Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Biomaterials ; 216: 119263, 2019 09.
Article in English | MEDLINE | ID: mdl-31220794

ABSTRACT

The development of effective treatment options for repair of peripheral nerves is complicated by lack of knowledge concerning the interactions between cells and implants. A promising device, the multichannel scaffold, incorporates microporous channels, aligning glia and directing axonal growth across a nerve gap. To enhance clinical outcomes of nerve repair, a platform, representative of current implant technology, was engineered which 1) recapitulated key device features (porosity and linearity) and 2) demonstrated remyelination of adult neurons. The in vitro platform began with the study of Schwann cells on porous polycaprolactone (PCL) and poly(lactide co-glycolide) (PLGA) substrates. Surface roughness determined glial cell attachment, and an additional layer of topography, 40 µm linear features, aligned Schwann cells and axons. In addition, direct co-culture of sensory neurons with Schwann cells significantly increased neurite outgrowth, compared to neurons cultured alone (naive or pre-conditioned). In contrast to the control substrate (glass), on porous PCL substrates, Schwann cells differentiated into a mature myelinating phenotype, expressing Oct-6, MPZ and MBP. The direct applicability of this platform to nerve implants, including its response to physiological cues, allows for optimization of cell-material interactions, close observation of the regeneration process, and the study of therapeutics, necessary to advance peripheral nerve repair technology.


Subject(s)
Nerve Regeneration , Neuroglia/cytology , Neurons/cytology , Polyesters/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Coculture Techniques , Guided Tissue Regeneration , Mice, Inbred C57BL , Neuroglia/physiology , Neurons/physiology , Porosity , Schwann Cells/cytology , Schwann Cells/physiology
2.
Hum Mol Genet ; 21(16): 3525-34, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22581779

ABSTRACT

FIG4 is a ubiquitously expressed phosphatase that, in complex with FAB1/PIKFYVE and VAC14, regulates the biosynthesis of the signaling lipid PI(3,5)P(2). Null mutation of Fig4 in the mouse results in spongiform degeneration of brain and peripheral ganglia, defective myelination and juvenile lethality. Partial loss-of-function of human FIG4 results in a severe form of Charcot-Marie-Tooth neuropathy. Neurons from null mice contain enlarged vacuoles derived from the endosome/lysosome pathway, and astrocytes accumulate proteins involved in autophagy. Other cellular defects include astrogliosis and microgliosis. To distinguish the contributions of neurons and glia to spongiform degeneration in the Fig4 null mouse, we expressed Fig4 under the control of the neuron-specific enolase promoter and the astrocyte-specific glial fibrillary acidic protein promoter in transgenic mice. Neuronal expression of Fig4 was sufficient to rescue cellular and neurological phenotypes including spongiform degeneration, gliosis and juvenile lethality. In contrast, expression of Fig4 in astrocytes prevented accumulation of autophagy markers and microgliosis but did not prevent spongiform degeneration or lethality. To confirm the neuronal origin of spongiform degeneration, we generated a floxed allele of Fig4 and crossed it with mice expressing the Cre recombinase from the neuron-specific synapsin promoter. Mice with conditional inactivation of Fig4 in neurons developed spongiform degeneration and the full spectrum of neurological abnormalities. The data demonstrate that expression of Fig4 in neurons is necessary and sufficient to prevent spongiform degeneration. Therapy for patients with FIG4 deficiency will therefore require correction of the deficiency in neurons.


Subject(s)
Brain/pathology , Flavoproteins/genetics , Neurons/physiology , Animals , Astrocytes/pathology , Astrocytes/physiology , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Flavoproteins/metabolism , Gene Expression , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Phosphoinositide Phosphatases , Phosphoric Monoester Hydrolases , Schwann Cells/metabolism , Schwann Cells/pathology
5.
J Comp Neurol ; 422(4): 579-93, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10861527

ABSTRACT

Geniculate (gustatory) and trigeminal (somatosensory) afferents take different routes to the tongue during rat embryonic development. To learn more about the mechanisms controlling neurite outgrowth and axon guidance, we are studying the roles of diffusible factors. We previously profiled the in vitro sensitivity of trigeminal axons to neurotrophins and target-derived diffusible factors and now report on these properties for geniculate axons. GDNF, BDNF, and NT-4, but not NT-3 or NGF, stimulate geniculate axon outgrowth during the ages investigated, embryonic days 12-14. Sensitivity to effective neurotrophins is developmentally regulated and different from that of the trigeminal ganglion. In vitro coculture studies revealed that geniculate axons were repelled by branchial arch explants that were previously shown to be repellent to trigeminal axons (Rochlin and Farbman [1998] J Neurosci 18:6840-6852). In addition, some branchial arch explants and untransfected COS7 cells repelled geniculate but not trigeminal axons. Sema3A, a ligand for neuropilin-1, is effective in repelling geniculate and trigeminal axons, and antineuropilin-1, but not antineuropilin-2, completely blocks the repulsion by arch explants that repel axon outgrowth from both ganglia. Sema3A mRNA is concentrated in branchial arch epithelium at the appropriate time to mediate the repulsion. In Sema3A knockout mice, geniculate and trigeminal afferents explore medial regions of the immature tongue and surrounding territories not explored in heterozygotes, supporting our previous hypothesis that Sema3A-based repulsion mediates the early restriction of sensory afferents away from midline structures.


Subject(s)
Axons/drug effects , Geniculate Ganglion/drug effects , Nerve Growth Factors/pharmacology , Tongue/innervation , Trigeminal Ganglion/drug effects , Animals , Axons/physiology , Brain-Derived Neurotrophic Factor/pharmacology , COS Cells , Cells, Cultured , Geniculate Ganglion/embryology , Geniculate Ganglion/physiology , Glial Cell Line-Derived Neurotrophic Factor , Mice , Mice, Knockout , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/pharmacology , Neurotrophin 3/pharmacology , Rats , Rats, Sprague-Dawley , Tongue/embryology , Trigeminal Ganglion/embryology , Trigeminal Ganglion/physiology
6.
Neuron ; 25(1): 29-41, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10707970

ABSTRACT

Neuropilins are receptors for class 3 secreted semaphorins, most of which can function as potent repulsive axon guidance cues. We have generated mice with a targeted deletion in the neuropilin-2 (Npn-2) locus. Many Npn-2 mutant mice are viable into adulthood, allowing us to assess the role of Npn-2 in axon guidance events throughout neural development. Npn-2 is required for the organization and fasciculation of several cranial nerves and spinal nerves. In addition, several major fiber tracts in the brains of adult mutant mice are either severely disorganized or missing. Our results show that Npn-2 is a selective receptor for class 3 semaphorins in vivo and that Npn-1 and Npn-2 are required for development of an overlapping but distinct set of CNS and PNS projections.


Subject(s)
Axons/physiology , Carrier Proteins/metabolism , Glycoproteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Age Factors , Animals , Axons/chemistry , Brain Chemistry/physiology , COS Cells , Gene Deletion , Gene Expression Regulation, Developmental , Habenula/chemistry , Habenula/embryology , Habenula/pathology , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/chemistry , Mossy Fibers, Hippocampal/embryology , Mossy Fibers, Hippocampal/pathology , Motor Neurons/chemistry , Motor Neurons/physiology , Motor Neurons/ultrastructure , Neuropilin-1 , Peripheral Nervous System/chemistry , Peripheral Nervous System/embryology , Peripheral Nervous System/pathology , Protein Binding/physiology , Rats , Semaphorin-3A , Spinal Nerves/chemistry , Spinal Nerves/pathology , Spinal Nerves/physiology , Superior Cervical Ganglion/chemistry , Superior Cervical Ganglion/embryology , Superior Cervical Ganglion/pathology , Thalamus/chemistry , Thalamus/embryology , Thalamus/pathology , Trochlear Nerve/chemistry , Trochlear Nerve/embryology , Trochlear Nerve/pathology
7.
Dev Biol ; 220(2): 129-41, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10753505

ABSTRACT

Sema3A (Sema III, SemD, collapsin-1) can induce neuronal growth cone collapse and axon repulsion of distinct neuronal populations. To study Sema3A function in patterning afferent projections into the developing spinal cord, we employed the recombinant adenoviral vector technique in embryonic rat spinal cord slices. Virus solution was injected in the dorsal aspect of organotypic spinal cord cultures with segmentally attached dorsal root ganglia (sc-DRG). In cultures grown in the presence of nerve growth factor (NGF), injected either with the control virus AdCMVLacZ or with vehicle only, afferent innervation patterns were similar to those of control. However, unilateral injection of AdCMVSema3A/AdCMVLacZ in sc-DRG slices revealed a strong inhibitory effect on NGF-dependent sensory afferent growth. Ectopic Sema3A in the dorsal spinal cord, the target area of NGF-responsive DRG fibers in vivo, created an exclusion zone for these fibers and as a result they failed to reach and innervate their appropriate target zones. Taken together, gain of Sema3A function in the dorsal aspect of sc-DRG cultures revealed a dominant inhibitory effect on NGF-dependent, nociceptive sensory DRG afferents, an observation in line with the model proposed by E. K. Messersmith et al. (1995, Neuron 14, 949-959), suggesting that Sema3A secreted by spinal cord cells can act to repel central sensory fibers during the formation of lamina-specific connections in the spinal cord.


Subject(s)
Ganglia, Spinal/embryology , Genetic Vectors/metabolism , Glycoproteins/metabolism , Spinal Cord/embryology , Adenoviridae/genetics , Animals , Blotting, Northern , COS Cells , Chemotaxis , Choristoma , Culture Techniques , Ganglia, Spinal/metabolism , Gene Transfer Techniques , Immunohistochemistry , In Situ Hybridization , Nerve Growth Factor/metabolism , Neurons, Afferent/physiology , Rats , Rats, Wistar , Semaphorin-3A , Spinal Cord/metabolism
8.
Mol Cell Neurosci ; 13(2): 143-66, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10192772

ABSTRACT

This study evaluates the expression of the chemorepellent semaphorin III (D)/collapsin-1 (sema III) following lesions to the rat CNS. Scar tissue, formed after penetrating injuries to the lateral olfactory tract (LOT), cortex, perforant pathway, and spinal cord, contained numerous spindle-shaped cells expressing high levels of sema III mRNA. The properties of these cells were investigated in detail in the lesioned LOT. Most sema III mRNA-positive cells were located in the core of the scar and expressed proteins characteristic for fibroblast-like cells. Neuropilin-1, a sema III receptor, was expressed in injured neurons with projections to the lesion site, in a subpopulation of scar-associated cells and in blood vessels around the scar. In contrast to lesions made in the mature CNS, LOT transection in neonates did not induce sema III mRNA expression within cells in the lesion and was followed by vigorous axonal regeneration. The concomitant expression of sema III and its receptor neuropilin-1 in the scar suggests that sema III/neuropilin-1-mediated mechanisms are involved in CNS scar formation. The expression of the secreted chemorepellent sema III following CNS injury provides the first evidence that chemorepulsive semaphorins may contribute to the inhibitory effects exerted by scars on the outgrowth of injured CNS neurites. The vigorous regrowth of injured axons in the absence of sema III following early neonatal lesions is consistent with this notion. The inactivation of sema III in scar tissue by either antibody perturbation or by genetic or pharmacological intervention could be a powerful means to promote long-distance regeneration in the adult CNS.


Subject(s)
Central Nervous System/injuries , Cicatrix/genetics , Gene Expression Regulation , Glycoproteins/genetics , Animals , Cerebral Cortex/injuries , Cicatrix/metabolism , Glycoproteins/biosynthesis , Nerve Regeneration , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuropilin-1 , Olfactory Pathways/injuries , Perforant Pathway/injuries , RNA, Messenger/biosynthesis , Rats , Semaphorin-3A , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism
9.
Neuron ; 21(5): 1079-92, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9856463

ABSTRACT

Neuropilins bind secreted members of the semaphorin family of proteins. Neuropilin-1 is a receptor for Sema III. Here, we show that neuropilin-2 is a receptor for the secreted semaphorin Sema IV and acts selectively to mediate repulsive guidance events in discrete populations of neurons. neuropilin-2 and semaIV are expressed in strikingly complementary patterns during neurodevelopment. The extracellular complement-binding (CUB) and coagulation factor domains of neuropilin-2 confer specificity to the Sema IV repulsive response, and these domains of neuropilin-1 are necessary and sufficient for binding of the Sema III semaphorin (sema) domain. The coagulation factor domains alone are necessary and sufficient for binding of the Sema III immunoglobulin- (Ig-) basic domain and the unrelated ligand, vascular endothelial growth factor (VEGF). Lastly, neuropilin-1 can homomultimerize and form heteromultimers with neuropilin-2. These results provide insight into how interactions between neuropilins and secreted semaphorins function to coordinate repulsive axon guidance during neurodevelopment.


Subject(s)
Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/physiology , Animals , Cells, Cultured , Coculture Techniques , Embryo, Mammalian , Ganglia, Spinal/cytology , Ganglia, Sympathetic/cytology , Glycoproteins/physiology , Humans , Nerve Growth Factors/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Neurons/metabolism , Neuropilin-1 , Rats , Receptors, Cell Surface/chemistry , Semaphorin-3A , Superior Cervical Ganglion/chemistry , Superior Cervical Ganglion/cytology
10.
Science ; 282(5395): 1904-6, 1998 Dec 04.
Article in English | MEDLINE | ID: mdl-9836643

ABSTRACT

Cortical neurons communicate with various cortical and subcortical targets by way of stereotyped axon projections through the white matter. Slice overlay experiments indicate that the initial growth of cortical axons toward the white matter is regulated by a diffusible chemorepulsive signal localized near the marginal zone. Semaphorin III is a major component of this diffusible signal, and cortical neurons transduce this signal by way of the neuropilin-1 receptor. These observations indicate that semaphorin-neuropilin interactions play a critical role in the initial patterning of projections in the developing cortex.


Subject(s)
Axons/physiology , Cerebral Cortex/cytology , Glycoproteins/physiology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/physiology , Neurons, Efferent/physiology , Animals , Cell Line , Cerebral Cortex/embryology , Coculture Techniques , Gene Targeting , Glycoproteins/genetics , Humans , Mice , Neurons, Efferent/cytology , Neuropilin-1 , Rats , Recombinant Proteins/metabolism , Semaphorin-3A , Signal Transduction
11.
Exp Neurol ; 153(2): 313-27, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9784290

ABSTRACT

The competence of neurons to regenerate depends on their ability to initiate a program of gene expression supporting growth and on the growth-permissive properties of glial cells in the distal stump of the injured nerve. Most studies on intrinsic molecular mechanisms governing peripheral nerve regeneration have focussed on the lesion-induced expression of proteins promoting growth cone motility, neurite extension, and adhesion. However, little is known about the expression of intrinsic chemorepulsive proteins and their receptors, after peripheral nerve injury and during nerve regeneration. Here we report the effect of peripheral nerve injury on the expression of the genes encoding sema III/coll-1 and its receptor neuropilin-1, which are known to be expressed in adult sensory and/or motor neurons. We have shown that peripheral nerve crush or transection results in a decline in sema III/coll-1 mRNA expression in injured spinal and facial motor neurons. This decline was paralleled by an induction in the expression of the growth-associated protein B-50/GAP-43. As sema III/coll-1 returned to normal levels following nerve crush, B-50/GAP-43 returned to precrush levels. Thus, the decline in sema III/coll-1 mRNA coincided with sensory and motor neuron regeneration. A sustained decline in sema III/coll-1 mRNA expression was found when regeneration was blocked by nerve transection and ligation. No changes were observed in neuropilin-1 mRNA levels after injury to sensory and motor neurons, suggesting that regenerating peripheral neurons continue to be sensitive to sema III/coll-1. Therefore we propose that a decreased expression of sema III/coll-1, one of the major ligands for neuropilin-1, during peripheral nerve regeneration is an important molecular event that is part of the adaptive response related to the success of regenerative neurite outgrowth occurring following peripheral nerve injury.


Subject(s)
Gene Expression Regulation , Glycoproteins/genetics , Nerve Regeneration/physiology , Sciatic Nerve/physiology , Transcription, Genetic , Animals , GAP-43 Protein/genetics , In Situ Hybridization , Male , Nerve Crush , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Neuropilin-1 , RNA Probes , RNA, Complementary , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Semaphorin-3A , Time Factors
12.
J Neurotrauma ; 15(6): 387-97, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9624624

ABSTRACT

Although much progress has been made, current treatments of peripheral nerve damage mostly result in only partial recovery. Local production of neurite outgrowth-promoting molecules, such as neurotrophins and/or cell adhesion molecules, at the site of damage may be used as a new means to promote the regeneration process. We have now explored the ability of an adenoviral vector encoding the reporter gene LacZ (Ad-LacZ) to direct the expression of a foreign gene to Schwann cells of intact and crushed rat sciatic nerves. Infusion of 8 x 10(7) PFU Ad-LacZ in the intact sciatic nerve resulted in the transduction of many Schwann cells with high levels of transgene expression lasting at least up to 12 days following viral vector administration. The efficacy of adenoviral vector delivery to a crushed nerve was investigated using three strategies. Injection of the adenoviral vector at the time of, or immediately after, a crush resulted in the transduction of only a few Schwann cells. Administration of the adenoviral vector the day after the crush resulted in the transduction of a similar number of Schwann cells 5 days after administration, as observed in uncrushed nerves. Regenerating nerve fibers were closely associated with beta-galactosidase-positive Schwann cells, indicating that the capacity of transduced Schwann cells to guide regenerating fibers was not altered. These results imply that the expression of growth-promoting proteins through adenoviral vector-mediated gene transfer may be a realistic option to promote peripheral nerve regeneration.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Sciatic Nerve/injuries , Wounds, Nonpenetrating/therapy , Animals , Genes, Reporter/genetics , Lac Operon/genetics , Male , Nerve Crush , Nerve Fibers/physiology , Nerve Regeneration/physiology , Rats , Rats, Wistar , Schwann Cells/physiology , Sciatic Nerve/pathology , Transduction, Genetic , Wounds, Nonpenetrating/pathology
13.
Anat Embryol (Berl) ; 197(3): 177-85, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9543336

ABSTRACT

TAG-1/axonin-1 is a neuronal cell adhesion molecule of the immunoglobulin superfamily. It is predominantly expressed during neural development and has been reported to be involved in axonal growth and pathfinding. Here, the expression of TAG-1/axonin-1 was investigated anatomically in the adult mouse brain by in situ hybridization using digoxigenin-labeled cRNA probes. Low levels of TAG-1/axonin-1 could be detected in cerebellar granule cells, in tufted and mitral cells of the olfactory bulb, and in pyramidal cells of area CA1 and CA3 of the hippocampus. We suspect that the expression of TAG-1/axonin-1 in these structures of the adult brain may serve neural plasticity.


Subject(s)
Brain Chemistry , Cell Adhesion Molecules, Neuronal/analysis , Animals , Cerebellum/chemistry , Contactin 2 , Hippocampus/chemistry , In Situ Hybridization , Male , Membrane Glycoproteins/analysis , Mice , Mice, Inbred ICR , Neuronal Plasticity/physiology , Olfactory Bulb/chemistry
14.
J Neurosci Res ; 52(1): 27-42, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9556027

ABSTRACT

Alterations in neuronal connectivity of the mature central nervous system (CNS) appear to depend on a delicate balance between growth-promoting and growth-inhibiting molecules. To begin to address a potential role of the secreted chemorepulsive protein semaphorin(D)III/collapsin-1 (semaIII/coll-1) in structural plasticity during adulthood, we used high-resolution nonradioactive in situ hybridization to identify neural structures that express semaIII/coll-1 mRNA in the mature rat and human brain. SemaIII/coll-1 was expressed in distinct but anatomically and functionally linked structures of the adult nervous system. The olfactory-hippocampal pathway displayed semaIII/coll-1 expression in a continuum of neuronal structures, including mitral and tufted cells of the olfactory bulb, olfactory tubercle, and piriform cortex; and distinct nuclei of the amygdaloid complex, the superficial layers of the entorhinal cortex, and the subiculum of the hippocampal formation. In addition, prominent labeling was found in neuronal components of the motor system, particularly in cerebellar Purkinje cells and in subpopulations of cranial and spinal motoneurons. Retrograde tracing combined with in situ hybridization also revealed that the staining of semaIII/coll-1 within the entorhinal cortex was present in the stellate neurons that project via the perforant path to the molecular layer of the dentate gyrus. Like in the rat, the human brain displayed discrete expression of semaIII/coll-1. Among the structures examined, the most prominent staining was observed in the cellular islands of the superficial layers of the human entorhinal cortex. The constitutive expression of the chemorepellent semaIII/coll-1 in discrete populations of neurons in the mature rat and human CNS raises the possibility that, in addition to its function as repulsive axon guidance cue during development, semaIII/coll-1 might be involved in restricting structural changes that occur in the wiring of the intact CNS.


Subject(s)
Aging/metabolism , Brain/metabolism , Gene Expression Regulation, Developmental , Glycoproteins/biosynthesis , Adult , Aged , Animals , Brain/cytology , Brain/growth & development , Cerebellum/cytology , Cerebellum/metabolism , DNA Primers , Female , Glycoproteins/analysis , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Middle Aged , Nerve Growth Factors/analysis , Nerve Growth Factors/biosynthesis , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Organ Specificity , Polymerase Chain Reaction , RNA Probes , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Semaphorin-3A , Transcription, Genetic
17.
J Neurosci ; 17(17): 6575-86, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9254670

ABSTRACT

B-50/GAP-43 is an intraneuronal membrane-associated growth cone protein with an important role in axonal growth and regeneration. By using adenoviral vector-directed expression of B-50/GAP-43 we studied the morphogenic action of B-50/GAP-43 in mature primary olfactory neurons that have established functional synaptic connections. B-50/GAP-43 induced gradual alterations in the morphology of olfactory synapses. In the first days after overexpression, small protrusions originating from the preterminal axon shaft and from the actual synaptic bouton were formed. With time the progressive formation of multiple ultraterminal branches resulted in axonal labyrinths composed of tightly packed sheaths of neuronal membrane. Thus, B-50/GAP-43 is a protein that can promote neuronal membrane expansion at synaptic boutons. This function of B-50/GAP-43 suggests that this protein may subserve an important role in ongoing structural synaptic plasticity in adult neurons and in neuronal membrane repair after injury to synaptic fields.


Subject(s)
Adenoviridae/genetics , Axons/physiology , Genetic Vectors , Membrane Glycoproteins/metabolism , Nerve Endings/physiology , Nerve Tissue Proteins/metabolism , Olfactory Pathways/physiology , Animals , Chlorocebus aethiops , Epithelium/physiology , GAP-43 Protein , Neuronal Plasticity , Synapses/ultrastructure , Synaptic Transmission , Vero Cells
18.
Cell ; 90(4): 753-62, 1997 Aug 22.
Article in English | MEDLINE | ID: mdl-9288754

ABSTRACT

The semaphorin family contains a large number of phylogenetically conserved proteins and includes several members that have been shown to function in repulsive axon guidance. Semaphorin III (Sema III) is a secreted protein that in vitro causes neuronal growth cone collapse and chemorepulsion of neurites, and in vivo is required for correct sensory afferent innervation and other aspects of development. The mechanism of Sema III function, however, is unknown. Here, we report that neuropilin, a type I transmembrane protein implicated in aspects of neurodevelopment, is a Sema III receptor. We also describe the identification of neuropilin-2, a related neuropilin family member, and show that neuropilin and neuropilin-2 are expressed in overlapping, yet distinct, populations of neurons in the rat embryonic nervous system.


Subject(s)
Antigens, Surface/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Antibodies/metabolism , COS Cells , Ganglia, Spinal/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neuropilin-1 , Rats , Receptors, Cell Surface/genetics , Semaphorin-3A , Spinal Cord/cytology , Spinal Cord/growth & development
19.
J Neurosci Methods ; 71(1): 85-98, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9125378

ABSTRACT

In this paper a detailed protocol is presented for neuroscientists planning to start work on first generation recombinant adenoviral vectors as gene transfer agents for the nervous system. The performance of a prototype adenoviral vector encoding the bacterial lacZ gene as a reporter was studied, following direct injection in several regions of the central and peripheral nervous system. The distribution of the cells expressing the transgene appears to be determined by natural anatomical boundaries and possibly by the degree of myelinization of a particular brain region. In highly myelinated areas with a compact cellular structure (e.g. the cortex and olfactory bulb) the spread of the viral vector is limited to the region close to the injection needle, while in areas with a laminar structure (e.g. the hippocampus and the eye) more widespread transgene expression is observed. Retrograde transport of the viral vector may serve as an attractive alternative route of transgene delivery. A time course of expression of beta-galactosidase in neural cells in the facial nucleus revealed high expression during the first week after AdLacZ injection. However, a significant decline in transgene expression during the second and third week was observed. This may be caused by an immune response against the transduced cells or by silencing of the cytomegalovirus promoter used to drive transgene expression. Taken together, the data underscore that for each application of adenoviral vectors as gene transfer agents in the nervous system it is important to examine vector spread in and infectability of the neural structure that is subject to genetic modification.


Subject(s)
Adenoviruses, Human/genetics , Defective Viruses/genetics , Genetic Therapy/methods , Genetic Vectors , Neuroglia , Neurons , Transfection/methods , Administration, Intranasal , Animals , Astrocytes/metabolism , Astrocytes/virology , Brain , Cell Line , Central Nervous System/cytology , Eye , Female , Gene Expression Regulation , Genes, Reporter , Genes, Synthetic , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Injections , Male , Neuroglia/metabolism , Neuroglia/virology , Neurons/metabolism , Neurons/virology , Oligodendroglia/metabolism , Oligodendroglia/virology , Peripheral Nervous System/cytology , Pregnancy , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Schwann Cells/metabolism , Schwann Cells/virology , Sciatic Nerve , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
20.
J Neurosci Methods ; 71(1): 99-111, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9125379

ABSTRACT

By homologous recombination, a first-generation adenovirus-based gene transfer vector, AdCMVax-1, was constructed as a means of manipulating the expression level of the axonal cell adhesion molecule axonin-1 in neurons and glial cells. AdCMVax-1 harbours the entire coding region of the chicken axonin-1 cDNA under the transcriptional control of the Cytomegalovirus enhancer/promoter in the early-region 1 of the viral genome. Characterization of AdCMVax-1 in vitro revealed highly efficient gene transfer and expression of recombinant axonin-1 in neurons and glial cells of dissociated rat dorsal root ganglia. Similar to its native counterpart, virus-derived axonin-1 was detected on the cell body, neurites, and growth cones of transduced neurons, occurred in a secreted and membrane-associated form, and could be cleaved from the membrane with phosphatidylinositol-specific phospholipase C. Functional characterization of recombinant axonin-1 revealed the same binding properties as previously reported for native axonin-1 isolated from the vitreous fluid of chicken embryos. In vivo gene transfer was studied by stereotactic injection of AdCMVax-1 in the dentate gyrus of the hippocampus and the facial nucleus in the brainstem of adult Wistar rats and revealed high level expression of recombinant axonin-1 in a subset of hippocampal neurons and motor neurons in the facial nucleus.


Subject(s)
Adenoviruses, Human/genetics , Cell Adhesion Molecules, Neuronal/genetics , Defective Viruses/genetics , Genetic Vectors , Neurons/metabolism , Animals , Cell Adhesion Molecules, Neuronal/biosynthesis , Contactin 2 , Cytomegalovirus/genetics , DNA, Complementary/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/virology , Enhancer Elements, Genetic , Facial Nerve/metabolism , Facial Nerve/virology , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Gene Expression Regulation , Genome, Viral , HeLa Cells , Humans , Injections , Mice , Neurons/virology , Promoter Regions, Genetic , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Stereotaxic Techniques , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL