Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neuroscience ; 141(1): 379-89, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16675138

ABSTRACT

Behavioral and neuronal abnormalities observed in mice exhibiting a reduced expression of the dopamine transporter model important aspects of schizophrenia, addiction, and attentional disorders. As the consequences of a chronic hyperdopaminergic tone for striatal output regulation have remained poorly understood, the present experiments were designed to determine the status of striatal interneuronal cholinergic neurotransmission in dopamine transporter knockdown animals. The high-affinity choline transporter represents the rate-limiting step of acetylcholine synthesis and release. Compared with wild type mice, striatal high-affinity choline transporter expression in dopamine transporter knockdown mice was significantly decreased. As in vivo basal striatal acetylcholine release did not differ between the strains, reduced high-affinity choline transporter expression in dopamine transporter knockdown mice was not due to reduced basal cholinergic activity. Furthermore, the proportion of high-affinity choline transporters expressed in plasma membrane-enriched versus vesicular membrane-enriched fractions did not differ from wild type animals, suggesting that changes in intracellular high-affinity choline transporter trafficking were not associated with lower overall levels of striatal high-affinity choline transporters. Synaptosomal choline uptake assays indicated a reduced capacity of striatal high-affinity choline transporters in dopamine transporter knockdown mice, and thus the functional significance of the reduced level of high-affinity choline transporter expression. Likewise, in vivo measures of the capacity of striatal high-affinity choline transporters to clear increases in extracellular choline concentrations, using choline-sensitive microelectrodes, revealed a 37-41% reduction in hemicholinium-sensitive clearance of exogenous choline in dopamine transporter knockdown mice. Furthermore, clearance of potassium-evoked choline signals was reduced in dopamine transporter knockdown mice (1.63+/-0.15 microM/s) compared with wild type animals (2.29+/-0.21 microM/s). Dysregulated striatal cholinergic neurotransmission is hypothesized to disrupt the integration of thalamic and cortical information at spiny projection neurons and thus to contribute to abnormal striatal information processing in dopamine transporter knockdown mice.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Gene Expression Regulation/physiology , Membrane Transport Proteins/metabolism , Acetylcholine/metabolism , Analysis of Variance , Animals , Blotting, Western/methods , Brain Chemistry/genetics , Choline/metabolism , Choline/pharmacology , Corpus Striatum/cytology , Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/deficiency , Dose-Response Relationship, Drug , Gene Expression Regulation/genetics , Hemicholinium 3/pharmacology , Mice , Mice, Knockout , Microdialysis/methods , Neurotransmitter Uptake Inhibitors/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism
2.
Biochem Soc Trans ; 29(Pt 6): 711-6, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11709061

ABSTRACT

In cholinergic neurons, a specific requirement for precursor choline in the biosynthesis of acetylcholine (ACh) is thought to be sustained by a presynaptic, hemicholinium-3 (HC-3)-sensitive choline transporter (CHT). This transporter exhibits micromolar affinity for choline and transport activity is Na(+)- and Cl(-)-dependent. Based on the sequence information available with the recent cloning of rat and human CHTs [Okuda, Haga, Kanai, Endou, Ishihara and Katsura (2000) Nat. Neurosci. 3, 120-125; Apparsundaram, Ferguson, George Jr and Blakely (2000) Biochem. Biophys. Res. Commun. 276, 862-867; Okuda and Haga (2000) FEBS Lett. 484, 92-97], we have identified a murine CHT orthologue (mCHT) by reverse transcriptase-PCR of spinal cord mRNA and confirmed this sequence using assembled mouse genomic DNA. Inferred splice junctions for mCHT exons are conserved with those of hCHT. The mCHT cDNA encodes a protein of 580 amino acids with 93% and 98% amino acid identity to human CHT and rat CHT1, respectively. Hydropathy analysis of the predicted amino acid sequence of mCHT indicates a protein containing 13 transmembrane domains (TMDs), with the N-terminus oriented extracellularly and the C-terminus oriented intracellularly. Northern blot analysis of mouse tissues reveals the expression of mCHT as a single transcript of approximately 5 kb with highest expression in regions that are rich in cholinergic cell bodies, e.g. the spinal cord, brainstem, mid-brain and striatum, whereas hybridization signals are absent in regions lacking cholinergic soma, e.g. the cerebellum and kidney. Expression of mCHT in COS-7 cells results in high-affinity [(3)H]HC-3-binding sites (K(d)=5 nM), and Na(+)- and Cl(-)-dependent HC-3-sensitive choline uptake (K(m)=2 microM), assessed in resealed membrane vesicles. The availability of cloned, functional mCHT and its cognate genomic DNA should prove useful for studies of mCHT regulation and should open possibilities for evaluation of CHT dysfunction in murine models.


Subject(s)
Hemicholinium 3/pharmacology , Membrane Transport Proteins/genetics , Animals , Blotting, Northern , CHO Cells , Cholinergic Agents/pharmacology , Cricetinae , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Kinetics , Mice , Protein Binding , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
J Pharmacol Exp Ther ; 299(2): 666-77, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602680

ABSTRACT

Presynaptic, cocaine- and antidepressant-sensitive norepinephrine (NE) transporters (NETs) dictate levels of extracellular NE after vesicular release. Recent studies suggest that G protein-coupled receptors linked to protein kinase C (PKC) down-regulate cell surface NET protein levels and diminish NE uptake capacity. We identified distinct phosphatidylinositol 3-OH kinase (PI3K)-linked pathways supporting basal and insulin-triggered NE transport in the human noradrenergic neuroblastoma, SK-N-SH. Acute (0-60 min) insulin treatments produced a time- and concentration-dependent stimulation of NE transport, resolved in kinetic studies as an enhancement of NE transport capacity (Vmax) without an alteration in NE Km. Basal and insulin-modulated NET activities were reduced by the tyrosine kinase inhibitor genistein and the PI3K inhibitors wortmannin and LY-294002, but not by the PKC inhibitor staurosporine. PI3K activation was found to support phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK). However, basal and insulin-stimulated NET activities were differentiated by their reliance on p38 MAPK activation. Thus, the p38 MAPK inhibitor SB203580 and SB202190 abolished insulin activation of NE transport yet failed to impact basal NET activity. Moreover, p38 MAPK activation and insulin activation of NETs were found to be sensitive to external Ca2+ depletion, blockade of voltage-sensitive Ca2+ channels, and inhibition of protein phosphatase 2A. Effects of tyrosine kinase and PI3K inhibitors on basal NET uptake appear to arise from a loss of cell surface NET protein, whereas the p38 MAPK-dependent enhancement of NE transport occurs without a detectable enhancement of surface NET. Our findings establish two distinct pathways for regulation of NE uptake involving PI3K, one linked to transporter trafficking and a second linked to Ca2+-dependent, p38 MAPK phosphorylation that promotes activation of cell surface NETs.


Subject(s)
Fluoxetine/analogs & derivatives , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Norepinephrine/metabolism , Symporters/metabolism , Blotting, Western , Calcium/metabolism , Cell Line , Fluoxetine/metabolism , Humans , Indicators and Reagents , Norepinephrine Plasma Membrane Transport Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Radioligand Assay , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases
4.
CNS Spectr ; 6(8): 671-4, 677-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-15520615

ABSTRACT

Presynaptic norepinephrine transporters (NETs) mediate the rapid clearance of norepinephrine from synaptic spaces. NET is a member of the Na+ and Cl- -coupled neurotransmitter transporter gene family, which also includes the serotonin and dopamine transporters. Recent studies reveal that these transporter molecules might be a dynamic component of synaptic plasticity, rather than a constitutive determinant of neurotransmitter levels in synaptic spaces. Recognition that cellular signaling molecules and transporter ligands, including cocaine, amphetamines, and antidepressants, can modify transporter intrinsic activity, trafficking, phosphorylation, and protein levels suggests opportunities for revealing unknown mechanisms of drug action. Control of these properties of transporter function may allow for the development of new strategies to modulate monoaminergic neurotransmission and identify regulatory pathways that may be compromised in psychiatric, neurologic, and neurodegenerative disorders.

5.
J Neurosci ; 20(20): 7571-8, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11027216

ABSTRACT

Presynaptic transporter proteins regulate the clearance of extracellular biogenic amines after release and are important targets for multiple psychoactive agents, including amphetamines, cocaine, and antidepressant drugs. Recent studies reveal that dopamine (DA), norepinephrine (NE), and serotonin (5-HT) transporters (DAT, NET, and SERT, respectively) are rapidly regulated by direct or receptor-mediated activation of cellular kinases, particularly protein kinase C (PKC). With SERTs, PKC activation results in activity-dependent transporter phosphorylation and sequestration. Protein phosphatase 1/2A (PP1/PP2A) inhibitors, such as okadaic acid (OA) and calyculin A, also promote SERT phosphorylation and functional downregulation. How kinase, phosphatase, and transporter activities are linked mechanistically is unclear. In the present study, we found that okadaic acid-sensitive phosphatase activity is enriched in SERT immunoprecipitates from human SERT stably transfected cells. Moreover, blots of these immunoprecipitates reveal the presence of PP2A catalytic subunit (PP2Ac), findings replicated using brain preparations. Whole-cell treatments with okadaic acid or calyculin A diminished SERT/PP2Ac associations. Phorbol esters, which trigger SERT phosphorylation, also diminish SERT/PP2Ac associations, effects that can be blocked by PKC antagonists as well as the SERT substrate 5-HT. Similar transporter/PP2Ac complexes were also observed in coimmunoprecipitation studies with NETs and DATs. Our findings provide evidence for the existence of regulated heteromeric assemblies involving biogenic amine transporters and PP2A and suggest that the dynamic stability of these complexes may govern transporter phosphorylation and sequestration.


Subject(s)
Antidepressive Agents/pharmacology , Biogenic Amines/metabolism , Cocaine/pharmacology , Membrane Transport Proteins , Nerve Tissue Proteins , Phosphoprotein Phosphatases/metabolism , Symporters , Animals , Biological Transport/drug effects , Carrier Proteins/metabolism , Cell Line , Dopamine Plasma Membrane Transport Proteins , Humans , Macromolecular Substances , Marine Toxins , Membrane Glycoproteins/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Phorbol Esters/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation/drug effects , Precipitin Tests , Protein Kinase C/antagonists & inhibitors , Protein Phosphatase 1 , Protein Phosphatase 2 , Protein Transport/drug effects , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Plasma Membrane Transport Proteins , Transfection
6.
Biochem Biophys Res Commun ; 276(3): 862-7, 2000 Oct 05.
Article in English | MEDLINE | ID: mdl-11027560

ABSTRACT

Under many physiological circumstances, Na(+)- and Cl(-)-dependent, hemicholinium-3 (HC-3)-sensitive, high-affinity choline uptake (HACU) in cholinergic neurons is thought to be rate-limiting in the biosynthesis of acetylcholine (ACh). Based on sequence information provided by the Human Genome Project and the recently reported rat CHT1 (rCHT1), we cloned a human CHT cDNA from spinal cord. The hCHT cDNA encodes a protein of 580 amino acids having 93% identity to rCHT1 and 51% identity to the Caenorhabditis elegans homolog CHO-1, and is distantly related to members of the Na(+)-coupled glucose transporter (SGLT) gene family of Na(+)-coupled glucose (SGLT), nucleoside and iodide transporters. Northern blot analysis reveals the expression of a approximately 5 kb transcript in human brain regions rich in cholinergic neurons including the putamen, spinal cord, and medulla. Expression of hCHT cDNA in COS-7 cells results in saturable, Na(+)/Cl(-)-dependent choline uptake (K(m) = 1.2 microM) in membrane vesicles and [(3)H] HC-3 binding (K(d) = 4 nM) in membrane fractions, consistent with characteristics reported in mammalian cholinergic neurons. Using radiation hybrid mapping techniques, we localized the hCHT gene to human chromosome 2q12. These studies elucidate the primary structure and chromosomal localization of hCHT and provide a basis for mechanistic analysis of HACU regulation and an investigation of the role of hCHT in disease states.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Hemicholinium 3/pharmacology , Membrane Transport Proteins , Amino Acid Sequence , Animals , Binding Sites , Biological Transport , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Choline/metabolism , Chromosomes, Human, Pair 2/genetics , Cloning, Molecular , Humans , Ligands , Molecular Sequence Data , Oocytes , Protein Binding , RNA, Messenger/analysis , RNA, Messenger/genetics , Radiation Hybrid Mapping , Sequence Homology, Amino Acid , Transfection , Xenopus laevis
7.
J Comp Neurol ; 420(2): 211-32, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10753308

ABSTRACT

Norepinephrine (NE) transporters (NETs) constitute the primary mechanism for inactivation of synaptically released NE, are targets for multiple antidepressants and psychostimulants, and have been reported to be deficient in affective and autonomic disorders. Although the regional distribution of NETs has been defined through synaptosomal transport and autoradiographic approaches, NET protein expression has yet to be characterized fully in the central nervous system (CNS). We identified a cytoplasmic NET epitope (amino acids 585-602) and corresponding antibody (43411) that permits cellular localization of endogenous NET expression in the CNS and periphery. In the adult rat brain, NET labeling was confined to noradrenergic neuronal somata, axons, and dendrites, including extensive arborizations within the hippocampus and cortex, but was absent from epinephrine- and dopamine-containing neurons. Intracerebroventricular anti-dopamine beta-hydroxylase/saporin, a treatment that destroys a majority of noradrenergic neurons and their projections, validated the specificity of the 43411 antibody. At the level of light microscopy, 43411 labeling colocalized with the axonal markers syntaxin, synaptophysin, and SNAP-25. Indirect immunofluorescence revealed a nonuniform pattern of NET expression along axons, particularly evident within sympathetic fibers of the vas deferens, reflecting a high degree of spatial organization of NE clearance. NET labeling in somata was intracellular and absent from plasma membranes. Among nonneuronal cells, glial cells lacked NET immunoreactivity, whereas CNS ependymal cells were an unexpected site of labeling. NET immunoreactivity was also evident in a subset of adrenal chromaffin cells where labeling appeared to be predominantly associated with intracellular vesicles. Initial ultrastructural evaluation via preembedding immunogold techniques also revealed substantial cytoplasmic NET immunoreactivity in axon terminals within the prelimbic prefrontal cortex, consistent with postulates of regulated trafficking controlling neurotransmitter clearance. NET visualization should be of significant benefit in evaluating neuronal injury resulting from chronic drug exposure and in disease states.


Subject(s)
Carrier Proteins/analysis , Symporters , Animals , Animals, Newborn , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Axons/metabolism , Axons/ultrastructure , Brain/cytology , Brain/drug effects , Brain/metabolism , Carrier Proteins/drug effects , Cell Culture Techniques , Cocaine/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/metabolism , Dopamine Uptake Inhibitors/pharmacology , Epitopes/chemistry , Epitopes/immunology , Immunohistochemistry , Male , Norepinephrine Plasma Membrane Transport Proteins , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
8.
J Pharmacol Exp Ther ; 287(2): 733-43, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808704

ABSTRACT

Using SK-N-SH cells, we observe that muscarinic acetylcholine receptor activation by methacholine (MCh) rapidly and selectively diminishes l-NE transport capacity (Vmax) with little or no change in norepinephrine (NE) Km and without apparent effects on membrane potential monitored directly under current clamp. Over the same time frame, MCh exposure reduces the density of [3H]nisoxetine binding sites (Bmax) in intact cells but not in total membrane fractions, consistent with a loss of transport capacity mediated by sequestration of transporters rather than changes in intrinsic transport activity or protein degradation. Similar changes in NE transport and [3H]nisoxetine binding capacity are observed after phorbol ester (beta-PMA) treatment. Inhibition of PKC by antagonists and downregulation of PKC by chronic treatment with phorbol esters abolishes beta-PMA-mediated effects but produce only a partial blockade of MCh-induced effects. Neither muscarinic acetylcholine receptor nor PKC activation require extracellular Ca++ to diminish NET activity. In contrast, treatment of cells with the Ca++/ATPase antagonist, thapsigargin in Ca++-free medium, eliminates the staurosporine-insensitive component of MCh regulation. These findings were further corroborated by the ability of [1, 2-bis(o-amino-phenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl)ester application in Ca++-free medium to abolish NET regulation by MCh. Although they may contribute to basal NET expression, we could not implicate CaMKII-, PKA- or nitric oxide-linked pathways in MCh regulation. Together, these findings 1) provide evidence in support of G-protein coupled receptor-mediated regulation of catecholamine transport, 2) reveal intracellular Ca++-sensitive, PKC-dependent and -independent pathways that serve to regulate NET expression and 3) indicate that the diminished capacity for NE transport evident after mAChR and PKC activation involves a redistribution of NET protein.


Subject(s)
Norepinephrine/metabolism , Protein Kinase C/metabolism , Receptors, Muscarinic/metabolism , Symporters , Biological Transport , Carrier Proteins/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Activation , Methacholine Chloride/pharmacology , Norepinephrine Plasma Membrane Transport Proteins , Thapsigargin/pharmacology
9.
J Pharmacol Exp Ther ; 287(2): 744-51, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808705

ABSTRACT

Norepinephrine (NE) transporters (NETs) found in the neuronal plasma membrane mediate the removal of NE from the extracellular space, limiting the activation of adrenoceptors at noradrenergic synapses. Our previous studies with the noradrenergic neuroblastoma SK-N-SH have revealed a muscarinic receptor-triggered regulation of NET surface density and transport capacity, mediated in part by protein kinase C activation. Low abundance of NET proteins in this native cell model, however, preclude direct confirmation of altered trafficking of NET proteins. In our study, we monitored the activity and surface distribution of human NET proteins in transient and stably-transfected cell lines after application of kinase activators and inhibitors. Using hNET stably transfected HEK-293 and LLC-PK1 cells, as well as transiently transfected COS-7 cells, we demonstrate that PKC-activating phorbol esters, beta-PMA or beta-PDBu selectively diminish l-NE transport capacity (Vmax) with little change in NE Km. Effects of phorbol esters are rapid, stereospecific and blocked by protein kinase C inhibitors, staurosporine and bisindolylmaleimide I. As in SK-N-SH cells, beta-PMA induces a reduction in intact cell [3H]nisoxetine binding sites with no change in nisoxetine Kd or total membrane NET density. Cell-surface biotinylation and confocal immunofluorescence techniques confirm that protein kinase C-dependent reductions in NE transport capacity and whole-cell antagonist binding density are accompanied by reductions in cell-surface human NET protein expression. Together these findings argue for kinase-modulated protein trafficking as a potential route for acute regulation of antidepressant-sensitive NE clearance.


Subject(s)
Carrier Proteins/metabolism , Norepinephrine/metabolism , Symporters , Binding Sites , Biological Transport , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Enzyme Activation , Fluoxetine/analogs & derivatives , Fluoxetine/metabolism , Humans , Norepinephrine Plasma Membrane Transport Proteins , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology
10.
Mol Pharmacol ; 54(4): 601-9, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9765501

ABSTRACT

A small subset of neurons in the nematode Caenorhabditis elegans utilizes the catecholamine dopamine (DA) as a neurotransmitter to control or modulate movement and egg-laying. Disruption of DA-mediated behaviors represents a potentially powerful strategy to identify genes that are likely to participate in dopaminergic systems in man. In vertebrates, extracellular DA is inactivated by presynaptic DA transport proteins (DATs) that are also major targets of addictive agents, including amphetamines and cocaine. We used oligonucleotides derived from the C. elegans genomic locus T23G5.5 to isolate and characterize T23G5.5 cDNAs. Our studies predict that mRNAs from this locus encode a 615-amino-acid polypeptide with twelve stretches of hydrophobicity suitable for transmembrane domains, similar to that found in vertebrate catecholamine transporters. The inferred translation product bears highest identity (43-47%) to catecholamine (DA, norepinephrine, epinephrine) transporters within the GAT1/NET gene family and possesses conserved residues implicated in amine substrate recognition. Consistent with these findings, HeLa cells transfected with the C. elegans cDNA exhibit saturable and high affinity DA transport (Km = 1.2 microM) that is dependent on extracellular Na+ and Cl- and blocked by inhibitors of mammalian catecholamine transporters, including norepinephrine transporter- and DAT-selective antagonists, tricyclic antidepressants, and the nonselective amine transporter antagonists cocaine and D-amphetamine. These studies validate the T23G5.5 locus as encoding a functional catecholamine transporter, providing important comparative sequence information for catecholamine transporter structure/function studies and a path to identify regulators of dopaminergic signaling via genetic or pharmacologic manipulation of C. elegans cDNA in vivo.


Subject(s)
Antidepressive Agents/pharmacology , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Carrier Proteins/drug effects , Carrier Proteins/genetics , Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Uptake Inhibitors/pharmacology , Membrane Glycoproteins , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Base Sequence , Carrier Proteins/antagonists & inhibitors , Cattle , DNA, Complementary/genetics , Exons , HeLa Cells , Humans , Kinetics , Mice , Molecular Sequence Data , Oligonucleotides/genetics , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity
11.
Biol Psychiatry ; 44(3): 169-78, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9693389

ABSTRACT

Presynaptic serotonin (5-hydroxytryptamine, 5-HT) transporters (SERTs) mediate antidepressant-sensitive clearance of 5-HT following release. Although we have been aware for decades that SERT-mediated 5-HT clearance can be modulated by exogenous agents including serotonin-selective reuptake inhibitors, amphetamines, and cocaine, we have had little reason to speculate that SERT activity was actively controlled through endogenous pathways. Recent studies indicate that SERTs are likely to be trafficked to specific plasma membrane subdomains to achieve localized clearance of 5-HT, and that the number of SERTs resident in the plasma membrane is controlled through kinase- and phosphatase-linked pathways. In particular, roles for protein kinase C and phosphatase 2A become apparent through studies with enzyme activators and inhibitors in SERT-transfected cells, where SERT proteins are rapidly phosphorylated in parallel with transporter redistribution and loss of functional uptake capacity. Based on our findings, and the studies of others in native tissues and transfected cells, we propose a model whereby SERTs are organized in a macromolecular complex in the plasma membrane that may serve to locate reuptake activity near release sites. Although many elements of this model remain hypothetical, our findings suggest a much more dynamic picture of transporter-mediated 5-HT reuptake than typically described and suggest opportunities both for the development of new SERT regulatory agents and for the identification of regulatory pathways that may be compromised in mental illness.


Subject(s)
Carrier Proteins/physiology , Membrane Glycoproteins/physiology , Membrane Transport Proteins , Nerve Tissue Proteins , Phosphoric Monoester Hydrolases/physiology , Protein Kinase C/physiology , Serotonin/blood , Animals , Antidepressive Agents/therapeutic use , Carrier Proteins/drug effects , Carrier Proteins/genetics , Cell Membrane/drug effects , Cell Membrane/physiology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Humans , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/genetics , Phosphorylation , Serotonin Plasma Membrane Transport Proteins
12.
J Neurochem ; 70(4): 1547-55, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9523572

ABSTRACT

The effects of continuous exposure of cultured cells expressing the human norepinephrine transporter (hNET) to the hNET inhibitor desipramine on hNET expression and function were studied. Exposure of HEK-293 cells transfected stably with the hNET cDNA (293-hNET cells) to desipramine for 3 days reduced the specific binding of [3H]nisoxetine in membrane homogenates in a concentration-dependent manner. The magnitude of the reductions in [3H]nisoxetine binding to hNET was dependent on the length of time of the exposure to desipramine, reaching 77% after a 21-day exposure. The reduction of [3H]nisoxetine binding returned to control levels within 72 h after a 3-day exposure to desipramine. Reductions in [3H]nisoxetine binding to hNET were accompanied by time-dependent and exposure concentration-dependent reductions in hNET protein levels as determined by western blotting. Similar to binding, hNET protein levels returned to control levels 72 h after cessation of desipramine exposure. Northern blotting indicated that exposure of 293-hNET cells to desipramine did not significantly alter hNET mRNA levels. Uptake of [3H]norepinephrine by 293-hNET cells was markedly reduced after a 3-day exposure to desipramine. However, desipramine exposure had no effect on uptake of [3H]glutamate or [3H]alanine. The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Carrier Proteins/antagonists & inhibitors , Desipramine/pharmacology , Symporters , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fluoxetine/analogs & derivatives , Fluoxetine/metabolism , Humans , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Norepinephrine/pharmacokinetics , Norepinephrine Plasma Membrane Transport Proteins , RNA, Messenger/metabolism
14.
J Neurosci ; 17(8): 2691-702, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9092590

ABSTRACT

Chemical signaling by dopamine (DA) and L-norepinephrine (L-NE) at synapses is terminated by uptake via specialized presynaptic transport proteins encoded by the DA transporter (DAT) and L-NE transporter (NET) genes, respectively. In some vertebrate neurons, particularly the sympathetic neurons of amphibians, L-NE is converted to L-epinephrine (L-Epi, adrenaline) and released as the primary neurotransmitter. Although evidence exists for a molecularly distinct L-Epi transporter (ET) in the vertebrate brain and peripheral nervous system, a transporter specialized for extracellular L-Epi clearance has yet to be identified. To pursue this issue, we cloned transporter cDNAs from bullfrog (Rana catesbiana) paravertebral sympathetic ganglia and characterized functional properties via heterologous expression in non-neuronal cells. A cDNA of 2514 bp (fET) was identified for which the cognate 3.1 kb mRNA is highly enriched in frog sympathetic ganglia. Sequence analysis of the fET cDNA reveals an open reading frame coding for a protein of 630 amino acids. Inferred fET protein sequence bears 75, 66, and 48% amino acid identity with human NET, DAT, and the 5-hydroxytryptamine transporter (SERT), respectively. Transfection of fET confers Na+- and Cl--dependent catecholamine uptake in HeLa cells. Uptake of [3H]-L-NE by fET is inhibited by catecholamines in a stereospecific manner. L-Epi and DA inhibit fET-mediated [3H]-L-NE uptake more potently than they inhibit [3H]-L-NE uptake by human NET (hNET), whereas L-NE exhibits equivalent potency between the two carriers. Moreover, fET exhibits a greater maximal velocity (Vmax) for the terminal products of catecholamine biosynthesis (L-Epi > L-NE >> DA), unlike hNET, in which a Vmax rank order of L-NE > DA > L-Epi is observed. fET-mediated transport of catecholamines is sensitive to cocaine and tricyclic antidepressants, with antagonist potencies significantly correlated with hNET inhibitor sensitivity. Amino acid conservation and divergence of fET with mammalian catecholamine transporters help define residues likely to be involved in catecholamine recognition and translocation as well as blockade by selective reuptake inhibitors.


Subject(s)
Amphibian Proteins , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Ganglia, Sympathetic/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Symporters , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/biosynthesis , Cattle , Cloning, Molecular , Conserved Sequence , DNA Primers , Dopamine Plasma Membrane Transport Proteins , Epinephrine/metabolism , Epinephrine/pharmacology , HeLa Cells , Humans , Kinetics , Membrane Glycoproteins/chemistry , Mice , Molecular Sequence Data , Norepinephrine Plasma Membrane Transport Proteins , Organ Specificity , Polymerase Chain Reaction , Rana catesbeiana , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Serotonin Plasma Membrane Transport Proteins , Transfection
15.
J Pharmacol Exp Ther ; 278(2): 862-70, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8768741

ABSTRACT

This study examines the effects of acute incorporation of epinephrine (EPI) into cardiac catecholamine stores and chronic elevation of plasma EPI concentrations on sympathetic neurotransmission and adrenoceptor modulation in the rat heart. Chronic elevation of plasma EPI was accomplished by infusion of EPI (100 micrograms/kg/hr s.c.) for 6 days via osmotic minipumps. Vehicle-treated animals served as controls. The cardiac catecholamine stores of the vehicle-treated group consisted of norepinephrine only. Chronic EPI treatment resulted in incorporation of EPI (90% of total catecholamines) without a significant change in the total cardiac catecholamine content. At 0.1 to 1 Hz, stimulus-induced catecholamine overflows in the EPI-treated group consisted of both norepinephrine and EPI and were 66 to 70% higher than those of the vehicle-treated group. A preferential beta-2 adrenoceptor antagonist, ICI 118,551 (1 nM-1 microM) did not alter catecholamine overflows in either group. In contrast, a preferential alpha-2 adrenoceptor antagonist, idazoxan (1 microM), significantly increased catecholamine overflow in the vehicle-treated group but not in the EPI-treated group. After idazoxan treatment, the significant difference between overflows in the chronic vehicle- and EPI-treated groups was abolished. Acute EPI treatment in vitro (1 microM, 1 hr in the presence of phentolamine 10 microM; nadolol, 30 microM) resulted in incorporation of EPI (75% of total catecholamine) into cardiac catecholamine stores and increased the stimulus-induced catecholamine overflow at 0.1 Hz. Blockade of prejunctional beta-2 adrenoceptors abolished the difference between the acute EPI- and vehicle-treated groups. Moreover, the increase in catecholamine overflow resulting from alpha-2 adrenoceptor blockade with idazoxan (1 microM) was comparable between the acute EPI- and vehicle-treated groups. These results suggest that 1) both chronic elevation of plasma EPI and acute incorporation of EPI into cardiac neurotransmitter stores result in facilitation of neurotransmitter release and 2) prejunctional beta adrenoceptor activation appears to be the cause of the increase after acute EPI incorporation, whereas desensitization of prejunctional alpha-2 adrenoceptors, not beta-2 adrenoceptor-mediated facilitation, is the cause of the enhanced overflow after chronic EPI treatment.


Subject(s)
Catecholamines/metabolism , Epinephrine/pharmacology , Heart/drug effects , Receptors, Adrenergic, alpha/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley
16.
Nature ; 374(6519): 272-6, 1995 Mar 16.
Article in English | MEDLINE | ID: mdl-7885448

ABSTRACT

G-protein-coupled receptors are thought to have an inactive conformation (R), requiring an agonist-induced conformational change for receptor/G-protein coupling. But new evidence suggests a two-state model in which receptors are in equilibrium between the inactive conformation (R), and a spontaneously active conformation (R*) that can couple to G protein in the absence of ligand (Fig. 1). Classic agonists have a high affinity for R* and increase the concentration of R*, whereas inverse agonists have a high affinity for R and decrease the concentration of R*. Neutral competitive antagonists have equal affinity for R and R* and do not displace the equilibrium, but can competitively antagonize the effects both of agonists and of inverse agonists. The lack of suitable in vivo model systems has restricted the evidence for the existence of inverse agonists to computer simulations and in vitro systems. We have used a transgenic mouse model in which there is such marked myocardial overexpression of beta 2-adrenoceptors that a significant population of spontaneously activated receptor (R*) is present, inducing a maximal response without agonist. We show that the beta 2-adrenoceptor ligand ICI-118,551 functions as an inverse agonist, providing evidence supporting the existence of inverse agonists and validating the two-state model of G-protein-coupled receptor activation.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Myocardium/metabolism , Propanolamines/pharmacology , Receptors, Adrenergic, beta/biosynthesis , Alprenolol/pharmacology , Animals , Cyclohexane Monoterpenes , GTP-Binding Proteins/metabolism , Heart/drug effects , Mice , Mice, Transgenic , Pindolol/analogs & derivatives , Pindolol/pharmacology
17.
J Pharmacol Exp Ther ; 272(2): 519-26, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7853165

ABSTRACT

This study examines the role of prejunctional beta adrenoceptors in the modulation of sympathetic neurotransmission in the rat heart. Under anesthesia, the heart with right cardiac sympathetic nerves attached was isolated and perfused with Krebs' bicarbonate buffer containing cocaine (10 microM), corticosterone (40 microM) and atropine (6 microM). Stimulation of sympathetic nerves at frequencies of 0.05 to 3 Hz (15 pulses, supramaximal voltage, 2-msec pulse duration) produced a pronounced frequency-dependent increase in heart rate. A maximal increase of 89 +/- 10 bpm was obtained at 3 Hz. Propranolol (0.1 microM) had no significant effect on the stimulus-induced NE overflow in the absence and presence of alpha adrenoceptor blockade alone or combined with adenosine receptor blockade. The beta-2 adrenoceptor agonist salbutamol increased stimulus-induced NE overflow (15 pulses) at 0.1 Hz but not at 0.5 Hz. Alpha adrenoceptor blockade did not alter the salbutamol-evoked facilitation at 0.1 Hz but revealed salbutamol-evoked facilitation at 0.5 Hz. Salbutamol did not alter NE overflow at 1 Hz even in the presence of alpha adrenoceptor blockade. In conclusion, prejunctional beta adrenoceptor facilitation of NE overflow is frequency and alpha adrenoceptor modulated. At low frequency (0.1 Hz), beta adrenoceptor-mediated facilitation is maximal and alpha adrenoceptor independent; at intermediate frequency (0.5 Hz) it is alpha adrenoceptor restrained and it is absent at 1 Hz. Finally, NE does not modulate its own release via the prejunctional beta adrenoceptors.


Subject(s)
Heart/innervation , Norepinephrine/metabolism , Receptors, Adrenergic, beta/physiology , Synaptic Transmission , Albuterol/pharmacology , Animals , Electric Stimulation , Male , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha/physiology
18.
J Pharmacol Exp Ther ; 256(2): 486-91, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1671595

ABSTRACT

There are reports of an increase in renin release after the administration of fenoldopam which probably results from the activation of dopamine (DA)-1 receptors located on juxtaglomerular cells in the kidney. However, the functional significance of this finding in terms of modulating the tubular response to DA-1 receptor stimulation remains to be determined. In this study we have examined the effect of an increase in renin-angiotensin system activity during the administration of fenoldopam on the natriuretic and diuretic action of this compound. Intravenous infusion of fenoldopam (0.5 microgram/kg/min) for 30 min produced significant increases in urine output and urinary sodium excretion without causing any changes in glomerular filtration rate, renal blood flow and mean arterial blood pressure, a phenomenon suggestive of a direct tubular site of action. In animals treated with the angiotensin converting enzyme inhibitor captopril, both the diuretic and natriuretic effects of fenoldopam were potentiated markedly. In comparison with fenoldopam infusion in control animals, urine output, urinary sodium excretion and potassium excretion increased by approximately 4-fold (375 +/- 24 vs. 97 +/- 3 microliters/30 min, P less than .01), 9-fold (50 +/- 5 vs. 6 +/- 1 microEq/30 min, P less than .001) and 2-fold (46 +/- 8 vs. 24 +/- 2 microEq/30 min, P less than .05), respectively, in animals receiving a bolus injection of captopril (1 mg/kg i.v.) 30 min before onset of fenoldopam infusion. Whereas no significant changes in renal blood flow occurred when fenoldopam was given to control rats, in animals treated with captopril, fenoldopam produced a modest but significant increase in renal blood flow.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives , Angiotensin II/physiology , Dopamine Agents/pharmacology , Kidney/drug effects , Receptors, Dopamine/drug effects , Sodium/urine , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Benzazepines/pharmacology , Captopril/pharmacology , Fenoldopam , Male , Rats , Rats, Inbred Strains , Receptors, Dopamine D1 , Renin-Angiotensin System/drug effects
19.
Naunyn Schmiedebergs Arch Pharmacol ; 343(1): 21-30, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1674360

ABSTRACT

Dopexamine hydrochloride (DPX) is a dopamine analog and it possesses agonistic action at DA-1 receptors and beta 2-adrenoceptors. It also is a weak agonist at DA-2 receptors. In the present study, we have examined the anatomical localization of DPX binding sites in rat kidney and their functional significance in terms of the renal effects of this compound. In receptor-ligand binding studies, [3H]-DPX was found to bind specifically to sections of rat kidney in a time (maximum binding at 60 min), temperature (optimal temperature 25 degrees C) and concentration (highest specific/non-specific ratio at 2 nmol/l) dependent manner. Autoradiographic studies revealed the presence of [3H]-DPX binding sites in renal tubules, glomerulus and various layers of small and large blood vessels. Inhibition studies with SCH 23390, ICI 118.551 and 1-sulpiride showed that DPX binds primarily to DA-1 receptors in tubules, only to beta 2-adrenoceptors in glomerulus and to beta 2-adrenoceptors, DA-1 and DA-2 receptors in blood vessels. Also, DPX caused concentration related increases in cyclic AMP levels in rat kidney membrane particles, which could be completely abolished by a combined presence of SCH 23390 and propranolol suggesting that both binding sites of DPX are linked to adenylate cyclase. In functional studies DPX (1 microgram/kg.min for 30 min) produced a modest fall in blood pressure, pronounced tachycardia and slight but significant increase in renal blood flow (11%). These responses were accompanied by increases in urine output (97%), urinary sodium excretion (89%), and fractional excretion of sodium (132%). There was no change in glomerular filtration rate. Propranolol pretreatment abolished DPX-induced hypotension and tachycardia but seemed to potentiate the natriuretic responses to DPX. On the other hand, SCH 23390, a DA-1 receptor antagonist completely abolished DPX-induced hypotension, natriuresis and diuresis without affecting tachycardia. These results indicate that (1) DPX binds predominantly to DA-1 receptors in renal tubules, to beta 2-adrenoceptors in glomerulus and to beta 2-adrenoceptors, as well as DA-1 and DA-2 receptors in renal blood vessels (2) DPX stimulates cAMP formation in the kidney by activating both DA-1 and beta 2-adrenoceptors and (3) DPX produces natriuresis and diuresis by selectively activating DA-1 receptors located on renal tubules.


Subject(s)
Adrenergic Agonists/pharmacology , Dopamine/analogs & derivatives , Kidney Tubules/ultrastructure , Receptors, Dopamine/physiology , Adenylyl Cyclases/metabolism , Adrenergic Agonists/metabolism , Animals , Autoradiography , Binding Sites , Cyclic AMP/metabolism , Dopamine/metabolism , Dopamine/pharmacology , Kidney/enzymology , Kidney/metabolism , Kidney/physiology , Kidney/ultrastructure , Kidney Tubules/metabolism , Kidney Tubules/physiology , Male , Natriuresis/drug effects , Rats , Rats, Inbred Strains , Receptors, Dopamine/metabolism , Renal Circulation , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...