Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 1715: 3-17, 2018.
Article in English | MEDLINE | ID: mdl-29188502

ABSTRACT

Adeno-associated viral vectors have numerous applications in neuroscience, including the study of gene function in health and disease, targeting of light-sensitive proteins to anatomically distinct sets of neurons to manipulate neuronal activity (optogenetics), and the delivery of fluorescent protein to study anatomical connectivity in the brain. Moreover several phase I/II clinical trials for gene therapy of eye and brain diseases with adeno-associated viral vectors have shown that these vectors are well tolerated by human patients. In this chapter we describe a detailed protocol for the small scale production of recombinant adeno-associated viral vectors. This protocol can be executed by investigators with experience in cell culture and molecular biological techniques in any well-equipped molecular neurobiology laboratory. With this protocol we typically obtain research batches of 100-200 µL that range in titer from 5 × 1012 to 2 × 1013 genomic copies/mL.


Subject(s)
Brain Diseases/therapy , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Brain Diseases/genetics , Eye Diseases/genetics , Eye Diseases/therapy , HEK293 Cells , Humans , Injections, Intraocular/methods , Nervous System/metabolism , Plasmids
2.
J Biol Chem ; 288(38): 27384-27395, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23940048

ABSTRACT

Chondroitin sulfate (CS) and the CS-rich extracellular matrix structures called perineuronal nets (PNNs) restrict plasticity and regeneration in the CNS. Plasticity is enhanced by chondroitinase ABC treatment that removes CS from its core protein in the chondroitin sulfate proteoglycans or by preventing the formation of PNNs, suggesting that chondroitin sulfate proteoglycans in the PNNs control plasticity. Recently, we have shown that semaphorin3A (Sema3A), a repulsive axon guidance molecule, localizes to the PNNs and is removed by chondroitinase ABC treatment (Vo, T., Carulli, D., Ehlert, E. M., Kwok, J. C., Dick, G., Mecollari, V., Moloney, E. B., Neufeld, G., de Winter, F., Fawcett, J. W., and Verhaagen, J. (2013) Mol. Cell. Neurosci. 56C, 186-200). Sema3A is therefore a candidate for a PNN effector in controlling plasticity. Here, we characterize the interaction of Sema3A with CS of the PNNs. Recombinant Sema3A interacts with CS type E (CS-E), and this interaction is involved in the binding of Sema3A to rat brain-derived PNN glycosaminoglycans, as demonstrated by the use of CS-E blocking antibody GD3G7. In addition, we investigate the release of endogenous Sema3A from rat brain by biochemical and enzymatic extractions. Our results confirm the interaction of Sema3A with CS-E containing glycosaminoglycans in the dense extracellular matrix of rat brain. We also demonstrate that the combination of Sema3A and PNN GAGs is a potent inhibitor of axon growth, and this inhibition is reduced by the CS-E blocking antibody. In conclusion, Sema3A binding to CS-E in the PNNs may be a mechanism whereby PNNs restrict growth and plasticity and may represent a possible point of intervention to facilitate neuronal plasticity.


Subject(s)
Axons/metabolism , Brain/metabolism , Chondroitin Sulfates/metabolism , Extracellular Matrix/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Semaphorin-3A/metabolism , Amino Acid Motifs , Animals , Brain Chemistry/physiology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/genetics , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , HEK293 Cells , Humans , Mice , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Binding , Rats , Semaphorin-3A/chemistry , Semaphorin-3A/genetics
3.
Mol Cell Neurosci ; 56: 186-200, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23665579

ABSTRACT

In the adult rodent brain, subsets of neurons are surrounded by densely organised extracellular matrix called perineuronal nets (PNNs). PNNs consist of hyaluronan, tenascin-R, chondroitin sulphate proteoglycans (CSPGs), and the link proteins Crtl1 and Bral2. PNNs restrict plasticity at the end of critical periods and can be visualised with Wisteria floribunda agglutinin (WFA). Using a number of antibodies raised against the different regions of semaphorin3A (Sema3A) we demonstrate that this secreted chemorepulsive axon guidance protein is localised to WFA-positive PNNs around inhibitory interneurons in the cortex and several other PNN-bearing neurons throughout the brain and co-localises with aggrecan, versican, phosphacan and tenascin-R. Chondroitinase ABC (ChABC) was injected in the cortex to degrade glycosaminoglycans (GAGs) from the CSPGs, abolishing WFA staining of PNNs around the injection site. Sema3A-positive nets were no longer observed in the area devoid of WFA staining. In mice lacking the link protein Crtl1 in the CNS only vestigial PNNs are present, and in these mice there were no Sema3A-positive PNN structures. A biochemical analysis shows that Sema3A protein binds with high-affinity to CS-GAGs and aggrecan and versican extracted from PNNs in the adult rat brain, and a significant proportion of Sema3A is retrieved in brain extracts that are enriched in PNN-associated GAGs. The Sema3A receptor components PlexinA1 and A4 are selectively expressed by inhibitory interneurons in the cortex that are surrounded by Sema3A positive PNNs. We conclude that the chemorepulsive axon guidance molecule Sema3A is present in PNNs of the adult rodent brain, bound to the GAGs of the CSPGs. These observations suggest a novel concept namely that chemorepulsive axon guidance molecules like Sema3A may be important functional attributes of PNNs in the adult brain.


Subject(s)
Cerebral Cortex/metabolism , Extracellular Matrix/metabolism , Semaphorin-3A/metabolism , Aggrecans/metabolism , Animals , Cerebral Cortex/cytology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glycosaminoglycans/metabolism , HEK293 Cells , Humans , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Protein Binding , Proteoglycans/genetics , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Semaphorin-3A/genetics , Versicans/metabolism
4.
PLoS One ; 5(8): e12387, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20808778

ABSTRACT

BACKGROUND: Müller cell gliosis occurs in various retinal pathologies regardless of the underlying cellular defect. Because activated Müller glial cells span the entire retina and align areas of injury, they are ideal targets for therapeutic strategies, including gene therapy. METHODOLOGY/PRINCIPAL FINDINGS: We used adeno-associated viral AAV2/6 vectors to transduce mouse retinas. The transduction pattern of AAV2/6 was investigated by studying expression of the green fluorescent protein (GFP) transgene using scanning-laser ophthalmoscopy and immuno-histochemistry. AAV2/6 vectors transduced mouse Müller glial cells aligning the retinal blood vessels. However, the transduction capacity was hindered by the inner limiting membrane (ILM) and besides Müller glial cells, several other inner retinal cell types were transduced. To obtain Müller glial cell-specific transgene expression, the cytomegalovirus (CMV) promoter was replaced by the glial fibrillary acidic protein (GFAP) promoter. Specificity and activation of the GFAP promoter was tested in a mouse model for retinal gliosis. Mice deficient for Crumbs homologue 1 (CRB1) develop gliosis after light exposure. Light exposure of Crb1(-/-) retinas transduced with AAV2/6-GFAP-GFP induced GFP expression restricted to activated Müller glial cells aligning retinal blood vessels. CONCLUSIONS/SIGNIFICANCE: Our experiments indicate that AAV2 vectors carrying the GFAP promoter are a promising tool for specific expression of transgenes in activated glial cells.


Subject(s)
Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Neuroglia/metabolism , Retinal Vessels/cytology , Transduction, Genetic/methods , Adenoviridae/genetics , Animals , Gene Expression , Humans , Injections , Mice , Promoter Regions, Genetic/genetics , Transgenes/genetics
5.
Mol Ther ; 18(4): 715-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20179682

ABSTRACT

For many experiments in the study of the peripheral nervous system, it would be useful to genetically manipulate primary sensory neurons. We have compared vectors based on adeno-associated virus (AAV) serotypes 1, 2, 3, 4, 5, 6, and 8, and lentivirus (LV), all expressing green fluorescent protein (GFP), for efficiency of transduction of sensory neurons, expression level, cellular tropism, and persistence of transgene expression following direct injection into the dorsal root ganglia (DRG), using histological quantification and qPCR. Two weeks after injection, AAV1, AAV5, and AAV6 had transduced the most neurons. The time course of GFP expression from these three vectors was studied from 1 to 12 weeks after injection. AAV5 was the most effective serotype overall, followed by AAV1. Both these serotypes showed increasing neuronal transduction rates at later time points, with some injections of AAV5 yielding over 90% of DRG neurons GFP(+) at 12 weeks. AAV6 performed well initially, but transduction rates declined dramatically between 4 and 12 weeks. AAV1 and AAV5 both transduced large-diameter neurons, IB4(+) neurons, and CGRP(+) neurons. In conclusion, AAV5 is a highly effective gene therapy vector for primary sensory neurons following direct injection into the DRG.


Subject(s)
Dependovirus/classification , Ganglia, Spinal , Genetic Therapy , Genetic Vectors , Animals , Dependovirus/genetics , Female , Plasmids , Rats , Rats, Wistar , Serotyping , Transduction, Genetic
6.
J Neurosci ; 27(52): 14260-4, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18160633

ABSTRACT

Neuroma formation after peripheral nerve injury is detrimental to functional recovery and is therefore a significant clinical problem. The molecular basis for this phenomenon is not fully understood. Here, we show that the expression of the chemorepulsive protein semaphorin 3A (sema3A), but not semaphorin 3F, is increased in human neuroma tissue that has formed in severe obstetric brachial plexus lesions. Sema3A is produced by fibroblasts in the epineurial space and appears to be secreted into the extracellular matrix. It surrounds fascicles, minifascicles, or single axons, suggesting a role in fasciculation and inhibition of neurite outgrowth. Lentiviral vector-mediated knock-down of Neuropilin 1, the receptor for sema3A, leads to increased neurite outgrowth of F11 cells cultured on neuroma tissue, but not of F11 cells cultured on normal nerve tissue. These findings demonstrate the putative inhibitory role of sema3A in human neuroma tissue. Our observations are the first demonstration of the expression of sema3A in human neural scar tissue and support a role for this protein in the inhibition of axonal regeneration in injured human peripheral nerves. These findings contribute to the understanding of the outgrowth inhibitory properties of neuroma tissue.


Subject(s)
Neurites/physiology , Neuroma/metabolism , Neuroma/pathology , Semaphorin-3A/metabolism , Animals , Cell Line, Transformed , Humans , In Situ Hybridization/methods , In Vitro Techniques , Infant , Mice , Nerve Fibers/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
7.
J Neurotrauma ; 23(3-4): 409-21, 2006.
Article in English | MEDLINE | ID: mdl-16629626

ABSTRACT

Regenerating axons stop growing when they reach the border of the glial-fibrotic scar, presumably because they encounter a potent molecular barrier inhibiting growth cone advance. Chemorepulsive axon guidance molecules provide a non-permissive environment restricting and channeling axon growth in the developing nervous system. These molecules could also act as growth-inhibitory molecules in the regenerating nervous system. The receptors for repulsive guidance cues are expressed in the mature nervous system, suggesting that adult neurons are sensitive to the activity of developmentally active repulsive proteins. In this review, we summarize recent observations on semaphorins, ephrins, and slits in the injured brain and spinal cord, providing evidence that these proteins are major players in inhibiting axonal regeneration and establishing the glial-fibrotic scar.


Subject(s)
Axons/physiology , Cell Movement/physiology , Intercellular Signaling Peptides and Proteins/physiology , Nerve Tissue Proteins/physiology , Spinal Cord Injuries/pathology , Animals , Ephrins/biosynthesis , Ephrins/physiology , Humans , Nerve Regeneration , Receptors, Eph Family/physiology , Semaphorins/physiology
8.
Mol Cell Neurosci ; 24(4): 902-12, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14697657

ABSTRACT

The neural scar that forms after injury to the mammalian central nervous system is a barrier to sprouting and regenerating axons. In addition to reactive astrocytes that are present throughout the lesion site, leptomeningeal fibroblasts invade the lesion core. When isolated in vitro, these cells form a very poor substrate for growing neurites, even more so than reactive astrocytes. Nevertheless the molecular mechanisms involved in this growth inhibition are not well understood. Semaphorins have been reported to be upregulated in meningeal cells (MCs) on mechanical injury to the brain and spinal cord. In the present study, we show that Sema3A mRNA and active protein are produced by cultured meningeal cells. A protein extract from these cells induces the collapse of embryonic dorsal root ganglion (DRG) growth cones. This collapsing activity is partially blocked by neuropilin-1 antibodies and is absent in meningeal cells derived from Sema3A-knockout mice. In addition to growth cone collapse, recombinant Sema3A but not Sema3C inhibits neurite outgrowth of embryonic DRGs. Consistent with this result we find that the inhibitory effect of meningeal cells on neurite outgrowth is partially overcome on Sema3A-deficient MCs. Furthermore we show that the inhibitory effect of MC-derived Sema3A on neurite outgrowth is modulated by nerve growth factor. Our results show that Sema3A, a chemorepellent during nervous system development, is a major neurite growth-inhibitory molecule in meningeal fibroblasts and is therefore likely to contribute to the inhibitory properties of the neural scar.


Subject(s)
Meninges/metabolism , Neural Inhibition/physiology , Neurites/metabolism , Semaphorin-3A/biosynthesis , Animals , Cells, Cultured , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Growth Cones/metabolism , Growth Inhibitors/biosynthesis , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Meninges/cytology , Meninges/embryology , Meninges/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Semaphorin-3A/deficiency , Semaphorin-3A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...