Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 180(2): 144-55, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12684028

ABSTRACT

Amyloid beta peptides (Abeta) may be neurotoxic during the progression of Alzheimer's disease by eliciting oxidative stress. Exposure of neuronally differentiated SK-N-BE cells to Abeta(25-35) fragment as well as to full-length Abeta(1-40) and Abeta(1-42) induces early and time-dependent generation of oxidative stress that has been evaluated by carefully monitoring generation of hydrogen peroxide (H(2)O(2)), 4-hydroxynonenal (HNE), thiobarbituric acid reactive substances (TBARS), and fluorescent chromolipids. Abeta treatment also results in the activation of c-Jun aminoterminal kinases (JNKs) and p38(MAPK) and is followed by characteristic nuclear changes of apoptosis as evaluated by DAPI staining and TUNEL technique. To reproduce the relationships between oxidative stress and Abeta apoptosis we found that only the simultaneous administration of HNE and H(2)O(2), at concentrations similar to those generated within the first 3 h of Abeta exposure, can fully mimic Abeta-dependent activation of JNKs and p38(MAPK) and occurrence of apoptosis. Antioxidants such as alpha-tocopherol and N-acetylcysteine prevent completely either neuronal apoptosis or activation of JNKs and p38(MAPK) elicited by Abeta or by simultaneous HNE and H(2)O(2) addition. Finally, direct evidence that activation of these kinases is required for cell death induced by Abeta has been obtained by pretreating cell with specific inhibitors of JNKs and p38(MAPK). These results suggest the existence of a sequence of events in Abeta-induced apoptosis involving simultaneous generation of HNE and H(2)O(2) and oxidative stress-dependent activation of JNKs and p38(MAPK).


Subject(s)
Aldehydes/metabolism , Amyloid beta-Peptides/toxicity , Hydrogen Peroxide/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Aldehydes/toxicity , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/toxicity , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/toxicity , p38 Mitogen-Activated Protein Kinases
4.
J Biol Chem ; 275(51): 40561-7, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11007794

ABSTRACT

Hepatic stellate cells (HSC) undergo activation toward myofibroblast-like cells during early stages of liver injury associated with fibrogenesis. Platelet-derived growth factor (PDGF), particularly its BB isoform, has been identified as the most potent mitogen for HSC. 4-Hydroxy-2,3-nonenal and related 4-hydroxy-2, 3-alkenals (HAKs) have been suggested to modulate the process of HSC activation. In this study we investigated the relationship between HAKs and PDGF receptor activation in human HSC. By employing noncytotoxic concentrations (10(-6) m) of HAKs, we observed a significant inhibition of PDGF-BB-dependent DNA synthesis. HAKs inhibited relevant pathways of PDGF-BB-dependent mitogenic signaling, including autophosphorylation of PDGF receptor (PDGF-R) beta subunits and activation of phosphatidylinositol 3-kinase and extracellular regulated kinases 1/2. Inhibition of DNA synthesis was reversible, and recovery of PDGF-mediated mitogenic signaling occurred within 24-48 h and was associated with HAKs-induced up-regulation of PDGF-R beta gene expression. 4-Hydroxy-2,3-nonenal, used as a model HAK, inhibited the intrinsic tyrosine kinase activity associated with the PDGF-R beta subunit, whereas binding of PDGF to its receptor was unaffected. This study identifies a novel regulatory mechanism of reactive aldehydes on PDGF receptor signaling and biologic actions, which may be relevant in several pathophysiological conditions, including liver fibrosis.


Subject(s)
Aldehydes/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Tyrosine/metabolism , Aldehydes/pharmacology , Cell Line , DNA Replication , Humans , Phosphorylation , Receptor, Platelet-Derived Growth Factor beta/chemistry , Up-Regulation/drug effects
5.
Gut ; 46(4): 553-61, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10716687

ABSTRACT

BACKGROUND: Liver/kidney microsomal antibody type 1 (LKM1) is the marker of type 2 autoimmune hepatitis (AIH) and is detected in up to 6% of patients with hepatitis C virus (HCV) infection. It recognises linear and conformational epitopes of cytochrome P450IID6 (CYP2D6) and may have liver damaging activity, provided that CYP2D6 is accessible to effector mechanisms of autoimmune attack. METHODS: The presence of LKM1 in the plasma membrane was investigated by indirect immunofluorescence and confocal laser microscopy of isolated rat hepatocytes probed with 10 LKM1 positive sera (five from patients with AIH and five from patients with chronic HCV infection) and a rabbit polyclonal anti-CYP2D6 serum. RESULTS: Serum from both types of patient stained the plasma membrane of non-permeabilised cells, where the fluorescent signal could be visualised as discrete clumps. Conversely, permeabilised hepatocytes showed diffuse submembranous/cytoplasmic staining. Adsorption with recombinant CYP2D6 substantially reduced plasma membrane staining and LKM1 immunoblot reactivity. Plasma membrane staining of LKM1 colocalised with that of anti-CYP2D6. Immunoprecipitation experiments showed that a single 50 kDa protein recognised by anti-CYP2D6 can be isolated from the plasma membrane of intact hepatocytes. CONCLUSIONS: AIH and HCV related LKM1 recognise CYP2D6 exposed on the plasma membrane of isolated hepatocytes. This observation supports the notion that anti-CYP2D6 autoreactivity may be involved in the pathogenesis of liver damage.


Subject(s)
Antibodies/immunology , Autoantibodies/analysis , Cytochrome P-450 CYP2D6/immunology , Liver/enzymology , Adult , Animals , Biomarkers/analysis , Cell Membrane/immunology , Cells, Cultured , Child , Female , Fluorescent Antibody Technique, Indirect , Hepatitis C, Chronic/immunology , Hepatitis, Autoimmune/immunology , Humans , Liver/immunology , Male , Microscopy, Confocal , Middle Aged , Rabbits , Rats , Rats, Sprague-Dawley
6.
Int J Mol Med ; 4(4): 425-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10493986

ABSTRACT

4-hydroxy-2,3-alkenals (HAKs) are major end products of oxidative decomposition of omega-3 and omega-6 polyunsaturated fatty acids of membrane phospholipids, a process usually referred to as lipid peroxidation. These reactive aldehydic compounds have been unequivocally detected in vivo in either clinical or experimental conditions of chronic liver damage, suggesting an involvement of lipid peroxidation processes, elicited by either reactive oxygen intermediates (ROI) or by pro-oxidant agents, in the pathogenesis of liver fibrosis. Literature data provided by experimental studies with animal models of liver fibrosis or by studies performed on primary culture of human hepatic stellate cells (hHSC), which are known to play a major role in liver fibrogenesis, indicate that HAKs may sustain at molecular level the fibrogenic development of chronic liver diseases. These compounds may act as ultimate mediators of oxidative stress able to up-regulate the synthesis of extracellular matrix components and of growth factors, chemokines and cytokines, as well as to modulate functional responses of hepatic cell types involved in the progression of chronic liver diseases and to sustain chronic hepatitis.


Subject(s)
Alkenes/metabolism , Fatty Acids, Unsaturated/metabolism , Liver Cirrhosis/etiology , Oxidative Stress , Humans , Liver Cirrhosis/metabolism , Oxidation-Reduction
7.
Antioxid Redox Signal ; 1(3): 255-84, 1999.
Article in English | MEDLINE | ID: mdl-11229439

ABSTRACT

Reactive oxygen intermediates (ROI) and other pro-oxidant agents are known to elicit, in vivo and in vitro, oxidative decomposition of omega-3 and omega-6 polyunsaturated fatty acids of membrane phospholipids (i.e, lipid peroxidation). This leads to the formation of a complex mixture of aldehydic end-products, including malonyldialdehyde (MDA), 4-hydroxy-2,3-nonenal (HNE), and other 4-hydroxy-2,3-alkenals (HAKs) of different chain length. These aldehydic molecules have been considered originally as ultimate mediators of toxic effects elicited by oxidative stress occurring in biological material. Experimental and clinical evidence coming from different laboratories now suggests that HNE and HAKs can also act as bioactive molecules in either physiological and pathological conditions. These aldehydic compounds can affect and modulate, at very low and nontoxic concentrations, several cell functions, including signal transduction, gene expression, cell proliferation, and, more generally, the response of the target cell(s). In this review article, we would like to offer an up-to-date review on this particular aspect of oxidative stress--dependent modulation of cellular functions-as well as to offer comments on the related pathophysiological implications, with special reference to human conditions of disease.


Subject(s)
Aldehydes/metabolism , Signal Transduction , Aldehydes/chemistry , Aldehydes/toxicity , Arteriosclerosis/chemically induced , Arteriosclerosis/physiopathology , Chemotactic Factors/physiology , Chronic Disease , Humans , Inflammation/physiopathology , Liver Diseases/physiopathology , Nervous System Diseases/physiopathology , Oxidative Stress , Proteins/metabolism , Reperfusion Injury/physiopathology
8.
J Clin Invest ; 102(11): 1942-50, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9835619

ABSTRACT

4-Hydroxy-2,3-nonenal (HNE) is an aldehydic end product of lipid peroxidation which has been detected in vivo in clinical and experimental conditions of chronic liver damage. HNE has been shown to stimulate procollagen type I gene expression and synthesis in human hepatic stellate cells (hHSC) which are known to play a key role in liver fibrosis. In this study we investigated the molecular mechanisms underlying HNE actions in cultured hHSC. HNE, at doses compatible with those detected in vivo, lead to an early generation of nuclear HNE-protein adducts of 46, 54, and 66 kD, respectively, as revealed by using a monoclonal antibody specific for HNE-histidine adducts. This observation is related to the lack of crucial HNE-metabolizing enzymatic activities in hHSC. Kinetics of appearance of these nuclear adducts suggested translocation of cytosolic proteins. The p46 and p54 isoforms of c-Jun amino-terminal kinase (JNKs) were identified as HNE targets and were activated by this aldehyde. A biphasic increase in AP-1 DNA binding activity, associated with increased mRNA levels of c-jun, was also observed in response to HNE. HNE did not affect the Ras/ERK pathway, c-fos expression, DNA synthesis, or NF-kappaB binding. This study identifies a novel mechanism linking oxidative stress to nuclear signaling in hHSC. This mechanism is not based on redox sensors and is stimulated by concentrations of HNE compatible with those detected in vivo, and thus may be relevant during chronic liver diseases.


Subject(s)
Aldehydes/pharmacology , JNK Mitogen-Activated Protein Kinases , Liver Cirrhosis/etiology , Liver Diseases/metabolism , Liver/cytology , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Genes, fos , Genes, jun , Histidine/chemistry , Histidine/drug effects , Humans , Lipid Peroxidation , Liver/metabolism , Liver Diseases/complications , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase 3 , Molecular Weight , Oxidative Stress , Protein Kinases/chemistry , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism
9.
J Hepatol ; 28(1): 40-5, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9537862

ABSTRACT

BACKGROUND/AIMS: Alcohol dehydrogenase, cytochrome P4502E1 (CYP2E1), and aldehyde dehydrogenase are known to play an important role in alcohol metabolism in the liver. Although the ethanol oxidation pathways are mainly localized in hepatocytes, we examine whether human hepatic stellate cells might also metabolize ethanol and acetaldehyde. METHODS: Hepatic stellate cells were isolated from normal human livers and exposed in vitro to 50 mmol/l ethanol or 85 micromol/l acetaldehyde for different periods of time. Alcohol dehydrogenase/aldehyde dehydrogenase activity and CYP2E1 protein expression were measured in hepatic stellate cells. Moreover, alcohol dehydrogenase and aldehyde dehydrogenase mRNA expression were evaluated in hepatic stellate cells. RESULTS: Exposure of hepatic stellate cells to ethanol for 24 h resulted in a 5-fold increase in cell alcohol dehydrogenase activity. The effect of ethanol on alcohol dehydrogenase activity was paralleled by a significant increase in the alcohol dehydrogenase mRNA expression in hepatic stellate cells. Acetaldehyde significantly increased the activity of high affinity aldehyde dehydrogenase in hepatic stellate cells, whereas ethanol was devoid of any effect. Acetaldehyde also induced high affinity aldehyde dehydrogenase mRNA expression in hepatic stellate cells. CYP2E1 was not expressed in hepatic stellate cells either in basal condition or after ethanol/acetaldehyde exposure. CONCLUSIONS: This study shows that human hepatic stellate cells have the capacity to metabolize both ethanol and acetaldehyde through a class I alcohol dehydrogenase- and an aldehyde dehydrogenase-oxidizing pathway. Conversely, no detectable levels of CYP2E1-associated proteins are expressed in these cells.


Subject(s)
Acetaldehyde/pharmacology , Alcohol Dehydrogenase/biosynthesis , Ethanol/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Liver/cytology , Liver/enzymology , Alcohol Dehydrogenase/analysis , Cells, Cultured , Cytochrome P-450 CYP2E1/analysis , Cytochrome P-450 CYP2E1/biosynthesis , Humans , RNA, Messenger/biosynthesis , Transcription, Genetic/drug effects
10.
Biochem Biophys Res Commun ; 238(2): 420-4, 1997 Sep 18.
Article in English | MEDLINE | ID: mdl-9299524

ABSTRACT

In this study we have investigated the occurrence of cytochrome P450 isoforms and of related cytochrome P450 reductase in human hepatic stellate cells (hHSC), a type of cell having relevant roles in physiopathological conditions of the liver. By performing immunoblotting of hHSC microsomes and immunofluorescence analysis associated to confocal laser microscopy we detected only P450 enzymes belonging to the cytochrome P450 3A subfamily (CYP3A) as well as cytochrome P450 reductase. The presence of CYP3A was further indicated by detection of testosterone 6beta-hydroxylase activity in hHSC microsomes. Other important human P450 forms were either undetectable (CYP1A2, CYP2E1, CYP2C8/9/19 and CYP4A) or bearly detectable (CYP1A1) in hHSC. This is the first study showing existence of active cytochrome P450 isoforms in human HSC.


Subject(s)
Adipocytes/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxidoreductases, N-Demethylating/metabolism , Cells, Cultured , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/analysis , Fluorescent Antibody Technique, Indirect , Humans , Liver/cytology , NADH, NADPH Oxidoreductases/analysis , NADPH-Ferrihemoprotein Reductase , Oxidoreductases, N-Demethylating/analysis
11.
Free Radic Biol Med ; 20(3): 351-9, 1996.
Article in English | MEDLINE | ID: mdl-8720905

ABSTRACT

Previous studies have suggested a possible involvement of free radical reactions in the pathogenesis of cholestatic liver injury as well as in the modulation of hepatic fibrogenesis. In this study we investigated whether lipid peroxidation is involved in the development of chronic liver damage induced by long-standing cholestasis. For this purpose we have used the rat model of bile duct ligation (BDL), which leads to liver fibrosis and cirrhosis. Using this model we observed that the development of chronic liver damage was associated with the onset of lipid peroxidation, as pointed out by detection of carbonyl compounds, 4-hydroxynonenal (HNE) and malondialdehyde (MDA), in BDL livers and of fluorescent adducts between MDA and serum proteins. Lipid peroxidation was a relatively late event (starting after 1-2 weeks of BDL) and was unrelated to the early development of liver necrosis and cholestasis (already evident after 72 h after BDL). A positive significant linear correlation between the kinetic of infiltration of neutrophils and of a monocyte/macrophage population in BDL livers and MDA and HNE generation in the same organs is presented, indicating a close link between lipid peroxidation and the activation of inflammatory cells. We also observed that a positive linear correlation exists between collagen deposition in these livers and hepatic production of MDA and HNE. This event, which is accompanied by an increase in the number of fat storing cells (FSC, the cells that produce collagen in fibrotic liver), suggests that lipid peroxidation in this model may contribute to stimulate collagen synthesis by proliferating FSC.


Subject(s)
Cholestasis/pathology , Lipid Peroxidation , Liver/pathology , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Analysis of Variance , Animals , Cholestasis/metabolism , Collagen/metabolism , Liver/metabolism , Liver Function Tests , Male , Rats , Rats, Wistar , Regression Analysis , Vitamin E/blood , Vitamin E/metabolism , gamma-Glutamyltransferase/blood
12.
Biochem Biophys Res Commun ; 214(2): 669-75, 1995 Sep 14.
Article in English | MEDLINE | ID: mdl-7677780

ABSTRACT

4-Hydroxy-2,3-nonenal is a major aldehydic end-product of lipid peroxidation known to exert several biological and cytotoxic effects and to be produced during conditions of chronic cholestasis. Here we report that viable hepatocytes isolated from cholestatic livers of bile duct-ligated rats (BDL hepatocytes) show a significantly lower rate of HNE metabolism than control cells. This feature is likely to be the consequence of a significant inhibition in the activity of HNE-metabolizing cytosolic glutathione-S-transferase and alcohol dehydrogenase in BDL hepatocytes. Particulate NADP-dependent aldehyde dehydrogenase was also inhibited. No significant change was found for aldehyde reductase activity. A decreased hepatocellular metabolism of HNE can expose liver parenchymal and non-parenchymal cells to cytotoxic as well as pro-inflammatory and pro-fibrogenic effects of HNE, contributing to the development of chronic cholestatic liver damage.


Subject(s)
Alcohol Dehydrogenase/metabolism , Aldehydes/metabolism , Cholestasis/metabolism , Liver/metabolism , Animals , Bile Ducts/physiology , Glutathione Transferase/metabolism , Kinetics , Lipid Peroxidation , Male , NAD/metabolism , NADP/metabolism , Rats , Rats, Wistar , Reference Values , Subcellular Fractions/enzymology
13.
J Bone Miner Res ; 9(8): 1239-49, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7976506

ABSTRACT

Differentiation of hypertrophic chondrocytes to an osteoblast-like phenotype occurs in vivo in the hypertrophic cartilage of chick embryo tibiae underneath early or prospective periosteum and in cartilage around vascular canals. Synthesis of type I collagen by hypertrophic chondrocytes was shown by immunolocalization of the C propeptide. By enzyme cytochemistry it was instead shown that, in vivo, further differentiating hypertrophic chondrocytes express alkaline phosphatase at the time of initial mineral deposition. Evidence that hypertrophic chondrocytes may resume proliferation was obtained by BrdU labeling. A monoclonal antibody (LA5) was isolated and characterized that recognizes a hypertrophic chondrocyte membrane protein. In addition to staining hypertrophic chondrocytes surrounded by a type II and type X collagen-stainable matrix, the LA5 antibodies also stained elongated chondrocytes at the cartilage/bone collar interface and cells incorporated in the first layer of bone and osteoid matrix.


Subject(s)
Bone Development/physiology , Growth Plate/cytology , Osteoblasts/cytology , Animals , Antigens, Surface/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Chick Embryo , Collagen/metabolism , Growth Plate/metabolism , Peptide Fragments/metabolism , Procollagen/metabolism
14.
Exp Cell Res ; 200(1): 26-33, 1992 May.
Article in English | MEDLINE | ID: mdl-1563490

ABSTRACT

Reduction of intercellular spaces in the areas of prospective cartilage and bone formation (precartilage condensation) precedes chondrogenesis and may represent an important step in the process of cartilage differentiation during limb skeletogenesis. We have attempted to clarify the role of the microenvironment established during cell condensation, taking advantage of a tissue culture model system that allows condensation (i.e., increased cell density due to cell aggregation) and chondrogenic differentiation (i.e., synthesis of cartilage-specific extracellular matrix proteins, such as type II collagen and acquisition of a chondrocyte morphology) of chick embryo cartilage-derived undifferentiated cells. To prevent condensation cells were grown in carboxymethylcellulose and changes in the differentiation pathway were evaluated. In another series of experiments, we have separated single cells from the aggregated cells and analyzed their differentiation properties. Morphological analyses and the evaluation of type II collagen expression, at both the protein and the mRNA level, show that a reduced rate of cell clustering and cell to cell contact parallels a reduction of cell recruitment into the differentiation program. On the basis of our results, we suggest that the following cascade of events regulates the early stages of chondrocyte differentiation: (a) the acquisition of the ability to establish cell to cell contacts, (b) the formation of a permissive environment capable of activating the differentiation program, and (c) the expression of differentiation markers.


Subject(s)
Cartilage/embryology , Cell Differentiation , Animals , Carboxymethylcellulose Sodium , Cartilage/cytology , Cartilage/ultrastructure , Cell Aggregation , Cell Communication , Cell Movement , Chick Embryo , Collagen/biosynthesis , Extracellular Space
15.
Dev Biol ; 149(1): 168-76, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1728587

ABSTRACT

The myc oncogene is expressed by proliferating quail embryo chondrocytes (QEC) grown as adherent cells and is repressed in QEC maintained in suspension culture. To investigate the interference of myc expression during chondrocyte differentiation, QEC were infected with a retrovirus carrying the v-myc oncogene (QEC-v-myc). Uninfected or helper virus-infected QEC were used as control. In adherent culture, QEC-v-myc displayed a chondrocytic phenotype and synthesized type II collagen and Ch21 protein, while control chondrocytes synthesized type I and type II collagen with no Ch21 protein detected as long as the attachment to the plastic was kept. In suspension culture, QEC-v-myc readily aggregated and within 1 week the cell aggregates released small single cells; still they secreted only type II collagen and Ch21 protein. In the same conditions control cell aggregates released hypertrophic chondrocytes producing type II and type X collagens and Ch21 protein. In the appropriate culture conditions, QEC-v-myc reconstituted a tissue defined as nonhypertrophic, noncalcifying cartilage by the high cellularity, the low levels of alkaline phosphatase enzymatic activity, and the absence of type X collagen synthesis and of calcium deposition. We conclude that the constitutive expression of the v-myc oncogene keeps chondrocytes in stage I (active proliferation and synthesis of type II collagen) and prevents these cells from reconstituting hypertrophic calcifying cartilage.


Subject(s)
Calcification, Physiologic , Cartilage/embryology , Gene Expression , Genes, myc , Animals , Cartilage/metabolism , Cartilage/pathology , Cell Division , Cells, Cultured , Chick Embryo , Collagen/chemistry , Fluorescent Antibody Technique , Hypertrophy/genetics , Hypertrophy/metabolism , Phenotype , Quail , RNA, Messenger/analysis , Tibia
16.
J Neurochem ; 52(6): 1854-8, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2542451

ABSTRACT

The effects of gamma-aminobutyric acid (GABA) on the spontaneous efflux of [3H]norepinephrine ([3H]NE) were studied in synaptosomes prepared from rat hippocampus and prelabelled with [3H]NE. It had been observed previously that, when synaptosomes were exposed in superfusion to GABA, the basal release of the tritiated catecholamine was enhanced, apparently with no involvement of the known GABA receptors. The mechanisms underlying this effect have now been investigated. The potency of GABA as a releaser of [3H]NE was decreased by lowering the Na+ content of the superfusion medium, and its effect disappeared at 23 mM Na+. The GABA-induced [3H]NE release was counteracted by the GABA uptake inhibitor N-(4,4-diphenyl-3-butenyl)nipecotic acid (SKF 89976A), but it was unaffected by the NE uptake blockers desmethylimipramine and nisoxetine. The GABA-induced release of [3H]NE was Ca2+-dependent and tetrodotoxin-sensitive. The data support the hypothesis that GABA provoked [3H]NE release by a novel mechanism which involves penetration into the noradrenergic nerve terminals through a GABA carrier located on the NE terminals themselves. This uptake process might be electrogenic and provoke depolarization of the nerve terminals, causing an exocytotic release of [3H]NE.


Subject(s)
Hippocampus/metabolism , Norepinephrine/metabolism , Proline/analogs & derivatives , Synapses/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Biomechanical Phenomena , Male , Nipecotic Acids/pharmacology , Rats , Rats, Inbred Strains , Receptors, Neurotransmitter/physiology , Sodium/pharmacology , Tetrodotoxin/pharmacology , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...