Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 13(4): 403-18, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23933087

ABSTRACT

Numerous transcriptional regulators of neurogenesis have been identified in the developing and adult brain, but how neurogenic fate is programmed at the epigenetic level remains poorly defined. Here, we report that the transcription factor Pax6 directly interacts with the Brg1-containing BAF complex in adult neural progenitors. Deletion of either Brg1 or Pax6 in the subependymal zone (SEZ) causes the progeny of adult neural stem cells to convert to the ependymal lineage within the SEZ while migrating neuroblasts convert to different glial lineages en route to or in the olfactory bulb (OB). Genome-wide analyses reveal that the majority of genes downregulated in the Brg1 null SEZ and OB contain Pax6 binding sites and are also downregulated in Pax6 null SEZ and OB. Downstream of the Pax6-BAF complex, we find that Sox11, Nfib, and Pou3f4 form a transcriptional cross-regulatory network that drives neurogenesis and can convert postnatal glia into neurons. Taken together, elements of our work identify a tripartite effector network activated by Pax6-BAF that programs neuronal fate.


Subject(s)
Adult Stem Cells/metabolism , Eye Proteins/metabolism , Gene Regulatory Networks , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Adult Stem Cells/cytology , Animals , Down-Regulation , Mice , Neural Stem Cells/cytology , PAX6 Transcription Factor , Transcription Factors/genetics
2.
J Neurosci Methods ; 209(2): 420-7, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22750652

ABSTRACT

Cells of the central nervous system are notoriously difficult to transfect. This is not only true for neurons and glial cells but also for dividing neural stem and progenitor cells (NSCs). About ten years ago a major advance was provided by introduction of the nucleofection technology that allowed for transfection of approximately half of the exposed NSCs. However, limitations were encountered with the need for large numbers of NSCs for a single transfection and compromised survival rates with typically only one-third of the cells surviving the pulse conditions. Here, we report the establishment of a pulse protocol that targets NSCs with high efficiency and twofold higher NSC survival rates using the 4D Nucleofector device. We demonstrate that the established protocol not only provides a clear and significant improvement over existing protocols with transfection rates above 80% and two-thirds of the NSCs surviving for at least 48h, but also their unaltered differentiation along neuronal and glial lineages. This improved protocol for the transfection of sensitive mouse central nervous system derived cells will provide an important step forward for studies of gene function by overexpression or knock-down of genes in cultured NSCs.


Subject(s)
Adult Stem Cells/metabolism , Electroporation/methods , Embryonic Stem Cells/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nestin
3.
Mol Cell Neurosci ; 48(3): 195-204, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820515

ABSTRACT

Y-P30, the 30 amino acid N-terminal peptide of the dermcidin gene, has been found to promote neuronal survival and differentiation. Its early presence in development and import to the fetal brain led to the hypothesis that Y-P30 has an influence on proliferation, differentiation and migration. Neurospheres derived from neural stem cells isolated from E13 mouse cortex and striatal ganglionic eminences were treated with Y-P30, however, the proportion of progenitors, neurons and astrocytes generated in differentiation assays was not altered. A short Y-P30 treatment of undifferentiated striatal and cortical neurospheres failed to alter the proportion of BrdU-positive cells. A longer treatment reduced the percentage of BrdU-positive cells and GABA-immunoreactive neurons only in striatal spheres. The presence of Y-P30 enhanced migration of T24 human bladder carcinoma cells in a wound-healing assay in vitro. Further, Y-P30 enhanced migration of T24 cells, rat primary cortical astrocytes and PC12 cells in chemotactic Boyden chamber assays. Together, these findings suggest that a major function of Y-P30 is to promote migration of neural and non-neural cell types.


Subject(s)
Cell Movement/drug effects , Neurons/drug effects , Peptides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Tissue Eng Part A ; 17(3-4): 475-86, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20819000

ABSTRACT

The best available treatment of peripheral nerve lesions involves transplantation of an autologous nerve. This approach, however, entails sensory deficits at the donor site and requires additional surgery. Such limitations have motivated the search for a bioengineering solution to design artificial implants. For this purpose we are producing orientated biodegradable microfibers of poly(ε-caprolactone) (PCL) with electrospinning. The present study describes the functionalization of these electrospun fibers with biologically active peptides to produce guidance structures for Schwann cell migration and axonal regeneration. For the chemical modification PCL was blended with star-shaped NCO-poly(ethylene glycol)-stat-poly(propylene glycol) (PCL/sPEG) as a covalent linker for the peptide GRGDS, derived from extracellular matrix proteins. To test biological functions of electrospun fibers, Schwann cell migration and axonal growth from dorsal root ganglia explants were investigated with time lapse video microscopy. Migrating Schwann cells as well as growing sensory axons closely followed the electrospun fibers with occasional leaps between adjacent fibers. Cell migration was characterized by frequent changes in velocity and direction reversals. Comparison of substrates showed that functionalized fibers caused more Schwann cells to move out of the explants, supported faster cell migration and axonal growth than the nonfunctional fibers. Using inhibitors of intracellular signaling kinases, we found that these biological effects required activation of the phosphatidyl inositol-3-kinase pathway. Since sPEG-containing fibers also showed low levels of nonspecific protein adsorption, which is desirable in the context of artificial implant design, the peptide modification of fibers appears to provide good substrates for nerve repair.


Subject(s)
Axons/physiology , Ganglia, Spinal/physiology , Guided Tissue Regeneration/instrumentation , Nerve Regeneration/physiology , Oligopeptides/chemistry , Polyesters/chemistry , Schwann Cells/physiology , Animals , Axons/ultrastructure , Biocompatible Materials/chemical synthesis , Cells, Cultured , Chick Embryo , Electrochemistry/methods , Ganglia, Spinal/cytology , Prosthesis Design , Rotation , Schwann Cells/cytology
5.
J Neurosci ; 30(37): 12310-22, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844127

ABSTRACT

Here, we present mechanisms for the inhibition of oligodendendrocyte precursor cell (OPC) differentiation, a biological function of neural extracellular matrix (ECM). The differentiation of oligodendrocytes is orchestrated by a complex set of stimuli. In the present study, we investigated the signaling pathway elicited by the ECM glycoprotein tenascin C (Tnc). Tnc substrates inhibit myelin basic protein (MBP) expression of cultured rat oligodendrocytes, and, conversely, we found that the emergence of MBP expression is accelerated in forebrains of Tnc-deficient mice. Mechanistically, Tnc interfered with phosphorylation of Akt, which in turn reduced MBP expression. At the cell surface, Tnc associates with lipid rafts in oligodendrocyte membranes, together with the cell adhesion molecule contactin (Cntn1) and the Src family kinase (SFK) Fyn. Depletion of Cntn1 in OPCs by small interfering RNAs (siRNAs) abolished the Tnc-dependent inhibition of oligodendrocyte differentiation, while Tnc exposure impeded the activation of the tyrosine kinase Fyn by Cntn1. Concomitant with oligodendrocyte differentiation, Tnc antagonized the expression of the signaling adaptor and RNA-binding molecule Sam68. siRNA-mediated knockdown or overexpression of Sam68 delayed or accelerated oligodendrocyte differentiation, respectively. Inhibition of oligodendrocyte differentiation with the SFK inhibitor PP2 could be rescued by Sam68 overexpression, which may indicate a regulatory role for Sam68 downstream of Fyn. Our study therefore uncovers the first signaling pathways that underlie Tnc-induced, ECM-dependent maintenance of the immature state of OPCs.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/metabolism , Growth Inhibitors/physiology , Neural Inhibition/physiology , Oligodendroglia/cytology , Signal Transduction/physiology , Stem Cells/cytology , Tenascin/physiology , Animals , Cells, Cultured , Central Nervous System/cytology , Down-Regulation/genetics , Female , Male , Mice , Mice, Knockout , Oligodendroglia/physiology , Rats , Stem Cells/physiology , Tenascin/genetics , Tenascin/metabolism
6.
Methods Enzymol ; 479: 37-71, 2010.
Article in English | MEDLINE | ID: mdl-20816159

ABSTRACT

The stem cell niche plays an important role for the maintenance and differentiation of neural stem/progenitor cells (NSPCs). It is composed of distinct cell types that influence NSCPs by the release of paracrine factors, and a specialized extracellular matrix that structures the NSPC environment. During the past years, several components of the neural stem cell (NSC) niche could be deciphered on the molecular level. One prominent constituent is the tenascin-C (Tnc) glycoprotein and its isoforms that intervene in NSPC proliferation and differentiation. Distinct chondroitin sulfate proteoglycans (CSPGs) associate with Tnc in the niche territory and we could show that these have functional connotations in the stem cell compartment in their own rights. In this chapter, we give an account of the tools and methods we developed to unravel the structures and functions of CSPGs in the NSC niche.


Subject(s)
Brain/cytology , Chondroitin Sulfate Proteoglycans/chemistry , Neural Stem Cells/chemistry , Stem Cell Niche/chemistry , Animals , Biomarkers, Tumor/chemistry , Cell Proliferation , Immunohistochemistry , Mice , Reverse Transcriptase Polymerase Chain Reaction
7.
Stem Cells ; 28(4): 775-87, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20087964

ABSTRACT

The neural stem cell niche of the embryonic and adult forebrain is rich in chondroitin sulfate glycosaminoglycans (CS-GAGs) that represent complex linear carbohydrate structures on the cell surface of neural stem/progenitor cells or in their intimate environment. We reported earlier that the removal of CS-GAGs with the bacterial enzyme chondroitinase ABC (ChABC) reduced neural stem/progenitor cell proliferation and self-renewal, whereas this treatment favored astroglia formation at the expense of neurogenesis. Here, we studied the consequences of CS-deglycanation further and revealed that CS-GAGs are selectively required for neurosphere formation, proliferation, and self-renewal of embryonic cortical neural stem/progenitor cells in response to fibroblast growth factor (FGF)-2. Consistently, the FGF-2-dependent activation of the MAPKinase in neural stem/progenitor cells was diminished after ChABC treatment, but unaltered after epidermal growth factor (EGF) stimulation. Upon EGF treatment, fewer radial glia were brain lipid-binding protein (BLBP)-positive, whereas more were glutamate aspartate transporter (GLAST)-positive after CS-GAG removal. Only in this latter situation, GLAST-positive radial glia cells extended processes that supported neuronal migration from differentiating neurospheres. CS-deglycanation also selectively increased astrocyte numbers and their migration in response to EGF. Thus, our approach revealed that CS-GAGs are essential for FGF-2-mediated proliferation and maintenance of neuron-generating neural stem/progenitor cells. Simultaneously, CS-GAGs act as a brake on the EGF-dependent maturation, migration, and gliogenesis of neural stem/progenitor cells. We conclude that neural stem/progenitor cell subpopulations reside in neurospheres that are distinguishable by their responsiveness to FGF-2 and EGF which is differentially regulated by CS-carbohydrate structures.


Subject(s)
Cell Movement , Cell Proliferation , Chondroitin Sulfates/metabolism , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cells, Cultured , Enzyme Activation , Mice , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Stem Cells/cytology
8.
Cell Tissue Res ; 338(2): 161-70, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19798513

ABSTRACT

Oligodendrocytes are the myelinating cells of the central nervous system. They differentiate from oligodendrocyte precursor cells through several intermediate states that can be followed by characteristic morphological changes and the expression of marker molecules. However, most oligodendrocyte lineage markers demarcate either the precursor or the differentiated oligodendrocyte in restricted subcellular compartments. Here, we describe a novel marker of the oligodendrocyte lineage recognised by the monoclonal antibody clone 4860. It selectively labels the surfaces of differentiated oligodendrocytes in culture and clearly differs from other oligodendrocyte markers. Importantly, the 4860 epitope highlights developing white matter tracts in rodent and avian brains and thus represents a useful and conserved feature. The 4860 epitope is not associated with protein backbones as revealed by the related 487/L5 antibody. Furthermore, the epitope disappears upon lipid extraction from cryosections or inhibition of sphingolipid synthesis in cultured oligodendrocytes. Thus, we conclude that mAb 4860 represents a novel lipid-based oligodendrocyte marker.


Subject(s)
Antigens, Differentiation/immunology , Brain/metabolism , Epitopes/immunology , Glycolipids/immunology , Oligodendroglia/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigens, Differentiation/metabolism , Axons/metabolism , Brain/cytology , Cell Differentiation , Cell Lineage , Cells, Cultured , Epitopes/metabolism , Glycolipids/metabolism , Mice , Oligodendroglia/cytology , Rats
9.
Glia ; 57(16): 1790-801, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19459213

ABSTRACT

Membrane formation and the initiation of myelin gene expression are hallmarks of the differentiation of oligodendrocytes from their precursors. Here, we compared the roles of the two related extracellular matrix (ECM) glycoproteins Tenascin C (Tnc) and Tenascin R (Tnr) in oligodendrocyte differentiation. Oligodendrocyte precursors from Tnr-deficient mice exhibited reduced differentiation, as revealed by retarded expression of myelin basic protein (MBP) in culture. This could be rescued with purified Tnr. In contrast, when we cultured oligodendrocytes on a Tnc-containing, astrocyte-derived ECM, they barely expressed MBP. This inhibition could be overcome when we used ECM from astrocytes deficient for Tnc, suggesting that Tnc inhibits differentiation. In contrast to their antagonistic effect on differentiation, both Tnc and Tnr similarly inhibited morphologic maturation. When oligodendrocytes were cultured on the purified glycoproteins, process elaboration and membrane expansion were reduced. Both Tnc and Tnr interfered with the activation of the small GTPase RhoA. Conversely, RhoA and Rac1 activation induced by cytotoxic necrotizing factor 1 (CNF1) increased the formation of myelin membranes, whereas Y27632-mediated inhibition of the Rho-cascade prevented it without, however, affecting the fraction of MBP-expressing cells. Because Tnc and Tnr play antagonistic roles for differentiation and comparably inhibit morphologic maturation, we conclude that independent molecular pathways regulate these processes.


Subject(s)
Myelin Basic Protein/metabolism , Myelin Sheath/metabolism , Tenascin/metabolism , rho GTP-Binding Proteins/metabolism , Amides/pharmacology , Animals , Astrocytes/metabolism , Blotting, Western , Brain/cytology , Brain/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Shape/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Myelin Basic Protein/genetics , Myelin Sheath/drug effects , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Tenascin/genetics
10.
Brain ; 132(Pt 8): 2252-64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19286696

ABSTRACT

CNS lesions stimulate adult neurogenic niches. Endogenous neural stem/progenitor cells represent a potential resource for CNS regeneration. Here, we investigate the response to unilateral focal laser-lesions applied to the visual cortex of juvenile rats. Within 3 days post-lesion, an ipsilateral increase of actively cycling cells was observed in cortical layer one and in the callosal white matter within the lesion penumbra. The cells expressed the neural stem/progenitor cell marker Nestin and the 473HD-epitope. Tissue prepared from the lesion area by micro-dissection generated self-renewing, multipotent neurospheres, while cells from the contralateral visual cortex did not. The newly formed neural stem/progenitor cells in the lesion zone might support neurogenesis, as suggested by the expression of Pax6 and Doublecortin, a marker of newborn neurons. We propose that focal laser-lesions may induce the emergence of stem/progenitor cells with neurogenic potential. This could underlie the beneficial effects of laser application in neurosurgery.


Subject(s)
Laser Therapy/methods , Nerve Regeneration/physiology , Stem Cells/physiology , Visual Cortex/surgery , Animals , Cell Proliferation , Cells, Cultured , Doublecortin Protein , Multipotent Stem Cells/pathology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Rats , Rats, Wistar , Visual Cortex/pathology , Visual Cortex/physiology
11.
Stem Cells ; 26(9): 2321-31, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18617690

ABSTRACT

Neural stem cells (NSCs) reside in a niche that abounds in extracellular matrix (ECM) molecules. The ECM glycoprotein tenascin-C (Tnc) that occurs in more than 25 isoforms represents a major constituent of the privileged NSC milieu. To understand its role for NSCs, the induction gene trap technology was successfully applied to mouse embryonic NSCs, and a library of more than 500 NSC lines with independent gene trap vector integrations was established. Our pilot screen identified Sam68 as a target of Tnc signaling in NSCs. The Tnc-mediated downregulation of Sam68, which we found expressed at low levels in the niche along with Tnc, was independently confirmed on the protein level. Sam68 is a multifunctional RNA-binding protein, and its potential significance for cultured NSCs was studied by overexpression. Increased Sam68 levels caused a marked reduction in NSC cell proliferation. In addition, Sam68 is a signal-dependent regulator of alternative splicing, and its overexpression selectively increased the larger Tnc isoforms, whereas a mutated phosphorylation-deficient Sam68 variant did not. This emphasizes the importance of Sam68 for NSC biology and implicates an instructive rather than a purely permissive role for Tnc in the neural stem cell niche.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Neurons/metabolism , RNA-Binding Proteins/biosynthesis , Stem Cells/metabolism , Tenascin/physiology , Adaptor Proteins, Signal Transducing/genetics , Alternative Splicing , Animals , Base Sequence , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mice , Molecular Sequence Data , Neurons/cytology , Protein Isoforms/physiology , RNA-Binding Proteins/genetics , Signal Transduction , Stem Cells/cytology
12.
Cells Tissues Organs ; 188(1-2): 170-7, 2008.
Article in English | MEDLINE | ID: mdl-18160825

ABSTRACT

The stem cell niche provides the specialized environment that is able to sustain the lifelong maintenance of stem cells in their discrete locations within organs. The niche is usually composed of several different cell types and a specialized extracellular matrix consisting of many different constituents. Additionally, a variety of growth factors are secreted into the extracellular space and contribute to the functional organization of the niche. Here, I will concentrate on the multimodular extracellular matrix glycoprotein tenascin C (Tnc) and discuss it as an exemplary molecule that is present in several stem cell niches. In spite of its intuitively suggestive presence, it has been difficult to provide functional evidence for the importance of Tnc in the context of stem cells. In the nervous system, the careful analysis of Tnc-deficient mice has revealed that the developmental program neural stem cell pass-through is delayed due to changes in growth factor responsiveness. To gain further insight, we have employed the gene trap technology in neural stem cells to identify potential Tnc target genes. This approach has surfaced 2 interesting candidates that may contribute to a better understanding of the signal(s) elicited by Tnc in neural stem/progenitor cells in the niche.


Subject(s)
Stem Cells/metabolism , Tenascin/metabolism , Animals , Neurons/cytology , Neurons/metabolism , Signal Transduction , Tenascin/chemistry
13.
Stem Cells ; 26(3): 798-809, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079434

ABSTRACT

Chondroitin/dermatan sulfotransferases (C/D-STs) underlie the synthesis of diverse sulfated structures in chondroitin/dermatan sulfate (CS/DS) chains. Recent reports have suggested that particular sulfated structures on CS/DS polymers are involved in the regulation of neural stem cell proliferation. Here, we examined the gene expression profile of C/D-STs in the neurogenic regions of embryonic and adult mouse central nervous system. Using reverse transcription-polymerase chain reaction analysis, all presently known C/D-STs were detected in the dorsal and ventral telencephalon of the embryonic day 13 (E13) mouse embryo, with the exception of chondroitin 4-O-sulfotransferase (C4ST)-3. In situ hybridization for C4ST-1, dermatan 4-O-sulfotransferase-1, chondroitin 6-O-sulfotransferase (C6ST)-1 and -2, and uronosyl 2-O-sulfotransferase revealed a cellular expression of these sulfotransferase genes in the embryonic germinal zones of the forebrain. The expression of multiple C/D-STs is maintained on cells residing in the adult neural stem cell niche. Neural stem cells cultured as neurospheres maintained the expression of these enzymes. Consistent with the gene expression pattern of C/D-STs, disaccharide analysis revealed that neurospheres and E13 mouse brain cells synthesized CS/DS chains containing monosulfated, but also significant amounts of disulfated, disaccharide units. Functionally, the inhibition of sulfation with sodium chlorate resulted in a significant, dose-dependent decrease in neurosphere number that could not be rescued by the addition of individual purified glycosaminoglycan (GAG) chains, including heparin. These findings argue against a simple charge-based mechanism of GAG chains in neural stem cell maintenance. The synergistic activities of C/D-STs might allow for the adaptive modification of CS/DS proteoglycans with diversely sulfated CS/DS chains in the extracellular microenvironment that surrounds neural stem cells.


Subject(s)
Central Nervous System/embryology , Central Nervous System/enzymology , Chondroitin Sulfates/metabolism , Neurons/cytology , Stem Cells/cytology , Sulfotransferases/metabolism , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Brain/cytology , Brain/drug effects , Brain/embryology , Brain/enzymology , Cell Movement/drug effects , Central Nervous System/cytology , Chlorates/pharmacology , Culture Media, Conditioned , Dermatan Sulfate/metabolism , Disaccharides , Epitopes , Gene Expression Regulation, Enzymologic/drug effects , In Situ Hybridization , Mice , Neurons/drug effects , Protein Isoforms/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Stem Cells/drug effects , Sulfotransferases/genetics
14.
Development ; 134(15): 2727-38, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17596283

ABSTRACT

Although the local environment is known to regulate neural stem cell (NSC) maintenance in the central nervous system, little is known about the molecular identity of the signals involved. Chondroitin sulfate proteoglycans (CSPGs) are enriched in the growth environment of NSCs both during development and in the adult NSC niche. In order to gather insight into potential biological roles of CSPGs for NSCs, the enzyme chondroitinase ABC (ChABC) was used to selectively degrade the CSPG glycosaminoglycans. When NSCs from mouse E13 telencephalon were cultivated as neurospheres, treatment with ChABC resulted in diminished cell proliferation and impaired neuronal differentiation, with a converse increase in astrocytes. The intrauterine injection of ChABC into the telencephalic ventricle at midneurogenesis caused a reduction in cell proliferation in the ventricular zone and a diminution of self-renewing radial glia, as revealed by the neurosphere-formation assay, and a reduction in neurogenesis. These observations suggest that CSPGs regulate neural stem/progenitor cell proliferation and intervene in fate decisions between the neuronal and glial lineage.


Subject(s)
Cell Differentiation , Cell Proliferation , Chondroitin Sulfates/physiology , Neuroglia/cytology , Neurons/cytology , Radial Nerve/cytology , Stem Cells/cytology , Animals , Cells, Cultured , Chondroitin ABC Lyase/metabolism , Chondroitin ABC Lyase/pharmacology , Chondroitin Sulfates/metabolism , Female , Mice , Mice, Inbred C57BL , Organogenesis/physiology , Pregnancy , Prosencephalon/cytology , Prosencephalon/embryology , Radial Nerve/embryology
15.
J Biol Chem ; 282(12): 9172-81, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17264084

ABSTRACT

Tenascin C (Tnc) is an alternatively spliced, multimodular extracellular matrix glycoprotein present in the ventricular zone of the developing brain. Pax6-deficient small eye (sey) mouse mutants show an altered Tnc expression pattern. Here, we investigated the expression of Tnc isoforms in neural stem/progenitor cells and their regulation by the paired-box transcription factor Pax6. Neural stem/progenitor cells cultured as neurospheres strongly expressed Tnc on the protein level. The Tnc isoform expression in neural stem/progenitor cells was analyzed by reverse transcriptase-PCR and dot blot Southern hybridization. In total, 20 different Tnc isoforms were detected in neurospheres derived from embryonic fore-brain cell suspensions. The Tnc isoform containing the fibronectin type III domains A1A4BD is novel and might be neural stem/progenitor cell-specific. Transient overexpression of Pax6 in neurospheres of the medial ganglionic eminence did not alter the total Tnc mRNA expression level but showed a pronounced regulative effect on different Tnc isoforms. The larger Tnc isoforms containing four, five, and six additional alternatively spliced fibronectin type III domains were up-regulated, whereas the small Tnc isoforms without any or with one additional domain were down-regulated. Thus, Pax6 is a homeodomain protein that also modulates the splicing machinery. We conclude that the combinatorial code of Tnc isoform expression in the neural stem/progenitor cell is complex and regulated by Pax6. These findings suggest a functional significance for individual Tnc isoforms in neural stem/progenitor cells.


Subject(s)
Eye Proteins/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Neurons/cytology , Paired Box Transcription Factors/physiology , Repressor Proteins/physiology , Stem Cells/cytology , Tenascin/chemistry , Alternative Splicing , Animals , Cell Line , Eye Proteins/metabolism , Fibroblasts/metabolism , Fibronectins/chemistry , Homeodomain Proteins/metabolism , Mice , Neurons/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Protein Isoforms , Protein Structure, Tertiary , Repressor Proteins/metabolism , Stem Cells/metabolism , Up-Regulation
16.
J Neurosci ; 26(15): 4082-94, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16611825

ABSTRACT

Neural stem cells have been documented in both the developing and the mature adult CNSs of mammals. This cell population holds a considerable promise for therapeutical applications in a wide array of CNS diseases. Therefore, universally applicable strategies for the purification of this population to further its cell biological characterization are sought. Here, we report that the unique chondroitin sulfate epitope recognized by the monoclonal antibody 473HD is surface expressed on actively cycling, multipotent progenitor cells of the developing telencephalon with radial glia-like properties. When used for immunopanning, the antibody enriched at least threefold for neural stem/progenitor cells characterized by the ability to self-renew as neurospheres that generated all major neural lineages in differentiation assays. In contrast, the 473HD-depleted cell fraction was mostly devoid of neurosphere-forming cells. The isolation of 473HD-positive adult multipotent progenitors from the subependymal zone of the lateral ventricle wall revealed a substantial overlap with the known adult neural stem cell marker LewisX. When the chondroitin sulfates were removed from immunoselected 473HD-positive neural stem/progenitor cell surfaces by chondroitinase ABC treatment or perturbed by the monoclonal antibody 473HD that recognizes the unique DSD-1 chondroitin sulfate epitope, the generation of neurospheres was significantly reduced. Thus, the 473HD epitope could not only be used for the isolation of multipotent neural progenitors during forebrain development as well as from the adult neurogenic niche but may also constitute a functionally important entity of the neural stem cell niche.


Subject(s)
Brain/embryology , Chondroitin Sulfates/chemistry , Epitopes/analysis , Neuroglia/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Brain/cytology , Cell Division , Chondroitin Sulfates/genetics , Chondroitin Sulfates/immunology , Chondroitin Sulfates/physiology , Embryonic Development , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Neuroglia/cytology , Pregnancy
17.
Dev Genes Evol ; 212(1): 43-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11875656

ABSTRACT

Here we describe the expression pattern of a previously unknown mouse gene mPet-1. The isolated cDNA codes for an ETS-domain transcription factor of 237 amino acids in length, which is localized to the nucleus. mPet-1 is a member of the winged helix transcription factor gene family like its rat homologue Pet-1 and the human homologue FEV. The start ATG of mPet-1 and the size of the predicted protein are identical to the human FEV. The mPet-1 protein is clearly smaller since it lacks the first 103 N-terminal amino acids of rat Pet-1. mPet- 1 is expressed in central serotonergic (5-hydroxytryptaminergic) neurons located in the mes-/metencephalic raphe nuclei from E11 on until adulthood. In these regions mPet-1 expression co-localizes precisely with the serotonin transporter (Sert),which it initially precedes. Interestingly, mPet-1 was not found in neurons transiently expressing Sert.


Subject(s)
Neurons/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Mice , Molecular Sequence Data , Proto-Oncogene Proteins/metabolism , Sequence Alignment , Serotonin/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...