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










Publication year range
1.
Sci Rep ; 6: 30504, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27465654

ABSTRACT

High harmonic generation (HHG) is an established means of producing coherent, short wavelength, ultrafast pulses from a compact set-up. Table-top high-harmonic sources are increasingly being used to image physical and biological systems using emerging techniques such as coherent diffraction imaging and ptychography. These novel imaging methods require coherent illumination, and it is therefore important to both characterize the spatial coherence of high-harmonic beams and understand the processes which limit this property. Here we investigate the near- and far-field spatial properties of high-harmonic radiation generated in a gas cell. The variation with harmonic order of the intensity profile, wavefront curvature, and complex coherence factor is measured in the far-field by the SCIMITAR technique. Using the Gaussian-Schell model, the properties of the harmonic beam in the plane of generation are deduced. Our results show that the order-dependence of the harmonic spatial coherence is consistent with partial coherence induced by both variation of the intensity-dependent dipole phase as well as finite spatial coherence of the driving radiation. These findings are used to suggest ways in which the coherence of harmonic beams could be increased further, which would have direct benefits to imaging with high-harmonic radiation.

2.
Exp Neurol ; 263: 150-60, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25448158

ABSTRACT

Sulf1 and Sulf2 are endosulfatases that cleave 6-O-sulphate groups from Heparan Sulphate Proteoglycans (HSPGs). Sulfation levels of HSPGs are critical for their role in modulating the activity of various growth factor receptors. Sulf1 and Sulf2 mRNAs were found to be widely expressed in the rodent nervous system and their full-length proteins were found in many types of neuronal perikarya and axons in the cerebral cortex, cerebellum, spinal cord and dorsal root ganglia (DRG) of adult rats. Sulf1/2 were also strongly expressed by cultured DRG neurons. To determine if blocking Sulf1 or Sulf2 activity affected neurite outgrowth in vitro, cultured DRG neurons were treated with neutralising antibodies to Sulf1 or Sulf2. Blocking Sulf1 and Sulf2 activity did not affect neurite outgrowth from cultured DRG neurons grown on a laminin/polylysine substrate but ameliorated the inhibitory effects of chondroitin sulphate proteoglycans (CSPGs) on neurite outgrowth. Blocking epidermal growth factor receptor (ErbB1) activity also improved neurite outgrowth in the presence of CSPGs, but the effects of ErbB1 antagonists and blocking SULFs were not additive. It is proposed that Sulf1, Sulf2 and ErbB1 are involved in the signalling pathway from CSPGs that leads to inhibition of neurite outgrowth and may regulate structural plasticity and regeneration in the nervous system.


Subject(s)
Nerve Regeneration/physiology , Neurites/metabolism , Sulfatases/biosynthesis , Sulfotransferases/biosynthesis , Animals , Blotting, Western , Brain/metabolism , ErbB Receptors/biosynthesis , Ganglia, Spinal/metabolism , Immunohistochemistry , In Vitro Techniques , Mice , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/metabolism , Spinal Cord/metabolism
3.
Exp Neurol ; 239: 82-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23022459

ABSTRACT

Pharmacological inhibitors of epidermal growth factor receptor (ErbB1) attenuate the ability of CNS myelin to inhibit axonal regeneration. However, it has been claimed that such effects are mediated by off-target interactions. We have tested the role of ErbB1 in axonal regeneration by culturing neurons from ErbB1 knockout mice in the presence of various inhibitors of axonal regeneration: CNS myelin, chondroitin sulfate proteoglycans (CSPG), fibrinogen or polyinosinic:polycytidylic acid (poly I:C). We confirmed that ErbB1 was activated in cultures of cerebellar granule cells exposed to inhibitors of axonal regeneration and that ErbB1 kinase inhibitors promoted neurite outgrowth under these conditions. In the presence of myelin, fibrinogen, CSPG and poly I:C ErbB1 -/- neurons grew longer neurites than neurons expressing ErbB1. Furthermore, inhibitors of ErbB1 kinase did not improve neurite outgrowth from ErbB1 -/- neurons, ruling out an off-target mechanism of action. ErbB1 kinase activity is therefore a valid target for promoting axonal elongation in the presence of many of the molecules believed to contribute to the failure of axonal regeneration in the injured CNS.


Subject(s)
Axons/drug effects , Genes, erbB-1/drug effects , Nerve Regeneration/drug effects , Animals , Blood-Brain Barrier/drug effects , Calcium Signaling/physiology , Cerebellum/cytology , Chondroitin Sulfates/pharmacology , Cytoplasmic Granules , Fibrinogen/pharmacology , Mice , Mice, Knockout , Myelin Sheath/physiology , Phosphorylation , Poly I-C/pharmacology , Proteoglycans/pharmacology , Quinazolines/pharmacology , RNA/metabolism , RNA, Double-Stranded/pharmacology , Sensory Receptor Cells/drug effects , Toll-Like Receptor 3/drug effects
4.
J Neurosci ; 31(18): 6809-6819, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21543611

ABSTRACT

In the developing spinal cord, most oligodendrocyte precursors (OLPs) arise from the ventral ventricular zone (VZ) under the influence of Sonic Hedgehog, but a minority are generated from the dorsal VZ in a Hedgehog-independent manner. In the developing forebrain too, OLPs arise from both the ventral and the dorsal VZ. It is not known whether dorsally and ventrally derived oligodendrocyte (OL) lineage cells have different properties. We generated a dual reporter mouse line to color code ventrally and dorsally derived OLPs (vOLPs and dOLPs) and their differentiated oligodendrocyte progeny (vOLs and dOLs) for functional studies. We found that ∼80% of OL lineage cells in the postnatal spinal cord and ∼20% in the corpus callosum are ventrally derived. In both spinal cord and corpus callosum, vOLPs and dOLPs had indistinguishable electrical properties, as did vOLs and dOLs. However, vOLPs and dOLPs had different migration and settling patterns. In the spinal cord, vOLPs appeared early and spread uniformly throughout the cord, whereas dOLPs arrived later and remained mainly in the dorsal and dorsolateral funiculi. During adulthood, corticospinal and rubrospinal tracts became myelinated mainly by dOLs, even though vOLs dominated these tracts during early postnatal life. Thus, dOLPs are electrically similar to vOLPs but appear to outcompete them for dorsal axons.


Subject(s)
Cell Lineage/physiology , Corpus Callosum/physiology , Myelin Sheath/physiology , Oligodendroglia/physiology , Spinal Cord/physiology , Animals , Electrophysiology , Immunohistochemistry , Mice , Mice, Transgenic
5.
Mol Cell Neurosci ; 46(1): 167-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20828614

ABSTRACT

Virus mediated RNA-interference (RNAi) is a powerful approach to study genes in vivo. Here we report a method using lentivirus-delivered RNAi to knockdown the glial enzyme, D-amino acid oxidase (DAO), in the mouse cerebellum. After initial characterisation in vitro, we achieved a 40-50% reduction of DAO mRNA in the cerebellum 7 and 28 days after a single injection of lentivirus encoding a DAO-specific, short-hairpin RNA. Injections also decreased DAO immunoreactivity (-33%). The major substrate for DAO is D-serine, an N-methyl-D-aspartate receptor (NMDAR) co-agonist. Thus, we also measured whether DAO knockdown impacted on d-serine, or expression of NMDAR subunits, and found that DAO RNAi led to increased cerebellar D-serine levels (+77%), and decreased NMDAR subunit NR2A mRNA (-22%), but did not affect NR1 or NR2C mRNAs. These data demonstrate the utility of lentiviruses to deliver RNAi to glial cells within the cerebellum, and confirm the role of DAO in D-serine metabolism. They also provide a tool to investigate DAO, an enzyme currently of considerable interest in the pathophysiology and therapy of schizophrenia.


Subject(s)
Cerebellum/enzymology , D-Amino-Acid Oxidase/genetics , D-Amino-Acid Oxidase/metabolism , Gene Knockdown Techniques , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cell Line , Genetic Vectors/genetics , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Male , Mental Disorders/physiopathology , Mice , Mice, Inbred C57BL , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/metabolism
6.
BMC Neurosci ; 11: 13, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20137064

ABSTRACT

BACKGROUND: Microglia/macrophages and lymphocytes (T-cells) accumulate around motor and primary sensory neurons that are regenerating axons but there is little or no microglial activation or T-cell accumulation around axotomised intrinsic CNS neurons, which do not normally regenerate axons. We aimed to establish whether there was an inflammatory response around the perikarya of CNS neurons that were induced to regenerate axons through a peripheral nerve graft. RESULTS: When neurons of the thalamic reticular nucleus (TRN) and red nucleus were induced to regenerate axons along peripheral nerve grafts, a marked microglial response was found around their cell bodies, including the partial enwrapping of some regenerating neurons. T-cells were found amongst regenerating TRN neurons but not rubrospinal neurons. Axotomy alone or insertion of freeze-killed nerve grafts did not induce a similar perineuronal inflammation. Nerve grafts in the corticospinal tracts did not induce axonal regeneration or a microglial or T-cell response in the motor cortex. CONCLUSIONS: These results strengthen the evidence that perineuronal microglial accumulation (but not T-cell accumulation) is involved in axonal regeneration by intrinsic CNS and other neurons.


Subject(s)
Axons/physiology , Microglia/physiology , Nerve Regeneration/physiology , Neurons/physiology , Red Nucleus/physiology , Thalamic Nuclei/physiology , Animals , Axotomy , Brain Tissue Transplantation , Cell Death , Facial Nerve/physiology , Facial Nerve/surgery , Female , Freezing , Male , Motor Cortex/physiology , Neurons/transplantation , Peripheral Nerves/surgery , Pyramidal Tracts/physiology , Pyramidal Tracts/surgery , Rats , Rats, Sprague-Dawley , Red Nucleus/surgery , T-Lymphocytes/physiology , Thalamic Nuclei/surgery
7.
J Comp Neurol ; 518(5): 699-721, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20034058

ABSTRACT

Generation of new axonal sprouts plays an important role in neural repair. In the current study, we examined the appearance, composition and effects of gene deletions on intrabrainstem sprouts following peripheral facial nerve axotomy. Axotomy was followed by the appearance of galanin(+) and calcitonin gene-related peptide (CGRP)(+) sprouts peaking at day 14, matching both large, neuropeptide(+) subpopulations of axotomized facial motoneurons, but with CGRP(+) sprouts considerably rarer. Strong immunoreactivity for vesicular acetylcholine transporter (VAChT) and retrogradely transported MiniRuby following its application on freshly cut proximal facial nerve stump confirmed their axotomized motoneuron origin; the sprouts expressed CD44 and alpha7beta1 integrin adhesion molecules and grew apparently unhindered along neighboring central white matter tracts. Quantification of the galanin(+) sprouts revealed a stronger response following cut compared with crush (day 7-14) as well as enhanced sprouting after recut (day 8 + 6 vs. 14; 14 + 8 vs. 22), arguing against delayed appearance of sprouting being the result of the initial phase of reinnervation. Sprouting was strongly diminished in brain Jun-deficient mice but enhanced in alpha7 null animals that showed apparently compensatory up-regulation in beta1, suggesting important regulatory roles for transcription factors and the sprout-associated adhesion molecules. Analysis of inflammatory stimuli revealed a 50% reduction 12-48 hours following systemic endotoxin associated with neural inflammation and a tendency toward more sprouts in TNFR1/2 null mutants (P = 10%) with a reduced inflammatory response, indicating detrimental effects of excessive inflammation. Moreover, the study points to the usefulness of the facial axotomy model in exploring physiological and molecular stimuli regulating central sprouting.


Subject(s)
Facial Nerve Injuries/physiopathology , Facial Nerve/physiology , Growth Cones/ultrastructure , Motor Neurons/physiology , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Animals , Axotomy , Calcitonin Gene-Related Peptide/metabolism , Cell Adhesion Molecules/metabolism , Facial Nerve/metabolism , Facial Nerve Injuries/metabolism , Galanin/metabolism , Gene Deletion , Growth Cones/metabolism , Immunohistochemistry , Integrins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Motor Neurons/metabolism , Oncogene Protein p65(gag-jun)/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Time Factors , Vesicular Acetylcholine Transport Proteins/metabolism
8.
Mol Cell Neurosci ; 39(2): 152-60, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18617007

ABSTRACT

After traumatic CNS injury, a cascade of secondary events expands the initial lesion. The gap-junction protein connexin43 (Cx43), which is transiently up-regulated, has been implicated in the spread of 'bystander' damage. We have used an antisense oligodeoxynucleotide (asODN) to suppress Cx43 up-regulation in two rat models of spinal cord injury. Within 24 h of compression injury, rats treated with Cx43-asODN scored higher than sense-ODN and vehicle-treated controls on behavioural tests of locomotion. Their spinal cords showed less swelling and tissue disruption, less up-regulation of astrocytic GFAP, and less extravasation of fluorescently-labelled bovine serum albumin and neutrophils. The locomotor improvement was sustained over at least 4 weeks. Following partial spinal cord transection, Cx43-asODN treatment reduced GFAP immunoreactivity, neutrophil recruitment, and the activity of OX42(+) microglia in and around the lesion site. Cx43 has many potential roles in the pathophysiology of CNS injury and may be a valuable target for therapeutic intervention.


Subject(s)
Connexin 43/metabolism , Down-Regulation/physiology , Inflammation/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Recovery of Function/physiology , Spinal Cord Injuries/complications , Animals , CD11b Antigen/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Connexin 43/antagonists & inhibitors , Disease Models, Animal , Down-Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Locomotion/drug effects , Male , Microglia/drug effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Time Factors
9.
BMC Neurosci ; 8: 80, 2007 Sep 27.
Article in English | MEDLINE | ID: mdl-17900358

ABSTRACT

BACKGROUND: The chondroitin sulphate proteoglycan NG2 blocks neurite outgrowth in vitro and has been proposed as a major inhibitor of axonal regeneration in the CNS. Although a substantial body of evidence underpins this hypothesis, it is challenged by recent findings including strong expression of NG2 in regenerating peripheral nerve. RESULTS: We studied axonal regeneration in the PNS and CNS of genetically engineered mice that do not express NG2, and in sex and age matched wild-type controls. In the CNS, we used anterograde tracing with BDA to study corticospinal tract (CST) axons after spinal cord injury and transganglionic labelling with CT-HRP to trace ascending sensory dorsal column (DC) axons after DC lesions and a conditioning lesion of the sciatic nerve. Injury to these fibre tracts resulted in no difference between knockout and wild-type mice in the ability of CST axons or DC axons to enter or cross the lesion site. Similarly, after dorsal root injury (with conditioning lesion), most regenerating dorsal root axons failed to grow across the dorsal root entry zone in both transgenic and wild-type mice. Following sciatic nerve injuries, functional recovery was assessed by analysis of the toe-spreading reflex and cutaneous sensitivity to Von Frey hairs. Anatomical correlates of regeneration were assessed by: retrograde labelling of regenerating dorsal root ganglion (DRG) cells with DiAsp; immunostaining with PGP 9.5 to visualise sensory reinnervation of plantar hindpaws; electron microscopic analysis of regenerating axons in tibial and digital nerves; and by silver-cholinesterase histochemical study of motor end plate reinnervation. We also examined functional and anatomical correlates of regeneration after injury of the facial nerve by assessing the time taken for whisker movements and corneal reflexes to recover and by retrograde labelling of regenerated axons with Fluorogold and DiAsp. None of the anatomical or functional analyses revealed significant differences between wild-type and knockout mice. CONCLUSION: These findings show that NG2 is unlikely to be a major inhibitor of axonal regeneration after injury to the CNS, and, further, that NG2 is unlikely to be necessary for regeneration or functional recovery following peripheral nerve injury.


Subject(s)
Axons/physiology , Central Nervous System/pathology , Nerve Regeneration/physiology , Nervous System Diseases/pathology , Peripheral Nervous System/pathology , Proteoglycans/deficiency , Animals , Antigens , Central Nervous System/physiopathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/physiopathology , Peripheral Nervous System/physiopathology , Pyridinium Compounds , Recovery of Function/physiology , Stilbamidines , Ubiquitin Thiolesterase/metabolism
10.
J Neurosci Res ; 84(2): 278-90, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16683235

ABSTRACT

To understand whether tissue inhibitors of metalloproteinase (TIMPs) contribute to the failure of regenerating sensory axons to enter the spinal cord, we used in situ hybridization and immunocytochemistry to examine the expression of TIMP1, TIMP2, and TIMP3 in the dorsal root, dorsal root entry zone (DREZ), and dorsal column after dorsal root injury in adult rats. We found that the three TIMPs and their mRNAs were up-regulated in a time-, region-, and cell-type-specific manner. Strong up-regulation of all three TIMPs was seen in the injured dorsal roots. TIMP2 was also significantly up-regulated in the DREZ and degenerating dorsal column, where TIMP1 and TIMP3 showed only moderate up-regulation. Most cells up-regulating the TIMPs in the DREZ and degenerating dorsal column were reactive astrocytes, but TIMP2 was also up-regulated by microglia/macrophages, especially at long postoperative survival times. These results suggest that TIMPs may be involved in controlling tissue remodelling following dorsal root injury and that manipulation of the expression of TIMPs may provide a means of promoting axonal regeneration into and within the injured spinal cord.


Subject(s)
Nerve Regeneration/physiology , Spinal Cord/enzymology , Spinal Nerve Roots/enzymology , Spinal Nerve Roots/injuries , Tissue Inhibitor of Metalloproteinases/metabolism , Animals , Female , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Time Factors , Up-Regulation
11.
Front Biosci ; 11: 2967-75, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16720368

ABSTRACT

The ability to down regulate the expression of a specific protein within the intact central nervous system (CNS) is highly desirable from both a research and therapeutic perspective. Antisense has the potential to do this. However, problems of invasive antisense delivery methods and short half life of remain problematic. We overcome this by using Pluronic gel to provide a sustained delivery antisense oligodeoxynucleotides (ODN's) to the intact central nervous system and achieving rapid penetration throughout the spinal cord in 2-3 hours and significant knockdown of our target protein connexin 43 (Cx43) in 4-8 hours (recovering at 48-72 hours). Interestingly CY3-siRNA probes could not be detected penetrating the intact CNS and no knockdown the Cx43 was found. This approach with conventional ODNs could provide a faster and cheaper alternative to knockout mice in the investigation of the functions of specific proteins within the CNS and may also have therapeutic implications for drug discovery and development.


Subject(s)
Connexin 43/biosynthesis , Oligoribonucleotides, Antisense/administration & dosage , Oligoribonucleotides, Antisense/pharmacokinetics , RNA, Small Interfering , Spinal Cord/metabolism , Animals , Connexin 43/drug effects , Down-Regulation , Fluorescence Resonance Energy Transfer , Male , Oligoribonucleotides, Antisense/physiology , Poloxamer , Rats , Rats, Sprague-Dawley , Spinal Cord/chemistry
12.
Nat Med ; 12(3): 348-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16491086

ABSTRACT

Retroviral and lentiviral vector integration into host-cell chromosomes carries with it a finite chance of causing insertional mutagenesis. This risk has been highlighted by the induction of malignancy in mouse models, and development of lymphoproliferative disease in three individuals with severe combined immunodeficiency-X1 (refs. 2,3). Therefore, a key challenge for clinical therapies based on retroviral vectors is to achieve stable transgene expression while minimizing insertional mutagenesis. Recent in vitro studies have shown that integration-deficient lentiviral vectors can mediate stable transduction. With similar vectors, we now show efficient and sustained transgene expression in vivo in rodent ocular and brain tissues. We also show substantial rescue of clinically relevant rodent models of retinal degeneration. Therefore, the high efficiency of gene transfer and expression mediated by lentiviruses can be harnessed in vivo without a requirement for vector integration. For therapeutic application to postmitotic tissues, this system substantially reduces the risk of insertional mutagenesis.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Lentivirus/genetics , Animals , Brain/cytology , Carrier Proteins , Electroretinography , Eye Proteins/metabolism , Female , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mice , Pigment Epithelium of Eye/cytology , Rats , Retina/cytology , Tumor Cells, Cultured , Virus Integration/genetics , cis-trans-Isomerases
13.
Proc Natl Acad Sci U S A ; 102(41): 14883-8, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16195382

ABSTRACT

Neuronal expression of growth-associated protein 43 (GAP-43) and the cell adhesion molecule L1 has been correlated with CNS axonal growth and regeneration, but it is not known whether expression of these molecules is necessary for axonal regeneration to occur. We have taken advantage of the fact that Purkinje cells do not express GAP-43 or L1 in adult mammals or regenerate axons into peripheral nerve grafts to test the importance of these molecules for axonal regeneration in vivo. Transgenic mice were generated in which Purkinje cells constitutively express L1 or both L1 and GAP-43 under the Purkinje cell-specific L7 promoter, and regeneration of Purkinje cell axons into peripheral nerve grafts implanted into the cerebellum was examined. Purkinje cells expressing GAP-43 or L1 showed minor enhancement of axonal sprouting. Purkinje cells expressing both GAP-43 and L1 showed more extensive axonal sprouting and axonal growth into the proximal portion of the graft. When a predegenerated nerve graft was implanted into double-transgenic mice, penetration of the graft by Purkinje cell axonal sprouts was strongly enhanced, and some axons grew along the entire intracerebral length of the graft (2.5-3.0 mm) and persisted for several months. The results demonstrate that GAP-43 and L1 coexpressed in Purkinje cells can act synergistically to switch these regeneration-incompetent CNS neurons into a regeneration-competent phenotype and show that coexpression of these molecules is a key regulator of the regenerative ability of intrinsic CNS neurons in vivo.


Subject(s)
Axons/physiology , GAP-43 Protein/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Purkinje Cells/physiology , Regeneration/physiology , Animals , Axons/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Purkinje Cells/metabolism
14.
Diabetes ; 54(1): 212-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15616031

ABSTRACT

Although autonomic neuropathy is recognized as an independent risk factor for stroke in diabetes, the mechanism by which autonomic nerves are involved in this pathology is unknown. Parasympathetic (cholinergic) nerves of the autonomic nervous system are known to innervate and to cause relaxation of cerebral arteries by releasing nitric oxide (NO); hence, they are called nitrergic nerves. However, the effect of diabetes on nitrergic nerves is unknown. Here, we show that perivascular nitrergic nerves around the cerebral arteries degenerate in two phases in streptozotocin-induced diabetic rats. In the first phase, perivascular nitrergic nerve fibers remain intact while they lose their neuronal NO synthase content. This phase is reversible with insulin treatment. In the second phase, nitrergic cell bodies in the ganglia are lost via apoptosis in an irreversible manner. Throughout the two phases, irreversible thickening of the smooth muscle layer of cerebral arteries is observed. This is the first demonstration of nitrergic degeneration in diabetic cerebral arteries, which could elucidate the link between diabetic autonomic neuropathy and stroke.


Subject(s)
Cerebral Arteries/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/physiopathology , Diabetic Neuropathies/physiopathology , Nerve Degeneration/physiopathology , Nitric Oxide/physiology , Stroke/physiopathology , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Disease Models, Animal , Insulin/therapeutic use , Male , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Rats , Rats, Wistar , Vesicular Acetylcholine Transport Proteins
15.
Mol Cell Neurosci ; 26(1): 34-49, 2004 May.
Article in English | MEDLINE | ID: mdl-15121177

ABSTRACT

Axonal regeneration in the adult CNS is limited by the presence of several inhibitory proteins associated with myelin. Nogo-A, a myelin-associated inhibitor, is responsible for axonal outgrowth inhibition in vivo and in vitro. Here we study the onset and maturation of Nogo-A and Nogo receptor in the entorhino-hippocampal formation of developing and adult mice. We also provide evidence that Nogo-A does not inhibit embryonic hippocampal neurons, in contrast to other cell types such as cerebellar granule cells. Our results also show that Nogo and Nogo receptor mRNA are expressed in the adult by both principal and local-circuit hippocampal neurons, and that after lesion, Nogo-A is also transiently expressed by a subset of reactive astrocytes. Furthermore, we analyzed their regulation after kainic acid (KA) treatment and in response to the transection of the entorhino-hippocampal connection. We found that Nogo-A and Nogo receptor are differentially regulated after kainic acid or perforant pathway lesions. Lastly, we show that the regenerative potential of lesioned entorhino-hippocampal organotypic slice co-cultures is increased after blockage of Nogo-A with two IN-1 blocking antibodies. In conclusion, our results show that Nogo and its receptor might play key roles during development of hippocampal connections and that they are implicated in neuronal plasticity in the adult.


Subject(s)
Entorhinal Cortex/physiology , Hippocampus/physiology , Myelin Proteins/metabolism , Nerve Regeneration/physiology , Perforant Pathway/physiology , Receptors, Cell Surface/metabolism , Animals , Animals, Newborn , Antibodies/pharmacology , Astrocytes/cytology , Astrocytes/metabolism , Brain Injuries/chemically induced , Brain Injuries/physiopathology , COS Cells , Entorhinal Cortex/embryology , Entorhinal Cortex/injuries , Fetus , GPI-Linked Proteins , Gene Expression Regulation, Developmental/genetics , Gliosis/metabolism , Gliosis/physiopathology , Growth Cones/metabolism , Growth Cones/ultrastructure , Hippocampus/embryology , Hippocampus/injuries , Kainic Acid , Mice , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Neuronal Plasticity/physiology , Nogo Proteins , Nogo Receptor 1 , Perforant Pathway/embryology , Perforant Pathway/injuries , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
16.
BMC Neurosci ; 5: 4, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-15005815

ABSTRACT

BACKGROUND: Inactivating genes in vivo is an important technique for establishing their function in the adult nervous system. Unfortunately, conventional knockout mice may suffer from several limitations including embryonic or perinatal lethality and the compensatory regulation of other genes. One approach to producing conditional activation or inactivation of genes involves the use of Cre recombinase to remove loxP-flanked segments of DNA. We have studied the effects of delivering Cre to the hippocampus and neocortex of adult mice by injecting replication-deficient adeno-associated virus (AAV) and lentiviral (LV) vectors into discrete regions of the forebrain. RESULTS: Recombinant AAV-Cre, AAV-GFP (green fluorescent protein) and LV-Cre-EGFP (enhanced GFP) were made with the transgene controlled by the cytomegalovirus promoter. Infecting 293T cells in vitro with AAV-Cre and LV-Cre-EGFP resulted in transduction of most cells as shown by GFP fluorescence and Cre immunoreactivity. Injections of submicrolitre quantities of LV-Cre-EGFP and mixtures of AAV-Cre with AAV-GFP into the neocortex and hippocampus of adult Rosa26 reporter mice resulted in strong Cre and GFP expression in the dentate gyrus and moderate to strong labelling in specific regions of the hippocampus and in the neocortex, mainly in neurons. The pattern of expression of Cre and GFP obtained with AAV and LV vectors was very similar. X-gal staining showed that Cre-mediated recombination had occurred in neurons in the same regions of the brain, starting at 3 days post-injection. No obvious toxic effects of Cre expression were detected even after four weeks post-injection. CONCLUSION: AAV and LV vectors are capable of delivering Cre to neurons in discrete regions of the adult mouse brain and producing recombination.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Integrases/administration & dosage , Integrases/genetics , Lentivirus/genetics , Neurons/metabolism , Viral Proteins/administration & dosage , Viral Proteins/genetics , Animals , Cell Line , Gene Expression , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/virology , Integrases/metabolism , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Neocortex/metabolism , Neocortex/virology , Neurons/virology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic/drug effects , Recombination, Genetic/genetics , Transgenes , Viral Proteins/metabolism
17.
Neuroreport ; 14(16): 2085-8, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14600502

ABSTRACT

Following a crush injury of the optic nerve in adult rats, the axons of retinal ganglion cells, stimulated to regenerate by a lens injury and growing within the optic nerve, are associated predominantly with astrocytes: they remain of small diameter (0.1-0.5 microm) and unmyelinated for > or = 2 months after the operation. In contrast, when the optic nerve is cut and a segment of a peripheral nerve is grafted to the ocular stump of the optic nerve, the regenerating retinal axons are associated predominantly with Schwann cells: they are of larger diameter than in the previous experiment and include unmyelinated axons (0.2-2.5 microm) and myelinated axons (mean diameter 2.3 microm). Thus, the grafted peripheral nerve, and presumably its Schwann cells, stimulate enlargement of the regenerating retinal axons leading to partial myelination, whereas the injured optic nerve itself, and presumably its astrocytes, does not. The result points to a marked difference of peripheral (Schwann cells) and central (astrocytes) glia in their effect on regenerating retinal axons.


Subject(s)
Astrocytes/physiology , Nerve Regeneration/physiology , Optic Nerve Injuries/surgery , Peripheral Nerves/transplantation , Retinal Ganglion Cells/physiology , Schwann Cells/transplantation , Animals , Astrocytes/ultrastructure , Axons/physiology , Axons/ultrastructure , Brain Tissue Transplantation , Female , Graft Survival/physiology , Male , Microscopy, Electron , Nerve Crush , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Myelinated/ultrastructure , Nerve Fibers, Unmyelinated/physiology , Nerve Fibers, Unmyelinated/ultrastructure , Optic Nerve/growth & development , Optic Nerve/surgery , Optic Nerve/ultrastructure , Optic Nerve Injuries/pathology , Optic Nerve Injuries/physiopathology , Peripheral Nerves/physiology , Peripheral Nerves/ultrastructure , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/ultrastructure , Schwann Cells/physiology , Schwann Cells/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...