Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Med Sci Monit ; 28: e933830, 2022 Mar 07.
Article in English | MEDLINE | ID: mdl-35250022

ABSTRACT

BACKGROUND Ischemic cerebrovascular disease leads to the activation and differentiation of neural stem cells (NSCs) into mature neurons and glia cells to repair nerve damage. Astragalus flavone (ASF) has shown its potential role in proliferation and differentiation into dopamine neurons of NSCs. MATERIAL AND METHODS Cerebral infarction models were constructed to determine the effects of ASF on NSCs in vivo and in vitro. RESULTS ASF therapy had the ability to reduce the neurologic function scores and the cerebral infarction volume of the cerebral infarction model. Moreover, ASF was able to increase BrdU-positive cells and promote the expression of Nestin, ß-Tubulin III, and O4, while decreasing the expression of GFAP. qRT-PCR and western blot assays showed ASF promoted the expression of Mash1, Math1, and Ngn2 mRNA and protein in cerebral infarction rats. Meanwhile, ASF (20 µg/ml) was able to increase EdU-positive cells and promote the expression of Nestin, ß-Tubulin III, and O4 of NSCs at day14 in vitro. In normoxia, ASF obviously promoted the expression of Mash1, Ngn1, and Ngn2 mRNA and proteins, but in hypoxia, ASF promoted the expression of Notch1 and Math1 mRNA and proteins and inhibited the expression of Ngn1 and Ngn2 mRNA and proteins. CONCLUSIONS ASF therapy can improve the neurologic functions and reduce the cerebral infarction volume in a cerebral infarction model. Moreover, ASF promoted the proliferation of NSCs and induced differentiation into neurons and oligodendrocytes, which might be involved in regulating factors in Notch signaling.


Subject(s)
Cerebral Infarction/pathology , Flavones/pharmacology , Neural Stem Cells/classification , Neurogenesis/drug effects , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cell Differentiation/drug effects , Cell Proliferation , Cells, Cultured , Cerebral Infarction/drug therapy , Disease Models, Animal , Male , Neural Stem Cells/drug effects , Rats , Rats, Wistar , Signal Transduction
2.
Biochem Soc Trans ; 49(5): 1997-2006, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34397081

ABSTRACT

The mammalian neocortex is the seat of higher cognitive functions, such as thinking and language in human. A hallmark of the neocortex are the cortical neurons, which are generated from divisions of neural progenitor cells (NPCs) during development, and which constitute a key feature of the well-organized layered structure of the neocortex. Proper formation of neocortex structure requires an orchestrated cellular behavior of different cortical NPCs during development, especially during the process of cortical neurogenesis. Here, we review the great diversity of NPCs and their contribution to the development of the neocortex. First, we review the categorization of NPCs into different classes and types based on their cell biological features, and discuss recent advances in characterizing marker expression and cell polarity features in the different types of NPCs. Second, we review the different modes of cell divisions that NPCs undergo and discuss the importance of the balance between proliferation and differentiation of NPCs in neocortical development. Third, we review the different proliferative capacities among different NPC types and among the same type of NPC in different mammalian species. Dissecting the differences between NPC types and differences among mammalian species is beneficial to further understand the development and the evolutionary expansion of the neocortex and may open up new therapeutic avenues for neurodevelopmental and psychiatric disorders.


Subject(s)
Neocortex/cytology , Neocortex/growth & development , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Biological Evolution , Cell Division/physiology , Cell Polarity/physiology , Humans , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction/physiology
3.
Elife ; 92020 08 07.
Article in English | MEDLINE | ID: mdl-32762844

ABSTRACT

Different subtypes of interneurons, destined for the olfactory bulb, are continuously generated by neural stem cells located in the ventricular and subventricular zones along the lateral forebrain ventricles of mice. Neuronal identity in the olfactory bulb depends on the existence of defined microdomains of pre-determined neural stem cells along the ventricle walls. The molecular mechanisms underlying positional identity of these neural stem cells are poorly understood. Here, we show that the transcription factor Vax1 controls the production of two specific neuronal subtypes. First, it is directly necessary to generate Calbindin expressing interneurons from ventro-lateral progenitors. Second, it represses the generation of dopaminergic neurons by dorsolateral progenitors through inhibition of Pax6 expression. We present data indicating that this repression occurs, at least in part, via activation of microRNA miR-7.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis , Neuropeptides/metabolism , Olfactory Bulb/physiology , PAX6 Transcription Factor/metabolism , Animals , Calbindins/genetics , Cell Differentiation , Female , Homeodomain Proteins/genetics , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neural Stem Cells/classification , Neuropeptides/genetics , PAX6 Transcription Factor/genetics
4.
Anal Bioanal Chem ; 411(21): 5423-5436, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31161326

ABSTRACT

It is necessary to characterize and classify neural stem cells (NSCs) and differentiated cells (DCs) for potential use of NSC to treat neurodegenerative diseases. We therefore performed an analysis of NSCs and DCs using gas chromatography mass spectrometry (GC-MS) and direct infusion mass spectrometry (DI-MS) with elaborate multivariate statistical analysis for the characterization and classification of rat NSCs and DCs. GC-MS and DI-MS detected a total of 92 metabolites and lipids in NSCs and DCs, and the levels of 72 of them differed significantly between NSCs and DCs. The optimal model for partial least squares (PLS) discriminant analysis was constructed by applying 3 and 2 PLS components with a unit-variance scaling method for classifying NSCs and DCs based on the data obtained in the GC-MS and DI-MS analyses, respectively. The obtained results from PCA and PLS-DA suggest that creatinine, lactic acid, lysine, glutamine, glycine, pyroglutamic acid, PG 18:1/20:2, PS 18:0/20:2, PI 18:0/20:3, PC 16:0/20:4, PI 16:0/20:4, and PI 18:1/20:4 were the main contributors that provided distinct characteristics of NSCs and DCs. The results of this study suggest objective and complementary criteria for the characterization and classification of NSCs and DCs for potential clinical applications. Graphical abstract.


Subject(s)
Cell Differentiation , Lipid Metabolism , Neural Stem Cells/classification , Neural Stem Cells/cytology , Animals , Cells, Cultured , Discriminant Analysis , Gas Chromatography-Mass Spectrometry/methods , Least-Squares Analysis , Mass Spectrometry/methods , Principal Component Analysis , Rats , Rats, Sprague-Dawley
5.
Epilepsy Res ; 150: 78-94, 2019 02.
Article in English | MEDLINE | ID: mdl-30735971

ABSTRACT

Neurogenesis persists throughout life in the hippocampi of all mammals, including humans. In the healthy hippocampus, relatively quiescent Type-1 neural stem cells (NSCs) can give rise to more proliferative Type-2a neural progenitor cells (NPCs), which generate neuronal-committed Type-2b NPCs that mature into Type-3 neuroblasts. Many Type-3 neuroblasts survive and mature into functionally integrated granule neurons over several weeks. In kindling models of epilepsy, neurogenesis is drastically upregulated and many new neurons form aberrant connections that could support epileptogenesis and/or seizures. We have shown that sustained vector-mediated hippocampal somatostatin (SST) expression can both block epileptogenesis and reverse seizure susceptibility in fully kindled rats. Here we test whether adeno-associated virus (AAV) vector-mediated sustained SST expression modulates hippocampal neurogenesis and microglial activation in fully kindled rats. We found significantly more dividing Type-1 NSCs and a corresponding increased number of surviving new neurons in the hippocampi of kindled versus sham-kindled rats. Increased numbers of activated microglia were found in the granule cell layer and hilus of kindled rats at both time points. After intrahippocampal injection with either eGFP or SST-eGFP vector, we found similar numbers of dividing Type-1 NSCs and -2 NPCs and surviving BrdU+ neurons and glia in the hippocampi of kindled rats. Upon observed variability in responses to SST-eGFP (2/4 rats exhibited Grade 0 seizures in the test session), we conducted an additional experiment. We found significantly fewer dividing Type-1 NSCs in the hippocampi of SST-eGFP vector-treated responder rats (5/13 rats) relative to SST-eGFP vector-treated non-responders and eGFP vector-treated controls that exhibited high-grade seizures on the test session. The number of activated microglia was upregulated in the GCL and hilus of kindled rats, regardless of vector treatment. These data support the hypothesis that sustained SST expression exerts antiepileptic effects potentially through normalization of neurogenesis and suggests that abnormally high proliferating Type-1 NSC numbers may be a cellular mechanism of epilepsy.


Subject(s)
Hippocampus/metabolism , Hippocampus/pathology , Kindling, Neurologic/pathology , Neural Stem Cells/physiology , Neurogenesis/physiology , Somatostatin/metabolism , Amygdala/metabolism , Animals , Antigens/metabolism , Bromodeoxyuridine/metabolism , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cell Count , Disease Models, Animal , Electric Stimulation/adverse effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Microfilament Proteins/metabolism , Microglia/pathology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/classification , Neurons/metabolism , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Somatostatin/genetics , Transduction, Genetic , Urea/analogs & derivatives , Urea/metabolism
6.
J Cell Biochem ; 119(4): 3394-3403, 2018 04.
Article in English | MEDLINE | ID: mdl-29130544

ABSTRACT

Adult neural stem cells (NSCs) are a group of multi-potent, self-renewing progenitor cells that contribute to the generation of new neurons and oligodendrocytes. Three subtypes of NSCs can be isolated based on the stages of the NSC lineage, including quiescent neural stem cells (qNSCs), activated neural stem cells (aNSCs) and neural progenitor cells (NPCs). Although it is widely accepted that these three groups of NSCs play different roles in the development of the nervous system, their molecular signatures are poorly understood. In this study, we applied the Monte-Carlo Feature Selection (MCFS) method to identify the gene expression signatures, which can yield a Matthews correlation coefficient (MCC) value of 0.918 with a support vector machine evaluated by ten-fold cross-validation. In addition, some classification rules yielded by the MCFS program for distinguishing above three subtypes were reported. Our results not only demonstrate a high classification capacity and subtype-specific gene expression patterns but also quantitatively reflect the pattern of the gene expression levels across the NSC lineage, providing insight into deciphering the molecular basis of NSC differentiation.


Subject(s)
Astrocytes/cytology , Gene Expression Profiling/methods , Gene Regulatory Networks , Neural Stem Cells/classification , Algorithms , Cell Lineage , Cells, Cultured , Humans , Monte Carlo Method , Support Vector Machine
7.
Development ; 144(23): 4313-4321, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29183942

ABSTRACT

Cellular reprogramming is a dedifferentiation process during which cells continuously undergo phenotypical remodeling. Although the genetic and biochemical details of this remodeling are fairly well understood, little is known about the change in cell mechanical properties during the process. In this study, we investigated changes in the mechanical phenotype of murine fetal neural progenitor cells (fNPCs) during reprogramming to induced pluripotent stem cells (iPSCs). We find that fNPCs become progressively stiffer en route to pluripotency, and that this stiffening is mirrored by iPSCs becoming more compliant during differentiation towards the neural lineage. Furthermore, we show that the mechanical phenotype of iPSCs is comparable with that of embryonic stem cells. These results suggest that mechanical properties of cells are inherent to their developmental stage. They also reveal that pluripotent cells can differentiate towards a more compliant phenotype, which challenges the view that pluripotent stem cells are less stiff than any cells more advanced developmentally. Finally, our study indicates that the cell mechanical phenotype might be utilized as an inherent biophysical marker of pluripotent stem cells.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Animals , Biomarkers/metabolism , Biomechanical Phenomena , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/physiology , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/classification , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Lewis X Antigen/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/classification , Phenotype , Single-Cell Analysis
8.
Sci Rep ; 6: 37540, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857203

ABSTRACT

The CRISPR/Cas9 system is a rapid and customizable tool for gene editing in mammalian cells. In particular, this approach has widely opened new opportunities for genetic studies in neurological disease. Human neurons can be differentiated in vitro from hPSC (human Pluripotent Stem Cells), hNPCs (human Neural Precursor Cells) or even directly reprogrammed from fibroblasts. Here, we described a new platform which enables, rapid and efficient CRISPR/Cas9-mediated genome targeting simultaneously with three different paradigms for in vitro generation of neurons. This system was employed to inactivate two genes associated with neurological disorder (TSC2 and KCNQ2) and achieved up to 85% efficiency of gene targeting in the differentiated cells. In particular, we devised a protocol that, combining the expression of the CRISPR components with neurogenic factors, generated functional human neurons highly enriched for the desired genome modification in only 5 weeks. This new approach is easy, fast and that does not require the generation of stable isogenic clones, practice that is time consuming and for some genes not feasible.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/classification , CRISPR-Cas Systems/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Silencing , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism
9.
Cell Transplant ; 25(4): 645-64, 2016.
Article in English | MEDLINE | ID: mdl-26720923

ABSTRACT

Intracerebral cell transplantation is increasingly finding a clinical translation. However, the number of cells surviving after implantation is low (5-10%) compared to the number of cells injected. Although significant efforts have been made with regard to the investigation of apoptosis of cells after implantation, very little optimization of cell preparation and administration has been undertaken. Moreover, there is a general neglect of the biophysical aspects of cell injection. Cell transplantation can only be an efficient therapeutic approach if an optimal transfer of cells from the dish to the brain can be ensured. We therefore focused on the in vitro aspects of cell preparation of a clinical-grade human neural stem cell (NSC) line for intracerebral cell implantation. NSCs were suspended in five different vehicles: phosphate-buffered saline (PBS), Dulbecco's modified Eagle medium (DMEM), artificial cerebral spinal fluid (aCSF), HypoThermosol, and Pluronic. Suspension accuracy, consistency, and cell settling were determined for different cell volume fractions in addition to cell viability, cell membrane damage, and clumping. Maintenance of cells in suspension was evaluated while being stored for 8 h on ice, at room temperature, or physiological normothermia. Significant differences between suspension vehicles and cellular volume fractions were evident. HypoThermosol and Pluronic performed best, with PBS, aCSF, and DMEM exhibiting less consistency, especially in maintaining a suspension and preserving viability under different storage conditions. These results provide the basis to further investigate these preparation parameters during the intracerebral delivery of NSCs to provide an optimized delivery process that can ensure an efficient clinical translation.


Subject(s)
Brain , Cell Culture Techniques/methods , Neural Stem Cells/classification , Stem Cell Transplantation , Cell Line , Humans , Neural Stem Cells/metabolism
10.
Neuron ; 87(5): 999-1007, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26299474

ABSTRACT

The mammalian neocortex is composed of two major neuronal cell types with distinct origins: excitatory pyramidal neurons and inhibitory interneurons, generated in dorsal and ventral progenitor zones of the embryonic telencephalon, respectively. Thus, inhibitory neurons migrate relatively long distances to reach their destination in the developing forebrain. The role of lineage in the organization and circuitry of interneurons is still not well understood. Utilizing a combination of genetics, retroviral fate mapping, and lineage-specific retroviral barcode labeling, we find that clonally related interneurons can be widely dispersed while unrelated interneurons can be closely clustered. These data suggest that migratory mechanisms related to the clustering of interneurons occur largely independent of their clonal origin.


Subject(s)
Cell Movement/physiology , Interneurons/physiology , Nerve Net/physiology , Neural Inhibition , Neural Stem Cells/physiology , Telencephalon/cytology , Animals , Cell Lineage , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Laser Capture Microdissection , Mice , Nerve Tissue Proteins/metabolism , Neural Stem Cells/classification , Organ Culture Techniques , Telencephalon/embryology , Transduction, Genetic
11.
Cell ; 161(7): 1644-55, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091041

ABSTRACT

Adult neural stem/progenitor (B1) cells within the walls of the lateral ventricles generate different types of neurons for the olfactory bulb (OB). The location of B1 cells determines the types of OB neurons they generate. Here we show that the majority of mouse B1 cell precursors are produced between embryonic days (E) 13.5 and 15.5 and remain largely quiescent until they become reactivated postnatally. Using a retroviral library carrying over 100,000 genetic tags, we found that B1 cells share a common progenitor with embryonic cells of the cortex, striatum, and septum, but this lineage relationship is lost before E15.5. The regional specification of B1 cells is evident as early as E11.5 and is spatially linked to the production of neurons that populate different areas of the forebrain. This study reveals an early embryonic regional specification of postnatal neural stem cells and the lineage relationship between them and embryonic progenitor cells.


Subject(s)
Adult Stem Cells/cytology , Cell Lineage , Embryo, Mammalian/cytology , Neural Stem Cells/cytology , Olfactory Bulb/cytology , Adult Stem Cells/classification , Animals , Mice , Neural Stem Cells/classification , Prosencephalon/cytology
12.
Brain Res ; 1628(Pt B): 327-342, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-25931262

ABSTRACT

Neural stem cells (NSCs) critical for the continued production of new neurons and glia are sequestered in distinct areas of the brain called stem cell niches. Until recently, only two forebrain sites, the subventricular zone (SVZ) of the anterolateral ventricle and the subgranular zone (SGZ) of the hippocampus, have been recognized adult stem cell niches (Alvarez-Buylla and Lim, 2004; Doetsch et al., 1999a, 1999b; Doetsch, 2003a, 2003b; Lie et al., 2004; Ming and Song, 2005). Nonetheless, the last decade has been witness to a growing literature suggesting that in fact the adult brain contains stem cell niches along the entire extent of the ventricular system. These niches are capable of widespread neurogenesis and gliogenesis, particularly after injury (Barnabé-Heider et al., 2010; Carlén et al., 2009; Decimo et al., 2012; Lin et al., 2015; Lindvall and Kokaia, 2008; Robins et al., 2013) or other inductive stimuli (Bennett et al., 2009; Cunningham et al., 2012; Decimo et al., 2011; Kokoeva et al., 2007, 2005; Lee et al., 2012a, 2012b; Migaud et al., 2010; Pencea et al., 2001b; Sanin et al., 2013; Suh et al., 2007; Sundholm-Peters et al., 2004; Xu et al., 2005; Zhang et al., 2007). This review focuses on the role of these novel and classic brain niches in maintaining adult neurogenesis and gliogenesis in response to normal physiological and injury-related pathological cues. This article is part of a Special Issue entitled SI: Neuroprotection.


Subject(s)
Brain/cytology , Homeostasis/physiology , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Humans , Neural Stem Cells/classification , Neurogenesis/physiology
13.
J Neurosci ; 35(15): 6142-52, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25878286

ABSTRACT

Several neural precursor populations contemporaneously generate neurons in the developing neocortex. Specifically, radial glial stem cells of the dorsal telencephalon divide asymmetrically to produce excitatory neurons, but also indirectly to produce neurons via three types of intermediate progenitor cells. Why so many precursor types are needed to produce neurons has not been established; whether different intermediate progenitor cells merely expand the output of radial glia or instead generate distinct types of neurons is unknown. Here we use a novel genetic fate mapping technique to simultaneously track multiple precursor streams in the developing mouse brain and show that layer 2 and 3 pyramidal neurons exhibit distinctive electrophysiological and structural properties depending upon their precursor cell type of origin. These data indicate that individual precursor subclasses synchronously produce functionally different neurons, even within the same lamina, and identify a primary mechanism leading to cortical neuronal diversity.


Subject(s)
Cell Lineage/physiology , Neocortex/cytology , Nerve Net/physiology , Neural Stem Cells/classification , Neural Stem Cells/physiology , Pyramidal Cells/physiology , Animals , Electroporation , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Luminescent Proteins/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Membrane Potentials/physiology , Mice , Mice, Transgenic , Neocortex/embryology , Patch-Clamp Techniques , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
14.
Neural Dev ; 10: 8, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25889070

ABSTRACT

BACKGROUND: The rostral patterning center (RPC) secretes multiple fibroblast growth factors (Fgfs) essential for telencephalon growth and patterning. Fgf expression patterns suggest that they mark functionally distinct RPC subdomains. We generated Fgf8(CreER) and Fgf17(CreER) mice and used them to analyze the lineages of Fgf8- versus Fgf17-expressing RPC cells. RESULTS: Both lineages contributed to medial structures of the rostroventral telencephalon structures including the septum and medial prefrontral cortex. In addition, RPC-derived progenitors were observed in other regions of the early telencephalic neuroepithelium and generated neurons in the olfactory bulb, neocortex, and basal ganglia. Surprisingly, Fgf8(+) RPC progenitors generated the majority of basal ganglia cholinergic neurons. Compared to the Fgf8 lineage, the Fgf17 lineage was more restricted in its early dispersion and its contributions to the telencephalon. Mutant studies suggested that Fgf8 and Fgf17 restrict spread of RPC progenitor subpopulations. CONCLUSIONS: We identified the RPC as an important source of progenitors that contribute broadly to the telencephalon and found that two molecularly distinct progenitor subtypes in the RPC make different contributions to the developing forebrain.


Subject(s)
Body Patterning/physiology , Fibroblast Growth Factor 8/physiology , Fibroblast Growth Factors/physiology , Neural Stem Cells/cytology , Telencephalon/cytology , Animals , Basal Ganglia/cytology , Basal Ganglia/embryology , Cell Lineage , Cholinergic Neurons/cytology , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Genes, Synthetic , Gestational Age , Mice , Neural Stem Cells/classification , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Prosencephalon/cytology , Prosencephalon/embryology , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/physiology , Telencephalon/embryology
15.
Cell Rep ; 9(6): 2139-51, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25497090

ABSTRACT

Evolutionary elaboration of tissues starts with changes in the genome and location of the stem cells. For example, GABAergic interneurons of the mammalian neocortex are generated in the ventral telencephalon and migrate tangentially to the neocortex, in contrast to the projection neurons originating in the ventricular/subventricular zone (VZ/SVZ) of the dorsal telencephalon. In human and nonhuman primates, evidence suggests that an additional subset of neocortical GABAergic interneurons is generated in the cortical VZ and a proliferative niche, the outer SVZ. The origin, magnitude, and significance of this species-specific difference are not known. We use a battery of assays applicable to the human, monkey, and mouse organotypic cultures and supravital tissue to identify neuronal progenitors in the cortical VZ/SVZ niche that produce a subset of GABAergic interneurons. Our findings suggest that these progenitors constitute an evolutionary novelty contributing to the elaboration of higher cognitive functions in primates.


Subject(s)
Cellular Microenvironment , Cerebral Cortex/cytology , GABAergic Neurons/cytology , Interneurons/cytology , Neural Stem Cells/cytology , Animals , Cells, Cultured , Cerebral Cortex/embryology , GABAergic Neurons/classification , GABAergic Neurons/metabolism , Humans , Interneurons/classification , Interneurons/metabolism , Macaca , Mice , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Neural Dev ; 9: 23, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25352248

ABSTRACT

BACKGROUND: Neural stem cell (NSC) differentiation is a complex multistep process that persists in specific regions of the postnatal forebrain and requires tight regulation throughout life. The transcriptional control of NSC proliferation and specification involves Class II (proneural) and Class V (Id1-4) basic helix-loop-helix (bHLH) proteins. In this study, we analyzed the pattern of expression of their dimerization partners, Class I bHLH proteins (E-proteins), and explored their putative role in orchestrating postnatal subventricular zone (SVZ) neurogenesis. RESULTS: Overexpression of a dominant-negative form of the E-protein E47 (dnE47) confirmed a crucial role for bHLH transcriptional networks in postnatal neurogenesis by dramatically blocking SVZ NSC differentiation. In situ hybridization was used in combination with RT-qPCR to measure and compare the level of expression of E-protein transcripts (E2-2, E2A, and HEB) in the neonatal and adult SVZ as well as in magnetic affinity cell sorted progenitor cells and neuroblasts. Our results evidence that E-protein transcripts, in particular E2-2 and E2A, are enriched in the postnatal SVZ with expression levels increasing as cells engage towards neuronal differentiation. To investigate the role of E-proteins in orchestrating lineage progression, both in vitro and in vivo gain-of-function and loss-of-function experiments were performed for individual E-proteins. Overexpression of E2-2 and E2A promoted SVZ neurogenesis by enhancing not only radial glial cell differentiation but also cell cycle exit of their progeny. Conversely, knock-down by shRNA electroporation resulted in opposite effects. Manipulation of E-proteins and/or Ascl1 in SVZ NSC cultures indicated that those effects were Ascl1 dependent, although they could not solely be attributed to an Ascl1-induced switch from promoting cell proliferation to triggering cell cycle arrest and differentiation. CONCLUSIONS: In contrast to former concepts, suggesting ubiquitous expression and subsidiary function for E-proteins to foster postnatal neurogenesis, this work unveils E-proteins as being active players in the orchestration of postnatal SVZ neurogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Neural Stem Cells/classification , Neural Stem Cells/physiology , Prosencephalon/cytology , Prosencephalon/growth & development , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle/genetics , Cell Differentiation/physiology , Cell Movement , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ki-67 Antigen/metabolism , Mice , Nerve Tissue Proteins , Neurogenesis , Transfection
17.
Proc Natl Acad Sci U S A ; 111(34): 12438-43, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114218

ABSTRACT

The apical domain of embryonic (radial glia) and adult (B1 cells) neural stem cells (NSCs) contains a primary cilium. This organelle has been suggested to function as an antenna for the detection of morphogens or growth factors. In particular, primary cilia are essential for Hedgehog (Hh) signaling, which plays key roles in brain development. Their unique location facing the ventricular lumen suggests that primary cilia in NSCs could play an important role in reception of signals within the cerebrospinal fluid. Surprisingly, ablation of primary cilia using conditional alleles for genes essential for intraflagellar transport [kinesin family member 3A (Kif3a) and intraflagellar transport 88 (Ift88)] and Cre drivers that are activated at early [Nestin; embryonic day 10.5 (E10.5)] and late [human glial fibrillary acidic protein (hGFAP); E13.5] stages of mouse neural development resulted in no apparent developmental defects. Neurogenesis in the ventricular-subventricular zone (V-SVZ) shortly after birth was also largely unaffected, except for a restricted ventral domain previously known to be regulated by Hh signaling. However, Kif3a and Ift88 genetic ablation also disrupts ependymal cilia, resulting in hydrocephalus by postnatal day 4. To directly study the role of B1 cells' primary cilia without the confounding effects of hydrocephalus, we stereotaxically targeted elimination of Kif3a from a subpopulation of radial glia, which resulted in ablation of primary cilia in a subset of B1 cells. Again, this experiment resulted in decreased neurogenesis only in the ventral V-SVZ. Primary cilia ablation led to disruption of Hh signaling in this subdomain. We conclude that primary cilia are required in a specific Hh-regulated subregion of the postnatal V-SVZ.


Subject(s)
Cilia/physiology , Neural Stem Cells/classification , Neural Stem Cells/ultrastructure , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Brain/metabolism , Cell Proliferation , Embryonic Stem Cells/classification , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Female , Gene Knockdown Techniques , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hedgehog Proteins/physiology , Humans , Kinesins/antagonists & inhibitors , Kinesins/genetics , Kinesins/metabolism , Mice , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Pregnancy , Signal Transduction , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
Annu Rev Cell Dev Biol ; 30: 465-502, 2014.
Article in English | MEDLINE | ID: mdl-25000993

ABSTRACT

Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.


Subject(s)
Neocortex/growth & development , Neurogenesis/physiology , Animals , Asymmetric Cell Division , Cell Compartmentation , Cell Lineage , Cell Membrane/physiology , Cell Nucleus/physiology , Cell Polarity , Cerebrospinal Fluid/physiology , Humans , Intercellular Junctions/physiology , Lateral Ventricles/embryology , Membrane Lipids/metabolism , Microglia/physiology , Mitosis , Neocortex/cytology , Neocortex/embryology , Neural Stem Cells/classification , Neural Stem Cells/physiology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Neurons/physiology , Organelles/physiology , Species Specificity
19.
Development ; 141(6): 1392-403, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24595292

ABSTRACT

In the ventral spinal cord, generation of neuronal and glial cell subtypes is controlled by Sonic hedgehog (Shh). This morphogen contributes to cell diversity by regulating spatial and temporal sequences of gene expression during development. Here, we report that establishing Shh source cells is not sufficient to induce the high-threshold response required to specify sequential generation of ventral interneurons and oligodendroglial cells at the right time and place in zebrafish. Instead, we show that Shh-producing cells must repeatedly upregulate the secreted enzyme Sulfatase1 (Sulf1) at two critical time points of development to reach their full inductive capacity. We provide evidence that Sulf1 triggers Shh signaling activity to establish and, later on, modify the spatial arrangement of gene expression in ventral neural progenitors. We further present arguments in favor of Sulf1 controlling Shh temporal activity by stimulating production of active forms of Shh from its source. Our work, by pointing out the key role of Sulf1 in regulating Shh-dependent neural cell diversity, highlights a novel level of regulation, which involves temporal evolution of Shh source properties.


Subject(s)
Hedgehog Proteins/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Sulfatases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Mice , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Signal Transduction , Spinal Cord/cytology , Sulfatases/genetics , Sulfotransferases/genetics , Sulfotransferases/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
20.
Development ; 141(6): 1260-71, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24553291

ABSTRACT

Axon guidance by commissural neurons has been well documented, providing us with a molecular logic of how midline crossing is achieved during development. Despite these advances, knowledge of the intrinsic genetic programs is still limited and it remains obscure whether the expression of a single transcription factor is sufficient to activate transcriptional programs that ultimately enable midline crossing. Here, we show in the mouse that the homeodomain transcription factor Dbx1 is expressed by a subset of progenitor cells that give rise to commissural neurons in the dorsal midbrain. Gain- and loss-of-function analyses indicate that the expression of Dbx1 alone is sufficient and necessary to trigger midline crossing in vivo. We also show that Robo3 controls midline crossing as a crucial downstream effector of the Dbx1-activated molecular programs. Furthermore, Dbx1 suppresses the expression of the transcriptional program for ipsilateral neuron differentiation in parallel. These results suggest that a single transcription factor, Dbx1, has an essential function in assigning midline-crossing identity, thereby contributing crucially to the establishment of the wiring laterality in the developing nervous system.


Subject(s)
Homeodomain Proteins/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Axons/metabolism , Body Patterning/genetics , Body Patterning/physiology , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesencephalon/cytology , Mice , Mice, Inbred ICR , Mice, Transgenic , Nerve Tissue Proteins/antagonists & inhibitors , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Pregnancy , Receptors, Cell Surface , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL