Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Nature ; 516(7529): 68-70, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25471881

ABSTRACT

Recent observations have revealed that starburst galaxies can drive molecular gas outflows through stellar radiation pressure. Molecular gas is the phase of the interstellar medium from which stars form, so these outflows curtail stellar mass growth in galaxies. Previously known outflows, however, involve small fractions of the total molecular gas content and have typical scales of less than a kiloparsec. In at least some cases, input from active galactic nuclei is dynamically important, so pure stellar feedback (the momentum return into the interstellar medium) has been considered incapable of rapidly terminating star formation on galactic scales. Molecular gas has been detected outside the galactic plane of the archetypal starburst galaxy M82 (refs 4 and 5), but so far there has been no evidence that starbursts can propel substantial quantities of cold molecular gas to the same galactocentric radius (about 10 kiloparsecs) as the warmer gas that has been traced by metal ion absorbers in the circumgalactic medium. Here we report observations of molecular gas in a compact (effective radius 100 parsecs) massive starburst galaxy at redshift 0.7, which is known to drive a fast outflow of ionized gas. We find that 35 per cent of the total molecular gas extends approximately 10 kiloparsecs, and one-third of this extended gas has a velocity of up to 1,000 kilometres per second. The kinetic energy associated with this high-velocity component is consistent with the momentum flux available from stellar radiation pressure. This demonstrates that nuclear bursts of star formation are capable of ejecting large amounts of cold gas from the central regions of galaxies, thereby strongly affecting their evolution by truncating star formation and redistributing matter.

2.
Transl Psychiatry ; 3: e259, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23695234

ABSTRACT

The SLC1A1 gene, which encodes the neuronal glutamate transporter, EAAC1, has consistently been implicated in obsessive-compulsive disorder (OCD) in genetic studies. Moreover, neuroimaging, biochemical and clinical studies support a role for glutamatergic dysfunction in OCD. Although SLC1A1 is an excellent candidate gene for OCD, little is known about its regulation at the genomic level. Here, we report the identification and characterization of three alternative SLC1A1/EAAC1 mRNAs: a transcript derived from an internal promoter, termed P2 to distinguish it from the transcript generated by the primary promoter (P1), and two alternatively spliced mRNAs: ex2skip, which is missing exon 2, and ex11skip, which is missing exon 11. All isoforms inhibit glutamate uptake from the full-length EAAC1 transporter. Ex2skip and ex11skip also display partial colocalization and interact with the full-length EAAC1 protein. The three isoforms are evolutionarily conserved between human and mouse, and are expressed in brain, kidney and lymphocytes under nonpathological conditions, suggesting that the isoforms are physiological regulators of EAAC1. Moreover, under specific conditions, all SLC1A1 transcripts were differentially expressed in lymphocytes derived from subjects with OCD compared with controls. These initial results reveal the complexity of SLC1A1 regulation and the potential clinical utility of profiling glutamatergic gene expression in OCD and other psychiatric disorders.


Subject(s)
Excitatory Amino Acid Transporter 3/genetics , Glutamic Acid/metabolism , Obsessive-Compulsive Disorder/genetics , Adolescent , Adult , Aged , Animals , Excitatory Amino Acid Transporter 3/physiology , Female , Glutamic Acid/physiology , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Promoter Regions, Genetic/genetics , Protein Isoforms , Young Adult
3.
Am J Physiol Cell Physiol ; 293(4): C1286-95, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17626242

ABSTRACT

KAAT1 is a lepidopteran neutral amino acid transporter belonging to the NSS super family (SLC6), which has an unusual cation selectivity, being activated by K(+) and Li(+) in addition to Na(+). We have previously demonstrated that Asp338 is essential for KAAT1 activation by K(+) and for the coupling of amino acid and driver ion fluxes. By comparing sequences of NSS family members, site-directed mutagenesis, and expression in Xenopus laevis oocytes, we identified Lys102 as a residue likely to interact with Asp338. Compared with wild type, the single mutants K102V and D338E each showed altered leucine uptake and transport-associated currents in the presence of both Na(+) and K(+). However, in K102V/D338E double mutant, the K102V mutation reversed both the inhibition of Na(+)-dependent transport and the block in K(+)-dependent transport that characterize the D338E mutant. K(+)-dependent leucine currents were not observed in any mutants with D338E. In the presence of the oxidant Cu(II) (1,10-phenanthroline)(3), we observed specific and reversible inhibition of K102C/D338C mutant, but not of the corresponding single cysteine mutants, suggesting that these residues are sufficiently close to form a disulfide bond. Thus both structural and functional evidence suggests that these two residues interact. Similar results have been obtained mutating the bacterial transporter homolog TnaT. Asp338 corresponds to Asn286, a residue located in the Na(+) binding site in the recently solved crystal structure of the NSS transporter LeuT(Aa) (41). Our results suggest that Lys102, interacting with Asp338, could contribute to the spatial organization of KAAT1 cation binding site and permeation pathway.


Subject(s)
Amino Acid Transport Systems, Neutral/metabolism , Aspartic Acid/metabolism , Insect Proteins/metabolism , Lysine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/genetics , Animals , Aspartic Acid/chemistry , Aspartic Acid/genetics , Binding Sites/genetics , Biological Transport/drug effects , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Dithiothreitol/chemistry , Dithiothreitol/pharmacology , Female , Insect Proteins/chemistry , Insect Proteins/genetics , Kinetics , Lepidoptera , Lysine/chemistry , Lysine/genetics , Models, Molecular , Molecular Sequence Data , Oocytes/metabolism , Phenanthrolines/chemistry , Phenanthrolines/pharmacology , Potassium/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sodium/metabolism , Tryptophan/chemistry , Tryptophan/genetics , Tryptophan/metabolism , Xenopus laevis
4.
Handb Exp Pharmacol ; (175): 59-73, 2006.
Article in English | MEDLINE | ID: mdl-16722230

ABSTRACT

Serotonin transporter (SERT) serves the important function of taking up serotonin (5-HT) released during serotonergic neurotransmission. It is the target for important therapeutic drugs and psychostimulants. SERT catalyzes the influx of 5-HT together with Na+ and Cl- in a 1:1:1 stoichiometry. In the same catalytic cycle, there is coupled efflux of one K+ ion. SERT is one member of a large family of amino acid and amine transporters that is believed to utilize similar mechanisms of transport. A bacterial member of this family was recently crystallized, revealing the structural basis of these transporters. In light of the new structure, previous results with SERT have been re-interpreted, providing new insight into the substrate binding site, the permeation pathway, and the conformational changes that occur during the transport cycle.


Subject(s)
Bacterial Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Binding Sites , Biological Transport , Humans , Molecular Sequence Data , Protein Binding , Protein Conformation , Serotonin Plasma Membrane Transport Proteins/chemistry , Structure-Activity Relationship
5.
Mol Psychiatry ; 8(11): 933-6, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14593431

ABSTRACT

Two common serotonin transporter (SERT) untranslated region gene variants have been intensively studied, but remain inconclusively linked to depression and other neuropsychiatric disorders. We now report an uncommon coding region SERT mutation, Ile425Val, in two unrelated families with OCD and other serotonin-related disorders. Six of the seven family members with this mutation had OCD (n=5) or obsessive-compulsive personality disorder (n=1) and some also met diagnostic criteria for multiple other disorders (Asperger's syndrome, social phobia, anorexia nervosa, tic disorder and alcohol and other substance abuse/dependence). The four most clinically affected individuals--the two probands and their two slbs--had the I425V SERT gene gain-of-function mutation and were also homozygous for 5'-UTR SERT gene variant with greater transcriptional efficacy.


Subject(s)
Anorexia Nervosa/genetics , Autistic Disorder/genetics , Carrier Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Transport Proteins , Mutation, Missense , Nerve Tissue Proteins/genetics , Obsessive-Compulsive Disorder/genetics , Amino Acid Sequence , Asperger Syndrome/genetics , Carrier Proteins/chemistry , Female , Genotype , Humans , Male , Membrane Glycoproteins/chemistry , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Pedigree , Phenotype , Phobic Disorders/genetics , Polymorphism, Single-Stranded Conformational , Protein Structure, Tertiary , Serotonin Plasma Membrane Transport Proteins
6.
J Neurochem ; 85(6): 1513-20, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12787070

ABSTRACT

Before this study, the human norepinephrine transporter (hNET) was the only member of the biogenic amine neurotransmitter transporter family that had not been demonstrated to be a functional homo-oligomer. Here, using two forms of the transporter, I155C and hNET-myc, with distinct antigenicity and inhibitor sensitivity, we demonstrated that hNET exists as a homo-oligomer. hNET I155C is a functional mutant and is sensitive to inactivation by the sulfhydryl reagent [2-(trimethylammonium)ethyl]methanethiosulfonate, while hNET-myc is resistant to inactivation by this reagent. Coimmunoprecipitation of these two forms demonstrated that a physical interaction exists between norepinephrine transporter monomers. Further characterization of this physical interaction has revealed that the activity of norepinephrine transporters depends on interactions between monomers. Because norepinephrine transporters and serotonin transporters are the only two members of the neurotransmitter transporter family endogenously expressed in the cell membrane of the same cells, placental syncytiotrophoblasts, we tested the ability of norepinephrine transporters and serotonin transporters to associate and function in a hetero-oligomeric form. Similarly, coexpression of hNET-myc with serotonin transporter-FLAG showed a physical interaction in coimmunoprecipitation assays. However, coexpression of serotonin and norepinephrine transporters did not sensitize norepinephrine transporter activity to inhibition by citalopram, a selective serotonin transport inhibitor. Thus, the norepinephrine transporter-serotonin transporter physical association did not produce functional consequences. Based on this, we propose that the transporters for biogenic amine neurotransmitters interact functionally in homo- but not hetero-oligomeric forms.


Subject(s)
Biogenic Monoamines/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Symporters/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Gene Expression/physiology , HeLa Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesylates/chemistry , Molecular Sequence Data , Mutation , Norepinephrine Plasma Membrane Transport Proteins , Precipitin Tests , Protein Binding/physiology , Proto-Oncogene Proteins c-myc/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins , Sulfhydryl Reagents/chemistry , Symporters/chemistry , Symporters/genetics , Transfection
7.
Mol Biol Cell ; 12(12): 3797-807, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739781

ABSTRACT

When expressed in epithelial cells, dopamine transporter (DAT) was detected predominantly in the apical plasma membrane, whereas norepinephrine transporter (NET) was found in the basolateral membrane, despite 67% overall amino acid sequence identity. To identify possible localization signals responsible for this difference, DAT-NET chimeras were expressed in MDCK cells and localized by immunocytochemistry and transport assays. The results suggested that localization of these transporters in MDCK cells depends on their highly divergent NH(2)-terminal regions. Deletion of the first 58 amino acids of DAT (preceding TM1) did not change its apical localization. However, the replacement of that region with corresponding sequence from NET resulted in localization of the chimeric protein to the basolateral membrane, suggesting that the NH(2)-terminus of NET, which contains two dileucine motifs, contains a basolateral localization signal. Mutation of these leucines to alanines in the context of a basolaterally localized NET/DAT chimera restored transporter localization to the apical membrane, indicating that the dileucine motifs are critical to the basolateral localization signal embodied within the NET NH(2)-terminal region. However, the same mutation in the context of wild-type NET did not disrupt basolateral localization, indicating the presence of additional signals in NET directing its basolateral localization within the plasma membrane.


Subject(s)
Cell Polarity , Epithelial Cells/cytology , Epithelial Cells/metabolism , Membrane Glycoproteins , Nerve Tissue Proteins , Protein Sorting Signals/physiology , Symporters/chemistry , Symporters/metabolism , Amino Acid Sequence , Animals , Cell Line , Dogs , Dopamine Plasma Membrane Transport Proteins , Humans , Membrane Transport Proteins/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , Norepinephrine Plasma Membrane Transport Proteins , Sequence Alignment , Symporters/genetics
8.
J Biol Chem ; 276(49): 45933-8, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11592963

ABSTRACT

Serotonin transporter (SERT) contains a single reactive external cysteine residue at position 109 (Chen, J. G., Liu-Chen, S., and Rudnick, G. (1997) Biochemistry 36, 1479-1486) and seven predicted cytoplasmic cysteines. A mutant of rat SERT (X8C) in which those eight cysteine residues were replaced by other amino acids retained approximately 32% of wild type transport activity and approximately 56% of wild type binding activity. In contrast to wild-type SERT or the C109A mutant, X8C was resistant to inhibition of high affinity cocaine analog binding by the cysteine reagent 2-(aminoethyl)methanethiosulfonate hydrobromide (MTSEA) in membrane preparations from transfected cells. Each predicted cytoplasmic cysteine residue was reintroduced, one at a time, into the X8C template. Reintroduction of Cys-357, located in the third intracellular loop, restored MTSEA sensitivity similar to that of C109A. Replacement of only Cys-109 and Cys-357 was sufficient to prevent MTSEA sensitivity. Thus, Cys-357 was the sole cytoplasmic determinant of MTSEA sensitivity in SERT. Both serotonin and cocaine protected SERT from inactivation by MTSEA at Cys-357. This protection was apparently mediated through a conformational change following ligand binding. Although both ligands bind in the absence of Na(+) and at 4 degrees C, their ability to protect Cys-357 required Na(+) and was prevented at 4 degrees C. The accessibility of Cys-357 to MTSEA inactivation was increased by monovalent cations. The K(+) ion, which is believed to serve as a countertransport substrate for SERT, was the most effective ion for increasing Cys-357 reactivity.


Subject(s)
Carrier Proteins/metabolism , Cysteine/metabolism , Cytoplasm/metabolism , Ethyl Methanesulfonate/analogs & derivatives , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Ethyl Methanesulfonate/metabolism , Ligands , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Rats , Serotonin Plasma Membrane Transport Proteins
9.
J Biol Chem ; 276(33): 30942-7, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11408487

ABSTRACT

Inactivation of serotonin transporter (SERT) expressed in HeLa cells by [2-(trimethylammonium)ethyl]methanethiosulfonate (MTSET) occurred much more readily when Na(+) in the reaction medium was replaced with Li(+). This did not result from a protective effect of Na(+) but rather from a Li(+)-specific increase in the reactivity of Cys-109 in the first external loop of the transporter. Li(+) alone of the alkali cations caused this increase in reactivity. Replacing Na(+) with N-methyl-d-glucamine (NMDG(+)) did not reduce the affinity of cocaine for SERT, as measured by displacement of a high affinity cocaine analog, but replacement of Na(+) with Li(+) led to a 2-fold increase in the K(D) for cocaine. The addition of either cocaine or serotonin (5-HT) protected SERT against MTSET inactivation. When SERT was expressed in Xenopus oocytes, inward currents were elicited by superfusing the cell with 5-HT (in the presence of Na(+)) or by replacing Na(+) with Li(+) but not NMDG(+). MTSET treatment of oocytes in Li(+) but not in Na(+) decreased both 5-HT and Li(+) induced currents, although 5-HT-induced currents were inhibited to a greater extent. Na(+) antagonized the effects of Li(+) on both inactivation and current. These results are consistent with Li(+) inducing a conformational change that exposes Cys-109, decreases cocaine affinity, and increases the uncoupled inward current.


Subject(s)
Carrier Proteins/chemistry , Cocaine/metabolism , Lithium/pharmacology , Membrane Glycoproteins/chemistry , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Cysteine , Glutamates/pharmacology , HeLa Cells , Humans , Membrane Potentials/drug effects , Mesylates/pharmacology , Protein Conformation , Serotonin Plasma Membrane Transport Proteins , Sodium/pharmacology , Xenopus
10.
J Biol Chem ; 276(6): 4038-45, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11058600

ABSTRACT

Mutations at critical residue positions in transmembrane span 7 (TM7) of the serotonin transporter affect the Na(+) dependence of transport. It was possible that these residues, which form a stripe along one side of the predicted alpha-helix, formed part of a water-filled pore for Na(+). We tested whether cysteine substitutions in TM7 were accessible to hydrophilic, membrane-impermeant methanethiosulfonate (MTS) reagents. Although all five cysteine-containing mutants tested were sensitive to these reagents, noncysteine control mutants at the same positions were in most cases equally sensitive. In all cases, MTS sensitivity could be traced to changes in accessibility of a native cysteine residue in extracellular loop 1, Cys-109. Moreover, none of the TM7 cysteines reacted with the biotinylating reagent MTSEA-biotin when tested in the C109A background. It is thus unlikely that the critical stripe forms part of a water-filled pore. Instead, studies of the ion dependence of the reaction between Cys-109 and MTS reagents lead to the conclusion that TM7 is involved in propagating conformational changes caused by ion binding, perhaps as part of the translocation mechanism. The critical stripe residues on TM7 probably represent a close contact region between TM7 and one or more other TMs in the transporter's three-dimensional structure.


Subject(s)
Carrier Proteins/metabolism , Lithium/chemistry , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Mesylates/chemistry , Nerve Tissue Proteins , Sodium/chemistry , Carrier Proteins/chemistry , Cell Membrane/metabolism , Membrane Glycoproteins/chemistry , Serotonin Plasma Membrane Transport Proteins
11.
Astrophys J ; 534(1): L27-L30, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10790063

ABSTRACT

We present spectroscopic observations from the Hubble Space Telescope that reveal for the first time the presence of a broad pedestal of Balmer line emission in the LINER galaxy NGC 4203. The emission-line profile is suggestive of a relativistic accretion disk and is reminiscent of double-peaked transient Balmer emission observed in a handful of other LINERs. The very broad line emission thus constitutes clear qualitative evidence for a black hole, and spatially resolved narrow-line emission in NGC 4203 can be used to constrain its mass, MBH

12.
Proc Natl Acad Sci U S A ; 97(7): 3106-11, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10716733

ABSTRACT

Two forms of serotonin transporter (SERT) were prepared with different epitope tags. When co-expressed in HeLa cells, the form containing a FLAG tag (Res-FLAG) was associated with the form containing a c-myc tag (Sens-myc). Antibody against c-myc precipitated Res-FLAG from detergent extracts of cells expressing both forms, but not when Res-FLAG was expressed alone. The specificity of the interaction was demonstrated by the observation that anti-myc antibodies did not precipitate the unrelated vesicular stomatitis virus coat glycoprotein when it was co-expressed with Sens-myc. Sens-myc contained a reactive cysteine at position 172, which reacted with both (2-aminoethyl)methanethiosulfonate and N-biotinylaminoethyl methanethiosulfonate on the surface of intact cells. Sens-myc, but not Res-FLAG, was inactivated by these reagents. When co-expressed with Sens-myc, functionally active Res-FLAG was precipitated by immobilized streptavidin from digitonin-solubilized cells that had been treated with N-biotinylaminoethyl methanethiosulfonate. In cells co-expressing mixtures of Sens-myc and Res-FLAG, the amount of inactivation by (2-aminoethyl)methanethiosulfonate was less than expected if the two forms were independent. The results are consistent with a dimeric form of SERT with functional interactions between subunits, and with association of dimers into a higher order complex, possibly a tetramer.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Biopolymers , Carrier Proteins/genetics , Genes, myc , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Precipitin Tests , Serotonin Plasma Membrane Transport Proteins
13.
Proc Natl Acad Sci U S A ; 97(3): 1044-9, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10655481

ABSTRACT

The third transmembrane domain (TM3) of serotonin transporter (SERT) contains two isoleucine residues previously proposed to be involved in binding and transport of serotonin. When Ile-172 was replaced with cysteine, SERT became sensitive to inactivation by externally added [2-(trimethylammonium)ethyl]methanethio-sulfonate (MTSET). The disulfide product of this inactivation was not sensitive to reduction by externally added sulfhydryl compounds, but apparently reacted with intracellular reducing agents to spontaneously regenerate active SERT. The apparent accessibility of this residue to both external and cytoplasmic reagents is consistent with its localization near a serotonin binding site that is alternately exposed to both internal and external media. In another SERT mutant, I179C, transport also was inactivated by MTSET but substrate binding was resistant. External substrate bound to the inactivated I179C and enhanced its reactivation by free thiols. In norepinephrine transporter (NET), cysteine replacement of Ile-155 (corresponding to SERT Ile-179) also rendered the transporter sensitive to MTSET inactivation. In NET I155C, cocaine enhanced this inactivation, and the substrate, dopamine, apparently protected against inactivation. The characteristics of this protection suggest that dopamine was transported, converting NET to a form in which Ile-155 was occluded. The results support the proposal that TM3 of SERT and NET constitute part of the substrate permeation pathway, and that Ile-172 in SERT resides close to the substrate binding site. They also suggest that Ile-179 in SERT (and Ile-155 in NET) is in a conformationally sensitive part of TM3, which may act as part of an external gate.


Subject(s)
Carrier Proteins/metabolism , Ion Channel Gating/physiology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Symporters , Allosteric Regulation , Amino Acid Substitution , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/drug effects , Carrier Proteins/genetics , Chlorides/metabolism , Cocaine/pharmacology , Humans , Hydrogen/metabolism , Ion Channel Gating/drug effects , Ion Transport , Isoleucine/physiology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/drug effects , Mesylates/pharmacology , Mutagenesis, Site-Directed , Norepinephrine Plasma Membrane Transport Proteins , Oxidation-Reduction , Potassium/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Reducing Agents/pharmacology , Serotonin/metabolism , Serotonin/pharmacology , Serotonin Plasma Membrane Transport Proteins , Sodium/metabolism , Sulfhydryl Compounds/pharmacology
14.
J Biol Chem ; 274(51): 36058-64, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593887

ABSTRACT

Chimeric transporters were constructed in which the predicted external loops of the serotonin transporter (SERT) were replaced one at a time with a corresponding sequence from the norepinephrine transporter (NET). All of the chimeric transporters were expressed at levels equal to or greater than those of wild type SERT, but the transport and binding activity of the mutants varied greatly. In particular, mutants in which the NET sequence replaced external loops 4 or 6 of SERT had transport activity 5% or less than that of wild type, and the loop 5 replacement was essentially inactive. In some of these mutants, binding of a high affinity cocaine analog was less affected than transport, suggesting that the mutation had less effect on the initial binding steps in transport than on subsequent conformational changes. The more severely affected mutants also displayed an altered response to Na(+). In contrast to the dramatic reduction in transport and binding, the specificity of ligand binding was essentially unchanged. Chimeric transporters did not gain affinity for dopamine, a NET substrate, or desipramine, an inhibitor, at the expense of affinity for serotonin or paroxetine, a selective SERT inhibitor. The results suggest that external loops are not the primary determinants of substrate and inhibitor binding sites. However, they are not merely passive structures connecting transmembrane segments but rather active elements responsible for maintaining the stability and conformational flexibility of the transporter.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Serotonin/metabolism , Base Sequence , Biological Transport/genetics , Carrier Proteins/genetics , HeLa Cells , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins , Transfection
15.
Brain Res Mol Brain Res ; 71(1): 120-6, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10407194

ABSTRACT

The serotonin transporter (SERT) is a member of a highly homologous family of sodium/chloride dependent neurotransmitter transporters responsible for reuptake of biogenic amines from the extracellular fluid. SERT constitutes the pharmacological target of several clinically important antidepressants. Here we report the molecular cloning of SERT from the bovine species. Translation of the nucleotide sequence revealed 44 amino acid differences compared to human SERT. When transiently expressed in HeLa cells and compared with rat and human SERTs the K(m) value for uptake was increased 2-fold. V(max) and B(max) were both increased about 4-fold indicating the turnover number is conserved. The pharmacological profile revealed a decreased sensitivity towards imipramine, desipramine, citalopram, fluoxetine and paroxetine compared with human SERT, while the sensitivity towards 3, 4-methylenedioxymethamphetamine (MDMA) was mainly unchanged. RT-PCR amplification of RNA from different tissues demonstrated expression of SERT in placenta, brain stem, bone marrow, kidney, lung, heart, adrenal gland, liver, parathyroid gland, thyroid gland, small intestine and pancreas.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Membrane Transport Proteins , Nerve Tissue Proteins , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cattle , Citalopram/pharmacology , Cloning, Molecular , Desipramine/pharmacology , Female , Fluoxetine/pharmacology , HeLa Cells , Humans , Imipramine/pharmacology , Kinetics , Membrane Glycoproteins/metabolism , Molecular Sequence Data , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Organ Specificity , Paroxetine/pharmacology , Phylogeny , Pregnancy , Rats , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins , Transfection
17.
J Biol Chem ; 273(43): 28098-106, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9774428

ABSTRACT

Transmembrane span 7 of the rat brain serotonin transporter was subjected to random mutagenesis. Of the 27 amino acid residues mutated, six were identified as functionally important by their sensitivity to nonconservative mutations. These residues were Asn-368 and Tyr-385, where substitutions that retained hydrogen-bonding ability were preferred; Gly-376 and Gly-384, where only glycine was accepted; Phe-380, where a phenyl ring was preferred; and Met-386, where hydrophobic substitutions were preferred. Mutations that did not preserve these structural characteristics were highly detrimental to serotonin transport activity. These six residues form a stripe that runs at an angle down the side of the putative alpha-helix, lending support to this structural prediction. Mutations at some of these positions also specifically impaired transport activity under low Na+ conditions. Other mutations at nearby positions in transmembrane span 7 also impaired activity in low Na+, although the activity of the mutants in high Na+ was similar to wild type. These results suggest that at least some of the six critical residues play a role in Na+ binding or perhaps in the coupling of Na+ binding to later steps in the transport cycle. These residues may be important in other aspects of the transporter's function as well.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins/metabolism , Serotonin/metabolism , Amino Acid Sequence , Animals , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/genetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Conformation , Rats , Serotonin Plasma Membrane Transport Proteins , Structure-Activity Relationship
18.
J Bioenerg Biomembr ; 30(2): 173-85, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9672239

ABSTRACT

Neurotransmitter transporters are essential components in the recycling of neurotransmitters released during neuronal activity. These transporters are the targets for important drugs affecting mood and behavior. They fall into at least four gene families, two encoding proteins in the plasma membrane and two in the synaptic vesicle membrane, although the known vesicular transporters have not all been cloned. Each of these transporters works by coupling the downhill movement of small ions such as Na+, Cl-, K+, and H+ to the uphill transport of neurotransmitter. Plasma membrane transporters move the transmitter into the cytoplasm by cotransport with Na+. Many transporters also couple Cl- cotransport to transmitter influx and these all belong to the NaCl-coupled family, although within the family the coupling stoichiometry can vary. Transporters for glutamate couple influx of this excitatory amino acid to Na+ and H+ influx and K+ efflux. Transporters in synaptic vesicles couple H+ efflux to neurotransmitter transport from the cytoplasm to the vesicle lumen.


Subject(s)
Carrier Proteins/metabolism , Energy Metabolism , Ion Transport , Membrane Proteins/metabolism , Neurotransmitter Agents/metabolism , Animals , Biological Transport, Active/physiology , Humans , Ion Channels , Membrane Potentials/physiology , Synaptic Vesicles/metabolism
19.
J Biol Chem ; 273(20): 12675-81, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9575231

ABSTRACT

The transmembrane topology of the serotonin transporter (SERT) has been examined by measuring the reactivity of selected lysine and cysteine residues with extracellular reagents. An impermeant biotinylating reagent, sulfosuccinimidyl 2-(biotinamido)ethyl-1, 3-dithiopropionate (NHS-SS-biotin), was shown to label SERT transiently expressed in cultured cells. Replacement of four lysine residues that were predicted to lie in external hydrophilic loops (eK-less) largely prevented the biotinylation reaction. Likewise, the cysteine-specific biotinylation reagent N-biotinylaminoethylmethanethiosulfonate (MTSEA-biotin) labeled wild type SERT but not a mutant in which Cys-109, predicted to lie in the first external loop, was replaced with alanine. These two mutant transporters reacted with the biotinylating reagents in digitonin-permeabilized cells, demonstrating that the abundant lysine and cysteine residues predicted to lie in intracellular hydrophilic domains were reactive but not accessible in intact cells. Mutants containing a single external lysine at positions 111, 194, 243, 319, 399, 490, and 571 reacted more readily with NHS-SS-biotin than did the eK-less mutant. Similarly, mutants with a single cysteine at positions 109, 310, 406, 489, and 564 reacted more readily with MTSEA-biotin than did the C109A mutant. All of these mutants were active and therefore likely to be folded correctly. These results support the original transmembrane topology and argue against an alternative topology proposed recently for the related glycine and gamma-aminobutyric acid transporters.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Biotin/analogs & derivatives , Biotin/chemistry , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cysteine/metabolism , Lysine/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mutagenesis, Site-Directed , Protein Conformation , Serotonin Plasma Membrane Transport Proteins , Succinimides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...