1
|
Gavid M, Coulomb L, Thomas J, Aouimeur I, Verhoeven P, Mentek M, Dumollard JM, Forest F, Prades JM, Thuret G, Gain P, He Z. Technique of flat-mount immunostaining for mapping the olfactory epithelium and counting the olfactory sensory neurons. PLoS One 2023; 18:e0280497. [PMID: 36649285 PMCID: PMC9844923 DOI: 10.1371/journal.pone.0280497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
The pathophysiology underlying olfactory dysfunction is still poorly understood, and more efficient biomolecular tools are necessary to explore this aspect. Immunohistochemistry (IHC) on cross sections is one of the major tools to study the olfactory epithelium (OE), but does not allow reliable counting of olfactory sensory neurons (OSNs) or cartography of the OE. In this study, we want to present an easy immunostaining technique to compensate for these defects of IHC. Using the rat model, we first validated and pre-screened the key OSN markers by IHC on cross sections of the OE. Tuj-1, OMP, DCX, PGP9.5, and N-cadherin were selected for immunostaining on flat-mounted OE because of their staining of OSN dendrites. A simple technique for immunostaining on flat-mounted septal OE was developed: fixation of the isolated septum mucosa in 0.5% paraformaldehyde (PFA) preceded by pretreatment of the rat head in 1% PFA for 1 hour. This technique allowed us to correctly reveal the olfactory areas using all the 5 selected markers on septum mucosa. By combining the mature OSN marker (OMP) and an immature OSN marker (Tuj-1), we quantified the mature (OMP+, Tuj-1-), immature (OMP-, Tuj-1+), transitory (OMP+, Tuj-1+) and total OSN density on septal OE. They were respectively 42080 ± 11820, 49384 ± 7134, 14448 ± 5865 and 105912 ± 13899 cells per mm2 (mean ± SD). Finally, the same immunostaining technique described above was performed with Tuj-1 for OE cartography on ethmoid turbinates without flat-mount.
Collapse
Affiliation(s)
- Marie Gavid
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
- Department of Otorhinolaryngology, CHU of Saint-Etienne, Saint-Etienne, France
| | - Louise Coulomb
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Justin Thomas
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Inès Aouimeur
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Paul Verhoeven
- CIRI, GIMAP Team, INSERM U1111, CNRS UMR5308, University of Lyon, University of Saint-Etienne, Saint-Etienne, France
| | - Marielle Mentek
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Jean-Marc Dumollard
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
- Department of Pathology, CHU of Saint-Etienne, Saint-Etienne, France
| | - Fabien Forest
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
- Department of Pathology, CHU of Saint-Etienne, Saint-Etienne, France
| | - Jean-Michel Prades
- Department of Otorhinolaryngology, CHU of Saint-Etienne, Saint-Etienne, France
| | - Gilles Thuret
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Philippe Gain
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
| | - Zhiguo He
- Laboratory BIIO (EA2521), Jean Monnet University, Saint-Etienne, France
- * E-mail:
| |
Collapse
|
2
|
Blits B, Boer GJ, Verhaagen J. Pharmacological, Cell, and Gene Therapy Strategies to Promote Spinal Cord Regeneration. Cell Transplant 2017. [DOI: 10.3727/000000002783985521] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review, recent studies using pharmacological treatment, cell transplantation, and gene therapy to promote regeneration of the injured spinal cord in animal models will be summarized. Pharmacological and cell transplantation treatments generally revealed some degree of effect on the regeneration of the injured ascending and descending tracts, but further improvements to achieve a more significant functional recovery are necessary. The use of gene therapy to promote repair of the injured nervous system is a relatively new concept. It is based on the development of methods for delivering therapeutic genes to neurons, glia cells, or nonneural cells. Direct in vivo gene transfer or gene transfer in combination with (neuro)transplantation (ex vivo gene transfer) appeared powerful strategies to promote neuronal survival and axonal regrowth following traumatic injury to the central nervous system. Recent advances in understanding the cellular and molecular mechanisms that govern neuronal survival and neurite outgrowth have enabled the design of experiments aimed at viral vector-mediated transfer of genes encoding neurotrophic factors, growth-associated proteins, cell adhesion molecules, and antiapoptotic genes. Central to the success of these approaches was the development of efficient, nontoxic vectors for gene delivery and the acquirement of the appropriate (genetically modified) cells for neurotransplantation. Direct gene transfer in the nervous system was first achieved with herpes viral and E1-deleted adenoviral vectors. Both vector systems are problematic in that these vectors elicit immunogenic and cytotoxic responses. Adeno-associated viral vectors and lentiviral vectors constitute improved gene delivery systems and are beginning to be applied in neuroregeneration research of the spinal cord. Ex vivo approaches were initially based on the implantation of genetically modified fibroblasts. More recently, transduced Schwann cells, genetically modified pieces of peripheral nerve, and olfactory ensheathing glia have been used as implants into the injured spinal cord.
Collapse
Affiliation(s)
- Bas Blits
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Gerard J. Boer
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| | - Joost Verhaagen
- Graduate School Neurosciences Amsterdam, Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ Amsterdam-ZO, The Netherlands
| |
Collapse
|
3
|
Song JN, Liu ZW, Sui L, Zhang BF, Zhao YL, Ma XD, Gu H. Dynamic expression of nerve growth factor and its receptor TrkA after subarachnoid hemorrhage in rat brain. Neural Regen Res 2016; 11:1278-84. [PMID: 27651776 PMCID: PMC5020827 DOI: 10.4103/1673-5374.189193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Delayed ischemic neurologic deficit after subarachnoid hemorrhage results from loss of neural cells. Nerve growth factor and its receptor TrkA may promote regeneration of neural cells, but their expression after subarachnoid hemorrhage remains unclear. In the present study, a rat model of subarachnoid hemorrhage was established using two injections of autologous blood into the cistern magna. Immunohisto-chemical staining suggested that the expression of nerve growth factor and TrkA in the cerebral cortex and brainstem increased at 6 hours, peaked at 12 hours and decreased 1 day after induction of subarachnoid hemorrhage, whereas the expression in the hippocampus increased at 6 hours, peaked on day 1, and decreased 3 days later. Compared with those for the rats in the sham and saline groups, neurobehavioral scores decreased significantly 12 hours and 3 days after subarachnoid hemorrhage (P < 0.05). These results suggest that the expression of nerve growth factor and its receptor TrkA is dynamically changed in the rat brain and may thus participate in neuronal survival and nerve regeneration after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Jin-Ning Song
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zun-Wei Liu
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Long Sui
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; Department of Neurosurgery, the 521 Hospital of China North Industries Group, Xi'an, Shaanxi Province, China
| | - Bin-Fei Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yong-Lin Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xu-Dong Ma
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hua Gu
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
4
|
van Niekerk EA, Tuszynski MH, Lu P, Dulin JN. Molecular and Cellular Mechanisms of Axonal Regeneration After Spinal Cord Injury. Mol Cell Proteomics 2015; 15:394-408. [PMID: 26695766 DOI: 10.1074/mcp.r115.053751] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Indexed: 12/28/2022] Open
Abstract
Following axotomy, a complex temporal and spatial coordination of molecular events enables regeneration of the peripheral nerve. In contrast, multiple intrinsic and extrinsic factors contribute to the general failure of axonal regeneration in the central nervous system. In this review, we examine the current understanding of differences in protein expression and post-translational modifications, activation of signaling networks, and environmental cues that may underlie the divergent regenerative capacity of central and peripheral axons. We also highlight key experimental strategies to enhance axonal regeneration via modulation of intraneuronal signaling networks and the extracellular milieu. Finally, we explore potential applications of proteomics to fill gaps in the current understanding of molecular mechanisms underlying regeneration, and to provide insight into the development of more effective approaches to promote axonal regeneration following injury to the nervous system.
Collapse
Affiliation(s)
- Erna A van Niekerk
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093;
| | - Mark H Tuszynski
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Paul Lu
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093; §Veterans Administration Medical Center, San Diego, CA 92161
| | - Jennifer N Dulin
- From the ‡Department of Neurosciences, University of California, San Diego, La Jolla, CA, 92093
| |
Collapse
|
5
|
Fagoe ND, van Heest J, Verhaagen J. Spinal cord injury and the neuron-intrinsic regeneration-associated gene program. Neuromolecular Med 2014; 16:799-813. [PMID: 25269879 DOI: 10.1007/s12017-014-8329-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/20/2014] [Indexed: 12/14/2022]
Abstract
Spinal cord injury (SCI) affects millions of people worldwide and causes a significant physical, emotional, social and economic burden. The main clinical hallmark of SCI is the permanent loss of motor, sensory and autonomic function below the level of injury. In general, neurons of the central nervous system (CNS) are incapable of regeneration, whereas injury to the peripheral nervous system is followed by axonal regeneration and usually results in some degree of functional recovery. The weak neuron-intrinsic regeneration-associated gene (RAG) response upon injury is an important reason for the failure of neurons in the CNS to regenerate an axon. This response consists of the expression of many RAGs, including regeneration-associated transcription factors (TFs). Regeneration-associated TFs are potential key regulators of the RAG program. The function of some regeneration-associated TFs has been studied in transgenic and knock-out mice and by adeno-associated viral vector-mediated overexpression in injured neurons. Here, we review these studies and propose that AAV-mediated gene delivery of combinations of regeneration-associated TFs is a potential strategy to activate the RAG program in injured CNS neurons and achieve long-distance axon regeneration.
Collapse
Affiliation(s)
- Nitish D Fagoe
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands,
| | | | | |
Collapse
|
6
|
de Winter F, Hoyng S, Tannemaat M, Eggers R, Mason M, Malessy M, Verhaagen J. Gene therapy approaches to enhance regeneration of the injured peripheral nerve. Eur J Pharmacol 2013; 719:145-152. [DOI: 10.1016/j.ejphar.2013.04.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/28/2013] [Accepted: 04/03/2013] [Indexed: 01/26/2023]
|
7
|
Gau P, Rodriguez S, De Leonardis C, Chen P, Lin DM. Air-assisted intranasal instillation enhances adenoviral delivery to the olfactory epithelium and respiratory tract. Gene Ther 2010; 18:432-6. [PMID: 21085195 DOI: 10.1038/gt.2010.153] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intranasal instillation is used to deliver adenoviral vectors to the olfactory epithelium and respiratory tract. The success of this approach, however, has been tempered by inconsistent infectivity in both the epithelium and lungs. Infection of the epithelium may be hampered in part by the convoluted structure of the cavity, the presence of mucus or poor airflow in the posterior cavity. Delivery of adenovirus to the lungs can be uneven in the various lobes and distal bronchioles may be poorly infected. Current approaches to circumvent these issues rely principally on intubation or intratracheal instillation. Here we describe a technique that significantly improves adenoviral infectivity rates without requiring surgical intervention. We use compressed air to increase circulation of instilled adenovirus, resulting in enhanced infection in both the epithelium and lungs. This procedure is straightforward, simple to perform and requires no specialized equipment. In the epithelium, neurons and sustentacular cells are both labeled. In the lungs, all lobes can be infected, with penetration to the most distal bronchioles. The use of compressed air will likely also be useful for enhancing the distribution of other, desired agents within the epithelium, central nervous system and respiratory tract.
Collapse
Affiliation(s)
- P Gau
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | |
Collapse
|
8
|
El Meskini R, Crabtree KL, Cline LB, Mains RE, Eipper BA, Ronnett GV. ATP7A (Menkes protein) functions in axonal targeting and synaptogenesis. Mol Cell Neurosci 2007; 34:409-21. [PMID: 17215139 PMCID: PMC1876716 DOI: 10.1016/j.mcn.2006.11.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Revised: 11/11/2006] [Accepted: 11/20/2006] [Indexed: 12/19/2022] Open
Abstract
Menkes disease (MD) is a neurodegenerative disorder caused by mutations in the copper transporter, ATP7A, a P-type ATPase. We previously used the olfactory system to demonstrate that ATP7A expression is developmentally, not constitutive, regulated, peaking during synaptogenesis when it is highly expressed in extending axons in a copper-independent manner. Although not known to be associated with axonal functions, we explored the possibility that the inability of mutant ATP7A to support axon outgrowth contributes to the neurodegeneration seen in MD. In vivo analysis of the olfactory system in mottled brindled (Atp7aMobr) mice, a rodent model for MD, demonstrates that ATP7A deficiency affects olfactory sensory neuron (OSN) maturation. Disrupted OSN axonal projections and mitral/tufted cell dendritic growth lead to altered synapse integrity and glomerular disorganization in the olfactory bulbs of Atp7aMobr mice. Our data indicate that the neuronal abnormalities observed in MD are a result of specific age-dependent developmental defects. This study demonstrates a role for ATP7A and/or copper in axon outgrowth and synaptogenesis, and will further help identify the cause of the neuropathology that characterizes MD.
Collapse
Affiliation(s)
- Rajaâ El Meskini
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | | | | | | | | | | |
Collapse
|
9
|
Rossi F, Gianola S, Corvetti L. Regulation of intrinsic neuronal properties for axon growth and regeneration. Prog Neurobiol 2006; 81:1-28. [PMID: 17234322 DOI: 10.1016/j.pneurobio.2006.12.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/04/2006] [Accepted: 12/05/2006] [Indexed: 01/29/2023]
Abstract
Regulation of neuritic growth is crucial for neural development, adaptation and repair. The intrinsic growth potential of nerve cells is determined by the activity of specific molecular sets, which sense environmental signals and sustain structural extension of neurites. The expression and function of these molecules are dynamically regulated by multiple mechanisms, which adjust the actual growth properties of each neuron population at different ontogenetic stages or in specific conditions. The neuronal potential for axon elongation and regeneration are restricted at the end of development by the concurrent action of several factors associated with the final maturation of neurons and of the surrounding tissue. In the adult, neuronal growth properties can be significantly modulated by injury, but they are also continuously tuned in everyday life to sustain physiological plasticity. Strict regulation of structural remodelling and neuritic elongation is thought to be required to maintain specific patterns of connectivity in the highly complex mammalian CNS. Accordingly, procedures that neutralize such mechanisms effectively boost axon growth in both intact and injured nervous system. Even in these conditions, however, aberrant connections are only formed in the presence of unusual external stimuli or experience. Therefore, growth regulatory mechanisms play an essentially permissive role by setting the responsiveness of neural circuits to environmental stimuli. The latter exert an instructive action and determine the actual shape of newly formed connections. In the light of this notion, efficient therapeutic interventions in the injured CNS should combine targeted manipulations of growth control mechanisms with task-specific training and rehabilitation paradigms.
Collapse
Affiliation(s)
- Ferdinando Rossi
- Rita Levi Montalcini Centre for Brain Repair, Department of Neuroscience, University of Turin, Corso Raffaello 30, I-10125 Turin, Italy.
| | | | | |
Collapse
|
10
|
Venkatraman G, Behrens M, Pyrski M, Margolis FL. Expression of Coxsackie-Adenovirus receptor (CAR) in the developing mouse olfactory system. ACTA ACUST UNITED AC 2006; 34:295-305. [PMID: 16841169 DOI: 10.1007/s11068-005-8359-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 11/14/2005] [Accepted: 11/14/2005] [Indexed: 10/24/2022]
Abstract
Interest in manipulating gene expression in olfactory sensory neurons (OSNs) has led to the use of adenoviruses (AdV) as gene delivery vectors. OSNs are the first order neurons in the olfactory system and the initial site of odor detection. They are highly susceptible to adenovirus infection although the mechanism is poorly understood. The Coxsackie-Adenovirus receptor (CAR) and members of the integrin family have been implicated in the process of AdV infection in various systems. Multiple serotypes of AdV efficiently bind to the CAR, leading to entry and infection of the host cell by a mechanism that can also involve integrins. Cell lines that do not express CAR are relatively resistant, but not completely immune to AdV infection, suggesting that other mechanisms participate in mediating AdV attachment and entry. Using in situ hybridization and western blot analyses, we show that OSNs and olfactory bulbs (OB) of mice express abundant CAR mRNA at embryonic and neonatal stages, with progressive diminution during postnatal development. By contrast to the olfactory epithelium (OE), CAR mRNA is still present in the adult mouse OB. Furthermore, despite a similar postnatal decline, CAR protein expression in the OE and OB of mice continues into adulthood. Our results suggest that the robust AdV infection observed in the postnatal olfactory system is mediated by CAR and that expression of even small amounts of CAR protein as seen in the adult rodent, permits efficient AdV infection and entry. CAR is an immunoglobulin domain-containing protein that bears homology to cell-adhesion molecules suggesting the possibility that it may participate in organization of the developing olfactory system.
Collapse
Affiliation(s)
- Giri Venkatraman
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, 21201, USA
| | | | | | | |
Collapse
|
11
|
Abstract
For regrowth of injured nerve fibers following spinal cord injury (SCI), the environment must be favorable for axonal growth. The delivery of a therapeutic gene, beneficial for axonal growth, into the central nervous system for repair can be accomplished in many ways. Perhaps the most simple and elegant strategy is the so-called direct gene therapy approach that uses a single injection for delivery of a gene therapy vehicle. Among the vectors that have been used to transduce neural tissue in vivo are non-viral, herpes simplex viral, adeno-associated viral, adenoviral, and lentiviral vectors, each with their own merits and limitations. Many studies have been undertaken using direct gene therapy, ranging from strategies for neuroprotection to axonal growth promotion at the injury site, dorsal root injury repair, and initiation of a growth-supporting genetic program. The limitations and successes of direct gene transfer for spinal cord repair are discussed in this review.
Collapse
Affiliation(s)
- Bas Blits
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida 33101, USA
| | | |
Collapse
|
12
|
Youngentob SL, Pyrski MM, Margolis FL. Adenoviral vector-mediated rescue of the OMP-null behavioral phenotype: enhancement of odorant threshold sensitivity. Behav Neurosci 2004; 118:636-42. [PMID: 15174942 DOI: 10.1037/0735-7044.118.3.636] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mice from which the olfactory marker protein (OMP) gene has been deleted demonstrate a number of neurophysiologic and behavioral defects that suggest OMP is an important component in olfactory signal transduction and is critically involved in odor processing. Recently, the potential pleiotropic effects of gene deletion were addressed by adenoviral vector-mediated rescue of the neurophysiologic defects, in vivo. As a complement to this study, the authors used a recombinant adenoviral vector to transiently introduce OMP into olfactory sensory neurons of adult OMP-null mice and, using psychophysical methods, demonstrated the resulting reacquisition of behavioral function subsequent to gene replacement. The rescue of the OMP-null behavioral phenotype further supports the hypothesis that OMP is an important component in olfactory signal amplification and/or transduction processing.
Collapse
Affiliation(s)
- Steven L Youngentob
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY, USA.
| | | | | |
Collapse
|
13
|
Schmidt JT. Activity-driven sharpening of the retinotectal projection: the search for retrograde synaptic signaling pathways. ACTA ACUST UNITED AC 2004; 59:114-33. [PMID: 15007831 DOI: 10.1002/neu.10343] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patterned visual activity, acting via NMDA receptors, refines developing retinotectal maps by shaping individual retinal arbors. Because NMDA receptors are postsynaptic but the retinal arbors are presynaptic, there must be retrograde signals generated downstream of Ca(++) entry through NMDA receptors that direct the presynaptic retinal terminals to stabilize and grow or to withdraw. This review defines criteria for retrograde synaptic messengers, and then applies them to the leading candidates: nitric oxide (NO), brain-derived neurotrophic factor (BDNF), and arachidonic acid (AA). NO is not likely to be a general mechanism, as it operates only in selected projections of warm blooded vertebrates to speed up synaptic refinement, but is not essential. BDNF is a neurotrophin with strong growth promoting properties and complex interactions with activity both in its release and receptor signaling, but may modulate rather than mediate the retrograde signaling. AA promotes growth and stabilization of synaptic terminals by tapping into a pre-existing axonal growth-promoting pathway that is utilized by L1, NCAM, N-cadherin, and FGF and acts via PKC, GAP43, and F-actin stabilization, and it shares some overlap with BDNF pathways. The actions of both are consistent with recent demonstrations that activity-driven stabilization includes directed growth of new synaptic contacts. Certain nondiffusible factors (synapse-specific CAMs, ephrins, neurexin/neuroligin, and matrix molecules) may also play a role in activity-driven synapse stabilization. Interactions between these pathways are discussed.
Collapse
Affiliation(s)
- John T Schmidt
- Department of Biological Sciences and Center for Neuroscience Research, University at Albany-SUNY, 1400 Washington Avenue, Albany, New York 12222, USA.
| |
Collapse
|
14
|
Gianola S, Rossi F. GAP-43 overexpression in adult mouse Purkinje cells overrides myelin-derived inhibition of neurite growth. Eur J Neurosci 2004; 19:819-30. [PMID: 15009129 DOI: 10.1111/j.0953-816x.2004.03190.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Up-regulation of growth-associated proteins in adult neurons promotes axon regeneration and neuritic elongation onto nonpermissive substrates. To investigate the interaction between these molecules and myelin-related inhibitory factors, we examined transgenic mice in which overexpression of the growth-associated protein GAP-43 is driven by the Purkinje cell-specific promoter L7. Contrary to their wild-type counterparts, which have extremely poor regenerative capabilities, axotomized transgenic Purkinje cells exhibit profuse sprouting along the intracortical neurite and at the severed stump [Buffo et al. (1997) J. Neurosci., 17, 8778-8791]. Here, we investigated the relationship between such sprouting axons and oligodendroglia to ask whether GAP-43 overexpression enables Purkinje neurites to overcome myelin-derived inhibition. Intact transgenic Purkinje axons display normal morphology and myelination. Following injury, however, many GAP-43-overexpressing neurite stumps are devoid of myelin cover and sprout into white matter regions containing densely packed myelin and Nogo-A- or MAG-immunopositive oligodendrocytes. The intracortical segments of these neurites show focal accumulations of GAP-43, which are associated with disrupted or retracted myelin sheaths. Numerous sprouts originate from such demyelinated segments and spread into the granular layer. Some myelin loss, though not axon sprouting, is also evident in wild-type mice, but this phenomenon is definitely more rapid and extensive in transgenic cerebella. Thus, GAP-43-overexpressing Purkinje axons are endowed with enhanced capabilities for growing into nonpermissive territories and show a pronounced tendency to lose myelin. Our observations suggest that accumulation of GAP-43 along precise axon segments disrupts the normal axon-glia interaction and enhances the retraction of oligodendrocytic processes to facilitate the outgrowth of neuritic sprouts.
Collapse
Affiliation(s)
- Sara Gianola
- Rita Levi Montalcini Centre for Brain Repair, Department of Neuroscience, University of Turin, Corso Raffaello 30, I-10125 Turin, Italy
| | | |
Collapse
|
15
|
Abstract
Nerve regeneration is a complex biological phenomenon. In the peripheral nervous system, nerves can regenerate on their own if injuries are small. Larger injuries must be surgically treated, typically with nerve grafts harvested from elsewhere in the body. Spinal cord injury is more complicated, as there are factors in the body that inhibit repair. Unfortunately, a solution to completely repair spinal cord injury has not been found. Thus, bioengineering strategies for the peripheral nervous system are focused on alternatives to the nerve graft, whereas efforts for spinal cord injury are focused on creating a permissive environment for regeneration. Fortunately, recent advances in neuroscience, cell culture, genetic techniques, and biomaterials provide optimism for new treatments for nerve injuries. This article reviews the nervous system physiology, the factors that are critical for nerve repair, and the current approaches that are being explored to aid peripheral nerve regeneration and spinal cord repair.
Collapse
Affiliation(s)
- Christine E Schmidt
- Department of Biomedical Engineering The University of Texas at Austin, Austin, Texas 78712, USA.
| | | |
Collapse
|
16
|
Abstract
Following injury to the CNS, severed axons undergo a phase of abortive sprouting in the vicinity of the wound, but do not spontaneously re-grow or regenerate. From a long history of attempts to stimulate regeneraion, a major strategy that has been developed clinically is the implantation of tissue into denervated target regions. Unfortunately trials have so far not borne out the promise that this would prove a useful therapy for disorders such as Parkinson's disease. Many strategies have also been developed to stimulate the regeneration of axons across sites of injury, particularly in the spinal cord. Animal data have demonstrated that some of these approaches hold promise and that the spinal cord has a remarkable degree of intrinsic plasticity. Attempts are now being made to utilize experimental techniques in spinal patients.
Collapse
Affiliation(s)
- Peter E Batchelor
- Departments of Medicine and Neurology, University of Melbourne, Austin and Repatriation Medical Centre, Vic. 3084, Heidelberg, Australia
| | | |
Collapse
|
17
|
Eaton MJ, Blits B, Ruitenberg MJ, Verhaagen J, Oudega M. Amelioration of chronic neuropathic pain after partial nerve injury by adeno-associated viral (AAV) vector-mediated over-expression of BDNF in the rat spinal cord. Gene Ther 2002; 9:1387-95. [PMID: 12365004 DOI: 10.1038/sj.gt.3301814] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2002] [Accepted: 05/07/2002] [Indexed: 11/09/2022]
Abstract
Changing the levels of neurotrophins in the spinal cord micro-environment after nervous system injury has been proposed to recover normal function, such that behavioral response to peripheral stimuli does not lead to chronic pain. We have investigated the effects of recombinant adeno-associated viral (rAAV)-mediated over-expression of brain-derived neurotrophic factor (BDNF) in the spinal cord on chronic neuropathic pain after unilateral chronic constriction injury (CCI) of the sciatic nerve. The rAAV-BDNF vector was injected into the dorsal horn at the thirteenth thoracic spinal cord vertebra (L(1) level) 1 week after CCI. Allodynia and hyperalgesia induced by CCI in the hindpaws were permanently reversed, beginning 1 week after vector injection, compared with a similar injection of a control rAAV-GFP vector (green fluorescent protein) or saline. In situ hybridization for BDNF demonstrated that both dorsal and ventral lumbar spinal neurons contained an intense signal for BDNF mRNA, at 1 to 8 weeks after vector injection. There was no similar BDNF mRNA over-expression associated with either injections of saline or rAAV-GFP. These data suggest that chronic neuropathic pain is sensitive to early spinal BDNF levels after partial nerve injury and that rAAV-mediated gene transfer could potentially be used to reverse chronic pain after nervous system injuries in humans.
Collapse
Affiliation(s)
- M J Eaton
- The Miami Project To Cure Paralysis, University of Miami School of Medicine, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
18
|
Piontek J, Régnier-Vigouroux A, Brandt R. Contact with astroglial membranes induces axonal and dendritic growth of human CNS model neurons and affects the distribution of the growth-associated proteins MAP1B and GAP43. J Neurosci Res 2002; 67:471-83. [PMID: 11835314 DOI: 10.1002/jnr.10094] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The development of morphological complexity of CNS neurons is thought to be regulated by extracellular factors and cellular contact. To analyze the role of contact with astroglia in this process and to determine the intraneuronal mechanisms involved, an in vitro system was developed where terminally differentiated and polar human CNS model neurons (NT2-N neurons) were cultured on a layer of mouse astrocytes or isolated membrane fractions in chemically defined medium. Morphometric analysis revealed that physical contact with living astrocytes increased the lengths of axonal and dendritic processes and lead to an increased number of branch points. Contact with astrocytes also resulted in a redistribution of the growth-associated proteins MAP1b and GAP-43 toward the growth cones of NT2-N neurons. Astrocyte-contact did not lead to a maturation of the neurons as would be detected by an increased expression of tau isoforms containing the adult-specific exons 2 and 3. Culture on immobilized membrane fractions prepared from astrocytes also increased the morphological complexity of the neurons in a qualitatively similar manner. The results indicate that physical contact with astrocyte membranes increases the morphological complexity of CNS model neurons through a mechanism that involves a redistribution of growth-associated proteins to neuronal growth cones. NT2-N neurons may provide a useful cellular model to analyze cytoskeletal mechanisms during the development of terminally differentiated and polar human neurons.
Collapse
Affiliation(s)
- Jörg Piontek
- Department of Neurobiology, IZN, University of Heidelberg, Im Neuenheimer Feld 364, D-16920 Heidelberg, Germany
| | | | | |
Collapse
|
19
|
Huber AB, Ehrengruber MU, Schwab ME, Brösamle C. Adenoviral gene transfer to the injured spinal cord of the adult rat. Eur J Neurosci 2000; 12:3437-42. [PMID: 10998127 DOI: 10.1046/j.1460-9568.2000.00255.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have investigated gene transfer to the injured adult rat spinal cord by the use of a recombinant adenovirus. 105 or 5 x 106 plaque-forming units (pfu) of a replication-defective adenoviral vector carrying the green fluorescent protein (GFP) reporter gene were injected into a dorsal hemisection lesion at spinal level T8. Gene expression and inflammatory responses were studied 4, 8 and 21 days after surgery. Numerous cells within 3 mm on each side of the lesion were found to express high levels of GFP at 4 days after infection as shown by GFP fluorescence and immunohistochemistry. At 8 days, expression was still strong although weaker than at 4 days. After 21 days, transgene expression had almost ceased. Expression was neither higher nor more prolonged in animals that had received the higher vector dose. Delayed injection 1 week after spinal injury also did not increase transgene expression. Infected cell types were identified immunohistochemically. The most prominent transduced cells were spinal motoneurons. Additionally, we could identify other neurons, astrocytes, oligodendrocytes and peripheral cells infiltrating the lesion site. The glial and inflammatory reaction at and around the lesion was studied by cresyl violet histology, alpha-GFAP, OX42 and alpha-CD-8 immunohistochemistry. No significant differences from controls were found in the low virus group; in the high virus group a strong invasion of CD-8-positive lymphocytes was found. Open-field locomotion analysis showed virus-infected animals performing as well as control animals. Adenoviral gene transfer may be an efficient way to introduce factors to the injured spinal cord in paradigms of research or therapy.
Collapse
Affiliation(s)
- A B Huber
- Brain Research Institute, Department of Neuromorphology, University of Zurich and Swiss Federal Institute of Technology, Winterthurer Str. 190, 8057 Zurich, Switzerland.
| | | | | | | |
Collapse
|
20
|
|
21
|
Griff ER, Greer CA, Margolis F, Ennis M, Shipley MT. Ultrastructural characteristics and conduction velocity of olfactory receptor neuron axons in the olfactory marker protein-null mouse. Brain Res 2000; 866:227-36. [PMID: 10825498 DOI: 10.1016/s0006-8993(00)02291-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Olfactory receptor neuron (ORN) axon diameters and the conduction velocity of the compound action potential along ORN axons were studied in olfactory marker protein (OMP)-null mice and genotypically matched controls. The compound action potential was distinguished from postsynaptic field potentials by its shorter latency, its persistence following application of cobalt or kynurenic acid that blocked postsynaptic responses, and its ability to follow paired-pulse stimulation at 300 Hz. Blockade of the postsynaptic field responses by kynurenic acid indicates that in the mouse, as in the rat, glutamate is the olfactory nerve transmitter. The mean conduction velocity of ORNs in wild-type control mice was 0. 47+/-0.19 (S.E.M.) m/s (n=5), similar to the conduction velocity reported for other mammals. The mean diameter of ORN axons in control mice was 0.202+/-0.005 and 0.261+/-0.006 microm in the OMP-null mice. This increase in fiber diameter in the OMP-nulls predicts an increase in impulse conduction velocity. However, the mean conduction velocity of OMP-null mice, 0.38+/-0.03 m/s (n=6), was not significantly different from control (P>0.1). The conduction velocity predicted by the increase in fiber diameter in OMP-null mice was within the 95% confidence interval of the measured value. Thus, OMP-null ORNs are normal with respect to the conduction velocity of their axons. The number of axodendritic synapses in the glomeruli of OMP-null mice is higher than in congenic wild-type mice.
Collapse
Affiliation(s)
- E R Griff
- Department of Biological Sciences, University of Cincinnati, ML 0006, Cincinnati, OH 45221-0006, USA.
| | | | | | | | | |
Collapse
|
22
|
Cho S, Dawson PE, Dawson G. In vitro depalmitoylation of neurospecific peptides: Implication for infantile neuronal ceroid lipofuscinosis. J Neurosci Res 2000. [DOI: 10.1002/(sici)1097-4547(20000101)59:1<32::aid-jnr5>3.0.co;2-a] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
23
|
Holtmaat AJ, Huizinga CT, Margolis FL, Gispen WH, Verhaagen J. Transgenic expression of B-50/GAP-43 in mature olfactory neurons triggers downregulation of native B-50/GAP-43 expression in immature olfactory neurons. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 74:197-207. [PMID: 10640691 DOI: 10.1016/s0169-328x(99)00263-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The adult mammalian olfactory neuroepithelium is an unusual neural tissue, since it maintains its capacity to form new neurons throughout life. Newly formed neurons differentiate in the basal layers of the olfactory neuroepithelium and express B-50/GAP-43, a protein implicated in neurite outgrowth. During maturation these neurons migrate into the upper portion of the epithelium, upregulate expression of olfactory marker protein (OMP) and concomitantly downregulate the expression of B-50/GAP-43. Transgenic mice that exhibit OMP-promoter directed expression of B-50/GAP-43 in mature olfactory neurons display an unexpected decrease in the complement of B-50/GAP-43-positive cells in the lower region of the olfactory epithelium [A.J.G.D. Holtmaat, P.A. Dijkhuizen, A.B. Oestreicher, H. J. Romijn, N.M.T. Van der Lugt, A. Berns, F.L. Margolis, W.H. Gispen, J. Verhaagen, Directed expression of the growth-associated protein B-50/GAP-43 to olfactory neurons in transgenic mice results in changes in axon morphology and extraglomerular growth, J. Neurosci. 15 (1995) 7953-7965]. We have investigated whether the decrement in B-50/GAP-43-positive cells in this region was due to a dislocation of the immature neurons to other regions of the olfactory epithelium or to a downregulation of B-50/GAP-43 synthesis in these immature neurons. In eight of nine independent transgenic mouse lines that express the transgene in different numbers of olfactory neurons, a decline in the number of B-50/GAP-43-expressing neurons in the basal portion of the olfactory neuroepithelium was observed, both at the protein level and the mRNA level. An alternative marker for immature cells, a juvenile form of tubulin, was normally expressed in this location, indicating that the olfactory epithelium of OMP-B-50/GAP-43 transgenic mice contains a normal complement of immature olfactory neurons and that most of these neurons display a downregulation of B-50/GAP-43 expression.
Collapse
Affiliation(s)
- A J Holtmaat
- Netherlands Institute for Brain Research, Meibergdreef 33, 1105 AZ, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
24
|
Cantallops I, Routtenberg A. Activity-dependent regulation of axonal growth: Posttranscriptional control of the GAP-43 gene by the NMDA receptor in developing hippocampus. ACTA ACUST UNITED AC 1999. [DOI: 10.1002/(sici)1097-4695(19991105)41:2<208::aid-neu4>3.0.co;2-v] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
25
|
Aarts LH, Verkade P, van Dalen JJ, van Rozen AJ, Gispen WH, Schrama LH, Schotman P. B-50/GAP-43 potentiates cytoskeletal reorganization in raft domains. Mol Cell Neurosci 1999; 14:85-97. [PMID: 10532807 DOI: 10.1006/mcne.1999.0775] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
B-50 (GAP-43) is a neural, membrane-associated protein that has been implicated in neurite outgrowth and guidance. Following stable transfection of Rat1 fibroblasts with B-50 cDNA we observed a dispersed distribution of B-50 immunoreactivity in flattened resting cells. In contrast, motile cells exhibited high concentrations of B-50 at the leading edge of ruffling membranes, coinciding with actin polymerization. Time-lapse studies on Rat1 fibroblasts transiently transfected with B-50/EGFP revealed that large vesicles originated from the ruffling membranes. These large vesicles (pinocytes) were found positive for Thy-1, a GPI-anchored protein, but negative for rab-5, an early endosome marker. In primary hippocampal neurons B-50 also colocalized completely with the raft marker Thy-1. Antibody-mediated cross-linking of Thy-1 in hippocampal neurons resulted in a redistribution of the intracellular protein B-50 to Thy-1-immunopositive membrane patches, whereas syntaxin was mainly excluded from the patches, showing that B-50 is associated with rafts. Academic Press.
Collapse
Affiliation(s)
- L H Aarts
- Rudolf Magnus Institute for Neurosciences, Department of Physiological Chemistry, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
26
|
Klein RL, McNamara RK, King MA, Lenox RH, Muzyczka N, Meyer EM. Generation of aberrant sprouting in the adult rat brain by GAP-43 somatic gene transfer. Brain Res 1999; 832:136-44. [PMID: 10375659 DOI: 10.1016/s0006-8993(99)01482-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The expression of GAP-43 was modulated genetically in the adult rat nigrostriatal or septohippocampal pathway using recombinant adeno-associated virus (rAAV) vectors incorporating the neuron specific enolase (NSE) promoter and either a rat GAP-43 cDNA or the corresponding antisense sequence. Bicistronic expression of green fluorescent protein (GFP) enabled us to evaluate transduced neurons selectively. Single injections of rAAV into the substantia nigra pars compacta (SNc) transduced both dopaminergic and non-dopaminergic neurons stably for the 3-month duration of the study. Transduction with the GAP-43 vector in this region: (1) increased GAP-43 mRNA levels 2-fold compared to controls; (2) led to GAP-43 immunoreactivity in neuronal perikarya, axons, and dendrites that was not observed otherwise; and (3) resulted in GAP-43/ GFP-positive axons that were traced to the striatum where they formed clusters of aberrant nets. The GAP-43 antisense vector, in contrast, decreased neuropil GAP-43 immunoreactivity compared to controls in the SNc. In septum, injections of the GAP-43 expressing vector also caused aberrant clusters of GAP-43 labelled fibers in terminal fields, i.e., fornix and hippocampus, that were not observed in control tissues. It therefore appears that rAAV vectors provide a novel approach for modulating intraneuronal GAP-43 expression in the adult brain.
Collapse
Affiliation(s)
- R L Klein
- Department of Pharmacology and Therapeutics, University of Florida, Campus Box 100267 JHMHC, Gainesville, FL 32610-0267, USA
| | | | | | | | | | | |
Collapse
|
27
|
Fernandez AM, Gonzalez de la Vega AG, Planas B, Torres-Aleman I. Neuroprotective actions of peripherally administered insulin-like growth factor I in the injured olivo-cerebellar pathway. Eur J Neurosci 1999; 11:2019-30. [PMID: 10336671 DOI: 10.1046/j.1460-9568.1999.00623.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Exogenous administration of insulin-like growth factor I (IGF-I) restores motor function in rats with neurotoxin-induced cerebellar deafferentation. We first determined that endogenous IGFs are directly involved in the recovery process because infusion of an IGF-I receptor antagonist into the lateral ventricle blocks gradual recovery of limb coordination that spontaneously occurs after partial deafferentation of the olivo-cerebellar circuitry. We then analysed mechanisms whereby exogenous IGF-I restores motor function in rats with complete damage of the olivo-cerebellar pathway. Treatment with IGF-I normalized several markers of cell function in the cerebellum, including calbindin, glutamate receptor 1 (GluR1), gamma-aminobutyric acid (GABA) and glutamate, which are all depressed after 3-acetylpyridine (3AP)-induced deafferentation. IGF-I also promoted functional reinnervation of the cerebellar cortex by inferior olive (IO) axons. In the IO, increased expression of bax in neurons and bcl-X in astrocytes after 3AP was significantly reduced by IGF-I treatment. On the contrary, IGF-I prevented the decrease in poly-sialic-acid neural cell adhesion molecule (PSA-NCAM) and GAP-43 expression induced by 3AP in IO cells. IGF-I also significantly increased the number of neurons expressing bcl-2 in brainstem areas surrounding the IO. Altogether, these results indicate that subcutaneous IGF-I therapy promotes functional recovery of the olivo-cerebellar pathway by acting at two sites within this circuitry: (i) by modulating death- and plasticity-related proteins in IO neurons; and (ii) by impinging on homeostatic mechanisms leading to normalization of cell function in the cerebellum. These results provide insight into the neuroprotective actions of IGF-I and may be of practical consequence in the design of new therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- A M Fernandez
- Laboratory of Cellular and Molecular Neuroendocrinology, Cajal Institute, CSIC, Madrid, Spain
| | | | | | | |
Collapse
|
28
|
Aarts LH, Schotman P, Verhaagen J, Schrama LH, Gispen WH. The role of the neural growth associated protein B-50/GAP-43 in morphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 446:85-106. [PMID: 10079839 DOI: 10.1007/978-1-4615-4869-0_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- L H Aarts
- Rudolf Magnus Institute for Neurosciences, Laboratory of Physiological Chemistry, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Schwaiger FW, Hager G, Raivich G, Kreutzberg GW. Cellular activation in neuroregeneration. PROGRESS IN BRAIN RESEARCH 1999; 117:197-210. [PMID: 9932410 DOI: 10.1016/s0079-6123(08)64017-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- F W Schwaiger
- Department of Neuromorphology, Max-Planck-Institute of Neurobiology, Martinsried, Germany.
| | | | | | | |
Collapse
|
30
|
Pasterkamp RJ, De Winter F, Giger RJ, Verhaagen J. Role for semaphorin III and its receptor neuropilin-1 in neuronal regeneration and scar formation? PROGRESS IN BRAIN RESEARCH 1999; 117:151-70. [PMID: 9932407 DOI: 10.1016/s0079-6123(08)64014-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- R J Pasterkamp
- Graduate School for Neurosciences Amsterdam, Netherlands Institute for Brain Research, The Netherlands
| | | | | | | |
Collapse
|
31
|
Affiliation(s)
- G M Smith
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas 75235, USA.
| | | |
Collapse
|
32
|
Pasterkamp RJ, De Winter F, Holtmaat AJ, Verhaagen J. Evidence for a role of the chemorepellent semaphorin III and its receptor neuropilin-1 in the regeneration of primary olfactory axons. J Neurosci 1998; 18:9962-76. [PMID: 9822752 PMCID: PMC6793295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
To explore a role for chemorepulsive axon guidance mechanisms in the regeneration of primary olfactory axons, we examined the expression of the chemorepellent semaphorin III (sema III), its receptor neuropilin-1, and collapsin response mediator protein-2 (CRMP-2) during regeneration of the olfactory system. In the intact olfactory system, neuropilin-1 and CRMP-2 mRNA expression define a distinct population of olfactory receptor neurons, corresponding to immature (B-50/GAP-43-positive) and a subset of mature (olfactory marker protein-positive) neurons located in the lower half of the olfactory epithelium. Sema III mRNA is expressed in pial sheet cells and in second-order olfactory neurons that are the target cells of neuropilin-1-positive primary olfactory axons. These data suggest that in the intact olfactory bulb sema III creates a molecular barrier, which helps restrict ingrowing olfactory axons to the nerve and glomerular layers of the bulb. Both axotomy of the primary olfactory nerve and bulbectomy induce the formation of new olfactory receptor neurons expressing neuropilin-1 and CRMP-2 mRNA. After axotomy, sema III mRNA is transiently induced in cells at the site of the lesion. These cells align regenerating bundles of olfactory axons. In contrast to the transient appearance of sema III-positive cells at the lesion site after axotomy, sema III-positive cells increase progressively after bulbectomy, apparently preventing regenerating neuropilin-1-positive nerve bundles from growing deeper into the lesion area. The presence of sema III in scar tissue and the concomitant expression of its receptor neuropilin-1 on regenerating olfactory axons suggests that semaphorin-mediated chemorepulsive signal transduction may contribute to the regenerative failure of these axons after bulbectomy.
Collapse
Affiliation(s)
- R J Pasterkamp
- Graduate School for Neurosciences Amsterdam, Netherlands Institute for Brain Research, 1105 AZ Amsterdam-ZO, The Netherlands
| | | | | | | |
Collapse
|
33
|
Schmidt JT. Up-regulation of protein kinase C in regenerating optic nerve fibers of goldfish: immunohistochemistry and kinase activity assay. JOURNAL OF NEUROBIOLOGY 1998; 36:315-24. [PMID: 9733068 DOI: 10.1002/(sici)1097-4695(19980905)36:3<315::aid-neu1>3.0.co;2-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein kinase C (PKC) activation has been associated with synaptic plasticity in many projections, and manipulating PKC in the retinotectal projection strongly affects the activity-driven sharpening of the retinotopic map. This study examined levels of PKC in the regenerating retinotectal projection via immunostaining and assay of activity. A polyclonal antibody to the conserved C2 (Ca2+ binding) domain of classical PKC isozymes (anti-panPKC) recognized a single band at 79-80 kD on Western blots of goldfish brain. It stained one class of retinal bipolar cells and the ganglion cells in normal retina, as shown previously. Strong staining was not present in the optic fiber layer of retina or in optic nerve, optic tract, or terminal zone in tectum, with the exception of a single fascicle of optic nerve fibers that by their location and by L1 (E587) staining were identified as those arising from newly added ganglion cells at the retinal margin. Normal tectal sections showed dark staining of a subclass of type XIV neuron with somas at the top of the periventricular layer and an apical dendrite ascending to stratum opticum. In regenerating retina, swollen ganglion cells stained darkly and stained axons were seen in the optic fiber layer. In regenerating optic nerve (2-11 weeks postcrush), all fascicles of optic fibers stained darkly for both PKC and L1(E587). At 5 weeks postcrush, PKC staining could also be seen in the medial and lateral optic tracts and stratum opticum at the front half of the tectum and very lightly over the terminal zones. PKC activity was measured in homogenized tissues dissected from a series of fish with unilateral nerve crush from 1 to 5 weeks previously. Activity levels stimulated by phorbols and Ca2+ were measured by phosphorylation of a specific peptide and referred to levels measured in the opposite control side. Regeneration did not increase overall PKC activity in retina or tectum, but in optic nerve there was an 80% rise after the first week. The increased activity verifies that the increased staining in nerve represented an up-regulation of functional PKC during nerve regeneration.
Collapse
Affiliation(s)
- J T Schmidt
- Department of Biological Sciences and Neurobiology Research Center, State University of New York at Albany, 12222, USA
| |
Collapse
|
34
|
Aarts LH, Schrama LH, Hage WJ, Bos JL, Gispen WH, Schotman P. B-50/GAP-43-induced formation of filopodia depends on Rho-GTPase. Mol Biol Cell 1998; 9:1279-92. [PMID: 9614174 PMCID: PMC25350 DOI: 10.1091/mbc.9.6.1279] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In the present study we show that expression of the neural PKC-substrate B-50 (growth-associated protein [GAP-43]) in Rat-1 fibroblasts induced the formation of filopodial extensions during spreading. This morphological change was accompanied by an enhanced formation of peripheral actin filaments and by accumulation of vinculin immunoreactivity in filopodial focal adhesions, colocalizing with B-50. In time lapse experiments, the B-50-induced filopodial extensions were shown to stay in close contact with the substratum and appeared remarkably stable, resulting in a delayed lamellar spreading of the fibroblasts. The morphogenetic effects of the B-50 protein were entirely dependent on the integrity of the two N-terminal cysteines involved in membrane association (C3C4), but were not significantly affected by mutations of the PKC-phosphorylation site (S41) or deletion of the C terminus (177-226). Cotransfection of B-50 with dominant negative Cdc42 or Rac did not prevent B-50-induced formation of filopodial cells, whereas this process could be completely blocked by cotransfection with dominant negative Rho or Clostridium botulinum C3-transferase. Conversely, constitutively active Rho induced a similar filopodial phenotype as B-50. We therefore propose that the induction of surface extensions by B-50 in spreading Rat-1 fibroblasts depends on Rho-guanosine triphosphatase function.
Collapse
Affiliation(s)
- L H Aarts
- Department of Physiological Chemistry, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
35
|
Holtmaat AJ, Oestreicher AB, Gispen WH, Verhaagen J. Manipulation of gene expression in the mammalian nervous system: application in the study of neurite outgrowth and neuroregeneration-related proteins. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1998; 26:43-71. [PMID: 9600624 DOI: 10.1016/s0165-0173(97)00044-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A fundamental issue in neurobiology entails the study of the formation of neuronal connections and their potential to regenerate following injury. In recent years, an expanding number of gene families has been identified involved in different aspects of neurite outgrowth and regeneration. These include neurotrophic factors, cell-adhesion molecules, growth-associated proteins, cytoskeletal proteins and chemorepulsive proteins. Genetic manipulation technology (transgenic mice, knockout mice, viral vectors and antisense oligonucleotides) has been instrumental in defining the function of these neurite outgrowth-related proteins. The aim of this paper is to provide an overview of the above-mentioned four approaches to manipulate gene expression in vivo and to discuss the progress that has been made using this technology in helping to understand the molecular mechanisms that regulate neurite outgrowth. We will show that work with transgenic mice and knockout mice has contributed significantly to the dissection of the function of several proteins with a key role in neurite outgrowth and neuronal survival. Recently developed viral vectors for gene transfer in postmitotic neurons have opened up new avenues to analyze the function of a protein following local expression in naive adult rodents. The initial results with viral vector-based gene transfer provide a conceptual framework for further studies on genetic therapy of neuroregeneration and neurodegenerative diseases.
Collapse
Affiliation(s)
- A J Holtmaat
- Graduate School of Neurosciences Amsterdam, Netherlands Institute for Brain Research
| | | | | | | |
Collapse
|
36
|
Gold BG, Yew JY, Zeleny-Pooley M. The immunosuppressant FK506 increases GAP-43 mRNA levels in axotomized sensory neurons. Neurosci Lett 1998; 241:25-8. [PMID: 9502207 DOI: 10.1016/s0304-3940(97)00960-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FK506, an immunosuppressant drug used to prevent allograft rejection in organ transplantations, accelerates functional recovery and nerve regeneration in the rat sciatic nerve crush model. While the mechanism by which FK506 increases regeneration is unknown, in contrast to immunosuppression, it does not involve calcineurin inhibition. Using the reverse-transcriptase polymerase chain reaction (RT-PCR) technique and a digoxigenin-labeled probe, we show that subcutaneous injections of FK506 (10 mg/kg/day) markedly increases the level of axotomy-induced growth-associated protein (GAP-43) mRNA in dorsal root ganglion (DRG) neurons. Quantitation of DRG neurons revealed that FK506 produced a 33% increase in the numbers of neurons exhibiting intense staining. Increased synthesis of GAP-43 may play a role in FK506's ability to speed nerve regeneration.
Collapse
Affiliation(s)
- B G Gold
- Center for Research on Occupational and Environmental Toxicology, Oregon Health Sciences University, Portland 97201, USA
| | | | | |
Collapse
|