Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Mol Med ; 18(7): 1444-59, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24779367

ABSTRACT

Members of the transforming growth factor (TGF)-ß family govern a wide range of mechanisms in brain development and in the adult, in particular neuronal/glial differentiation and survival, but also cell cycle regulation and neural stem cell maintenance. This clearly created some discrepancies in the field with some studies favouring neuronal differentiation/survival of progenitors and others favouring cell cycle exit and neural stem cell quiescence/maintenance. Here, we provide a unifying hypothesis claiming that through its regulation of neural progenitor cell (NPC) proliferation, TGF-ß signalling might be responsible for (i) maintaining stem cells in a quiescent stage, and (ii) promoting survival of newly generated neurons and their functional differentiation. Therefore, we performed a detailed histological analysis of TGF-ß1 signalling in the hippocampal neural stem cell niche of a transgenic mouse that was previously generated to express TGF-ß1 under a tetracycline regulatable Ca-Calmodulin kinase promoter. We also analysed NPC proliferation, quiescence, neuronal survival and differentiation in relation to elevated levels of TGF-ß1 in vitro and in vivo conditions. Finally, we performed a gene expression profiling to identify the targets of TGF-ß1 signalling in adult NPCs. The results demonstrate that TGF-ß1 promotes stem cell quiescence on one side, but also neuronal survival on the other side. Thus, considering the elevated levels of TGF-ß1 in ageing and neurodegenerative diseases, TGF-ß1 signalling presents a molecular target for future interventions in such conditions.


Subject(s)
Cell Differentiation , Hippocampus/cytology , Neurogenesis/physiology , Neurons/cytology , Stem Cell Niche , Stem Cells/cytology , Transforming Growth Factor beta/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Cellular Senescence , Doublecortin Protein , Electrophysiology , Female , Gene Expression Profiling , Hippocampus/metabolism , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Transforming Growth Factor beta/genetics
2.
Glia ; 61(11): 1767-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038377

ABSTRACT

The differentiation of adult neural progenitors (NPCs) into functional neurons is still a limiting factor in the neural stem cell field but mandatory for the potential use of NPCs in therapeutic approaches. Neuronal function requires the appropriate electrophysiological properties. Here, we demonstrate that priming of NPCs using transforming growth factor (TGF)-ß1 under conditions that usually favor NPCs' proliferation induces electrophysiological neuronal properties in adult NPCs. Gene chip array analyses revealed upregulation of voltage-dependent ion channel subunits (Kcnd3, Scn1b, Cacng4, and Accn1), neurotransmitters, and synaptic proteins (Cadps, Snap25, Grik4, Gria3, Syngr3, and Gria4) as well as other neuronal proteins (doublecortin [DCX], Nrxn1, Sept8, and Als2cr3). Patch-clamp analysis demonstrated that control-treated cells expressed only voltage-dependent K(+) -channels of the delayed-rectifier type and the A-type channels. TGF-ß1-treated cells possessed more negative resting potentials than nontreated cells owing to the presence of delayed-rectifier and inward-rectifier channels. Furthermore, TGF-ß1-treated cells expressed voltage-dependent, TTX-sensitive Na(+) channels, which showed increasing current density with TGF-ß1 treatment duration and voltage-dependent (+)BayK8644-sensitive L-Type Ca(2+) channels. In contrast to nontreated cells, TGF-ß1-treated cells responded to current injections with action-potentials in the current-clamp mode. Furthermore, TGF-ß1-treated cells responded to application of GABA with an increase in membrane conductance and showed spontaneous synaptic currents that were blocked by the GABA-receptor antagonist picrotoxine. Only NPCs, which were treated with TGF-ß1, showed Na(+) channel currents, action potentials, and GABAergic currents. In summary, stimulation of NPCs by TGF-ß1 fosters a functional neuronal phenotype, which will be of relevance for future cell replacement strategies in neurodegenerative diseases or acute CNS lesions.


Subject(s)
Action Potentials/physiology , Cell Differentiation/physiology , Cell Proliferation , Potassium Channels/metabolism , Stem Cells/cytology , Transforming Growth Factor beta1/metabolism , Action Potentials/drug effects , Aging , Animals , Cell Differentiation/drug effects , Cells, Cultured , Doublecortin Protein , Female , Membrane Potentials/physiology , Potassium Channels/drug effects , Rats , Rats, Inbred F344 , Stem Cells/metabolism , Tetrodotoxin/pharmacology
3.
Cell Tissue Res ; 345(3): 313-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21837406

ABSTRACT

5-Bromo-2'-deoxyuridin (BrdU) is frequently used in anaylsis of neural stem cell biology, in particular to label and to fate-map dividing cells. However, up to now, only a few studies have addressed the question as to whether BrdU labeling per se affects the cells to be investigated. Here, we focused on the potential impact of BrdU on neurosphere cultures derived from the adult rat brain and on proliferation of progenitors in vivo. In vitro, neurospheres were pulsed for 48 h with BrdU, and cell proliferation, cell cycle, differentiation, survival and adhesion properties were subsequently analyzed. BrdU inhibited the expansion of neural progenitors as assessed by MTS assay and increased the fraction of cells in the G0/G1-phase of the cell cycle. Moreover, BrdU increased cell death and dose-dependently induced adherence of NPCs. Cell adherence was accompanied by a reduced amount of active matrix-metalloproteinase-2 (MMP-2). Furthermore, BrdU repressed neuronal and oligodendroglial differentiation, whereas astroglial fate was not affected. In contrast to the in vitro situation, BrdU apparently did not influence endogenous proliferation of NPCs or neurogenesis in concentrations that are typically used for labeling of neural progenitors in vivo. Our results reveal so far uncharacterized effects of BrdU on adult NPCs. We conclude that, because of its ubiquitous use in stem cell biology, any potential effect of BrdU of NPCs has to be scrutinized prior to interpretation of data.


Subject(s)
Bromodeoxyuridine/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Animals , Cell Adhesion/drug effects , Cell Aggregation/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Male , Matrix Metalloproteinase 2/metabolism , Neural Stem Cells/enzymology , Neural Stem Cells/transplantation , Phenotype , Rats , Rats, Inbred F344 , Staining and Labeling , Stem Cell Transplantation
4.
Stem Cell Rev Rep ; 7(4): 815-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21431886

ABSTRACT

It is commonly accepted that adult neurogenesis and gliogenesis follow the same principles through the mammalian class. However, it has been reported that neurogenesis might differ between species, even from the same order, like in rodents. Currently, it is not known if neural stem/progenitor cells (NSPCs) from various species differ in their cell identity and potential. NSPCs can be expanded ex vivo as neurospheres (NSph), a model widely used to study neurogenesis in vitro. Here we demonstrate that rat (r) and mouse (m) NSph display different cell identities, differentiation fate, electrophysiological function and tumorigenic potential. Adult rNSph consist mainly of oligodendroglial progenitors (OPCs), which after repeated passaging proliferate independent of mitogens, whereas adult mNSph show astroglial precursor-like characteristics and retain their mitogen dependency. Most of the cells in rNSph express OPC markers and spontaneously differentiate into oligodendrocytes after growth factor withdrawal. Electrophysiological analysis confirmed OPC characteristics. mNSph have different electrophysiological properties, they express astrocyte precursor markers and spontaneously differentiate primarily into astrocytes. Furthermore, rNSph have the potential to differentiate into oligodendrocytes and astrocytes, whereas mNSph are restricted to the astrocytic lineage. The phenotypic differences between rNSph and mNSph were not due to a distinct response to species specific derived growth factors and are probably not caused by autocrine mechanisms. Our findings suggest that NSph derived from adult rat and mouse brains display different cell identities. Thus, results urge for caution when data derived from NSph are extrapolated to other species or to the in vivo situation, especially when aimed towards the clinical use of human NSph.


Subject(s)
Cell Differentiation , Neural Stem Cells/cytology , Oligodendroglia/cytology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers/metabolism , Cell Adhesion , Cell Aggregation , Cell Count , Cell Culture Techniques , Cell Lineage , Cell Proliferation , Cells, Cultured , Chromosomes, Mammalian/genetics , Culture Media, Conditioned , Electrophysiology , Female , Membrane Potentials , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Patch-Clamp Techniques , Rats , Rats, Inbred F344 , Species Specificity
5.
J Neuropathol Exp Neurol ; 69(7): 717-28, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20535034

ABSTRACT

Cellular proliferation, differentiation, integration, and survival within the adult neural stem cell niche are altered under pathological conditions, but the molecular cues regulating the biology of this niche are mostly unknown. We examined the hippocampal neural stem cell niche in a transgenic rat model of Huntington disease. In this model, progressive cognitive deficits develop at the age of 9 months, suggesting possible hippocampal dysfunction. We found a disease-associated progressive decline in hippocampal progenitor cell proliferation accompanied by an expansion of the pool of 5-bromo-2-deoxyuridine label-retaining Sox-2-positive quiescent stem cells in the transgenic animals. Increments in quiescent stem cells occurred at the expense of cAMP-responsive element-binding protein-mediated neuronal differentiation and survival. Because elevated levels of transforming growth factor-beta1 (TGF-beta1) impair neural progenitor proliferation, we investigated hippocampal TGF-beta signaling and determined that TGF-beta1 induces the neural progenitors to exit the cell cycle. Although phospho-Smad2, an effector of TGF-beta signaling, is normally absent in subgranular stem cells, it accumulated progressively in Sox2/glial fibrillary acidic protein-expressing cells of the subgranular zone in the transgenic rats. These results indicate that alterations in neurogenesis in transgenic Huntington disease rats occur in successive phases that are associated with increasing TGF-beta signaling. Thus, TGF-beta1 signaling seems to be a crucial modulator of neurogenesis in Huntington disease and may represent a target for future therapy.


Subject(s)
Hippocampus/pathology , Huntington Disease/pathology , Neurogenesis/genetics , Signal Transduction/physiology , Stem Cell Niche/physiopathology , Transforming Growth Factor beta/metabolism , Age Factors , Animals , Animals, Genetically Modified , Bromodeoxyuridine/metabolism , CREB-Binding Protein/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Doublecortin Domain Proteins , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Huntingtin Protein , Male , Microtubule-Associated Proteins/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neuropeptides/metabolism , Nuclear Proteins , Proliferating Cell Nuclear Antigen/metabolism , Rats , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad2 Protein/metabolism , Stem Cell Niche/drug effects , Transforming Growth Factor beta/pharmacology , Trinucleotide Repeat Expansion/genetics
6.
Cell Physiol Biochem ; 24(5-6): 397-406, 2009.
Article in English | MEDLINE | ID: mdl-19910680

ABSTRACT

We recently demonstrated that prolactin (PRL) prevents chronic stress-induced inhibition of adult hippocampal neurogenesis. It remained unsettled, however, whether PRL is acting directly on neural stem and progenitors cells (NPCs) or if neurogenesis is affected by an indirect mechanism, for example through the extensively described effects of PRL on the HPA axis. To address this point, we used neurosphere cultures derived from the adult rat hippocampus as an in vitro model for NPCs. Dexamethasone (DEX) was applied to stress the NPCs, and proliferation, survival and differentiation of cells were examined. DEX markedly inhibited proliferation of NPCs and cells entered the G(0) phase of cell cycle. Moreover, DEX reduced NPC survival and repressed astroglial differentiation, which is normally induced by serum or bone morphogenetic protein application. Even though we could demonstrate that NPCs express the PRL receptor and ERK1/2 signaling is induced by PRL, we did not observe any effect of PRL on NPCs proliferation, differentiation or survival, neither in the presence nor during absence of DEX. In summary, our results indicate that PRL action on NPCs and neurogenesis in vivo occurs via an indirect mechanism.


Subject(s)
Glucocorticoids/pharmacology , MAP Kinase Signaling System/drug effects , Neurogenesis/drug effects , Prolactin/pharmacology , Stem Cells/cytology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dexamethasone/pharmacology , Female , Hippocampus/cytology , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Receptors, Prolactin/metabolism , Resting Phase, Cell Cycle
7.
Stem Cells ; 27(2): 420-3, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18988707

ABSTRACT

In vivo visualization of endogenous neural progenitor cells (NPCs) is crucial to advance stem cell research and will be essential to ensure the safety and efficacy of neurogenesis-based therapies. Magnetic resonance spectroscopic imaging (i.e., spatially resolved spectroscopy in vivo) is a highly promising technique by which to investigate endogenous neurogenesis noninvasively. A distinct feature in nuclear magnetic resonance spectra (i.e., a lipid signal at 1.28 ppm) was recently attributed specifically to NPCs in vitro and to neurogenic regions in vivo. Here, we demonstrate that although this 1.28-ppm biomarker is present in NPC cultures, it is not specific for the latter. The 1.28-ppm marker was also evident in mesenchymal stem cells and in non-stem cell lines. Moreover, it was absent in freshly isolated NPCs but appeared under conditions favoring growth arrest or apoptosis; it is initiated by induction of apoptosis and correlates with the appearance of mobile lipid droplets. Thus, although the 1.28-ppm signal cannot be considered as a specific biomarker for NPCs, it might still serve as a sensor for processes that are tightly associated with neurogenesis and NPCs in vivo, such as apoptosis or stem cell quiescence. However, this requires further experimental evidence. The present work clearly urges the identification of additional biomarkers for NPCs and for neurogenesis.


Subject(s)
Biomarkers/analysis , Magnetic Resonance Imaging , Neurogenesis/physiology , Neurons/metabolism , Stem Cells/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Mice , Neurons/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...