Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain Res ; 1367: 130-45, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-20869353

ABSTRACT

Synaptic vesicle 2 proteins (SV2), SV2A, SV2B and SV2C, are integral proteins localized on the surface of synaptic vesicles in all neurons. SV2 proteins appear to play an important, but not yet fully understood role in synaptic vesicle exocytosis and neurotransmitter release. Moreover, SV2 seems to be the receptor of the botulinum neurotoxin A. In the present study, using single and double-labeling fluorescent immunohistochemistry and in situ hybridization we have identified the brain pattern of SV2C mRNA and protein expression in mice. Our results indicated that SV2C protein was expressed in a small subset of brain regions including the olfactory bulb, olfactory tubercle, nucleus accumbens, caudate-putamen, ventral pallidum, globus pallidus, substantia nigra and the ventral tegmental area. These results were confirmed by means of in situ hybridization, except for the globus pallidus and the substantia nigra pars reticulata, in which no labeling was found, suggesting that SV2C-positive fibers in these areas are terminals of striatal projecting neurons. In the striatum, we found that, in addition to its presence in the projection neurons, SV2C was densely expressed in a fraction (around 45%) of cholinergic interneurons. In addition, our data also showed that SV2C was densely expressed in most dopaminergic neurons in the substantia nigra pars compacta and the ventral tegmental area (more than 70% of the dopaminergic neurons analyzed were SV2C-positive). Altogether, our results suggest that SV2C may contribute to the regulation of neurotransmitter release and synaptic transmission in the basal ganglia including cholinergic striatal interneurons and nigro-striatal/mesolimbic dopamine neurons.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , Animals , Basal Ganglia/cytology , Basal Ganglia/metabolism , Gene Expression Regulation/genetics , Male , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Neuropeptide Y/metabolism
2.
Neurology ; 61(11 Suppl 6): S24-9, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14663005

ABSTRACT

A2A receptor is highly coexpressed with enkephalin and D2 receptor in striatopallidal neurons. A2A antagonists acutely enhance motor behavior in animal models of Parkinson's disease (PD) and are therefore considered potential PD therapeutic agents. Analysis of gene expression regulation using pharmacologic tools or A2A receptor-deficient mice (A2A-/-) shows that the A2A receptor positively and tonically controls the expression of enkephalin and immediate early genes in striatopallidal neurons. Because this regulation strictly mirrors the effect of D2 receptor, these data strongly support the hypothesis that A2A antagonists reduce the activity of striatopallidal neurons in models of PD. However, analysis of A2A-/- mice suggests additional effects of A2A receptor in the control of striatal physiology. Unexpectedly, these animals exhibited a reduction in exploratory activity and a 50% reduction in substance P expression. This was associated with a 45% decrease in the striatal extracellular dopamine concentration, suggesting that chronic absence of A2A receptor results in a functional hypodopaminergic state in the striatum. The A2A receptor controls inhibitory synaptic transmission negatively in the striatum and positively in the globus pallidus; this further supports the efficacy of A2A antagonists in reducing the activity of striatopallidal neurons in PD. The A2A receptor does not modulate basal alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA)-mediated excitatory corticoaccumbal synaptic transmission during normal physiologic conditions. However, genetic inactivation or pharmacologic blockade of the A2A receptor significantly reduced long-term potentiation (LTP) at this synapse. Therefore, this receptor is implicated in the induction of corticoaccumbal LTP, an effect that could be related to its involvement in long-term behavioral sensitization to repeated dopaminergic treatment.


Subject(s)
Corpus Striatum/metabolism , Gene Expression Regulation/physiology , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Synaptic Transmission/physiology , Animals , Corpus Striatum/cytology , Genes, Immediate-Early/physiology , Humans , Ion Channels/metabolism , Neuropeptides/genetics
3.
Synapse ; 42(2): 63-76, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11574941

ABSTRACT

In order to assess for the respective involvement of adenosine A(1) and A(2A) receptors (A(2A)-R) in the consequences of short- and long-term caffeine exposure on gene expression, the effects of acute caffeine administration on striatal, cortical, and hippocampal expression of immediate early genes (IEG), zif-268 and arc, and the effects of long-term caffeine or 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) exposure (once daily for 15 days) on striatal gene expression of substance P, enkephalin, and glutamic acid decarboxylase isoforms, GAD65 and GAD67, were evaluated in wild-type and A(2A)-R-deficient (A(2A)-R(-/-)) mice. In situ hybridization histochemistry was performed using oligonucleotides followed by quantitative image analysis. Our results demonstrated that a biphasic response of IEG expression to acute caffeine observed in the wild-type striatum was resumed in a monophasic response in the mutant striatum. In the cerebral cortex and hippocampus, the effect of caffeine was weak in wild-type, whereas in mutant mice it induced a 2-3-fold increase in the IEG expression to restore a level similar to the wild-type basal expression. Chronic caffeine and DPCPX-mediated regulation in neuropeptide and GADs striatal gene expression typically showed the mimicking of alterations resulting from the A(2A)-R genetic deficiency in 25 mg/kg caffeine-treated wild-type mice as well as the dose-dependent normalization of substance P and enkephalin expression in A(2A)-R(-/-) mice. These results indicate that, depending on the dose, the blockade of A(2A)-R or A(1) receptors by caffeine is preferentially revealed leading to highly differential alterations in striatal gene expression and they also suggested the central role of these two receptors on the control of dopaminergic functions.


Subject(s)
Caffeine/pharmacology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Immediate-Early Proteins , Neostriatum/drug effects , Neuropeptides/drug effects , Phosphodiesterase Inhibitors/pharmacology , Receptors, Purinergic P1/deficiency , Animals , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Early Growth Response Protein 1 , Enkephalins/genetics , Gene Expression Regulation/physiology , Genes, Immediate-Early/physiology , Glutamate Decarboxylase/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Isoenzymes/genetics , Male , Mice , Mice, Knockout , Neostriatum/metabolism , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Purinergic P1 Receptor Antagonists , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Adenosine A2A , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Substance P/genetics , Transcription Factors/genetics , Xanthines/pharmacology
4.
Neuroscience ; 105(4): 1019-30, 2001.
Article in English | MEDLINE | ID: mdl-11530239

ABSTRACT

The germinative ventricular zone of embryonic brain contains neural lineage progenitor cells that give rise to neurons, astrocytes and oligodendrocytes. The ability to generate neurons persists at adulthood in restricted brain areas. During development, many growth factors exert their effects by interacting with tyrosine kinase receptors and activate the phosphatidylinositol 3-kinase and the Ras/MAP kinase pathways. By its ability to modulate these pathways, the recently identified Src homology 2 domain-containing inositol polyphosphate 5-phosphatase 2, SHIP2, has the potential to regulate neuronal development. Using in situ hybridization technique with multiple synthetic oligonucleotides, we demonstrated that SHIP2 mRNA was highly expressed in the ventricular zone at early embryonic stages and subventricular zones at latter stages of brain and spinal cord and in the sympathetic chain. No significant expression was seen in differentiated fields. This restricted expression was maintained from embryonic day 11.5 to birth. In the periphery, large expression was detected in muscle and kidney and moderate expression in thyroid, pituitary gland, digestive system and bone. In the adult brain, SHIP2 was mainly restricted in structures containing neural stem cells such as the anterior subventricular zone, the rostral migratory stream and the olfactory tubercle. SHIP2 was also detected in the choroid plexuses and the granular layer of the cerebellum. The specificity of SHIP2 expression in neural stem cells was further demonstrated by (i) the dramatic increase in SHIP2 mRNA signal in neural cell adhesion molecule (N-CAM)-deficient mice, which present an accumulation of progenitor cells in the anterior subventricular zone and the rostral migratory stream, (ii) the abundant expression of 160-kDa SHIP2 by western blotting in proliferating neurospheres in culture and its downregulation in non-proliferating differentiated neurospheres. In conclusion, the close correlation between the pattern of SHIP2 expression in the brain and the proliferative and early differentiative events suggests that the phosphatase SHIP2 may have important roles in neural development.


Subject(s)
Aging/metabolism , Brain/embryology , Brain/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Cells, Cultured , Cytological Techniques , Female , Fetus/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Probes , Neural Cell Adhesion Molecules/deficiency , Neurons/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , RNA, Messenger/metabolism , Tissue Distribution
5.
J Neurochem ; 78(1): 183-98, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11432985

ABSTRACT

Adenosine and caffeine modulate locomotor activity and striatal gene expression, partially through the activation and blockade of striatal A(2A) receptors, respectively. The elucidation of the roles of these receptors benefits from the construction of A(2A) receptor-deficient mice (A(2A)-R(-/-)). These mice presented alterations in locomotor behaviour and striatal expression of genes studied so far, which are unexpected regarding the specific expression of A(2A) receptor by striatopallidal neurones. To clarify the functions of A(2A) receptors in the striatum and to identify the mechanisms leading to these unexpected modifications, we studied the basal expression of immediate early and constitutive genes as well as dopamine and glutamate neurotransmission in the striatum. Basal zif268 and arc mRNAs expression was reduced in mutant mice by 60-80%, not only in the striatum but also widespread in the cerebral cortex and hippocampus. Striatal expression of substance P and enkephalin mRNAs was reduced by about 50% and 30%, respectively, whereas the expression of GAD67 and GAD65 mRNAs was slightly increased and unaltered, respectively. In vivo microdialysis in the striatum revealed a 45% decrease in the extracellular dopamine concentration and three-fold increase in extracellular glutamate concentration. This was associated with an up-regulation of D(1) and D(2) dopamine receptors expression but not with changes in ionotropic glutamate receptors. The levels of tyrosine hydroxylase and of striatal and cortical glial glutamate transporters as well as adenosine A(1) receptors expression were indistinguishable between A(2A)-R(-/-) and wild-type mice. Altogether these results pointed out that the lack of A(2A) receptors leads to a functional hypodopaminergic state and demonstrated that A(2A) receptors are necessary to maintain a basal level in immediate early and constitutive genes expression in the striatum and cerebral cortex, possibly via their control of dopamine pathways.


Subject(s)
Corpus Striatum/metabolism , Dopamine/deficiency , Receptors, Purinergic P1/physiology , ATP-Binding Cassette Transporters/metabolism , Amino Acid Transport System X-AG , Animals , Dopamine/metabolism , Enkephalins/metabolism , Extracellular Space/metabolism , Gene Expression , Genes, Immediate-Early/physiology , Glutamate Decarboxylase/metabolism , Glutamic Acid/metabolism , Isoenzymes/metabolism , Male , Mice , Mice, Knockout/genetics , Neuroglia/metabolism , Receptor, Adenosine A2A , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P1/deficiency , Substance P/metabolism , Tyrosine 3-Monooxygenase/metabolism
6.
Neuroreport ; 11(14): 3243-6, 2000 Sep 28.
Article in English | MEDLINE | ID: mdl-11043556

ABSTRACT

Most effects of nociceptin are related to blockade of stress and anxiolytic-like effects. This neuropeptide is highly expressed in septal nuclei, which are involved in response to stressful situations. Dopamine and adenosine may have modulatory effects on stress behaviour by acting on septal neurons. We therefore analysed the regulation of septal nociceptin expression using quantitative in situ hybridization following manipulations of adenosine and dopamine neurotransmission. No difference was observed between wild-type and A2A receptor-deficient mice. In both genotypes, chronic treatments with caffeine, an equipotent A1 and A2A adenosine receptor antagonist, did not significantly modify nociceptin expression. 6-Hydroxydopamine-induced dopamine depletion was also without effect. These results demonstrate that dopamine and adenosine are not involved in the regulation of septal nociceptin expression in spite of the involvement of these three neurotransmitters in stress and anxiety behaviours.


Subject(s)
Adenosine/metabolism , Dopamine/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Opioid Peptides/genetics , Septal Nuclei/metabolism , Stress, Physiological/metabolism , Animals , Caffeine/pharmacology , Denervation/adverse effects , Dose-Response Relationship, Drug , Male , Mice , Mice, Knockout , Neural Pathways/cytology , Neural Pathways/drug effects , Neurons/cytology , Neurons/drug effects , Oxidopamine/adverse effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Adenosine A2A , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/genetics , Receptors, Purinergic P1/metabolism , Septal Nuclei/cytology , Septal Nuclei/drug effects , Stress, Physiological/physiopathology , Nociceptin
8.
Neuroreport ; 10(14): 2993-7, 1999 Sep 29.
Article in English | MEDLINE | ID: mdl-10549811

ABSTRACT

We have analyzed the effects of a small cortical infarct which is known to induce dramatic changes in gene expression in the entire cerebral cortex, on the gene expression in the striatum, a target structure of cortical neurons. Striatal glutamic acid decarboxylase (GAD67) and enkephalin expressions were increased in the striatum ipsilateral to the lesion. Conversely, neuropeptide Y- and somatostatin-like immunoreactivity were decreased in the ipsilateral striatum and this decrease was only related to a decrease in the labeling of processes with no changes in the number of labeled neurons. A minimal cortical lesion may therefore induce changes in gene expression in a subcortical structure through hyperactivity of glutamatergic synaptic inputs. One should therefore remember these extensive and long-lasting effects when surgical manipulations are performed on rat brain for stereotaxic surgery and placement of electrodes or probes.


Subject(s)
Brain Injury, Chronic/metabolism , Cerebral Cortex/injuries , Glutamate Decarboxylase/biosynthesis , Neostriatum/metabolism , Neuropeptides/biosynthesis , Animals , Enkephalins/biosynthesis , Immunohistochemistry , In Situ Hybridization , Male , Neostriatum/enzymology , Neuropeptide Y/biosynthesis , Oligonucleotide Probes , Protein Precursors/biosynthesis , RNA, Messenger/biosynthesis , Radiography , Rats , Rats, Wistar , Somatostatin/biosynthesis
9.
Eur J Neurosci ; 11(9): 3101-14, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10510174

ABSTRACT

Adenosine and the adenosine receptor antagonist, caffeine, modulate locomotor activity and striatal neuropeptide expression through interactions with the dopaminergic system by mechanisms which remain partially undetermined. We addressed this question by using quantitative immunocytochemistry and in situ hybridization, combined with retrograde tracing of striatal neurons, to characterize the mechanism(s) leading to the striatal increase in the immediate early genes (IEG), c-fos, zif-268 and arc, following a single injection of caffeine or the A1 antagonist, 1,3-dipropyl-8-cyclopentylxanthine (DPCPX). Caffeine and DPCPX induced c-fos, zif-268 and arc expression, both at mRNA and protein levels, in large proportions of striatonigral and striatopallidal neurons. The involvement of dopamine systems was evaluated by manipulations of the dopaminergic transmission. Quinpirole, a D2 agonist, almost completely blocked the caffeine-induced IEG increase in both striatopallidal and striatonigral neurons. Conversely, the lesion of the nigrostriatal pathway and the D1 antagonist SCH23390 abolished the caffeine effects in striatonigral neurons but had no or slight effect, respectively, on its action in striatopallidal neurons. These observations demonstrate that caffeine- and DPCPX-mediated IEG inductions involved different mechanisms in striatonigral and striatopallidal neurons through blockade of A1 receptors. Immediate early gene inductions result from a stimulation of dopamine release in striatonigral neurons and from activation of glutamate release and probably also acetylcholine release in striatopallidal neurons. These results also support the idea that, besides A2A receptors, adenosine acting at the A1 receptor plays pivotal functions in the basal ganglia physiology and that blockade of these receptors by specific or nonspecific antagonists, DPCPX and caffeine, may influence a broad range of neuronal functions in the striatum.


Subject(s)
Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , DNA-Binding Proteins/biosynthesis , Dopamine/physiology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/genetics , Genes, fos/genetics , Immediate-Early Proteins , Muscle Proteins/biosynthesis , Neostriatum/metabolism , Receptors, Purinergic P1/drug effects , Transcription Factors/biosynthesis , Adenosine/physiology , Animals , Apoptosis Regulatory Proteins , Cell Count , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Fluorescent Antibody Technique , Genes, Immediate-Early/drug effects , Genes, fos/drug effects , Hydroxydopamines/pharmacology , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Male , Muscle Proteins/genetics , Neostriatum/cytology , Neostriatum/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Transcription Factors/genetics
10.
Am J Pathol ; 155(4): 1105-13, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10514393

ABSTRACT

Pterin-4a-carbinolamine dehydratase (PCD) is a bifunctional protein also known as DCoH (dimerization co-factor of hepatocyte nuclear factor 1 (HNF1)). PCD/DCoH modulates the DNA binding specificity of HNF1, thus acting on its transcriptional activity. In addition, it participates in the recycling of tetrahydrobiopterin (BH(4)), an essential cofactor of several metabolic reactions. We investigated colorectal tumors and colorectal tumor cell lines as compared to normal colon samples in search of a potential differential expression of PCD/DCoH. Immunohistochemistry was conducted on 20 human colorectal tumors and 20 normal samples using a specific polyclonal antibody. Immunoblotting and RT-PCR analysis for PCD/DCoH and HNF1 were also performed on both human tissues and CACO-2 and HT-29 cell lines. All of the 20 tumors and both colon cancer cell lines presented a strong and widespread immunoreactivity for PCD/DCoH, contrasting with the absence of expression in the normal epithelia. We thus report the massive overexpression of PCD/DCoH in colon tumors, which is in striking contrast with the absence of staining in normal counterparts. The sharp contrast in the expression of a modulator of transcriptional activity between tumoral and normal cells may have a physiopathological role. PCD/DCoH could potentially be a new marker of malignant colon cells in vivo.


Subject(s)
Adenocarcinoma/enzymology , Colonic Neoplasms/enzymology , Hydro-Lyases/biosynthesis , Aged , Aged, 80 and over , Colon/enzymology , Female , Humans , Immunohistochemistry , Intestinal Mucosa/enzymology , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Tumor Cells, Cultured
11.
Brain Res ; 786(1-2): 181-8, 1998 Mar 09.
Article in English | MEDLINE | ID: mdl-9555004

ABSTRACT

Axotomy induces a profound modification of Ca2+ homeostasis in injured neurons which may lead to neuronal death. Remarkably, after axotomy and resection of the hypoglossal nerve, 65-75% of the hypoglossal motoneurons survive in the long term and this suggests some adaptive mechanisms compensating the massive calcium influx. As potential components of this adaptation, we have examined calmodulin and calbindin-D28k by in situ hybridisation and immunohistochemistry in motoneurons of the rat after hypoglossal nerve transection. Neuronal calbindin mRNA and protein content was low in normal state, transiently increased to 200% of the basal expression at 8 days post-operation (dpo), then declined to normal again until 28 dpo. Calmodulin mRNA was highly expressed in normal hypoglossal motoneurons and remained constant after axotomy. Calmodulin protein immunoreactivity, however, was transiently decreased in axotomised motoneurons suggesting post-transcriptional modification. The upregulation of calbindin expression may facilitate the survival of injured motoneurons.


Subject(s)
Axotomy , Calmodulin/metabolism , Hypoglossal Nerve/physiology , Motor Neurons/metabolism , S100 Calcium Binding Protein G/metabolism , Animals , Calbindin 1 , Calbindins , Calmodulin/genetics , Female , Immunohistochemistry , In Situ Hybridization , RNA, Messenger/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein G/genetics
12.
Cell Calcium ; 22(5): 367-72, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9448943

ABSTRACT

Calbindin D28K, an intracellular calcium-binding protein, acts as Ca2+ buffering system in the cytoplasm. By means of this property, calbindin may protect neurons against large fluctuations in free intracellular Ca2+ and, hence, may prevent cell death. Although axotomy causes a massive influx of calcium into the lesioned neurons, resection of the hypoglossal nerve does not induce extensive neuronal cell death in rats. Even several weeks after axotomy, about 70% of the motoneurons survive despite permanent target deprivation. The mechanisms responsible for this remarkable survival rate are unknown. In this study, we have looked at the modification of calbindin immunoreactivity in axotomized hypoglossal motoneurons. In non-axotomized motoneurons, no calbindin is detectable by immunocytochemistry. Axotomy induced an increase of calbindin immunoreactivity in lesioned motoneurons. This increase, visualised by the number of calbindin-immunoreactive neurons extended from 1 day to 28 days. At this time most, but not all, motoneurons located on the side of the lesion were calbindin-positive as shown by retrograde labeling and immunoquenching. From 14 days post operation, calbindin immunoreactivity decreased and reached its basal value after 35 days post operation. At that time, only fibres were still calbindin immunoreactive. Interestingly, calbindin-immunoreactivity was also increased in almost all cell nuclei, compatible with a nuclear regulation. These data are consistent with the hypothesis that, as a reaction to axotomy, motoneurons trigger an increase in calbindin expression which acts as a compensatory Ca(2+)-buffering system, enabling neurons to maintain Ca2+ homeostasis and the survival of many motoneurons after axotomy.


Subject(s)
Hypoglossal Nerve/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Stilbamidines , Animals , Axotomy , Calbindin 1 , Calbindins , Female , Fluorescent Dyes , Hypoglossal Nerve/surgery , Rats , Rats, Wistar
13.
Brain Res ; 777(1-2): 187-201, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9449428

ABSTRACT

GTPCH-I immunoreactive structures in the rat brain were studied using a polyclonal antibody raised in the chick. General mapping was made using the avidin-biotin-peroxidase technique and compared with the distribution of tyrosine hydroxylase and serotonin immunoreactivities. Double immunofluorescence was performed in order to establish real intracellular colocalization. GTPCH-I immunoreactivity was generally found to be low. Immunostained neurons were present in all the serotonin cell groups. In catecholaminergic neurons, although tyrosine hydroxylase immunoreactivity was always very high, GTPCH-I immunoreactivity was extremely variable, from relatively strong (substantia nigra, ventral tegmental area) to low (locus coeruleus, caudal part of the hypothalamus), extremely low (rostral hypothalamus, ventral brainstem) or almost absent (dorsal brainstem, some hypothalamic nuclei). When feasible, double immunolabeling revealed that all the serotonin cells and most of the tyrosine hydroxylase cells were also expressing GTPCH-I. Our results argue in favor of a regulation of tyrosine hydroxylase activity by the intracellular synthesis of BH4.


Subject(s)
Brain Chemistry , GTP Cyclohydrolase/analysis , GTP Cyclohydrolase/immunology , Neurons/enzymology , Animals , Antibody Specificity , Antioxidants , Biopterins/analogs & derivatives , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Locus Coeruleus/chemistry , Locus Coeruleus/cytology , Locus Coeruleus/enzymology , Male , Neurons/chemistry , Norepinephrine/physiology , Paraventricular Hypothalamic Nucleus/chemistry , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/enzymology , Precipitin Tests , Raphe Nuclei/chemistry , Raphe Nuclei/cytology , Raphe Nuclei/enzymology , Rats , Rats, Wistar , Serotonin/analysis , Serotonin/immunology , Substantia Nigra/chemistry , Substantia Nigra/cytology , Substantia Nigra/enzymology , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/immunology , Ventral Tegmental Area/chemistry , Ventral Tegmental Area/cytology , Ventral Tegmental Area/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...