Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Amino Acids ; 52(6-7): 1033-1041, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32696177

ABSTRACT

Increasing evidence indicates that the enzyme creatine kinase (CK) is intimately involved in microvascular contractility. The mitochondrial isoenzyme catalyses phosphocreatine synthesis from ATP, while cytoplasmic CK, predominantly the BB isoenzyme in vascular tissue, is tightly bound near myosin ATPase, where it favours ATP production from phosphocreatine to metabolically support vascular contractility. However, the effect of CK gene inactivation on microvascular function is hitherto unknown. We studied functional and structural parameters of mesenteric resistance arteries isolated from 5 adult male mice lacking cytoplasmic BB-CK and ubiquitous mitochondrial CK (CK-/-) vs 6 sex/age-matched controls. Using a Mulvany Halpern myograph, we assessed the acute maximum contractile force with 125 mM K+ and 10-5 M norepinephrine, and the effect of two inhibitors, dinitrofluorobenzene, which inhibits phosphotransfer enzymes (0.1 µM), and the specific adenylate kinase inhibitor P1, P5-di(adenosine 5') pentaphosphate (10-6 to 10-5 M). WT and CK-/- did not significantly differ in media thickness, vascular elasticity parameters, or acute maximum contractile force. CK-/- arteries displayed greater reduction in contractility after dinitrofluorobenzene 38%; vs 14% in WT; and after AK inhibition, 14% vs 5.5% in WT, and displayed abnormal mitochondria, with a partial loss of the inner membrane. Thus, CK-/- mice display a surprisingly mild phenotype in vascular dysfunction. However, the mitochondrial abnormalities and greater effect of inhibitors on contractility may reflect a compromised energy metabolism. In CK-/- mice, compensatory mechanisms salvage energy metabolism, as described for other CK knock-out models.


Subject(s)
Arterioles/metabolism , Arterioles/physiology , Creatine Kinase, BB Form/deficiency , Creatine Kinase, Mitochondrial Form/deficiency , Vasoconstriction/physiology , Animals , Dinitrofluorobenzene/administration & dosage , Dinucleoside Phosphates/administration & dosage , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Norepinephrine/administration & dosage
2.
Neurobiol Dis ; 33(3): 482-98, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19130883

ABSTRACT

Cholesterol and docosahexenoic acid (DHA) may affect degenerative processes in Alzheimer's Disease (AD) by influencing Abeta metabolism indirectly via the vasculature. We investigated whether DHA-enriched diets or cholesterol-containing Typical Western Diets (TWD) alter behavior and cognition, cerebral hemodynamics (relative cerebral blood volume (rCBV)) and Abeta deposition in 8- and 15-month-old APP(swe)/PS1(dE9) mice. In addition we investigated whether changes in rCBV precede changes in Abeta deposition or vice versa. Mice were fed regular rodent chow, a TWD-, or a DHA-containing diet. Behavior, learning and memory were investigated, and rCBV was measured using contrast-enhanced MRI. The Abeta load was visualized immunohistochemically. We demonstrate that DHA altered rCBV in 8-month-old APP/PS1 and wild type mice[AU1]. In 15-month-old APP/PS1 mice DHA supplementation improved spatial memory, decreased Abeta deposition and slightly increased rCBV, indicating that a DHA-enriched diet can diminish AD-like pathology. In contrast, TWD diets decreased rCBV in 15-month-old mice. The present data indicate that long-term dietary interventions change AD-like pathology in APP/PS1 mice. Additionally, effects of the tested diets on vascular parameters were observed before effects on Abeta load were noted. These data underline the importance of vascular factors in the APP/PS1 mouse model of AD pathology.


Subject(s)
Alzheimer Disease/pathology , Brain/blood supply , Cholesterol, Dietary/administration & dosage , Cognition , Docosahexaenoic Acids/administration & dosage , Aging , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Atrophy , Behavior, Animal , Blood Volume , Brain/pathology , Brain Chemistry , Diet , Disease Models, Animal , Fatty Acids/analysis , Male , Maze Learning , Memory , Mice , Mice, Transgenic
3.
J Neurochem ; 90(6): 1321-30, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15341516

ABSTRACT

Creatine kinase (CK)-catalysed ATP-phosphocreatine (PCr) exchange is considered to play a key role in energy homeostasis of the brain. This study assessed the metabolic and anatomical consequences of partial or complete depletion of this system in transgenic mice without cytosolic B-CK (B-CK-/-), mitochondrial ubiquitous CK (UbCKmit-/-), or both isoenzymes (CK -/-), using non-invasive quantitative magnetic resonance (MR) imaging and spectroscopy. MR imaging revealed an increase in ventricle size in a subset of B-CK-/- mice, but not in animals with UbCKmit or compound CK mutations. Mice lacking single CK isoenzymes had normal levels of high-energy metabolites and tissue pH. In the brains of CK double knockouts pH and ATP and Pi levels were also normal, even though PCr had become completely undetectable. Moreover, a 20-30% decrease was observed in the level of total creatine and a similar increase in the level of neuronal N-acetyl-aspartate compounds. Although CKs themselves are not evenly distributed throughout the CNS, these alterations were uniform and concordant across different brain regions. Changes in myo-inositol and glutamate peaks did appear to be mutation type and brain area specific. Our results challenge current models for the biological significance of the PCr-CK energy system and suggest a multifaceted role for creatine in the brain.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Creatine Kinase/deficiency , Isoenzymes/deficiency , Magnetic Resonance Imaging/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Animals , Brain/pathology , Creatine/metabolism , Creatine Kinase/genetics , Creatine Kinase/physiology , Creatine Kinase, BB Form , Creatine Kinase, Mitochondrial Form , Isoenzymes/genetics , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorus Isotopes/metabolism , Tritium/metabolism
4.
Eur J Neurosci ; 17(5): 991-1005, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12653975

ABSTRACT

Cell adhesion molecule-like receptor-type protein tyrosine phosphatases have been shown to be important for neurite outgrowth and neural development in several animal models. We have previously reported that in leucocyte common antigen-related (LAR) phosphatase deficient (LAR-deltaP) mice the number and size of basal forebrain cholinergic neurons, and their innervation of the hippocampal area, is reduced. In this study we compared the sprouting response of LAR-deficient and wildtype neurons in a peripheral and a central nervous system lesion model. Following sciatic nerve crush lesion, LAR-deltaP mice showed a delayed recovery of sensory, but not of motor, nerve function. In line with this, neurofilament-200 immunostaining revealed a significant reduction in the number of newly outgrowing nerve sprouts in LAR-deltaP animals. Morphometric analysis indicated decreased axonal areas in regenerating LAR-deltaP nerves when compared to wildtypes. Nonlesioned nerves in wildtype and LAR-deltaP mice did not differ regarding myelin and axon areas. Entorhinal cortex lesion resulted in collateral sprouting of septohippocampal cholinergic fibres into the dentate gyrus outer molecular layer in both genotype groups. However, LAR-deltaP mice demonstrated less increase in acetylcholinesterase density and fibre number at several time points following the lesion, indicating a delayed collateral sprouting response. Interestingly, a lesion-induced reduction in number of (septo-entorhinal) basal forebrain choline acetyltransferase-positive neurons occurred in both groups, whereas in LAR-deltaP mice the average cell body size was reduced as well. Thus, regenerative and collateral sprouting is significantly delayed in LAR-deficient mice, reflecting an important facilitative role for LAR in peripheral and central nervous system axonal outgrowth.


Subject(s)
Central Nervous System/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/deficiency , Peripheral Nervous System/physiology , Protein Tyrosine Phosphatases , Receptors, Cell Surface/deficiency , Acetylcholinesterase/metabolism , Animals , Cell Count , Choline O-Acetyltransferase/metabolism , Entorhinal Cortex/injuries , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Nerve Crush , Neurofilament Proteins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Recovery of Function , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
5.
Neuroscience ; 110(4): 641-51, 2002.
Article in English | MEDLINE | ID: mdl-11934472

ABSTRACT

The p75 low-affinity neurotrophin receptor (p75(LNTR)) appears to have various functions that include enhancing nerve growth factor (NGF)-mediated survival by increasing TrkA (high-affinity NGF receptor) efficiency, and mediating apoptosis by acting as a ligand-regulated pro-apoptotic receptor. Here, we investigated the role of p75(LNTR) for adult cholinergic basal forebrain neurons by comparing neuronal responses to injury in control and p75(LNTR)-deficient mice. In both types of mice, approximately 70% of the cholinergic neurons in the ipsilateral medial septum had lost their markers choline acetyltransferase and tyrosine kinase A by 28 days following unilateral transection of the dorsal septohippocampal pathway (fimbria fornix). A 7-day delayed infusion of NGF that started 28 days after the injury resulted in reversal of choline acetyltransferase expression and cell atrophy in control, but not in p75(LNTR)-deficient, mice. This lack of response to delayed NGF treatment in p75(LNTR)-deficient mice was most likely not due to cell death, as all of the septohippocampal neurons, labeled with Fluorogold before the lesion, were present at 28 days post-lesion, similar to control mice. p75(LNTR)-deficient cholinergic neurons can respond to NGF as they were protected by NGF infusions that started immediately after the injury. These observations, the fact that lesioned p75(LNTR)-deficient neurons atrophy faster, and that non-lesioned neurons hypertrophy in response to NGF in control but not in p75(LNTR)-deficient mice, suggest that p75(LNTR) is needed for tyrosine kinase A and NGF signaling efficiency.In conclusion, during adulthood p75(LNTR) appears to play a beneficial role in the response of cholinergic neurons to injury, consistent with the proposed role of p75(LNTR) in the enhancement of TrkA signaling and the transport of neurotrophins by these neurons.


Subject(s)
Cell Survival/genetics , Cholinergic Fibers/metabolism , Nerve Growth Factor/metabolism , Neurons/metabolism , Receptor, Nerve Growth Factor/deficiency , Retrograde Degeneration/genetics , Septal Nuclei/metabolism , Animals , Atrophy/drug therapy , Atrophy/genetics , Atrophy/metabolism , Axotomy , Cell Death/drug effects , Cell Death/genetics , Cell Size/drug effects , Cell Size/genetics , Cell Survival/physiology , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/drug effects , Cholinergic Fibers/ultrastructure , Drug Administration Schedule , Female , Male , Mice , Mice, Knockout , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/pathology , Receptor, Nerve Growth Factor/genetics , Receptor, trkA/metabolism , Retrograde Degeneration/drug therapy , Retrograde Degeneration/metabolism , Septal Nuclei/drug effects , Septal Nuclei/pathology
6.
Neuroscience ; 102(4): 833-41, 2001.
Article in English | MEDLINE | ID: mdl-11182246

ABSTRACT

The leukocyte common antigen-related (LAR) receptor, composed of an extracellular region with three immunoglobulin-like and eight fibronectin type III-like domains, and a cytoplasmic region containing two protein tyrosine phosphatase domains, is thought to play a role in axonal outgrowth and guidance during neural development. LAR mutant mice were generated completely lacking the two cytoplasmic protein tyrosine phosphatase domains, resulting in the loss of ability to bind intracellular associating proteins, but (may be) still containing the ability to perform extracellular functions. A reduction in size of basal forebrain cholinergic neurons and diminished hippocampal innervation reported for knockout mice that contain a leaky gene trap inserted into the 5' part of the LAR gene [Yeo T. T. et al. (1997) J. Neurosci. Res. 47, 348-360] warranted a computer-assisted quantitative image analysis throughout the basal forebrain and hippocampus of our LAR mutant mice. The total number, longest diameter and cell body area were calculated for the choline acetyltransferase-positive neurons in the medial septum and vertical diagonal band, and optical density measurements were performed to determine the extent of acetyl cholinesterase-positive fibre innervation of the different layers in the dentate gyrus. In LAR mutant mice, the number of cholinergic cells was significantly reduced (approximately 25%) in the vertical diagonal band. Also, the cross-sectional area of the cholinergic neurons in the medial septum and vertical diagonal band was reduced (5%). These findings were paralleled by a diminished cholinergic innervation of the supragranular (18%) and molecular (4%) layers of the dentate gyrus. Thus, LAR protein tyrosine phosphatase activity appears crucial for size, number and target projection of basal forebrain cholinergic neurons, further strengthening a role for LAR in CNS development.


Subject(s)
Cholinergic Fibers/enzymology , Dentate Gyrus/cytology , Diagonal Band of Broca/cytology , Protein Tyrosine Phosphatases/genetics , Receptors, Cell Surface , Septal Nuclei/cytology , Acetylcholine/physiology , Animals , Cell Adhesion/physiology , Cell Count , Cell Size/physiology , Extracellular Matrix/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways , Neurons/enzymology , Neurons/ultrastructure , Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Signal Transduction/physiology
7.
Hum Mol Genet ; 9(11): 1615-22, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861288

ABSTRACT

Bent tail is a mouse model for human neural tube defects. Bent tail mice are characterized by a shortened, kinked tail. We have observed numerous aberrations in Bent tail embryos including exencephaly, rotation defects and occasionally omphalocele, orofacial schisis and situs abnormalities. Exencephaly was seen in >10% of all embryos and resulted from a closure defect of the hindbrain. Bent tail maps to the proximal part of the X chromosome. By haplotype analysis we have appointed the Bent tail locus to a 1.1 cM interval between markers DXMit159 and DXMit143. Subsequent analysis has revealed the presence of a deletion in all affected animals. The deletion is approximately 1 Mb in size and encompasses the gene for ZIC:3, a zinc finger transcription factor expressed in murine neuroectoderm and dorsal axial mesoderm during neurulation. ZIC:3 is a homolog of the Drosophila segmentation gene odd-paired. Although the Bent tail phenotype probably is the result of the deletion of several genes, combining data on ZIC:3 expression and function of ZIC: genes in the mouse shows that deletion of Zic3 alone is compatible with a major role of this gene in the congenital malformations of the Bent tail mouse. In man, mutations in ZIC3 are associated with situs abnormalities. These patients occasionally also show spina bifida, indicating that genetic variation in human ZIC3 may contribute to other congenital malformations, including neural tube defects.


Subject(s)
Homeodomain Proteins/genetics , Neural Tube Defects/genetics , Tail/abnormalities , Transcription Factors/genetics , X Chromosome/genetics , Animals , Chromosome Mapping , DNA/genetics , Disease Models, Animal , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Female , Gene Deletion , Genetic Linkage , Guanine Nucleotide Exchange Factors , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, Inbred Strains , Phenotype , Proto-Oncogene Proteins/genetics , Tail/embryology
8.
Neuroscience ; 94(4): 1163-72, 1999.
Article in English | MEDLINE | ID: mdl-10625055

ABSTRACT

The role of the p75 nerve growth factor receptor in the retrograde transport of neurotrophins in the adult CNS was investigated by comparing the transport of 125I-labeled neurotrophins by normal and p75 nerve growth factor receptor-deficient cholinergic septohippocampal neurons. In control mice, nerve growth factor was selectively transported from the hippocampal formation to the cholinergic neurons in the septum. Nerve growth factor labeling was found in three to four times as many septal cholinergic neuronal cell bodies than labeling for neurotrophin-3 or neurotrophin-4/5, and transported brain-derived neurotrophic factor was barely detectable. Cells were considered as labeled when the number of grains per cell exceeded five times background. In p75 nerve growth factor receptor-deficient mice, the number of cholinergic neurons labeled with each of the neurotrophins was reduced by 85-95%. Retrograde labeling of septohippocampal neurons with Fluorogold was not obviously reduced in p75 nerve growth factor receptor-deficient mice, suggesting that general transport mechanisms were not impaired. Despite the reduced neurotrophin transport, cholinergic neurons of p75 nerve growth factor receptor-deficient mice were larger than controls and had an apparently normal density of immunostaining for choline acetyltransferase. Since nerve growth factor is reportedly involved in size regulation and choline acetyltransferase expression, this raises the possibility that the retrograde transport itself is not essential for these events. Thus, p75 nerve growth factor receptor plays an important, although not exclusive, role in the transport of neurotrophins by cholinergic basal forebrain neurons, and retrograde transport of nerve growth factor may not be needed for regulating certain cellular processes.


Subject(s)
Cholinergic Fibers/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Receptor, Nerve Growth Factor/physiology , Animals , Biological Transport/physiology , Biological Transport, Active/physiology , Cell Size , Hippocampus/metabolism , Injections , Mice , Mice, Knockout/genetics , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacokinetics , Neurons/cytology , Prosencephalon/cytology , Receptor, Nerve Growth Factor/genetics , Reference Values , Septum Pellucidum/metabolism
9.
J Chem Neuroanat ; 14(3-4): 129-40, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9704891

ABSTRACT

We have previously shown that p75 nerve growth factor receptor (p75NGFR) mediates apoptosis of approximately 25% of the cholinergic basal forebrain neurons in normal control mice between postnatal day 6 and 15, but only of cholinergic neurons that lacked the nerve growth factor receptor TrkA. Here, we investigated whether and when the cholinergic neurons of the neostriatum, which express TrkA and p75NGFR during early postnatal times, undergo p75NGFR-mediated death. The cholinergic neurons in the lateral neostriatal regions expressed choline acetyltransferase (ChAT) earlier (postnatal day 3-6) than those of the medial regions and TrkA appeared before ChAT in all regions. Between postnatal day 6 and 10, approximately 40% of the ChAT-positive neurons in the most lateral regions disappeared in control mice but not in p75NGFR-deficient mice. During this time, the neostriatum of control, but not p75NGFR-deficient, mice contained many apoptotic cells. This suggests that, similar to the cholinergic neurons of the basal forebrain, the neostriatal cholinergic neurons of control mice die and that this process is mediated by p75NGFR. However, the roles of p75NGFR and TrkA appear to be more complicated in the neostriatum where relatively few neurons express p75NGFR during the death phase (and predominantly in the lateral neostriatum where the neuronal loss is greatest), and TrkA-positive as well as TrkA-negative neurons may be lost.


Subject(s)
Apoptosis , Neostriatum/physiology , Receptors, Nerve Growth Factor/physiology , Animals , Biomarkers , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/physiology , Interneurons/physiology , Mice , Neurons/physiology , Receptor, Nerve Growth Factor , Receptor, trkA/physiology
10.
J Neurosci ; 17(14): 5288-96, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9204913

ABSTRACT

Recurrent seizure activity induced during kindling has been reported to produce a functional synaptic reorganization of the mossy fibers in the hippocampus. To date, it is unclear whether this kindling-induced growth is secondary to decreases in hilar neuron density, which are presumed to reflect hilar neuronal cell loss, or whether it is related specifically to an activation-dependent plasticity. We recently demonstrated that blocking nerve growth factor (NGF) biological activity retards seizure development and inhibits the sprouting of mossy fibers. We now demonstrate that intraventricular administration of NGF itself accelerates the progression of kindling epileptogenesis, increases mossy fiber sprouting in the CA3 region and in the inner molecular layer (IML), but reduces seizure-induced decreases in hilar cell density. These findings provide support for a role of NGF in kindling and kindling-induced mossy fiber sprouting. In addition, the results dissociate this form of epileptogenesis from hilar cell loss or decreases in hilar cell density attributable to increases in hilar area, thereby supporting seizure-induced mossy fiber sprouting as being primarily attributable to the combined effects of neuronal activation and the activation-induced upregulation of growth factors.


Subject(s)
Cell Count/drug effects , Epilepsy/chemically induced , Kindling, Neurologic/physiology , Nerve Growth Factors/pharmacology , Nerve Regeneration/drug effects , Animals , Disease Models, Animal , Male , Rats
11.
Science ; 274(5293): 1729-32, 1996 Dec 06.
Article in English | MEDLINE | ID: mdl-8939868

ABSTRACT

The functions of the low-affinity p75 nerve growth factor receptor (p75(NGFR)) in the central nervous system were explored in vivo. In normal mice, approximately 25 percent of the cholinergic basal forebrain neurons did not express TrkA and died between postnatal day 6 and 15. This loss did not occur in p75(NGFR)-deficient mice or in normal mice systemically injected with a p75(NGFR)-inhibiting peptide. Control, but not p75(NGFR)-deficient, mice also had fewer cholinergic striatal interneurons. Apparently, p75(NGFR) mediates apoptosis of these developing neurons in the absence of TrkA, and modulation of p75(NGFR) can promote neuronal survival. Cholinergic basal forebrain neurons are involved in learning and memory.


Subject(s)
Apoptosis , Neurons/cytology , Parasympathetic Nervous System/cytology , Prosencephalon/cytology , Receptors, Nerve Growth Factor/physiology , Animals , Cell Survival , Choline O-Acetyltransferase/metabolism , DNA Fragmentation , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neostriatum/cytology , Neurons/enzymology , Oligopeptides/pharmacology , Phosphorylation , Proto-Oncogene Proteins/metabolism , Purkinje Cells/cytology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Nerve Growth Factor , Receptor, trkA , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/metabolism
12.
Proc Natl Acad Sci U S A ; 92(21): 9495-9, 1995 Oct 10.
Article in English | MEDLINE | ID: mdl-7568161

ABSTRACT

Kindling, an animal model of epilepsy wherein seizures are induced by subcortical electrical stimulation, results in the upregulation of neurotrophin mRNA and protein in the adult rat forebrain and causes mossy fiber sprouting in the hippocampus. Intraventricular infusion of a synthetic peptide mimic of a nerve growth factor domain that interferes with the binding of neurotrophins to their receptors resulted in significant retardation of kindling and inhibition of mossy fiber sprouting. These findings suggest a critical role for neurotrophins in both kindling and kindling-induced synaptic reorganization.


Subject(s)
Epilepsy/veterinary , Ganglia/drug effects , Hippocampus/drug effects , Nerve Growth Factors/pharmacology , Peptide Fragments/pharmacology , Receptors, Nerve Growth Factor/metabolism , Animals , Biological Assay , Brain-Derived Neurotrophic Factor , Electric Stimulation , Epilepsy/metabolism , Epilepsy/prevention & control , Ganglia/cytology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Hippocampus/cytology , Histocytochemistry , Kindling, Neurologic/drug effects , Male , Nerve Fibers/drug effects , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurotrophin 3 , Peptide Fragments/administration & dosage , Rats , Seizures/prevention & control , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects
13.
J Neurosci ; 15(7 Pt 2): 5316-23, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7623154

ABSTRACT

Repeated subconvulsive electrical stimulation of certain areas of the forebrain leads to kindling, a progressive and permanent amplification of evoked epileptiform activity, which is a model for human temporal lobe epilepsy. Recent studies have shown that kindling induces synthesis of nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) but not neurotrophin-3 (NT-3) in the hippocampus and cortex. Kindling also elicits mossy fiber sprouting and functional synaptogenesis in the supragranular layer, the hilus, and the CA3 region of the hippocampus. Intraventricular administration of antibodies to NGF has been shown to effectively block septohippocampal sprouting in the adult rat, and has been reported to retard amygdaloid kindling. In the present study, we have investigated the possible role of NGF in both kindling and kindling-associated sprouting. We have confirmed a kindling-induced sprouting of the mossy fibers into the stratum oriens of the CA3 region of the hippocampus, utilizing a new semiquantitative method of analysis based on Timm staining. Previous studies found no overt signs of hippocampal damage with this kindling paradigm, indicating that the increased Timm staining likely reflects a purely activity-induced sprouting. Intraventricular infusion of affinity-purified anti-NGF IgGs (which cross-react with NT-3 but not BDNF) resulted in both significant retardation of kindling and inhibition of the kindling-induced mossy fiber sprouting. The findings suggest a role for NGF in both these phenomena.


Subject(s)
Brain/physiology , Kindling, Neurologic , Nerve Growth Factors/physiology , Neural Inhibition/physiology , Animals , Antibodies/immunology , Brain/growth & development , Brain-Derived Neurotrophic Factor , Choline O-Acetyltransferase/metabolism , Hippocampus/growth & development , Immunoglobulin G/immunology , Immunohistochemistry , Injections, Intraventricular , Male , Nerve Fibers/physiology , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/immunology , Nerve Tissue Proteins/pharmacology , Neurites/physiology , Rats , Rats, Sprague-Dawley , Seizures/etiology , Seizures/prevention & control
14.
Eur J Neurosci ; 7(1): 160-8, 1995 Jan 01.
Article in English | MEDLINE | ID: mdl-7711931

ABSTRACT

Cholinergic hypofunction has often been correlated with a variety of behavioural impairments. In the present study, adult Wistar rats were intraventricularly infused with antibodies to nerve growth factor (anti-NGF) to examine the effects on cholinergic neurons of the basal forebrain, and on behavioural performance. Immunocytochemical techniques indicated that chronically infused anti-NGF penetrates into the basal forebrain, cortex, striatum, corpus callosum and hippocampus, confirming previous findings after a single injection. Treatment with anti-NGF for 1 or 2 weeks resulted in a significant decrease of 27-33% in density of choline acetyltransferase immunostaining of the cholinergic cell bodies in the medial septum and vertical diagonal band, and a 26% reduction in choline acetyltransferase enzyme activity in the septal area. An array of spatial learning Morris water maze tasks was used to distinguish between acquisition skills and the flexible use of learned information in novel tests. Rats subjected to the spatial learning paradigm received anti-NGF infusion for 2 weeks prior to and for another 2 weeks during the behavioural testing. The anti-NGF-treated animals were found to be no different from those receiving control serum in the Morris water maze acquisition task, either in the latency to find the platform or in the time spent searching in the training quadrant when the platform was removed. However, in consecutive extinction trials, anti-NGF rats continued to search in the empty training quadrant, suggesting the occurrence of perseveration; control rats expanded their search over other areas of the pool.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Brain/physiology , Cholinergic Fibers/physiology , Animals , Female , Immunohistochemistry , Learning , Nerve Growth Factors , Rats , Rats, Wistar , Spatial Behavior
15.
Neuroreport ; 4(5): 487-90, 1993 May.
Article in English | MEDLINE | ID: mdl-8513123

ABSTRACT

Pyramidal neurones of the rat neocortex do not normally express NADPH-diaphorase reactivity. However, after stab lesions which extended through the entire depth of the neocortex, strong NADPH-diaphorase reactivity was observed in pyramidal neurones at 7 and 14 days post-lesion. At 3 and 21 days post-lesion fewer and less reactive pyramidal neurones were observed, and no reactive pyramidal neurones were seen at 2 and 26 days post-lesion. The great majority of reactive pyramidal neurones were in layers V and VI and most were situated medial to the lesion. The induction of NADPH-diaphorase implies that the capability to synthesize nitric oxide may be a component of the pyramidal neurones' response to traumatic injury.


Subject(s)
Cerebral Cortex/injuries , NADPH Dehydrogenase/metabolism , Neurons/metabolism , Pyramidal Tracts/injuries , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Female , Histocytochemistry , Pyramidal Tracts/cytology , Pyramidal Tracts/metabolism , Rats , Rats, Wistar
16.
J Comp Neurol ; 326(1): 91-100, 1992 Dec 01.
Article in English | MEDLINE | ID: mdl-1479072

ABSTRACT

We have used an antiserum raised against mouse 2.5S NGF to examine the involvement of endogenous neurotrophins in the collateral sprouting of septohippocampal fibers in the adult rat brain. The antiserum was administered intraventricularly. Immunocytochemical techniques indicated that the injected antibodies penetrated into brain tissue that included the basal forebrain, cortex, striatum, corpus callosum, and hippocampus. Unilateral lesioning of the entorhinal cortex was done to evoke the sprouting of the cholinergic septohippocampal fibers. At 8 days postlesion, the sprouting was much advanced, as evidenced by an increase in density of the acetylcholinesterase (AChE) staining in the outer molecular layer (OML) of the dentate gyrus and by the associated increase in the absolute number of AChE-positive fibers in the OML. As well, there was a widening of the inner molecular layer (IML), interpreted as being due to sprouting of noncholinergic axons in that region. In rats injected daily with anti-NGF or anti-NGF Fab fragments, no increase in AChE density, or in the population of AChE-positive fibers, was observed in the OML. In contrast, the widening of the IML seemed to be unaffected by the anti-NGF treatment. No changes were observed in the AChE related parameters in the dentate gyrus of nonlesioned animals treated similarly for 8 days with anti-NGF; there was, however, a decrease of choline acetyltransferase (ChAT) immunostaining in the ChAT-positive cells of the basal forebrain. Our findings and the confirmation that our polyclonal anti-NGF also recognizes other members of the NGF neurotrophin family, specifically brain-derived neurotrophic factor and neurotrophin-3, indicate that at least one of these neurotrophins plays a key role in the collateral sprouting of the cholinergic septohippocampal fibers (but not that presumed to occur within the IML) following an entorhinal cortex lesion.


Subject(s)
Antibodies/immunology , Cerebral Cortex/physiology , Hippocampus/cytology , Nerve Fibers/immunology , Nerve Growth Factors/immunology , Acetylcholinesterase/immunology , Acetylcholinesterase/metabolism , Animals , Axons/immunology , Axons/metabolism , Choline O-Acetyltransferase/immunology , Choline O-Acetyltransferase/metabolism , Female , Hippocampus/immunology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/isolation & purification , Immunohistochemistry , Injections, Intraventricular , Nerve Fibers/ultrastructure , Rats , Rats, Wistar , Silver Staining
17.
Br J Pharmacol ; 103(1): 1041-6, 1991 May.
Article in English | MEDLINE | ID: mdl-1678980

ABSTRACT

1. In rats, recovery of sensory-motor function following a crush lesion of the sciatic or tibial nerve was monitored by measuring foot reflex withdrawal from a local noxious stimulation of the foot sole. 2. Putative neurotrophic compounds were tested on this functional recovery model: melanocortins (peptides derived from ACTH (corticotropin) and alpha-MSH (melanotropin], gangliosides and nimodipine were effective whereas isaxonine and TRH (thyrotropin releasing hormone) were not. 3. Structure-activity studies with melanocortins revealed a similar effectiveness of alpha-MSH, [N-Leu4, D-Phe7]-alpha-MSH, desacetyl-alpha-MSH and the ACTH analogue ORG 2766, questioning the validity of the previously suggested notion that the melanotrophic properties of these peptides are responsible for their neurotrophic effect. 4. As recovery of function after peripheral nerve damage follows a similar time course in hypophysectomized (five days post operation) and sham-operated rats, effective melanocortin therapy does not mimic an endogenous peptide signal in the repair process from pituitary origin. 5. Subcutaneous treatment with ORG 2766 (7.5 micrograms kg-1 48 h-1) facilitates recovery of function following peripheral nerve damage in young (6-7 weeks old), mature (5 month old) and old (20 month old) rats. 6. In view of the diversity in structure of the effective neurotrophic factors and the complexity of nerve repair, the present data support the notion that peripheral nerve repair may be facilitated by different humoral factors likely to be active on different aspects of the recovery process.


Subject(s)
Neurotransmitter Agents/physiology , Peripheral Nerves/drug effects , Adrenocorticotropic Hormone/analogs & derivatives , Adrenocorticotropic Hormone/pharmacology , Aging/physiology , Animals , Anticonvulsants/pharmacology , Female , Hypophysectomy , Melanocyte-Stimulating Hormones/analogs & derivatives , Melanocyte-Stimulating Hormones/pharmacology , Nimodipine/pharmacology , Peptide Fragments/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Inbred Strains , Reflex/drug effects , Sciatic Nerve/drug effects , Structure-Activity Relationship
18.
Neurobiol Aging ; 11(4): 451-6, 1990.
Article in English | MEDLINE | ID: mdl-2381504

ABSTRACT

In aged rats neuromuscular function and motor coordination is gradually impaired. Major motor deficits were seen in rats of more than 2 years of age; with increasing age, the incidence of abnormal footprints increased sharply. Oral nimodipine, a Ca2(+)-entry blocker of the dihydropyridine type, treatment suppressed and/or delayed the appearance of these abnormal footprints. In aged rats that already displayed a considerable amount of abnormal footprints in the free walking pattern, oral nimodipine treatment was similarly effective. Nimodipine not only delays the onset of age-related motor deficits, but also may counteract these deficits once already present. In aged rats the nerve conduction velocities were severely diminished. Nimodipine treatment resulted in an enhancement of the sciatic and caudal nerve conduction velocities. Histological analysis revealed a lower fiber density in aged rats compared to aged nimodipine-treated rats. Whether nimodipine acts directly on the peripheral nervous system is currently unclear. Nevertheless, the present study lends further support for the beneficial effects of nimodipine in age-related motor deficits in the rat.


Subject(s)
Aging/pathology , Motor Activity/drug effects , Neural Conduction/drug effects , Nimodipine/pharmacology , Peripheral Nerves/drug effects , Administration, Oral , Animals , Male , Motor Activity/physiology , Neural Conduction/physiology , Nimodipine/administration & dosage , Peripheral Nerves/pathology , Rats , Rats, Inbred Strains , Sciatic Nerve/drug effects , Sciatic Nerve/pathology
20.
J Neurosci ; 9(10): 3505-12, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2552034

ABSTRACT

Recently it has been shown that B-50 is identical to the neuron-specific, growth-associated protein GAP43. The present study reports on the fate of B-50/GAP43 mRNA and B-50/GAP43 protein, determined by radioimmunoassay, in a rat model of peripheral nerve regeneration (sciatic nerve crush) over a period of 37 and 312 d, respectively. Moreover, the effects of repeated subcutaneous injection of the neurotrophic peptide Org.2766 (an ACTH4-9 analog) and of a conditioning lesion on B-50/GAP43 protein levels in the regenerating nerve and dorsal root ganglia (DRG) were investigated. Both treatments enhanced the functional recovery as evidenced by a foot-flick withdrawal test. Immunocytochemical analysis using antineurofilament antibodies revealed a peptide-induced increase in the number of outgrowing sprouts in the sciatic nerve. Both the peptide and the conditioning lesion amplified the crush lesion-induced increase in B-50 protein content in the nerve as determined by radioimmunoassay. B-50 protein levels seem to correlate proportionally with the number of sprouts. In the DRG of the crushed sciatic nerve, the time course of B-50 expression was studied. B-50 mRNA was quantified from Northern blots. A linear increase up to 10 times the basal level of B-50 mRNA was observed 2 d postsurgery, followed by a gradual decline to normal levels at day 37. The first significant rise in B-50 mRNA level became apparent between 8 and 16 hr after placement of the crush lesion. The first significant rise in B-50 protein level occurred 40 hr after the crush lesion, reaching a plateau of 3 times the basal level between day 6 and 20. B-50 protein levels in DRG cell bodies remained elevated up to 60 d after crush, a period much longer than that observed for B-50 mRNA. Thus, during a later phase of peripheral axonal regeneration, the presence of B-50 appears to be prolonged, probably by an increase in half-life and not so much by enhanced transcription. Treatment with Org.2766 did not affect the B-50/GAP43 levels in DRG cell bodies during the first 6 d following crush. Conditioning lesion resulted in a DRG B-50/GAP43 protein amount at the same level as in rats 14 d after the test lesion. B-50/GAP43 levels in DRG are probably influenced by the rapid axonal transport of the protein, as has been reported by others.


Subject(s)
Ganglia, Spinal/metabolism , Membrane Proteins/metabolism , Nerve Regeneration , Nerve Tissue Proteins/metabolism , Sciatic Nerve/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Female , GAP-43 Protein , Growth Substances/metabolism , Membrane Proteins/genetics , Motor Activity/physiology , Nerve Crush , Nerve Tissue Proteins/genetics , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Sensation , Tibial Nerve/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...