capacity of the putative stem cells to form new neurons under different disease conditions cannot be tested. More recently, new technologies have been specifically developed for the detection and quantification of neural stem cells in the living human brain. These technologies rely on the use of magnetic resonance imaging, available in hospitals worldwide. Although they require further validation in rodents and primates, these new methods hold the potential to test the contribution of adult human neurogenesis to brain function in both health and disease. This review reports on the current knowledge on adult human neurogenesis. We first review the different methods available to assess human neurogenesis, both ex vivo and in vivo and then appraise the changes of adult neurogenesis in human diseases.
Collapse
Affiliation(s)
- Amanda Sierra
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHouston, TX, USA
| | - Juan M. Encinas
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHouston, TX, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHouston, TX, USA
| |
Collapse
|
552
|
Bento AR, Baptista S, Malva JO, Silva AP, Agasse F. Methamphetamine exerts toxic effects on subventricular zone stem/progenitor cells and inhibits neuronal differentiation. Rejuvenation Res 2011; 14:205-14. [PMID: 21453012 DOI: 10.1089/rej.2010.1109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Methamphetamine (METH) is a potent and widely consumed psychostimulant drug that causes brain functional and structural abnormalities. However, there is little information regarding METH impact on adult neurogenic niches and, indeed, nothing is known about its consequences on the subventricular zone (SVZ). Thus, this work aims to clarify the effect of METH on SVZ stem/progenitor cells dynamics and neurogenesis. For that purpose, SVZ neurospheres were obtained from early postnatal mice and treated with increasing concentrations of METH (1 μM to 500 μM). Exposure to 100, 250, or 500 μM METH for 24 h triggered cell death both by necrosis and apoptosis, as assessed by propidium iodide uptake, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, and quantification of the proapoptotic caspase-3 activity. Furthermore, we showed that METH inhibited SVZ progenitor cells proliferation as it decreased BrdU incorporation. Interestingly, at non-toxic concentrations (1 and 10 μM), METH decreased neuronal differentiation and maturation, which were evaluated by quantification of the number of neuronal nuclei-positive neurons and measurements of phospho-c-Jun-NH(2)-terminal kinase signal in growing axons, respectively. Altogether, our data demonstrate that METH has a negative impact on SVZ stem/progenitor cells, inducing cell death and inhibiting neurogenesis, effects that in vivo may challenge the cell replacement capacities displayed by endogenous populations of brain stem/progenitor cells.
Collapse
Affiliation(s)
- Ana Rita Bento
- Neuroprotection and Neurogenesis in Brain Repair Group, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | |
Collapse
|
553
|
Sajad M, Zargan J, Sharma J, Chawla R, Arora R, Umar S, Khan HA. Increased spontaneous apoptosis of rat primary neurospheres in vitro after experimental autoimmune encephalomyelitis. Neurochem Res 2011; 36:1017-26. [PMID: 21448597 DOI: 10.1007/s11064-011-0441-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2011] [Indexed: 12/25/2022]
Abstract
Survival of neuronal progenitors (NPCs) is a critical determinant of the regenerative capacity of brain following cellular loss. Herein, we report for the first time, the increased spontaneous apoptosis of the first acute phase of Experimental Autoimmune Encephalomyelitis (EAE) derived neurospheres in vitro. Neuronal as well as oligodendroglial loss occurs during experimental autoimmune encephalomyelitis (EAE). This loss is replenished spontaneously by the concomitant increase in the NPC proliferation evidenced by the presence of thin myelin sheaths in the remodeled lesions. However, remyelination depends upon the survival of NPCs and their lineage specific differentiation. We observed significant increase (P < 0.001) in number of BrdU (+) cells in ependymal subventricular zone (SVZ) in EAE rats. EAE derived NPCs showed remarkable increase in S-phase population which was indeed due to the decrease in G-phase progeny suggesting activation of neuronal progenitor cells (NPCs) from quiescence. However, EAE derived neurospheres showed limited survival in vitro which was mediated by the significantly (P < 0.01) depolarized mitochondria, elevated Caspase-3 (P < 0.001) and fragmentation of nuclear DNA evidenced by single cell gel electrophoresis. Our results suggest EAE induced spontaneous apoptosis of NPCs in vitro which may increase the possibility of early stage cell death in the negative regulation of the proliferative cell number and may explain the failure of regeneration in human multiple sclerosis.
Collapse
Affiliation(s)
- Mir Sajad
- Clinical Toxicology Laboratory Formerly Developmental Toxicology Laboratory, Department of Medical Elementology and Toxicology, Jamia Hamdard, Hamdard University, New Delhi 110062, India
| | | | | | | | | | | | | |
Collapse
|
554
|
Abstract
The function of adult tissue-specific stem cells declines with age, which may contribute to the physiological decline in tissue homeostasis and the increased risk of neoplasm during aging. Old stem cells can be 'rejuvenated' by environmental stimuli in some cases, raising the possibility that a subset of age-dependent stem cell changes is regulated by reversible mechanisms. Epigenetic regulators are good candidates for such mechanisms, as they provide a versatile checkpoint to mediate plastic changes in gene expression and have recently been found to control organismal longevity. Here, we review the importance of chromatin regulation in adult stem cell compartments. We particularly focus on the roles of chromatin-modifying complexes and transcription factors that directly impact chromatin in aging stem cells. Understanding the regulation of chromatin states in adult stem cells is likely to have important implications for identifying avenues to maintain the homeostatic balance between sustained function and neoplastic transformation of aging stem cells.
Collapse
Affiliation(s)
- E A Pollina
- Department of Genetics, Stanford University, CA, USA
| | | |
Collapse
|
555
|
Pugliese M, Rodríguez MJ, Gimeno-Bayon J, Pujadas L, Billett EE, Wells C, Mahy N. Alzheimer's disease modifies progenitor cell expression of monoamine oxidase B in the subventricular zone. J Neurosci Res 2011; 88:2588-97. [PMID: 20648649 DOI: 10.1002/jnr.22423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In the adult brain, progenitor cells remaining in the subventricular zone (SVZ) are frequently identified as glial fibrillary acidic protein (GFAP)-positive cells that retain attributes reminiscent of radial glia. Because the very high expression of monoamine oxidase B (MAO-B) in the subventricular area has been related to epithelial and astroglial expression, we sought to ascertain whether it was also expressed by progenitor cells of human control and Alzheimer's disease (AD) patients. In the SVZ, epithelial cells and astrocyte-like cells presented rich MAO-B activity and immunolabeling. Nestin-positive cells were found in the same area, showing a radial glia-like morphology. When coimmunostaining and confocal microscopy were performed, most nestin-positive cells showed MAO-B activity and labeling. The increased progenitor activity in SVZ proposed for AD patients was confirmed by the positive correlation between the SVZ nestin/MAO-B ratio and the progression of the disease. Nestin/GFAP-positive cells, devoid of MAO-B, can represent a distinct subpopulation of an earlier phase of maturation. This would indicate that MAO-B expression takes place in a further step of nestin/GFAP-positive cell differentiation. In the early AD stages, the discrete MAO-B reduction, different from the severe GFAP decrease, would reflect the capacity of this population of MAO-B-positive progenitor cells to adapt to the neurodegenerative process.
Collapse
Affiliation(s)
- Marco Pugliese
- Unitat de Bioquímica i Biologia Molecular, Facultat de Medicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
556
|
Abstract
Adult neurogenesis occurs in two privileged microenvironments, the hippocampal subgranular zone of the dentate gyrus and the subventricular zone (SVZ) along the lateral ventricle. This review focuses on accumulating evidence suggesting that the activity of specific brain regions or bodily states influences SVZ cell proliferation and neurogenesis. Neuromodulators such as dopamine and serotonin have been shown to have long-range effects through neuronal projections into the SVZ. Local γ-aminobutyric acid and glutamate signaling have demonstrated effects on SVZ proliferation and neurogenesis, but an extra-niche source of these neurotransmitters remains to be explored and options will be discussed. There is also accumulating evidence that diseases and bodily states such as Alzheimer's disease, seizures, sleep and pregnancy influence SVZ cell proliferation. With such complex behavior and environmentally-driven factors that control subregion-specific activity, it will become necessary to account for overlapping roles of multiple neurotransmitter systems on neurogenesis when developing cell therapies or drug treatments.
Collapse
Affiliation(s)
- Stephanie Z Young
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, FMB 422, New Haven, CT 06520-8082, USA
| | | | | |
Collapse
|
557
|
A radiotherapy technique to limit dose to neural progenitor cell niches without compromising tumor coverage. J Neurooncol 2011; 104:579-87. [PMID: 21327710 DOI: 10.1007/s11060-011-0530-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 01/31/2011] [Indexed: 12/13/2022]
Abstract
Radiation therapy (RT) for brain tumors is associated with neurocognitive toxicity which may be a result of damage to neural progenitor cells (NPCs). We present a novel technique to limit the radiation dose to NPC without compromising tumor coverage. A study was performed in mice to examine the rationale and another was conducted in humans to determine its feasibility. C57BL/6 mice received localized radiation using a dedicated animal irradiation system with on-board CT imaging with either: (1) Radiation which spared NPC containing regions; (2) Radiation which did not spare these niches; or (3) Sham irradiation. Mice were sacrificed 24 h later and the brains were processed for immunohistochemical Ki-67 staining. For the human component of the study, 33 patients with primary brain tumors were evaluated. Two intensity modulated radiotherapy (IMRT) plans were retrospectively compared: a standard clinical plan and a plan which spares NPC regions while maintaining the same dose coverage of the tumor. The change in radiation dose to the contralateral NPC-containing regions was recorded. In the mouse model, non-NPC-sparing radiation treatment resulted in a significant decrease in the number of Ki67(+) cells in dentate gyrus (DG) (P = 0.008) and subventricular zone (SVZ) (P = 0.005) compared to NPC-sparing radiation treatment. In NPC-sparing clinical plans, NPC regions received significantly lower radiation dose with no clinically relevant changes in tumor coverage. This novel radiation technique should significantly reduce radiation doses to NPC containing regions of the brain which may reduce neurocognitive deficits following RT for brain tumors.
Collapse
|
558
|
Brain cooling-stimulated angiogenesis and neurogenesis attenuated traumatic brain injury in rats. ACTA ACUST UNITED AC 2011; 69:1467-72. [PMID: 21150525 DOI: 10.1097/ta.0b013e3181f31b06] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Although brain cooling has been reported to be effective in improving the outcome after traumatic brain injury (TBI) in rats, the mechanisms of brain cooling-induced neuroprotective actions remain unclear. This study was to test whether angiogenesis and neurogenesis attenuating TBI could be brain cooling stimulated. METHODS Anesthetized rats, immediately after the onset of TBI, were divided into two groups and given the brain cooling (infusing 5 mL of 4°C saline via the external jugular vein) or no brain cooling (infusing 5 mL of 37°C saline via the external jugular vein). RESULTS Brain cooling without interference with the core temperature in rats significantly attenuated TBI-induced cerebral infarction (90 mm³ vs. 250 mm³) and motor (61 degrees vs. 57 degrees maximal angle) and proprioceptive (14% vs. 42% maximal possible effect) function deficits, significantly reduced TBI-induced neuronal (24 vs. 62 neuronal-specific nuclear [NeuN]-TUNEL double-positive cells) and glial (5 vs. 35 GFAP-TUNEL double-positive cells) apoptosis (increased TUNEL-positive and caspase-3-positive cells), neuronal loss (102 vs. 66 NeuN-positive cells), and gliosis (40 vs. 66 GFAP-positive cells; 66 vs. 89 Iba1-positive cells), and significantly promoted angiogenesis (5-bromodeoxyuridine [BrdU]/endothelial cells vs. 1-BrdU/endothelial cell; 58 vs. 31 vascular endothelial growth factor-positive cells), and neurogenesis (33 vs. 14 BrdU/NeuN positive cells). CONCLUSIONS Brain cooling-stimulated angiogenesis and neurogenesis attenuated a fluid percussion TBI in rats.
Collapse
|
559
|
Prospects and limitations of using endogenous neural stem cells for brain regeneration. Genes (Basel) 2011; 2:107-30. [PMID: 24710140 PMCID: PMC3924842 DOI: 10.3390/genes2010107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 12/06/2010] [Accepted: 01/04/2011] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells (NSCs) are capable of producing a variety of neural cell types, and are indispensable for the development of the mammalian brain. NSCs can be induced in vitro from pluripotent stem cells, including embryonic stem cells and induced-pluripotent stem cells. Although the transplantation of these exogenous NSCs is a potential strategy for improving presently untreatable neurological conditions, there are several obstacles to its implementation, including tumorigenic, immunological, and ethical problems. Recent studies have revealed that NSCs also reside in the adult brain. The endogenous NSCs are activated in response to disease or trauma, and produce new neurons and glia, suggesting they have the potential to regenerate damaged brain tissue while avoiding the above-mentioned problems. Here we present an overview of the possibility and limitations of using endogenous NSCs in regenerative medicine.
Collapse
|
560
|
Markiewicz I, Sypecka J, Domanska-Janik K, Wyszomirski T, Lukomska B. Cellular environment directs differentiation of human umbilical cord blood-derived neural stem cells in vitro. J Histochem Cytochem 2011; 59:289-301. [PMID: 21378283 DOI: 10.1369/0022155410397997] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cord blood-derived neural stem cells (NSCs) are proposed as an alternative cell source to repair brain damage upon transplantation. However, there is a lack of data showing how these cells are driven to generate desired phenotypes by recipient nervous tissue. Previous research indicates that local environment provides signals driving the fate of stem cells. To investigate the impact of these local cues interaction, the authors used a model of cord blood-derived NSCs co-cultured with different rat brain-specific primary cultures, creating the neural-like microenvironment conditions in vitro. Neuronal and astro-, oligo-, and microglia cell cultures were obtained by the previously described methods. The CMFDA-labeled neural stem cells originated from, non-transformed human umbilical cord blood cell line (HUCB-NSCs) established in a laboratory. The authors show that the close vicinity of astrocytes and oligodendrocytes promotes neuronal differentiation of HUCB-NSCs, whereas postmitotic neurons induce oligodendrogliogenesis of these cells. In turn, microglia or endothelial cells do not favor any phenotypes of their neural commitment. Studies have confirmed that HUCB-NSCs can read cues from the neurogenic microenvironment, attaining features of neurons, astrocytes, or oligodendrocytes. The specific responses of neurally committed cord blood-derived cells, reported in this work, are very much similar to those described previously for NSCs derived from other "more typical" sources. This further proves their genuine neural nature. Apart from having a better insight into the neurogenesis in the adult brain, these findings might be important when predicting cord blood cell derivative behavior after their transplantation for neurological disorders.
Collapse
Affiliation(s)
- Inga Markiewicz
- Neurorepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | |
Collapse
|
561
|
Paradisi M, Fernández M, Del Vecchio G, Lizzo G, Marucci G, Giulioni M, Pozzati E, Antonelli T, Lanzoni G, Bagnara GP, Giardino L, Calzà L. Ex vivo study of dentate gyrus neurogenesis in human pharmacoresistant temporal lobe epilepsy. Neuropathol Appl Neurobiol 2011; 36:535-50. [PMID: 20609110 DOI: 10.1111/j.1365-2990.2010.01102.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
AIMS Neurogenesis in adult humans occurs in at least two areas of the brain, the subventricular zone of the telencephalon and the subgranular layer of the dentate gyrus in the hippocampal formation. We studied dentate gyrus subgranular layer neurogenesis in patients subjected to tailored antero-mesial temporal resection including amygdalohippocampectomy due to pharmacoresistant temporal lobe epilepsy (TLE) using the in vitro neurosphere assay. METHODS Sixteen patients were enrolled in the study; mesial temporal sclerosis (MTS) was present in eight patients. Neurogenesis was investigated by ex vivo neurosphere expansion in the presence of mitogens (epidermal growth factor + basic fibroblast growth factor) and spontaneous differentiation after mitogen withdrawal. Growth factor synthesis was investigated by qRT-PCR in neurospheres. RESULTS We demonstrate that in vitro proliferation of cells derived from dentate gyrus of TLE patients is dependent on disease duration. Moreover, the presence of MTS impairs proliferation. As long as in vitro proliferation occurs, neurogenesis is maintained, and cells expressing a mature neurone phenotype (TuJ1, MAP2, GAD) are spontaneously formed after mitogen withdrawal. Finally, formed neurospheres express mRNAs encoding for growth (vascular endothelial growth factor) as well as neurotrophic factors (brain-derived neurotrophic factor, ciliary neurotrophic factor, glial-derived neurotrophic factor, nerve growth factor). CONCLUSION We demonstrated that residual neurogenesis in the subgranular layer of the dentate gyrus in TLE is dependent on diseases duration and absent in MTS.
Collapse
Affiliation(s)
- M Paradisi
- BioPharmaNet-DIMORFIPA, and Department of Histology, Embryology and Applied Biology, University of Bologna, Via Tolara di Sopra 50, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
562
|
Abstract
AbstractGlioblastoma Multiforme (GBM) is the most malignant and devastating primary brain tumour with a median survival of ∼12–16 months. Although recent large scale sequencing projects have shed considerable light into the complexity of the disease, there remains much to be elucidated in the hopes of generating effective therapeutic strategies. Although these studies investigate the mutations and expression of bulk tumour they have limits with respect to cell of origin and the concept of brain tumour initiating cells (BTIC). Current research has challenged the old paradigm of the stochastic model as recent evidence suggests that a subset of cancer cells within a tumor is responsible for tumor initiation, maintenance, and resistance to therapy. To gain a better understanding of the different compartment of cells that GBM comprise of require careful and elegant experiments. In addition to studying GBM, exploring the role of normal neural stem cells and progenitors cells is essential to partially explain whether these GBM BTIC behave similarly or differently then their non transformed counterparts. Here we discuss the recent literature between the two models, candidate regions of glioma genesis, candidate cells of origin for GBM, and possible therapeutic avenues to explore.
Collapse
|
563
|
Alcantara Llaguno SR, Chen Y, McKay RM, Parada LF. Stem Cells in Brain Tumor Development. Curr Top Dev Biol 2011; 94:15-44. [DOI: 10.1016/b978-0-12-380916-2.00002-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
564
|
Chopp M, Zhang ZG. Enhancing Brain Reorganization and Recovery of Function after Stroke. Stroke 2011. [DOI: 10.1016/b978-1-4160-5478-8.10011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
565
|
Martinez-Molina N, Kim Y, Hockberger P, Szele FG. Rostral migratory stream neuroblasts turn and change directions in stereotypic patterns. Cell Adh Migr 2011; 5:83-95. [PMID: 21045564 DOI: 10.4161/cam.5.1.13788] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neuroblasts generated in the adult subventricular zone (SVZ) migrate through the rostral migratory stream (RMS) to the olfactory bulb (OB). Previous work uncovered motility ranging from straight to complex, but it was unclear if directional changes were stochastic or exhibited stereotypical patterns. Here, we provide the first in-depth two-photon time-lapse microscopy study of morphological and dynamic features that accompany turning and direction reversals in the RMS. We identified three specific kinds of turning (30-90 degrees): bending of the leading process proximal to the cell body (P-bending 47% of cases), bending of the distal leading process (D-bending 30%) or branching of the leading process or lamellipodium (23%). Bending and branching angles were remarkably constrained and were significantly different from one another. Cells reversed direction (> 90 degrees) through D-bendings (54%), branching (11%) or de novo growth of processes from the soma (23%), but not P-bending. Direction reversal was often composed of several iterations of D-bending or branching as opposed to novel modalities. Individual neuroblasts could turn or change direction in multiple patterns suggesting that the patterns are not specific for different lineages. These findings show that neuroblasts in the RMS use a limited number of distinct and constrained modalities to turn or reverse direction.
Collapse
|
566
|
Stem Cells in the Developing and Adult Nervous System. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
567
|
Kane AJ, Sughrue ME, Rutkowski MJ, Tihan T, Parsa AT. The molecular pathology of central neurocytomas. J Clin Neurosci 2011; 18:1-6. [DOI: 10.1016/j.jocn.2010.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 06/09/2010] [Indexed: 11/29/2022]
|
568
|
Christensen K, Schrøder HD, Kristensen BW. CD133+ niches and single cells in glioblastoma have different phenotypes. J Neurooncol 2010; 104:129-43. [PMID: 21184132 DOI: 10.1007/s11060-010-0488-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 12/02/2010] [Indexed: 12/19/2022]
Abstract
Putative CD133(+) brain tumor stem cells have been shown to be located in niches and as single cells. This is the first study providing insight into the different phenotypes of CD133(+) cells in glioblastoma according to localization. Paraffin sections were stained by double immunofluorescence with CD133 and the candidate stem cell markers Sox2, Bmi-1, EGFR, podoplanin and nestin, the proliferation marker Ki67 and the endothelial cell markers CD31, CD34, and VWF. Cell counting showed that the CD133(+) cells in the niches had a significantly higher expression of Sox2, EGFR and nestin compared to CD133(+) single cells, but only a 3% Ki67 labeling index versus 14% found for CD133(+) single cells. Only low endothelial cell marker expression was found in the niches or the CD133(-) tumor areas, while 43% CD133(+)/CD31(+) and 25% CD133(+)/CD34(+) single cells were found. CD133(+) blood vessels within CD133(+) niches were less proliferative and more often Bmi-1(+) than CD133(+) blood vessels outside niches. In conclusion, different CD133(+) cell phenotypes exist according to the in situ localization, and also the phenotype of CD133(+) blood vessels vary according to the localization. CD133(+) niches contain stem-like cells with a lower proliferation index than CD133(+) single cells, which have an endothelial differentiation profile suggesting a role in angiogenesis.
Collapse
Affiliation(s)
- Karina Christensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 5000 Odense C, Denmark
| | | | | |
Collapse
|
569
|
Sawaishi Y. Periventricular calcification added to the phenotypic repertoire of Alexander disease. Dev Med Child Neurol 2010; 52:1081-2. [PMID: 21175465 DOI: 10.1111/j.1469-8749.2010.03818.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Yukio Sawaishi
- Department of Pediatrics, Akita Prefectural Center on Development and Disability, Akita, Japan
| |
Collapse
|
570
|
Sajad M, Zargan J, Chawla R, Umar S, Khan HA. Upregulation of CSPG3 accompanies neuronal progenitor proliferation and migration in EAE. J Mol Neurosci 2010; 43:531-40. [PMID: 21107918 DOI: 10.1007/s12031-010-9476-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 11/07/2010] [Indexed: 01/26/2023]
Abstract
The molecular identities of signals that regulate the CNS lesion remodeling remain unclear. Herein, we report for the first time that extracellular matrix chondroitin sulphate proteoglycan, CSPG3 (neurocan) is upregulated after primary inflammatory injury. EAE was induced using myelin oligodendrocyte glycoprotein (MOG) (35-55) which was characterized by massive polymorphonuclear cell infiltration and loss of myelin basic protein expression along with steep decrease of CNPase. Periventricular white matter (PVWM) and cortex presented with astrogliosis evidenced by increased Glial fibrillary acidic protein (GFAP) immunoreactivity 20 days post immunization (p.i). Neuronal progenitor cell (NPC) proliferation increased after first acute episode in the subventricular zone (SVZ), corpus callosum, and cortex, indicating migration of cells to structures other than rostral migration stream and olfactory bulb, which is indicative of cell recruitment for repair process and was confirmed by presence of thin myelin sheaths in the shadow plaques. Earlier CSPG3 has been demonstrated to impede regeneration. We observed neuroinflammation-induced up-regulation of the CSPG3 expression in two most affected regions viz. PVWM and cortex after proliferation and migration of NPCs. Our results show possible role of reactive astrogliosis in lesion remodeling and redefine the relation between inflammation and endogenous cellular repair which can aid in designing of newer therapeutic strategies.
Collapse
Affiliation(s)
- Mir Sajad
- Developmental Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | | | | | | | | |
Collapse
|
571
|
van Tijn P, Kamphuis W, Marlatt MW, Hol EM, Lucassen PJ. Presenilin mouse and zebrafish models for dementia: focus on neurogenesis. Prog Neurobiol 2010; 93:149-64. [PMID: 21056616 DOI: 10.1016/j.pneurobio.2010.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/27/2010] [Accepted: 10/31/2010] [Indexed: 01/18/2023]
Abstract
Autosomal dominant mutations in the presenilin gene PSEN cause familial Alzheimer's disease (AD), a neurological disorder pathologically characterized by intraneuronal accumulation and extracellular deposition of amyloid-β in plaques and intraneuronal, hyperphosphorylated tau aggregation in neurofibrillary tangles. Presenilins (PS/PSENs) are part of the proteolytic γ-secretase complex, which cleaves substrate proteins within the membrane. Cleavage of the amyloid precursor protein (APP) by γ-secretase releases amyloid-β peptides. Besides its role in the processing of APP and other transmembrane proteins, presenilin plays an important role in neural progenitor cell maintenance and neurogenesis. In this review, we discuss the role of presenilin in relation to neurogenesis and neurodegeneration and review the currently available presenilin animal models. In addition to established mouse models, zebrafish are emerging as an attractive vertebrate model organism to study the role of presenilin during the development of the nervous system and in neurodegenerative disorders involving presenilin. Zebrafish is a suitable model organism for large-scale drug screening, making this a valuable model to identify novel therapeutic targets for AD.
Collapse
Affiliation(s)
- Paula van Tijn
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
572
|
Capilla-Gonzalez V, Gil-Perotin S, Garcia-Verdugo JM. Postnatal exposure to N-ethyl-N-nitrosurea disrupts the subventricular zone in adult rodents. Eur J Neurosci 2010; 32:1789-99. [PMID: 21044178 DOI: 10.1111/j.1460-9568.2010.07450.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
N-ethyl-N-nitrosurea (ENU), a type of N-nitrous compound (NOC), has been used as inductor for brain tumours due to its mutagenic effect on the rodent embryo. ENU also affected adult neurogenesis when administered during pregnancy. However, no studies have investigated the effect of ENU when exposured during adulthood. For this purpose, three experimental groups of adult mice were injected with ENU at different doses and killed shortly after exposure. When administered in adult mice, ENU did not form brain tumours but led to a disruption of the subventricular zone (SVZ), an adult neurogenic region. Analyses of the samples revealed a reduction in the numbers of neural progenitors compared with control animals, and morphological changes in ependymal cells. A significant decrease in proliferation was tested in vivo with 5-bromo-2-deoxyuridine administration and confirmed in vitro with a neurosphere assay. Cell death, assessed as active-caspase-3 reactivity, was more prominent in treated animals and cell death-related populations increased in parallel. Two additional groups were maintained for 45 and 120 days after five doses of ENU to study the potential regeneration of the SVZ, but only partial recovery was detected. In conclusion, exposure to ENU alters the organization of the SVZ and causes partial exhaustion of the neurogenic niche. The functional repercussion of these changes remains unknown, but exposure to NOCs implies a potential risk that needs further evaluation.
Collapse
Affiliation(s)
- V Capilla-Gonzalez
- Instituto Cavanilles de Biodiversidad y Biologia Evolutiva, Universidad de Valencia, Valencia, Spain
| | | | | |
Collapse
|
573
|
Engh JA. Identification of a novel marker of brain tumor stem cells. Neurosurgery 2010; 67:N16. [PMID: 20881541 DOI: 10.1227/01.neu.0000389732.26611.f8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
574
|
Kohl Z, Regensburger M, Aigner R, Kandasamy M, Winner B, Aigner L, Winkler J. Impaired adult olfactory bulb neurogenesis in the R6/2 mouse model of Huntington's disease. BMC Neurosci 2010; 11:114. [PMID: 20836877 PMCID: PMC2945356 DOI: 10.1186/1471-2202-11-114] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Accepted: 09/13/2010] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder linked to expanded CAG-triplet nucleotide repeats within the huntingtin gene. Intracellular huntingtin aggregates are present in neurons of distinct brain areas, among them regions of adult neurogenesis including the hippocampus and the subventricular zone/olfactory bulb system. Previously, reduced hippocampal neurogenesis has been detected in transgenic rodent models of HD. Therefore, we hypothesized that mutant huntingtin also affects newly generated neurons derived from the subventricular zone of adult R6/2 HD mice. RESULTS We observed a redirection of immature neuroblasts towards the striatum, however failed to detect new mature neurons. We further analyzed adult neurogenesis in the granular cell layer and the glomerular layer of the olfactory bulb, the physiological target region of subventricular zone-derived neuroblasts. Using bromodeoxyuridine to label proliferating cells, we observed in both neurogenic regions of the olfactory bulb a reduction in newly generated neurons. CONCLUSION These findings suggest that the striatal environment, severely affected in R6/2 mice, is capable of attracting neuroblasts, however this region fails to provide sufficient signals for neuronal maturation. Moreover, in transgenic R6/2 animals, the hostile huntingtin-associated microenvironment in the olfactory bulb interferes with the survival and integration of new mature neurons. Taken together, endogenous cell repair strategies in HD may require additional factors for the differentiation and survival of newly generated neurons both in neurogenic and non-neurogenic regions.
Collapse
Affiliation(s)
- Zacharias Kohl
- Division of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany
| | | | - Robert Aigner
- Division of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany
| | - Mahesh Kandasamy
- Department of Neurology, University of Regensburg, Regensburg, Germany
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Beate Winner
- Department of Neurology, University of Regensburg, Regensburg, Germany
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Jürgen Winkler
- Division of Molecular Neurology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
575
|
Affiliation(s)
| | - Jonathan H. Sherman
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia
| | - Roberto Salvatori
- Department of Medicine, Division of Endocrinology, Johns Hopkins University, Baltimore, Maryland
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery and Oncology, Brain Tumor Stem Cell Laboratory and Neurosurgical Outcomes Laboratory, The Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
576
|
Wan F, Zhang S, Xie R, Gao B, Campos B, Herold-Mende C, Lei T. The utility and limitations of neurosphere assay, CD133 immunophenotyping and side population assay in glioma stem cell research. Brain Pathol 2010; 20:877-89. [PMID: 20331619 PMCID: PMC8094830 DOI: 10.1111/j.1750-3639.2010.00379.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 02/01/2010] [Indexed: 01/07/2023] Open
Abstract
The newly proposed glioma stem cell (GSC) hypothesis may re-model the way we diagnose and treat the tumor, which highlights the need for a complete knowledge on the genetic and epigenetic "blueprints" of GSCs. To identify the true "stemness" signatures, pure GSC populations are primarily needed. Reliable in vitro methods enriching for GSCs and thereby identifying the key stem-like characteristics constitute the preliminary step forward. We discuss in this review the current widely used methods for enriching and isolating GSCs, namely neurosphere assay, CD133 Immunophenotyping and side population assay, and detail their limitations and potential pitfalls that could complicate interpretation of corresponding results.
Collapse
Affiliation(s)
- Feng Wan
- Department of Neurosurgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | |
Collapse
|
577
|
Hamilton LK, Aumont A, Julien C, Vadnais A, Calon F, Fernandes KJL. Widespread deficits in adult neurogenesis precede plaque and tangle formation in the 3xTg mouse model of Alzheimer's disease. Eur J Neurosci 2010; 32:905-20. [PMID: 20726889 DOI: 10.1111/j.1460-9568.2010.07379.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) affects cognitive modalities that are known to be regulated by adult neurogenesis, such as hippocampal- and olfactory-dependent learning and memory. However, the relationship between AD-associated pathologies and alterations in adult neurogenesis has remained contentious. In the present study, we performed a detailed investigation of adult neurogenesis in the triple transgenic (3xTg) mouse model of AD, a unique model that generates both amyloid plaques and neurofibrillary tangles, the hallmark pathologies of AD. In both neurogenic niches of the brain, the hippocampal dentate gyrus and forebrain subventricular zone, we found that 3xTg mice had decreased numbers of (i) proliferating cells, (ii) early lineage neural progenitors, and (iii) neuroblasts at middle age (11months old) and old age (18months old). These decreases correlated with major reductions in the addition of new neurons to the respective target areas, the dentate granule cell layer and olfactory bulb. Within the subventricular zone niche, cytological alterations were observed that included a selective loss of subependymal cells and the development of large lipid droplets within the ependyma of 3xTg mice, indicative of metabolic changes. Temporally, there was a marked acceleration of age-related decreases in 3xTg mice, which affected multiple stages of neurogenesis and was clearly apparent prior to the development of amyloid plaques or neurofibrillary tangles. Our findings indicate that AD-associated mutations suppress neurogenesis early during disease development. This suggests that deficits in adult neurogenesis may mediate premature cognitive decline in AD.
Collapse
Affiliation(s)
- Laura K Hamilton
- Département de pathologie et biologie cellulaire, Université de Montréal, C.P. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | | | | | | | | | | |
Collapse
|
578
|
Dirks PB. Brain tumor stem cells: the cancer stem cell hypothesis writ large. Mol Oncol 2010; 4:420-30. [PMID: 20801091 DOI: 10.1016/j.molonc.2010.08.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 01/06/2023] Open
Abstract
Brain tumors, which are typically very heterogeneous at the cellular level, appear to have a stem cell foundation. Recently, investigations from multiple groups have found that human as well as experimental mouse brain tumors contain subpopulations of cells that functionally behave as tumor stem cells, driving tumor growth and generating tumor cell progeny that form the tumor bulk, but which then lose tumorigenic ability. In human glioblastomas, these tumor stem cells express neural precursor markers and are capable of differentiating into tumor cells that express more mature neural lineage markers. In addition, modeling brain tumors in mice suggests that neural precursor cells more readily give rise to full blown tumors, narrowing potential cells of origin to those rarer brain cells that have a proliferative potential. Applying stem cell concepts and methodologies is giving fresh insight into brain tumor biology, cell of origin and mechanisms of growth, and is offering new opportunities for development of more effective treatments. The field of brain tumor stem cells remains very young and there is much to be learned before these new insights are translated into new patient treatments.
Collapse
Affiliation(s)
- Peter B Dirks
- Division of Neurosurgery, Developmental and Stem Cell Biology Program, Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
579
|
Gonzalez-Perez O, Jauregui-Huerta F, Galvez-Contreras AY. Immune system modulates the function of adult neural stem cells. CURRENT IMMUNOLOGY REVIEWS 2010; 6:167-173. [PMID: 21037937 PMCID: PMC2964894 DOI: 10.2174/157339510791823772] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
New neurons are continuously produced in most, if not all, mammals. This Neurogenesis occurs only in discrete regions of the adult brain: the subventricular zone (SVZ) and the subgranular zone (SGZ). In these areas, there are neural stem cells (NSCs), multipotent and selfrenewing, which are regulated by a number of molecules and signaling pathways that control their cell fate choices, survival and proliferation rates. It was believed that growth and morphogenic factors were the unique mediators that controlled NSCs in vivo. Recently, chemokines and cytokines have been identified as important regulators of NSCs functions. Some of the most studied immunological effectors are leukemia inhibitory factor (LIF), ciliary neurotrophic factor (CNTF), interferon-gamma (IFN-γ), insulin-like growth factor-1 (IGF-1), tumor necrosis factor alpha (TNF-α), and the chemokines MCP-1 and SDF-1. These substances exert a considerable regulation on proliferation, cell-fate choices, migration and survival of NSCs. Hence, the immune system is emerging as an important regulator of neurogenic niches in the adult brain, but further studies are necessary to fully establish the biological meaning of these neural effects.
Collapse
Affiliation(s)
- Oscar Gonzalez-Perez
- Neuroscience Laboratory, Psychology School, University of Colima, Colima, Mexico 28040
- Neuroscience Department, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara. Guadalajara, Jal. Mexico 44340
| | - Fernando Jauregui-Huerta
- Neuroscience Department, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara. Guadalajara, Jal. Mexico 44340
| | - Alma Yadira Galvez-Contreras
- Neuroscience Department, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara. Guadalajara, Jal. Mexico 44340
| |
Collapse
|
580
|
Theus MH, Ricard J, Bethea JR, Liebl DJ. EphB3 limits the expansion of neural progenitor cells in the subventricular zone by regulating p53 during homeostasis and following traumatic brain injury. Stem Cells 2010; 28:1231-42. [PMID: 20496368 PMCID: PMC2967180 DOI: 10.1002/stem.449] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ephrins and Eph receptor(s) have recently been implicated in regulating neurogenesis in the adult subventricular zone (SVZ) and rostral migratory stream. Here, we examined the role of ephrinB3-EphB3 signaling in mediating the SVZ response to traumatic brain injury (TBI). Analysis of EphB3 expression showed colocalization with glial fibrillary acidic protein-positive neural stem progenitor cells (NSPCs) and doublecortin-positive neuroblasts, whereas ephrinB3 was expressed outside the neurogenic region. TBI resulted in a significant reduction in EphB3 expression, which coincided with enhanced NSPC survival and proliferation at 3 and 7 days postinjury. Analysis of mice lacking either ephrinB3 (ephrinB3(-/-)) or EphB3 (EphB3(-/-)) showed a significant increase in bromodeoxyuridine (BrdU) incorporation and Ki67 immunoreactivity in the SVZ. Interestingly, cell death was dissimilar between knockout mice, where cell death was reduced in EphB3(-/-) but increased in ephrinB3(-/-) mice. Lateral ventricle infusion of soluble preclustered ephrinB3-Fc reversed the proliferative and cell death defects in ephrinB3(-/-) but not EphB3(-/-) mice and prevented TBI-induced proliferation in wild-type NSPCs. Coincidently, tumor suppressor p53 expression was increased following EphB3 stimulation and is reduced in the absence of either EphB3 or ephrinB3. Furthermore, pharmacological inhibition and siRNA knockdown of p53-attenuated ephrinB3-Fc-mediated growth suppression while having no effect on cell death in cultured NSPCs. These data demonstrate that EphB3 signaling suppresses NSPC proliferation in a p53-dependent manner, induces cell death in the absence of ligand stimulation and is transiently reduced in the SVZ to initiate the expansion and survival of endogenous adult NSPCs following TBI.
Collapse
Affiliation(s)
- Michelle H Theus
- The Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami, Miami, Florida 33136, USA
| | | | | | | |
Collapse
|
581
|
Barami K. Oncomodulatory mechanisms of human cytomegalovirus in gliomas. J Clin Neurosci 2010; 17:819-23. [DOI: 10.1016/j.jocn.2009.10.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Accepted: 10/25/2009] [Indexed: 11/15/2022]
|
582
|
Characterization of neural stem/progenitor cells expressing VEGF and its receptors in the subventricular zone of newborn piglet brain. Neurochem Res 2010; 35:1455-70. [PMID: 20552272 DOI: 10.1007/s11064-010-0207-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2010] [Indexed: 12/11/2022]
Abstract
Neural stem/progenitor cell (NSP) biology and neurogenesis in adult central nervous system (CNS) are important both towards potential future therapeutic applications for CNS repair, and for the fundamental function of the CNS. In the present study, we report the characterization of NSP population from subventricular zone (SVZ) of neonatal piglet brain using in vivo and in vitro systems. We show that the nestin and vimentin-positive neural progenitor cells are present in the SVZ of the lateral ventricles of neonatal piglet brain. In vitro, piglet NSPs proliferated as neurospheres, expressed the typical protein of neural progenitors, nestin and a range of well-established neurodevelopmental markers. Upon dissociation and subculture, piglet NSPs differentiated into neurons and glial cells. Clonal analysis demonstrates that piglet NSPs are multipotent and retain the capacity to generate both glia and neurons. These cells expressed VEGF, VEGFR1, VEGFR2 and Neuropilin-1 and -2 mRNAs. Real time PCR revealed that SVZ NSPs from newborn piglet expressed total VEGF and all VEGF splice variants. These findings show that piglet NSPs may be helpful to more effectively design growth factor based strategies to enhance endogenous precursor cells for cell transplantation studies potentially leading to the application of this strategy in the nervous system disease and injury.
Collapse
|
583
|
Hedner M, Nilsson LG, Olofsson JK, Bergman O, Eriksson E, Nyberg L, Larsson M. Age-Related Olfactory Decline is Associated with the BDNF Val66met Polymorphism: Evidence from a Population-Based Study. Front Aging Neurosci 2010; 2:24. [PMID: 20589104 PMCID: PMC2893376 DOI: 10.3389/fnagi.2010.00024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 05/18/2010] [Indexed: 11/30/2022] Open
Abstract
The present study investigates the effect of the brain-derived neurotrophic factor (BDNF) val66met polymorphism on change in olfactory function in a large scale, longitudinal population-based sample (n = 836). The subjects were tested on a 13 item force-choice odor identification test on two test occasions over a 5-year-interval. Sex, education, health-related factors, and semantic ability were controlled for in the statistical analyses. Results showed an interaction effect of age and BDNF val66met on olfactory change, such that the magnitude of olfactory decline in the older age cohort (70–90 years old at baseline) was larger for the val homozygote carriers than for the met carriers. The older met carriers did not display larger age-related decline in olfactory function compared to the younger group. The BDNF val66met polymorphism did not affect the rate of decline in the younger age cohort (45–65 years). The findings are discussed in the light of the proposed roles of BDNF in neural development and maintenance.
Collapse
Affiliation(s)
- Margareta Hedner
- Department of Psychology, Stockholm University Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
584
|
Van Den Berge SA, Middeldorp J, Zhang CE, Curtis MA, Leonard BW, Mastroeni D, Voorn P, Van De Berg WDJ, Huitinga I, Hol EM. Longterm quiescent cells in the aged human subventricular neurogenic system specifically express GFAP-delta. Aging Cell 2010; 9:313-26. [PMID: 20121722 DOI: 10.1111/j.1474-9726.2010.00556.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A main neurogenic niche in the adult human brain is the subventricular zone (SVZ). Recent data suggest that the progenitors that are born in the human SVZ migrate via the rostral migratory stream (RMS) towards the olfactory bulb (OB), similar to what has been observed in other mammals. A subpopulation of astrocytes in the SVZ specifically expresses an assembly-compromised isoform of the intermediate filament protein glial fibrillary acidic protein (GFAP-delta). To further define the phenotype of these GFAP-delta expressing cells and to determine whether these cells are present throughout the human subventricular neurogenic system, we analysed SVZ, RMS and OB sections of 14 aged brain donors (ages 74-93). GFAP-delta was expressed in the SVZ along the ventricle, in the RMS and in the OB. The GFAP-delta cells in the SVZ co-expressed the neural stem cell (NSC) marker nestin and the cell proliferation markers proliferating cell nuclear antigen (PCNA) and Mcm2. Furthermore, BrdU retention was found in GFAP-delta positive cells in the SVZ. In the RMS, GFAP-delta was expressed in the glial net surrounding the neuroblasts. In the OB, GFAP-delta positive cells co-expressed PCNA. We also showed that GFAP-delta cells are present in neurosphere cultures that were derived from SVZ precursors, isolated postmortem from four brain donors (ages 63-91). Taken together, our findings show that GFAP-delta is expressed in an astrocytic subpopulation in the SVZ, the RMS and the OB. Importantly, we provide the first evidence that GFAP-delta is specifically expressed in longterm quiescent cells in the human SVZ, which are reminiscent of NSCs.
Collapse
|
585
|
Deroide N, Nih LR, Dinh RYT, Lévy B, Kubis N. [Cerebral plasticity: from bench to bedside in stroke treatment]. Rev Med Interne 2010; 31:486-92. [PMID: 20494495 DOI: 10.1016/j.revmed.2009.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 08/22/2009] [Indexed: 11/25/2022]
Abstract
It has long been believed that cerebral lesions were irreversible in the adult human brain. However, the spontaneous improvement in functional outcome observed in the following weeks after cerebral ischemia suggests plasticity phenomenons involving postischemic neuronal network reorganization. Regarding the large prevalence of stroke in industrialized countries, and the few available treatments, the understanding of cerebral plasticity has become an important issue but also a potential source of new therapeutic approaches in stroke. Thus, "constraint induced therapy" and repetitive transcranial magnetic stimulation (rTMS) are based on the concept of local but also remote consequences of the ischemic focal lesion. Cell-therapy is based on the capacity of stem cells to respond to hypoxic signals and adapt their phenotype to the host organ, but above all to release cytokines locally and boost endogeneous repair mechanisms. We could consider to perform in the future a sequential treatment with fibrinolysis, stem cell therapy, repetitive transcranial magnetic stimulation and constraint-induced therapy in the same patient.
Collapse
Affiliation(s)
- N Deroide
- Service de physiologie explorations fonctionnelles, hôpital Lariboisière, 2, rue Ambroise-Paré, 75010 Paris, France
| | | | | | | | | |
Collapse
|
586
|
Chaichana K, Parker S, Olivi A, Quiñones-Hinojosa A. A proposed classification system that projects outcomes based on preoperative variables for adult patients with glioblastoma multiforme. J Neurosurg 2010; 112:997-1004. [PMID: 19817542 DOI: 10.3171/2009.9.jns09805] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Glioblastoma multiforme (GBM) is the most common and aggressive type of primary brain tumor in adults. Although the average survival is approximately 12 months, individual survival is heterogeneous. The ability to predict short- and long-term survivors is limited. Therefore, the aims of this study were to ascertain preoperative risk factors associated with survival, develop a preoperative prognostic grading system, and evaluate the utility of this grading system in predicting survival for patients undergoing resection of a primary intracranial GBM. METHODS Cases involving adult patients who underwent surgery for an intracranial primary (de novo) GBM between 1997 and 2007 at The Johns Hopkins Hospital, an academic tertiary-care institution, were retrospectively reviewed. Multivariate proportional hazards regression analysis was used to identify preoperative factors associated with survival, after controlling for extent of resection and adjuvant therapies. The identified associations with survival were then used to develop a grading system based on preoperative variables. Survival as a function of time was plotted using the Kaplan-Meier method, and survival rates were compared using Log-rank analysis. Associations with p < 0.05 were considered statistically significant. RESULTS Of the 393 patients in this study, 310 (79%) had died as of most recent follow-up (median time from surgery to death 11.9 months). The preoperative factors, independent of extent of resection and adjuvant therapies (carmustine wafers, temozolomide, and radiation), found to be negatively associated with survival were: age > 60 years (p < 0.0001), Karnofsky performance status score < or = 80 (p < 0.0001), motor deficit (p = 0.02), language deficit (p = 0.001), and periventricular tumor location (p = 0.04). Patients possessing 0-1, 2, 3, and 4-5 of these variables were assigned a preoperative grade of 1, 2, 3, and 4, respectively. Patients with a preoperative grade of 1, 2, 3, and 4 had a median survival of 16.6, 10.2, 6.8, and 6.1 months, respectively. CONCLUSIONS The present study found that older age, poor performance status, motor deficit, language deficit, and periventricular tumor location independently predicted poorer survival in patients undergoing GBM resection. A grading system based on these factors was able to identify 4 distinct groups of patients with different survival rates. This grading system, based only on preoperative variables, may provide patients and physicians with prognostic information that may guide medical and surgical therapy before any intervention is pursued.
Collapse
Affiliation(s)
- Kaisorn Chaichana
- Department of Neurosurgery, Johns Hopkins School of Medicine, and The Johns Hopkins Neuro-Oncology Surgical Outcomes Research Laboratory, Baltimore, Maryland 21231, USA
| | | | | | | |
Collapse
|
587
|
Richardson RM, Singh A, Sun D, Fillmore HL, Dietrich DW, Bullock MR. Stem cell biology in traumatic brain injury: effects of injury and strategies for repair. J Neurosurg 2010; 112:1125-38. [PMID: 19499984 DOI: 10.3171/2009.4.jns081087] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Approximately 350,000 individuals in the US are affected annually by severe and moderate traumatic brain injuries (TBI) that may result in long-term disability. This rate of injury has produced approximately 3.3 million disabled survivors in the US alone. There is currently no specific treatment available for TBI other than supportive care, but aggressive prehospital resuscitation, rapid triage, and intensive care have reduced mortality rates. With the recent demonstration that neurogenesis occurs in all mammals (including man) throughout adult life, albeit at a low rate, the concept of replacing neurons lost after TBI is now becoming a reality. Experimental rodent models have shown that neurogenesis is accelerated after TBI, especially in juveniles. Two approaches have been followed in these rodent models to test possible therapeutic approaches that could enhance neuronal replacement in humans after TBI. The first has been to define and quantify the phenomenon of de novo hippocampal and cortical neurogenesis after TBI and find ways to enhance this (for example by exogenous trophic factor administration). A second approach has been the transplantation of different types of neural progenitor cells after TBI. In this review the authors discuss some of the processes that follow after acute TBI including the changes in the brain microenvironment and the role of trophic factor dynamics with regard to the effects on endogenous neurogenesis and gliagenesis. The authors also discuss strategies to clinically harness the factors influencing these processes and repair strategies using exogenous neural progenitor cell transplantation. Each strategy is discussed with an emphasis on highlighting the progress and limiting factors relevant to the development of clinical trials of cellular replacement therapy for severe TBI in humans.
Collapse
Affiliation(s)
- R Mark Richardson
- Department of Neurological Surgery, University of California San Francisco, California, USA
| | | | | | | | | | | |
Collapse
|
588
|
Katz J, Keenan B, Snyder EY. Culture and manipulation of neural stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 671:13-22. [PMID: 20455492 DOI: 10.1007/978-1-4419-5819-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Despite advances in the treatment of cancer, the prognosis of patient diagnosed with metastatis cancer to the brain remains poor. The role of neural stem cells as a viable tool in the treatment of metastatic cancer to the brain alone or in conjuction with current therapeutic modalities is promising. Both murine and human neural stem cells (NSCs) have been shown to migrate through the central nervous system (CNS) and infiltrate tumors and other pathological disease states of the brain. Genetic modification of NSCs to produce cytotoxic or immunomodulatory agents in the vicinity of a primary tumor and/or satellite lesion or has proven instrumental to the reduction of tumor bulk in murine models. Although the use of stem cells proves to be a volatile social topic, scientists have discovered that NSCs are present in the adult brain and continue to propagate and differentiate. These cells may be isolated and cultured to produce clonal NSC lines that are capable of self renewal and differentiation when introduced into the CNS. In this chapter, we describe protocols currently used in ourlab for the successful maintenance of NSCs in vitro advancing the role of neural stem cells in the treatment of brain tumors.
Collapse
Affiliation(s)
- Jennifer Katz
- Del E. Webb Neuroscience, Aging and Stem Cell Research Center, Burnham Institute for Regenerative Madicine, La Jolla, California, USA.
| | | | | |
Collapse
|
589
|
Prestegarden L, Svendsen A, Wang J, Sleire L, Skaftnesmo KO, Bjerkvig R, Yan T, Askland L, Persson A, Sakariassen PØ, Enger PØ. Glioma Cell Populations Grouped by Different Cell Type Markers Drive Brain Tumor Growth. Cancer Res 2010; 70:4274-9. [DOI: 10.1158/0008-5472.can-09-3904] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
590
|
Platel JC, Stamboulian S, Nguyen I, Bordey A. Neurotransmitter signaling in postnatal neurogenesis: The first leg. BRAIN RESEARCH REVIEWS 2010; 63:60-71. [PMID: 20188124 PMCID: PMC2862802 DOI: 10.1016/j.brainresrev.2010.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 02/16/2010] [Accepted: 02/18/2010] [Indexed: 12/27/2022]
Abstract
Like the liver or other peripheral organs, two regions of the adult brain possess the ability of self-renewal through a process called neurogenesis. This raises tremendous hope for repairing the damaged brain, and it has stimulated research on identifying signals controlling neurogenesis. Neurogenesis involves several stages from fate determination to synaptic integration via proliferation, migration, and maturation. While fate determination primarily depends on a genetic signature, other stages are controlled by the interplay between genes and microenvironmental signals. Here, we propose that neurotransmitters are master regulators of the different stages of neurogenesis. In favor of this idea, a description of selective neurotransmitter signaling and their functions in the largest neurogenic zone, the subventricular zone (SVZ), is provided. In particular, we emphasize the interactions between neuroblasts and astrocyte-like cells that release gamma-aminobutyric acid (GABA) and glutamate, respectively. However, we also raise several limitations to our knowledge on neurotransmitters in neurogenesis. The function of neurotransmitters in vivo remains largely unexplored. Neurotransmitter signaling has been viewed as uniform, which dramatically contrasts with the cellular and molecular mosaic nature of the SVZ. How neurotransmitters are integrated with other well-conserved molecules, such as sonic hedgehog, is poorly understood. In an effort to reconcile these differences, we discuss how specificity of neurotransmitter functions can be provided through their multitude of receptors and intracellular pathways in different cell types and their possible interactions with sonic hedgehog.
Collapse
Affiliation(s)
- Jean-Claude Platel
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520-8082, USA
| | | | | | | |
Collapse
|
591
|
Alunni A, Hermel JM, Heuzé A, Bourrat F, Jamen F, Joly JS. Evidence for neural stem cells in the medaka optic tectum proliferation zones. Dev Neurobiol 2010; 70:693-713. [DOI: 10.1002/dneu.20799] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
592
|
Bradford D, Faull R, Curtis M, Cooper H. Characterization of the Netrin/RGMa receptor neogenin in neurogenic regions of the mouse and human adult forebrain. J Comp Neurol 2010; 518:3237-53. [DOI: 10.1002/cne.22397] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
593
|
Sotnikov OS. Interpretation of some mechanisms of evolutional neuromorphogenesis with aid of experiments in neuronal culture. J EVOL BIOCHEM PHYS+ 2010. [DOI: 10.1134/s0022093009060088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
594
|
Targeting A20 decreases glioma stem cell survival and tumor growth. PLoS Biol 2010; 8:e1000319. [PMID: 20186265 PMCID: PMC2826371 DOI: 10.1371/journal.pbio.1000319] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 01/20/2010] [Indexed: 12/20/2022] Open
Abstract
The A20 protein is a known inhibitor of apoptosis that here is shown to be a novel cancer stem cell-promoting factor associated with poor glioma patient survival. Glioblastomas are deadly cancers that display a functional cellular hierarchy maintained by self-renewing glioblastoma stem cells (GSCs). GSCs are regulated by molecular pathways distinct from the bulk tumor that may be useful therapeutic targets. We determined that A20 (TNFAIP3), a regulator of cell survival and the NF-κB pathway, is overexpressed in GSCs relative to non-stem glioblastoma cells at both the mRNA and protein levels. To determine the functional significance of A20 in GSCs, we targeted A20 expression with lentiviral-mediated delivery of short hairpin RNA (shRNA). Inhibiting A20 expression decreased GSC growth and survival through mechanisms associated with decreased cell-cycle progression and decreased phosphorylation of p65/RelA. Elevated levels of A20 in GSCs contributed to apoptotic resistance: GSCs were less susceptible to TNFα-induced cell death than matched non-stem glioma cells, but A20 knockdown sensitized GSCs to TNFα-mediated apoptosis. The decreased survival of GSCs upon A20 knockdown contributed to the reduced ability of these cells to self-renew in primary and secondary neurosphere formation assays. The tumorigenic potential of GSCs was decreased with A20 targeting, resulting in increased survival of mice bearing human glioma xenografts. In silico analysis of a glioma patient genomic database indicates that A20 overexpression and amplification is inversely correlated with survival. Together these data indicate that A20 contributes to glioma maintenance through effects on the glioma stem cell subpopulation. Although inactivating mutations in A20 in lymphoma suggest A20 can act as a tumor suppressor, similar point mutations have not been identified through glioma genomic sequencing: in fact, our data suggest A20 may function as a tumor enhancer in glioma through promotion of GSC survival. A20 anticancer therapies should therefore be viewed with caution as effects will likely differ depending on the tumor type. Glioblastomas are the most common and aggressive primary brain tumors in adults, with a median survival of only 12–15 months. Glioblastomas display a cellular hierarchy with a subset of cells having stem cell–like properties, including the capacity to self-renew and propagate tumors. Specific ablation of cancer stem cells is widely thought to be critical for effective and long-lasting treatment of cancers. We report the identification of the antiapoptotic protein A20 (which is also known as TNFAIP3) as a novel regulator of glioma stem cell survival. Glioma stem cells overexpress A20 relative to non-stem glioma cells, and this protects them from cell death, whereas depletion of A20 attenuates glioma stem cell survival and tumor growth. Interrogation of a molecular glioma database reveals that A20 levels correlate with decreased survival in patients. These data indicate that A20 is a tumor enhancer in the context of glioma, which importantly contrasts with its known function as a tumor suppressor in the context of lymphoma. Therefore, A20 may be a context-specific regulator of cancer stem cell survival and growth.
Collapse
|
595
|
Michell AW, Phillips W, Barker RA. Can endogenous stem cells be stimulated to repair the degenerating brain? J Pharm Pharmacol 2010; 56:1201-10. [PMID: 15482633 DOI: 10.1211/0022357044517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- A W Michell
- Department of Neurology, Cambridge Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 2PY, UK.
| | | | | |
Collapse
|
596
|
Fishell G, Goldman JE. A silver lining to stroke: does ischemia generate new cortical interneurons? Nat Neurosci 2010; 13:145-6. [PMID: 20104204 DOI: 10.1038/nn0210-145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gord Fishell
- Gord Fishell is in the Department of Cell Biology and the Smilow Neuroscience Program, Smilow Research Center, New York University School of Medicine, New York, New York, USA.
| | | |
Collapse
|
597
|
Growth factors improve neurogenesis and outcome after focal cerebral ischemia. J Neural Transm (Vienna) 2010; 116:1397-402. [PMID: 19826899 DOI: 10.1007/s00702-009-0329-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2009] [Accepted: 09/22/2009] [Indexed: 12/12/2022]
Abstract
Stem cells have been proposed as a new form of cell-based therapy in a variety of disorders, including acute and degenerative brain diseases. Endogenous neural stem cells (eNSC) reside in the subventricular zone and in the subgranular zone of the hippocampus. eNSC are capable of self-renewal and differentiation into functional glia and neurons. Unfortunately, spontaneous brain regeneration is inefficient for clinically significant improvement following brain injury. However, eNSC responses may be augmented considerably by perturbing the pathways governing cell proliferation, migration and differentiation by application of exogenous growth factors. Importantly, current evidence suggests that such perturbations may lead to better functional outcome after stroke. This article summarizes the progress made in this field.
Collapse
|
598
|
Bordey A. The stem cell journey: from paradise to purgatory. Neuropharmacology 2010; 58:833-4. [PMID: 20146927 DOI: 10.1016/j.neuropharm.2010.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
599
|
Maire CL, Buchet D, Kerninon C, Deboux C, Baron-Van Evercooren A, Nait-Oumesmar B. Directing human neural stem/precursor cells into oligodendrocytes by overexpression of Olig2 transcription factor. J Neurosci Res 2010; 87:3438-46. [PMID: 19739249 DOI: 10.1002/jnr.22194] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Multipotential neural stem/precursor cells of the central nervous system were extensively studied for their properties of generating myelinating oligodendrocytes both in vitro and in vivo upon engraftment in animal models of myelin disorders, such as leucodystrophy and multiple sclerosis. These studies provided proof-of-principle that efficient myelination can be achieved by cell transplantation. However, one major drawback of cell-based therapy of myelin diseases is the difficulty in generating oligodendrocytes efficiently from human fetal neural stem/precursor cells (hNPC). Here we explored whether overexpression of the basic helix-loop-helix (bHLH) transcription factor Olig2 in fetal hNPC could enhance the generation of oligodendrocytes both in vitro and in vivo. We report that transduction of hNPC with Olig2-encoding lentiviral vectors enhances their commitment toward an oligodendroglial fate. Moreover, Olig2-transduced hNPC, grafted into the dysmyelinated shiverer mouse brain, survived up to 9 weeks, migrated extensively, and differentiated into MBP(+) myelinating oligodendrocytes. In contrast, control hNPC remained at a less mature stage and generated very few myelinating oligodendrocytes. Our study indicates that bHLH transcription factors, such as Olig2, are interesting targets for directing hNPC into myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Cécile L Maire
- Université Pierre et Marie Curie-Paris 6, UMR S 975, Paris, France
| | | | | | | | | | | |
Collapse
|
600
|
Achanta P, Roman NIS, Quiñones-Hinojosa A. Gliomagenesis and the use of neural stem cells in brain tumor treatment. Anticancer Agents Med Chem 2010; 10:121-30. [PMID: 20184546 PMCID: PMC2981502 DOI: 10.2174/187152010790909290] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 12/29/2009] [Indexed: 01/08/2023]
Abstract
The role of neural stem cells (NSCs) in both the physiological and pathological processes in the brain has been refined through recent studies within the neuro-oncological field. Alterations in NSC regulatory mechanisms may be fundamental for the development and progression of malignant gliomas. A subpopulation of cells within the tumor known as brain tumor stem cells (BTSCs) have been shown to share key properties with NSCs. The BTSC hypothesis has significantly contributed to a potential understanding as to why brain tumors hold such dismal prognosis. On the other hand, the normal NSCs possess the capacity to migrate extensively towards the tumor bulk as well as to lingering neoplastic regions of the brain. The tropism of NSCs towards brain tumors may provide an additional tool for the treatment of brain cancer. The creation of potential therapies through the use of NSCs has been studied and includes the delivery of gene products to specific locations of the central nervous system selectively targeting malignant brain tumor cells and maximizing the efficiency of their delivery. Here, the proposed mechanisms of how brain tumors emerge, the molecular pathways interrupted in NSC pathogenesis and the most recent preclinical results in the use of NSCs for glioma treatment are reviewed.
Collapse
Affiliation(s)
- Pragathi Achanta
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
| | - Neda I Sedora Roman
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
- University of Puerto Rico School of Medicine, Office A-873, PO BOX 365067, San Juan PR 00936-5067
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University, School of Medicine, CRB II, Room 272, 1550 Orleans Street, Baltimore, MD, 21231, USA
| |
Collapse
|