Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Neuroscience ; 142(1): 175-85, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16844309

ABSTRACT

Spatially and temporally restricted populations of neurogenic astrocytes can generate multipotent neurospheres in vitro. To examine the ability of neurogenic astrocytes to respond to in vivo differentiation cues within a germinal matrix, we provided cultured neonatal cerebellar astrocytes access to the subependymal zone (SEZ) by grafting them directly into the lateral ventricle of adult mice. Here we report three events that follow such transplants. 1) Donor cells attach to periventricular structures, and form "neoplastic-like" spheres that penetrate the ventricular wall. These attached spheres can persist for months, as they give rise to "clones" of cells that infiltrate forebrain parenchyma. 2) Many donor cells enter the rostral migratory stream and migrate into the olfactory bulb where a small percentage differentiates as olfactory interneurons. 3) Finally, within the SEZ, some donor cells formed cell clusters that appear to interact with the SEZ neuronal precursor chains, and some donor cells differentiate into distinctive neurons with extensive, beady projections precisely confined between the ependymal layer and the striatum. Further analysis of normal SEZ anatomy reveals indigenous neurons with identical morphologies--some of which are contacted by 5-HT+ fibers--that we propose represent a heretofore uncharacterized, intrinsic SEZ neuron of unknown function. These results suggest that cultured astrocytes derived from non-SEZ brain regions can respond in different ways to in vivo cues provided by the adult lateral ventricle and SEZ by differentiating into neurons that eventually inhabit both the olfactory bulb and SEZ proper.


Subject(s)
Astrocytes/physiology , Ependyma/cytology , Neurons/physiology , Olfactory Bulb/cytology , Organogenesis , Stem Cell Transplantation , Animals , Animals, Newborn , Cell Differentiation , Cell Growth Processes , Cell Movement , Cells, Cultured , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/radiation effects
2.
Exp Neurol ; 182(1): 240-4, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821394

ABSTRACT

There is a paucity of information on the roles of extracellular matrix (ECM) and substrate molecules in general with regard to the growth and differentiation of neural stem and progenitor cells. There are well-established findings of a dense, presumably astrocyte-derived ECM in the persistently neurogenic subependymal zone and its migratory extension the rostral migratory stream. Cells cultured from this region, as well as from early postnatal cerebellum, generate multipotent neurospheres, but at present there is little information as to the ECM regulation of these neural stem cell populations. The present study examined the behavior of cerebellar-derived neurospheres on the matrix components laminin, fibronectin, and chondroitin sulfate proteoglycan. The results showed that laminin and fibronectin significantly increase cell migration velocity as compared to CSPG. Fibronectin effected a maximal velocity after 48 h, whereas maximal velocity on laminin and CSPG was not reached until 72 h. Both laminin and fibronectin were very permissive substrates for cellular outgrowth. Chondroitin sulfate proteoglcyan showed a significant inhibition of migratory outgrowth and velocity. These ECM molecules did not appear to affect the fate choice of neurons and glia, thus their role in neuropoietic structures may be to facilitate or deter cell movement and process outgrowth.


Subject(s)
Cell Movement/physiology , Extracellular Matrix/physiology , Neurons/physiology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Count , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cerebellum/cytology , Chondroitin Sulfate Proteoglycans/pharmacology , Fibronectins/pharmacology , Laminin/pharmacology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology
3.
J Comp Neurol ; 430(4): 485-500, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11169482

ABSTRACT

A transgenic mouse containing the first exon of the human Huntington's disease (HD) gene has revealed a variety of behavioral and pathophysiological anomalies reminiscent of certain aspects of human Huntington's disease (HD). The present study has found that expression of the extracellular matrix glycoprotein tenascin-C appears to be unaffected in astroglial cells in wild-type and R6/2 transgenic mice that express the mutant huntingtin protein but that it is conspicuously absent in two neuronal populations within the cerebral cortex and thalamus of the R6/2 mice. Loss of tenascin-C expression begins between the fourth and eighth postnatal weeks, coincidental with the onset of abnormal behavioral phenotype and the appearance of intranuclear inclusion bodies and neuropil aggregates. By 12 weeks, R6/2 mice exhibit a complete absence of tenascin-C neuronal immunolabeling, a disappearance of cRNA probe-positive neurons across discrete cytoarchitectonic regions of the dorsal thalamus (e.g., the ventromedial, parafascicular, lateral posterior, and posterior thalamic groups) and frontal cortex, and an accompanying thalamic astrogliosis. The loss of neuronal tenascin-C expression includes structures that are known to send converging excitatory axonal projections to the caudate-putamen, the structure that is most at risk for neurodegeneration in HD. Altered neuronal expression of tenascin-C in R6/2 mice implicates altered transcriptional activities of the mutant huntingtin protein. The abnormal biochemistry and possibly abnormal activity of thalamostriate and corticostriate projection neurons may also affect abnormal neuronal activities in their primary connectional target, the neostriatum, which is severely compromised in HD.


Subject(s)
Cerebral Cortex/physiology , Huntington Disease/physiopathology , Mice, Knockout/physiology , Tenascin/genetics , Thalamus/physiology , Animals , Brain Chemistry/genetics , Cerebral Cortex/cytology , Disease Models, Animal , Exons , Female , Gene Expression/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Lac Operon , Male , Mice , Mice, Inbred C57BL , Neuroglia/physiology , Neurons/physiology , RNA, Messenger/analysis , Tenascin/analysis , Thalamus/cytology
4.
Proc Natl Acad Sci U S A ; 97(25): 13883-8, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11095732

ABSTRACT

The mammalian brain contains a population of neural stem cells (NSC) that can both self-renew and generate progeny along the three lineage pathways of the central nervous system (CNS), but the in vivo identification and localization of NSC in the postnatal CNS has proved elusive. Recently, separate studies have implicated ciliated ependymal (CE) cells, and special subependymal zone (SEZ) astrocytes as candidates for NSC in the adult brain. In the present study, we have examined the potential of these two NSC candidates to form multipotent spherical clones-neurospheres-in vitro. We conclude that CE cells are unipotent and give rise only to cells within the glia cell lineage, although they are capable of forming spherical clones when cultured in isolation. In contrast, astrocyte monolayers from the cerebral cortex, cerebellum, spinal cord, and SEZ can form neurospheres that give rise both to neurons and glia. However, the ability to form neurospheres is restricted to astrocyte monolayers derived during the first 2 postnatal wk, except for SEZ astrocytes, which retain this capacity in the mature forebrain. We conclude that environmental factors, simulated by certain in vitro conditions, transiently confer NSC-like attributes on astrocytes during a critical period in CNS development.


Subject(s)
Astrocytes/cytology , Brain/cytology , Stem Cells/cytology , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Brain/metabolism , Brain/ultrastructure , Cell Lineage , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Mice , Microscopy, Electron , Stem Cells/metabolism , Stem Cells/ultrastructure
5.
J Neurosci Methods ; 96(1): 57-61, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10704671

ABSTRACT

A method is described that allows cDNA production from individual brain cell clones or 'neurospheres'. These culture-generated spheres of stem, progenitor, and differentiated cells have been the focus of interest because they represent an in vitro model of neurogenesis. However, because neurospheres are somewhat resistant, in part due to their enclosure by a dense extracellular matrix, to methods attempting to disrupt them and isolate nucleic acids, there is a need for new technology that affords the simple and efficient RT-PCR for studies of neural gene expression and discovery. A method is described here that uses sonication and an all-in-one approach for the construction of cDNA from single neurospheres. The generation of cDNA from individual adult brain stem/progenitor cell neurospheres is useful for future studies of neurogenic gene expression.


Subject(s)
Brain/cytology , Cell Culture Techniques/methods , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells/physiology , Animals , Clone Cells , DNA Primers , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/physiology , Sonication , Stem Cells/cytology
6.
Trends Neurosci ; 22(8): 348-57, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10407420

ABSTRACT

There are pluripotent stem cells in the adult brain that might not be very different from those found in bone marrow. Recent and profound advances in the field of neuropoiesis, which often rely on insights from studies of hematopoiesis and in some instances use cross-paradigms with this field, have already revealed that bone marrow has much in common with so-called 'brain marrow'. Proliferative primogenitors and developmentally regulated molecules are hallmarks of both neuropoiesis and hematopoiesis. This article will focus on recent advances in neuropoiesis within a central core of the mature brain that is referred to as brain marrow, discussing its pluripotency and proliferative capacity, in vitro and molecular assays used in its study, and markers of neuropoietic stem/progenitor cells. As hematopoiesis research has led to the discovery of numerous morphogenetic factors, it is anticipated that studies of neuropoiesis should also uncover many new factors and genes that affect the growth and differentiation of neural cells. Recent breakthroughs in the stem-cell field prompt an inclusion of rationale for the persistence of normal stem/progenitor cells even in the aged brain. By analogy with hematopoiesis research, a thorough investigation of brain marrow should provide basic insights into developmental and persistent neurogenesis while anticipating cell-transplant and gene therapies for debilitating neurological diseases.


Subject(s)
Bone Marrow Cells/cytology , Brain/cytology , Stem Cells/cytology , Stem Cells/physiology , Animals , Bone Marrow Cells/physiology , Cell Division/physiology , Genetic Therapy , Humans , Nerve Tissue/transplantation
7.
Exp Neurol ; 156(2): 333-44, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328940

ABSTRACT

Recent in vitro studies have shown that the periventricular subependymal zone (SEZ) of the rodent brain is capable of de novo generation of neurons and glia. There is less information available on neurogenesis in the adult human brain, and no study has shown the clonal generation of neurons and glia from in vitro-generated "neurospheres." Here we describe the isolation of proliferative stem/progenitor cells within neurospheres from two different regions, the SEZ and the hippocampus, from surgical biopsy specimens of adult (24-57 years) human brain. Using light and electron microscopy; immunocytochemistry for a variety of neuronal, glial, and developmental (including extracellular matrix; ECM) markers; and the reverse transcriptase polymerase chain reaction to demonstrate different gene transcripts found in neurospheres, it is shown that the adult human brain harbors a complex population of stem/progenitor cells that can generate neuronal and glial progeny under particular in vitro growth conditions. These methods also show that these neurospheres contain both neurons and glia and demonstrate regional similarities at the mRNA level, indicating common stem/progenitor cell types within two different neurogenic regions of the adult human brain. In addition to the synthesis of developmentally regulated molecules such as the ECM protein tenascin-C, a variety of other genes (e.g., Pax 6) and proteins (e.g. , Bcl-2) involved in cell survival and differentiation are expressed by adult human brain neurospheres.


Subject(s)
Brain/cytology , Homeodomain Proteins , Stem Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers , Cell Lineage , Cells, Cultured , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Eye Proteins , Female , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/genetics , Hippocampus/cytology , Humans , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/genetics , Male , Middle Aged , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Nestin , Neurofilament Proteins/analysis , Neurofilament Proteins/genetics , Neuroglia/cytology , Neurons/cytology , PAX6 Transcription Factor , Paired Box Transcription Factors , Phosphopyruvate Hydratase/analysis , Phosphopyruvate Hydratase/genetics , RNA, Messenger/analysis , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/analysis , Tenascin/genetics
8.
Exp Neurol ; 156(2): 430-3, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328947

ABSTRACT

The adult mammalian CNS harbors a population of multipotent stem/progenitor cells that can be induced to grow as proliferative neurospheres in vitro. We demonstrate here that neurosphere-generating cells can be isolated from adult mouse spinal cord and forebrain subependymal zone after postmortem intervals of up to 140 h, when kept at 4 degrees C, and up to 30 h when kept at room temperature. Although there was an inverse relationship between postmortem interval and the number of neurospheres generated, neurospheres derived under these conditions were proliferative and could give rise to both neurons and glia.


Subject(s)
Neuroglia/cytology , Neurons/cytology , Prosencephalon/cytology , Spinal Cord/cytology , Stem Cells/cytology , Animals , Cell Survival , Cells, Cultured , DNA Replication , Mice , Mice, Inbred ICR , Postmortem Changes , Temperature , Time Factors
10.
J Neurosci ; 17(22): 8767-77, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9348346

ABSTRACT

Analysis of the molecular basis of neuronal migration in the mammalian CNS relies critically on the discovery and identification of genetic mutations that affect this process. Here, we report the detailed cerebellar phenotype caused by a new autosomal recessive neurological mouse mutation, scrambler (gene symbol scm). The scrambler mutation results in ataxic mice that exhibit several neuroanatomic defects reminiscent of reeler. The most obvious of these lies in the cerebellum, which is small and lacks foliation. Granule cells, although normally placed in an internal granule cell layer, are greatly reduced in number ( approximately 20% of normal). Purkinje cells are also reduced in number, and the majority are located ectopically in deep cerebellar masses. There is a small population of Purkinje cells ( approximately 5% of the total) that occupy a Purkinje cell layer between the molecular and granule cell layers. Despite this apparent disorganization of Purkinje cells, zebrin-positive and zebrin-negative parasagittal zones can be delineated. The ectopic masses of Purkinje cells are bordered by the extracellular matrix protein tenascin and by processes containing glial fibrillary acidic protein. Antibodies specific for these proteins also identify a novel midline raphe structure in both scrambler and reeler cerebellum that is not present in wild-type mice. Thus, in many respects, the scrambler cerebellum is identical to that of reeler. However, the scrambler locus has been mapped to a site distinct from that of reelin (Reln), the gene responsible for the reeler defect. Here we find that there are normal levels of Reln mRNA in scrambler brain and that reelin protein is secreted normally by scrambler cerebellar cells. These findings imply that the scrambler gene product may function in a molecular pathway critical for neuronal migration that is tightly linked to, but downstream of, reelin.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cerebellum/pathology , Extracellular Matrix Proteins/genetics , Mice, Neurologic Mutants/physiology , Animals , Ataxia/genetics , Cell Adhesion Molecules, Neuronal/analysis , Cell Movement/physiology , Cerebellum/chemistry , Cerebellum/physiopathology , Extracellular Matrix/pathology , Extracellular Matrix Proteins/analysis , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C3H , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Neuroglia/pathology , Phenotype , Purkinje Cells/chemistry , Purkinje Cells/cytology , RNA, Messenger/analysis , Reelin Protein , Serine Endopeptidases
11.
Glia ; 21(4): 399-407, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9419015

ABSTRACT

Using a novel suspension culture approach, previously undescribed populations of neural precursor cells have been isolated from the adult mouse brain. Recent studies have shown that neuronal and glial precursor cells proliferate within the subependymal zone of the lateral ventricle throughout life, and a persistent expression of developmentally regulated surface and extracellular matrix molecules implicates cell-cell and cell-substrate interactions in the proliferation, migration, and differentiation of these cells. By using reagents that may affect cell-cell interactions, dissociated adult brain yields two types of cell aggregates, type I and type II spheres. Both sphere types are proliferative, and type I spheres evolve into type II spheres. Neurons and glia arise from presumptive stem cells of type II spheres, and they can survive transplantation to the adult brain.


Subject(s)
Brain/cytology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins , Neuroglia/physiology , Neurons/physiology , Animals , Antimetabolites/pharmacology , Brain/metabolism , Bromodeoxyuridine/pharmacology , Cell Transplantation , DNA Probes , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Graft Survival , Mice , Mice, Inbred ICR , Mice, Transgenic , Microscopy, Electron , Nestin , Neuroglia/ultrastructure , Neurons/ultrastructure , Rats
12.
J Comp Neurol ; 374(1): 41-51, 1996 Oct 07.
Article in English | MEDLINE | ID: mdl-8891945

ABSTRACT

We have addressed the issue of whether or not peripherally expressed nerve growth factor (NGF) influences the formation of whisker-specific patterns in the brain by regulating the survival of sensory neurons. Transgenic mice that overexpress an NGF cDNA in the skin were examined. In these animals, excess NGF expression is controlled by promoter and enhancer sequences of a keratin gene, thus restricting the higher levels of NGF expression to basal keratinocytes of the epidermis. Twice the number of trigeminal sensory neurons survive in transgenic mice as in normal animals, and a corresponding hyperinnervation of the whisker pad is noted, both around the vibrissa follicles and along the intervibrissal epidermis. However, the increased survival of sensory neurons and the enhanced peripheral projections do not interfere with the development of whisker-specific patterns in the trigeminal brainstem, in the ventrobasal thalamic complex or in the face-representation region of the primary somatosensory (SI) cortex. These results demonstrate that vibrissa-related central patterns are able to form in the virtual absence of trigeminal ganglion cell death and suggest that mechanisms other than a selective elimination of sensory neurons control the development of whisker-specific neural patterns in the brain.


Subject(s)
DNA, Complementary/biosynthesis , Keratinocytes/metabolism , Nerve Growth Factors/biosynthesis , Peripheral Nerves/metabolism , Trigeminal Ganglion/metabolism , Vibrissae/innervation , Animals , Brain Stem/metabolism , Cell Count , Enhancer Elements, Genetic , Mice , Mice, Transgenic , Nerve Growth Factors/physiology , Neurons, Afferent/cytology , Promoter Regions, Genetic , Somatosensory Cortex/metabolism , Thalamus/metabolism
13.
Neuroscience ; 74(2): 579-97, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8865207

ABSTRACT

Transplantation of embryonic neurons to the adult mammalian central nervous system (CNS) offers the possibility of re-establishing neural functions lost after traumatic injuries or neurodegenerative disease. In the adult CNS, however, transplanted neurons and their growing neurites can become confined to the graft region, and there may also be a relative paucity of afferents innervating grafted neurons. Because glia may influence the development and regeneration of CNS neurons, the present study has characterized the distribution of astrocytes and developmentally regulated glycoconjugates (chondroitin-6-sulfate proteoglycan and tenascin) within regions of the embryonic mouse CNS used as donor tissues, and in and around these grafts to the adult striatum and substantia nigra. Both chondroitin-6-sulfate proteoglycan and tenascin are present in the embryonic ventral mesencephalon (in association with radial glia and their endfeet, and glial boundaries that cordon off the ventral mesencephalon dopamine neuron migratory zone) and lateral ganglionic eminence before transplantation, and they are conserved within grafts of these tissues to the adult mouse. Neostriatal grafts exhibit a heterogeneous pattern of astrocyte and extracellular matrix molecule distribution, unlike ventral mesencephalon grafts, which are rather homogeneous. There is evidence to suggest that, in addition to variation in astroglial/extracellular matrix immunostaining within different compartments in striatal grafts to either adult striatum or substantia nigra, there are also boundaries between these compartments that are rich in glial fibrillary acidic protein/extracellular matrix components. Substantia nigra grafts, with cells immunoreactive for tyrosine hydroxylase, are also rich in immature astroglia (RC-2-immunopositive), and as the astroglia mature (to glial fibrillary acidic protein-positive) over time the expression of chondroitin-6-sulfate proteoglycan and tenascin is also reduced. These same extracellular matrix constituents, however, are only slightly up-regulated in an area of the adult host which surrounds the grafted tissue. Glial scar components exhibit no obvious differences between grafts from different sources to homotopic (e.g., striatum to striatum) or heterotopic (e.g., substantia nigra to striatum) sites, and likewise grafts of non-synaptically associated structures (e.g., cerebellum to striatum), needle lesions or vehicle injections all yield astroglial/extracellular matrix scars in the host that are indistinguishable. Studies utilizing the ROSA-26 transgenic (beta-galactosidase-positive) mouse as a host for non-5-bromo-4-chloro-3-indolyl-beta-d-galactopyranoside-labeled grafts indicate that the early astroglial/extracellular matrix response to the graft is derived from the surrounding host structures. Furthermore, biochemical analysis of one of the "boundary molecules", tenascin, from the developing ventral mesencephalon versus adult striatal lesions, suggests that different forms of the molecule predominate in the embryonic versus lesioned adult brain. Such differences in the nature and distribution of astroglia and developmentally regulated extracellular matrix molecules between donor and host regions may affect the growth and differentiation of transplanted neurons. The present study suggests that transplanted neurons and their processes may flourish within graft versus host regions, in part due to a confining glial scar, but also because the extracellular milieu within the graft site remains more representative of the developmental environment from which the donor neurons were obtained [Gates M. A., et al. (1994) Soc. Neurosci. Abstr. 20, 471].


Subject(s)
Astrocytes/physiology , Brain Tissue Transplantation , Embryo Transfer , Extracellular Matrix/physiology , Mesencephalon/transplantation , Animals , Immunohistochemistry , Mice , Mice, Inbred ICR
14.
J Neurosci Methods ; 66(2): 99-108, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8835793

ABSTRACT

Previous studies describing the use of cryoculture methods have focused on the efficacy of the method for studying neuron attachment and neurite outgrowth on intact sections of nerve, and rodent and even human brain. The cryoculture method has shown promise for determining the presence of cell attachment- and neurite-growth-inhibiting molecules in such specimens, and some studies have also attempted to neutralize such molecules with antibodies to myelin inhibitory proteins, nerve growth factor, or factors present in conditioned media that may counteract the repulsiveness of some of these molecules preserved in sections of, for example, myelinated nerves or adult brain white matter. The present study describes the novel use of lesioned central nervous system cryocultures as substrates for investigating the attachment of embryonic neurons and PC12 cells. In addition to demonstrating the use of this novel scar substrate to extend previous 'scar-in-a-dish' models (David et al. (1990) Neuron, 5:463-469; Rudge and Silver (1990) J. Neurosci., 10: 3594-3603; Rudge et al. (1989) Exp. Neurol., 103: 1-16), the present study also describes antibody and lectin perturbations of putative inhibitory molecules that result in an enhanced attachment of cells to cryosection cultures of brain and spinal cord wounds.


Subject(s)
Brain Injuries/metabolism , Cell Culture Techniques/methods , Extracellular Matrix Proteins/metabolism , Lectins/pharmacology , Tenascin/immunology , Animals , Antibody Specificity , Carbocyanines , Cell Adhesion/physiology , Cells, Cultured/chemistry , Cells, Cultured/cytology , Cells, Cultured/physiology , Cryopreservation , Extracellular Matrix Proteins/immunology , Female , Fluorescent Dyes , Frozen Sections , Immunohistochemistry , Mice , Mice, Inbred ICR , Neutralization Tests , PC12 Cells/chemistry , PC12 Cells/cytology , PC12 Cells/physiology , Pregnancy , Rats , Wound Healing/drug effects , Wound Healing/physiology
15.
J Comp Neurol ; 361(2): 249-66, 1995 Oct 16.
Article in English | MEDLINE | ID: mdl-8543661

ABSTRACT

The subventricular zone (SVZ) of the lateral ventricle remains mitotically active in the adult mammalian central nervous system (CNS). Recent studies have suggested that this region may contain neuronal precursors (neural stem cells) in adult rodents. A variety of neuronal and glial markers as well as three extracellular matrix (ECM) markers were examined with the hope of understanding factors that may affect the growth and migration of neurons from this region throughout development and in the adult. This study has characterized the subventricular zone of late embryonic, postnatal, and adult mice using several neuronal markers [TuJ1, nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d), neuron-specific enolase (NSE)], glial markers [RC-2, vimentin, glial fibrillary acidic protein (GFAP), galactocerebroside (Gal-C)], ECM markers [tenascin-C (TN-C), chondroitin sulfate, a chondroitin sulfate proteoglycan termed dermatan sulfate-dependent proteoglycan-1 (DSD-1-PG)], stem-cell marker (nestin), and proliferation-specific marker [bromodeoxyuridine (BrdU)]. TuJ1+ and nestin+ cells (neurons and stem cells, respectively) persist in the region into adulthood, although the numbers of these cells become more sparse as the animal develops, and they appear to be immature compared to the cells in surrounding forebrain structures (e.g., not expressing NSE and having few, if any, processes). Likewise, NADPH-d+ cells are found in and around the SVZ during early postnatal development but become more sparse in the proliferative zone through maturity, and, by adulthood, only a few labeled cells can be found at the border between the SVZ and surrounding forebrain structures (e.g., the striatum), and even smaller numbers of positive cells can be found within the adult SVZ proper. BrdU labeling also seems to decrease significantly after the first postnatal week, but it still persists in the SVZ of adult animals. The disappearance of RC-2+ (radial) glia during postnatal development and the persistence of glial-derived ECM molecules such as tenascin and chondroitin sulfate proteoglycans (as well as other "boundary" molecules) in the adult SVZ may be associated with a persistence of immaturity, cell death, and a lack of cell emigration from the SVZ in the adult.


Subject(s)
Brain/embryology , Brain/growth & development , Embryo, Mammalian/metabolism , Aging/metabolism , Animals , Animals, Newborn , Biomarkers , Brain/cytology , Cell Division , Cerebral Ventricles , Embryo, Mammalian/cytology , Extracellular Matrix/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Neurons/cytology , Neurons/metabolism
16.
J Neurosci ; 15(3 Pt 1): 1971-83, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7534342

ABSTRACT

In light of a previous report suggesting that the brains of tenascin-deficient animals are grossly normal, we have studied the somatosensory cortical barrel field and injured cerebral cortex in postnatal homozygous tenascin knockout, heterozygote, and normal wild-type mice. Nissl staining, cytochrome oxidase, and Dil axonal tracing of thalamocortical axonal projections to the somatosensory cortex, all reveal the formation of normal barrels in the first postnatal week in homozygous knockout mice that cannot be distinguished from heterozygote or normal wild-type barrels. In addition to confirming the absence of tenascin in knockout animals, and reporting apparently reduced levels of the glycoprotein in barrel boundaries of heterozygote animals using well-characterized antibodies and immunocytochemistry, we also studied the DSD-1-PG proteoglycan, another developmentally regulated molecule known to be associated with transient glial/glycoconjugate boundaries that surround developing barrels; DSD-1-PG was also found to be expressed in barrel boundaries in apparently normal time frames in tenascin knockout mice. Peanut agglutinin (PNA) binding of galactosyl-containing glycoconjugates also revealed barrel boundaries in all three genotypes. We also examined the expression of tenascin-R, a paralog of tenascin-C (referred to here simply as tenascin). As previously reported, tenascin-R is prominently expressed in subcortical white matter, and we found it was not expressed in the barrel boundaries in any of the genotypes. Thus, the absence of tenascin does not result in a compensatory expression of tenascin-R in the barrel boundaries. Finally, we studied wounds of the cerebral cortex in the late postnatal mouse. The astroglial scar formed, for the most part, in the same time course and spatial distribution in the wild-type and tenascin knockout mice. However, there may be some differences in the extent of gliosis between the knockout and the wild type that warrant further study. Roles for boundary molecules like tenascin during brain pattern formation and injury are reconsidered in light of these findings on barrel development and cortical lesions in tenascin-deficient mice.


Subject(s)
Brain Mapping , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Somatosensory Cortex/metabolism , Vibrissae/anatomy & histology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Cell Adhesion Molecules, Neuronal/analysis , Electron Transport Complex IV , Extracellular Matrix Proteins/analysis , Mice , Mice, Knockout , Proteoglycans/analysis , Proteoglycans/metabolism , Somatosensory Cortex/cytology , Tenascin
17.
J Neurosurg ; 82(1): 106-12, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7529300

ABSTRACT

Tenascin is an extracellular matrix glycoprotein expressed during both normal development and neoplastic growth in both neural and nonneural tissues. During development of the central nervous system (CNS), tenascin is synthesized by glial cells, in particular by immature astrocytes, and is concentrated in transient boundaries around emerging groups of functionally distinct neurons. In the mature CNS, only low levels of the glycoprotein can be detected. The present study demonstrates that following trauma to the adult human cerebral cortex, discrete populations of reactive astrocytes upregulate their expression of tenascin and dramatically increase their transcription of the tenascin gene. The enhanced expression of tenascin may be involved in CNS wound healing, and may also affect neurite growth within and around a brain lesion.


Subject(s)
Brain Injuries/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Up-Regulation , Wounds, Gunshot , Adult , Brain Injuries/genetics , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Gene Expression , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , Male , Tenascin , Wound Healing/genetics
18.
Exp Neurol ; 123(2): 251-70, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7691650

ABSTRACT

We review the growing list of molecules that may be involved in wound healing in the central nervous system (CNS). It is known that many of these molecules are present during normal development and neoplastic growth in both neural and nonneural tissues, often in areas where pattern formation or tissue remodeling is evident; however, their functional roles are often quite elusive. In order to understand the changes that occur in and around a brain wound, we review proposed functions of neuroregeneration-related molecules in in vitro and in vivo preparations, as well as note their expression in other healing tissues including skin. A hypothesis that wound healing events in the CNS supersede neuritic growth around a lesion is presented. In contrast to the classical view of failed regeneration, there may be significant amounts of circuit reorganization that occur following injury, and such plasticity may be further enhanced by manipulating the molecular environment around a brain wound and in synaptically related structures.


Subject(s)
Central Nervous System/physiology , Extracellular Matrix Proteins/physiology , Nerve Regeneration , Wound Healing , Animals , Astrocytes/physiology , Cell Adhesion Molecules, Neuronal/physiology , Cell Movement , Central Nervous System/metabolism , Fibronectins/physiology , Growth Substances/physiology , Laminin/physiology , Neurites/physiology , Neuronal Plasticity , Peripheral Nervous System/metabolism , Peripheral Nervous System/physiology , Proteoglycans/physiology , Skin/metabolism , Skin Physiological Phenomena , Synapses/physiology , Tenascin
19.
Proc Natl Acad Sci U S A ; 89(7): 2634-8, 1992 Apr 01.
Article in English | MEDLINE | ID: mdl-1372985

ABSTRACT

Tenascin is an extracellular matrix molecule synthesized and released by young astrocytes during embryonic and early postnatal development of the nervous system, and it is concentrated in boundaries around emerging functional neuronal units. In the adult nervous system, tenascin can be detected only in very low levels. Distinct spatial and temporal distributions of tenascin during developmental events suggest a role in the guidance and/or segregation of neurons and their processes within incipient functional patterns. We show here, using in situ hybridization and immunocytochemistry, that stab wounds of the adult mouse cerebellar and cerebral cortices result in an enhanced expression of tenascin in a discrete region around the lesion site that is associated with a subset of glial fibrillary acidic protein-positive astrocytes. Tenascin up-regulation in the lesioned adult brain may be directly involved in failed regeneration or indirectly involved through its interactions with other glycoconjugates that either inhibit or facilitate neurite growth.


Subject(s)
Brain Injuries/physiopathology , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , Age Factors , Animals , Cerebellar Cortex/metabolism , Cerebral Cortex/metabolism , Gene Expression , Glial Fibrillary Acidic Protein/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred Strains , Nucleic Acid Hybridization , RNA, Messenger/genetics , Tenascin
20.
Ann N Y Acad Sci ; 633: 122-41, 1991.
Article in English | MEDLINE | ID: mdl-1789543

ABSTRACT

During brain development, transient partitions of glia and glycoconjugates (glycoproteins, glycolipids, and glycosaminoglycans) surround forming functional units (e.g., nuclear divisions, whisker-related barrels, and neostriatal striosomes). These partitions, which we think of as boundaries, consist of dense aggregates of glial fibrillary acidic protein (GFAP)-positive radial glia, young astrocytes and their processes, and developmentally regulated glycoconjugates (e.g., J1/tenascin and the 473 proteoglycan) that can be thought of as recognition molecules present on membranes or perhaps within the extracellular matrix. When functional patterns have formed and appear to be stabilized, these boundaries are no longer detectable. Lesions of the developing brain show the existence of a more global astrocytic distribution suggestive of biochemically distinct subsets of astrocytes that reside within boundary versus nonboundary positions. Lesions of the adult brain, in addition to showing gliosis, reveal a reexpression of some of the same macromolecules present in transient brain boundaries during development. It is postulated that developmental boundaries and wounds in the adult brain possess some of the same inhibitory and possibly alluring molecular substrates for neuritic expansion.


Subject(s)
Brain Diseases/physiopathology , Brain/growth & development , Glycoconjugates/physiology , Nerve Regeneration , Neuroglia/physiology , Aging/metabolism , Aging/physiology , Animals , Brain/cytology , Brain/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Extracellular Matrix/metabolism , Glycoconjugates/metabolism , Tissue Distribution , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...