Skip to main content
Erschienen in: Molecular Autism 1/2014

Open Access 01.12.2014 | Research

N-ethylmaleimide-sensitive factor interacts with the serotonin transporter and modulates its trafficking: implications for pathophysiology in autism

verfasst von: Keiko Iwata, Hideo Matsuzaki, Taro Tachibana, Koji Ohno, Saori Yoshimura, Hironori Takamura, Kohei Yamada, Shinsuke Matsuzaki, Kazuhiko Nakamura, Kenji J Tsuchiya, Kaori Matsumoto, Masatsugu Tsujii, Toshirou Sugiyama, Taiichi Katayama, Norio Mori

Erschienen in: Molecular Autism | Ausgabe 1/2014

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

Changes in serotonin transporter (SERT) function have been implicated in autism. SERT function is influenced by the number of transporter molecules present at the cell surface, which is regulated by various cellular mechanisms including interactions with other proteins. Thus, we searched for novel SERT-binding proteins and investigated whether the expression of one such protein was affected in subjects with autism.

Methods

Novel SERT-binding proteins were examined by a pull-down system. Alterations of SERT function and membrane expression upon knockdown of the novel SERT-binding protein were studied in HEK293-hSERT cells. Endogenous interaction of SERT with the protein was evaluated in mouse brains. Alterations in the mRNA expression of SERT (SLC6A4) and the SERT-binding protein in the post-mortem brains and the lymphocytes of autism patients were compared to nonclinical controls.

Results

N-ethylmaleimide-sensitive factor (NSF) was identified as a novel SERT-binding protein. NSF was co-localized with SERT at the plasma membrane, and NSF knockdown resulted in decreased SERT expression at the cell membranes and decreased SERT uptake function. NSF was endogenously co-localized with SERT and interacted with SERT. While SLC6A4 expression was not significantly changed, NSF expression tended to be reduced in post-mortem brains, and was significantly reduced in lymphocytes of autistic subjects, which correlated with the severity of the clinical symptoms.

Conclusions

These data clearly show that NSF interacts with SERT under physiological conditions and is required for SERT membrane trafficking and uptake function. A possible role for NSF in the pathophysiology of autism through modulation of SERT trafficking, is suggested.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​2040-2392-5-33) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HM and TK co-designed the study. KI and HM collected blood samples; collected, analyzed and interpreted the data and prepared the manuscript. TT and SY produced the SERT antibody. KO, HT, KY and SM collected, analyzed and interpreted the data. KN recruited participants, collected blood samples and obtained post-mortem brain samples. KJT and KM collected blood samples and undertook clinical evaluations. MT recruited participants. TS recruited participants and diagnosed ASD. TK collected, analyzed and interpreted the data and prepared the manuscript. NM analyzed and interpreted the data, and prepared the manuscript. All authors read and approved the final manuscript.
Abkürzungen
5-HT
5-hydroxytryptamine
ADI-R
autism diagnostic interview-revised
ANOVA
analysis of variance
ASD
autism spectrum disorder
cDNA
complementary DNA
CNS
central nervous system
C-SERT
C-terminal domain of SERT
DMEM
Dulbecco's modified Eagle's medium
GST
glutathione S-transferase
HBSS
Hank’s balanced salt solution
Hic-5
hydrogen peroxide-inducible clone 5 protein
hSERT
human serotonin transporter
IQ
intelligence quotient
LC-MS/MS
liquid chromatography-tandem mass spectrometry
MacMARCKS
macrophage myristoylated alanine-rich C kinase substrate
MTT
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide
MW
molecular weight
nNOS
nitric oxide synthase
NSF
N-ethylmaleimide-sensitive factor
N-SERT
N-terminal domain of SERT
PBS
phosphate-buffered saline
PCR
polymerase chain reaction
PMI
post-mortem interval
PP2A
phosphatase 2A
qRT-PCR
quantitative real-time reverse-transcription-polymerase chain reaction
RT
room temperature
RT-PCR
reverse-transcription-polymerase chain reaction
SCAMP2
secretory carrier membrane protein 2
SCID
structured clinical interview for DSM-IV
SERT
serotonin transporter
siRNA
small interfering RNA
SLC6A4
member 4 of solute carrier family 6 (neurotransmitter transporter)
SNARE
soluble NSF attachment protein–receptor
tcTPC
time-controlled transcardiac perfusion cross-linking.

Background

Autism is a pervasive developmental disorder characterized by severe and sustained impairment of social interaction and communication, and restricted or stereotyped patterns of behavior and interest. Many studies on the pathophysiological mechanisms of autism have focused on the serotonergic system. Prior studies have consistently found elevated serotonin levels in the whole blood cells and platelets of autism patients [15] and their relatives [68]. Short-term dietary depletion of tryptophan (the precursor of serotonin) has been shown to exacerbate repetitive behavior and to elevate anxiety and feelings of unhappiness in autistic adults [9]. Accordingly, many genetic studies have examined the associations between autism and genetic mutations of human serotonin transporter (SERT; solute carrier family 6 (neurotransmitter transporter), member 4 (SLC6A4)), especially the short allele of a polymorphism in the promoter region of the serotonin transporter gene. Although some positive relationships have been found, the results to date are inconsistent [1015]. A single photon emission computed tomography study showed that autistic children, under light sedation, exhibit a reduction in SERT binding in the medial frontal cortex, midbrain and temporal lobe areas [16]. Importantly, our colleagues recently reported that binding of SERT and its radioligand was significantly lower throughout the brain in autistic individuals compared with controls [17]. The reduction in the anterior and posterior cingulate cortices was associated with an impairment of social cognition in autistic subjects, and a significant correlation was also found between repetitive and/or obsessive behavior and interests and a reduction in SERT binding in the thalamus [17]. These results suggested that SERT protein levels and/or its transport capacity were decreased in the brains of autistic patients. Despite this prediction, Azmitia and colleagues reported increased immunoreactivity to a SERT antibody of serotonin axons in the post-mortem cortices of autism patients [18].
SERT is an integral plasma membrane glycoprotein that regulates neurotransmission through the reuptake of 5-hydroxytryptamine (5-HT), also known as serotonin, from the synaptic cleft. SERT transport capacity is known to be regulated through mechanisms that involve subcellular redistribution of the transporter, which are regulated by various cellular mechanisms, including interactions with other proteins [19, 20]. Indeed, several SERT-binding proteins have been reported. Syntaxin-1A [2123] and secretory carrier membrane protein 2 (SCAMP2) have been reported to be associated with the N-terminal tail of SERT [24]. Macrophage myristoylated alanine-rich C kinase substrate (MacMARCKS) [25], integrin β3 [26] and nitric oxide synthase (nNOS) [27] have been reported to be associated with the C-terminal tail of SERT. SERT also forms complexes with hydrogen peroxide-inducible clone 5 protein (Hic-5) [28, 29], phosphatase 2A (PP2A) [30], and α- and γ-synuclein [31, 32]. By interacting with SERT, SCAMP2, MacMARCKS, nNOS, Hic-5, PP2A and α/γ-synuclein reduce the efficacy of serotonin reuptake because of a reduction in surface expression of SERT or promotion of SERT dephosphorylation [24, 25, 27, 2932]. Loss of integrin β3 results in decreased SERT function and surface expression in platelets [26]. Syntaxin-1A regulates the electrophysiological properties of SERT [23].
In this study, we sought to identify novel proteins interacting with the N- and C-terminal portions of SERT, and which thereby regulate SERT function. We also measured the levels of mRNAs for SERT and SERT-interacting proteins in post-mortem brains and lymphocytes from autism patients to assess their involvement in autism.

Methods

Animal experiments

Experiments using mice were approved by the Committee on Animal Research of Hamamatsu University School of Medicine and University of Fukui. These experiments were performed in accordance with the Guide for Animal Experimentation at the Hamamatsu University School of Medicine and the University of Fukui.

Glutathione S-transferase pull-down assays

Full-length rat SERT complementary DNA (cDNA) was obtained from Dr Heinrich Betz (Max Planck Institute) [25, 33]. PCR fragments corresponding to the N-terminal domain of the rat SERT (N-SERT; residues 1 to 85 amino acids) and the C-terminal domain of the rat SERT (C-SERT; residues 595 to 630 amino acids) were fused to glutathione S-transferase (GST) by subcloning into the pGEX-5X-1 bacterial expression vector (Amersham Bioscience, Uppsala, Sweden), to produce vectors containing GST-N-SERT and GST-C-SERT. Plasmids were transformed into Escherichia coli (BL21 (DE3), Stratagene, La Jolla, CA, USA) and were cultured and induced with isopropyl-β-D-thiogalactopyranoside (IPTG) at 37°C for 4 h. Mouse brain tissue was homogenized on ice using a homogenizer (Iuchi, Osaka, Japan), in 5 ml of homogenization buffer (50 mM NH4Cl, 40 mM Tris–HCl pH 8.0) supplemented with a 1× complete protease inhibitor cocktail (Roche Applied Science, Indianapolis, IN, USA) per brain. The same amount of extraction buffer (20 mM NaCl, 20 mM Tris–HCl pH 8.0, 1% NP-40, 1% deoxycholate) was added, and homogenates were incubated at 4°C for 30 min with rotation. Insoluble cellular debris was removed by centrifugation, and the supernatants were collected. Then, the extracts were diluted up to tenfold in homogenization buffer plus extraction buffer without detergents. Extracts were incubated with glutathione agarose bound to GST, GST-N-SERT or GST-C-SERT at 4°C for 3 h. Beads were washed five times with TBS buffer (50 mM Tris–HCl pH 7.4, 150 mM NaCl and 1 mM ethylenediaminetetraacetic acid) and boiled in SDS-PAGE sample buffer for 5 min to elute bound proteins. These samples were subjected to SDS-PAGE, which was followed by silver staining using a Silver Stain MS Kit (Wako Pure Chemical Industries, Ltd, Osaka, Japan) to visualize protein bands for mass spectrometry analysis. The samples were also used for Western blotting experiments.

Western blot analysis

Western blotting was performed following a previously published protocol [34]. Antibodies against SERT (1:400 to 2,000; C-20, Santa Cruz Biotechnology, Inc, CA, USA), N-ethylmaleimide-sensitive fusion protein (NSF; 1:500; Cell Signaling Technology, Inc, Danvers, MA, USA), syntaxin-1A (1:500; Santa Cruz Biotechnology, Inc, CA, USA) or β-actin (1:1,000; Abcam Inc, Cambridge, MA, USA) were used. Immunoreactive bands were scanned and quantified using ImageJ software (ImageJ 1.44, National Institutes of Health, USA).

In-gel digestion and mass spectrometry analysis

Protein bands were excised from SDS-polyacrylamide gels. The bands were processed in destaining solutions included in the Silver Stain MS Kit. Disulfide bonds were reduced with dithiothreitol (DTT) and the proteins were alkylated with iodoacetamide. The proteins were then treated with 50 μl (25 ng/μl) of Trypsin Gold (Promega, Madison, WI, USA) in 50 mM ammonium bicarbonate for 45 min on ice, and then overnight at 37°C. After enzymatic digestion, the peptides were eluted from the gel by treatment (twice, for 30 min each time) with 50 μl of a mixture containing 50% acetonitrile and 5% trifluoroacetic acid. The two eluates were pooled and evaporated to dryness in a vacuum centrifuge. Prior to mass spectrometric analysis, peptides were re-dissolved in 50 μl of 0.1% formic acid. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) of the peptide mixtures was performed on a QSTAR XL (ESI-QqTOF; AB Sciex, Foster City, CA, USA) mass spectrometer. Product ion (MS/MS) spectra of the peptides separated by high-performance liquid chromatography (HPLC) were recorded and then submitted to the Mascot database search engine (Matrix Science [35]) for protein identification. The SwissProt database was used with ‘all entries’ for taxonomy. The tolerance was ±0.1 Da, and only one error was considered for the enzyme’s cutoff point.

Production of a stable cell line (HEK293-hSERT cells)

The human SERT (hSERT) protein was transcribed from the human SERT gene. The cDNA for hSERT was isolated by RT-PCR. The PCR fragments were cloned into pcDNA3.1(+) (Invitrogen Carlsbad, CA, USA) resulting in the construct pcDNA-hSERT. To generate stably transfected cells, pcDNA-hSERT was transfected into the human embryonic kidney cell line HEK293 using Transfectamine 2000 (Invitrogen) in accordance with the manufacturer’s instructions. After 24 h, transfected cells were switched to a medium containing 1 mg/ml geneticin (G418); 1 week later, resistant colonies were isolated from culture plates using sterile clone rings. Individual cells were used to generate clonal lines. Multiple lines tested positive for immunostaining using SERT Ab (Santa Cruz Biotechnology, Inc) and a fluorescence-based uptake assay, and clonal line #7 (termed HEK293-hSERT cells) was used in all experiments reported here. The HEK293-hSERT cells were cultured in DMEM (Invitrogen) supplemented with 10% fetal bovine serum (Invitrogen), penicillin (100 U/ml), streptomycin (100 μg/ml) and G418 (0.2 mg/ml) at 37°C in 5% CO2.

Primary culture of serotonergic raphe neurons

Primary culturing of serotonergic raphe neurons was performed using mouse neurons as described previously [36]. Pregnant BL6 mice (E16.5) were euthanized by cervical dislocation. Embryos were removed and placed in Hank’s balanced salt solution (HBSS) without Ca2+ (Life Technologies Co, Carlsbad, CA, USA). Rostral raphe neurons were dissected from the midbrain according to a method described previously [37]. Briefly, heads were removed from the embryos under a dissecting microscope (SMZ645; Nikon, Tokyo, Japan), and the midbrain/brainstem was gently dissociated. The neural tube was opened ventrally and flattened in a Petri dish containing HBSS without Ca2+. A strip of tissue of approximately 0.5 mm in width was dissected at the midline of the rostral rhombencephalon. Raphe tissue was resuspended in 5 ml of HBSS without Ca2+ and triturated ten times; the homogenate was strained through a cell strainer (BD Biosciences, Mississauga, ON, Canada) to remove debris, and an equal amount of HBSS containing Ca2+ was added. Cells were centrifuged (500 g, 5 min), and the pellet was resuspended in 5 ml of Neurobasal media (Invitrogen) containing B27 supplement (Invitrogen), penicillin (100 U/ml), streptomycin (100 μg/ml), and 0.4% l-Glutamine (Invitrogen) and plated onto eight-well slide chambers coated with poly-d-lysine (BD Biosciences). Two days after plating, 0.3 ml of medium from each well was replaced with fresh medium. Cells were cultured for 7 days in vitro[36].

siRNA-mediated gene knockdown

The duplexed oligonucleotides of siRNA used in this study were based on the sequence of the human cDNA encoding NSF. NSF siRNAs and a non-silencing control siRNA were obtained from Integrated DNA Technologies (Coralville, IA, USA). The targeted sequences of the human NSF siRNAs were as follows: 5′-GGAATGCAATAAAGAGTAAATATAC-3′ (siRNA-1) and 5′-GGATAGGAATCAAGAAGTTACTAAT-3′ (siRNA-2). Transfection was performed using Lipofectamine RNAiMAX (Invitrogen) in accordance with the manufacturer’s instructions, and cells were processed 48 h after transfection.

Immunocytochemistry and microscopy

HEK293-hSERT cells were grown on poly-d-lysine-coated glass coverslips. Raphe neurons were plated onto eight-well slide chambers coated with poly-d-lysine (BD Biosciences) and cultured for 7 days in vitro[36]. Cells were washed with PBS (-) and fixed with 2% paraformaldehyde in PBS (-), pH 7.4, for 15 min at room temperature (RT). Cells were washed with PBS (-) and incubated with ice cold 100% methanol for 10 min at -20°C to permeabilize them. Cells were washed with PBS (-) and incubated with blocking solution (5% skimmed milk in PBS (-)) at RT for 1 h followed by incubation with primary antibody against SERT (1:400; C-20, Santa Cruz Biotechnology, Inc), NSF (1:500; Cell Signaling Technology, Inc), cadherin (1:50; Abcam Inc, Cambridge, MA, USA) or serotonin (1:50; Gene Tex, Inc, Irvine, CA, USA) diluted in 1% skimmed milk in PBS (-) for 2 h at RT. Cells were washed in PBS (-) and incubated with the appropriate fluorophore-conjugated secondary antibody diluted in 1% skimmed milk in PBS for 60 min at RT. After washing, the cells were mounted onto microscope slides in 50% glycerol in PBS (-). Samples were imaged on a fluorescence microscope (BX53; Olympus, Tokyo, Japan) or a laser scanning confocal microscope (FluoView FV1000; Olympus).

Fluorescence-based uptake assay

The fluorescence-based uptake assay employed a fluorescent substrate that mimics the biogenic amine neurotransmitters and is taken up by the cell through their specific transporters, resulting in increased fluorescence intensity [38]. The corresponding fluorescence-based potencies (FL pIC50 values) were determined in a similar manner to the [3H]-neurotransmitter uptake protocols [39]. HEK293-hSERT cells were plated in black, 96-well optical bottom assay plates coated with poly-d-lysine (#3882, Corning Life Sciences, Lowell, MA, USA) and transfected with siRNAs as described above. Fluorescent substrate uptake assays were performed using the Neurotransmitter Transporter Uptake Assay Kit (Molecular Devices Co, Sunnyvale, CA, USA) in accordance with the manufacturer’s instructions. Kinetic measurements of relative fluorescence units (integrated over 0.5 ms) were made using a cycle time of 5 min in a fluorescence microplate reader (SpectraMax M5; Molecular Devices Co). Data were normalized to cell number using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay described below. Non-specific uptake was determined in the presence of 10 μM fluoxetine, a selective serotonin reuptake inhibitor.

MTT assay

Cell proliferation was measured with a MTT assay. Cells were incubated with MTT solution at 37 ºC for 6 h. Following removal of the solution, dimethyl sulfoxide was added, and the amount of formazan formed was measured spectrophotometrically at 550 nm using a microplate reader (Bio-Rad, Hercules, CA, USA).

Biotinylation

Biotinylation experiments were performed using the Cell Surface Protein Isolation Kit (Pierce, Rockford, IL, USA) in accordance with the manufacturer’s instructions. The cells were incubated with sulfo-NHS-SS-biotin solution for 30 min at 4°C, and the biotinylation of membrane proteins was stopped by adding quenching solution. The cells were washed and lysed in lysis buffer containing 1× complete protease inhibitor cocktail (Roche Applied Science). Cell lysates were incubated with NeutrAvidin Agarose beads for 1 h at RT. Beads were washed and biotinylated proteins were eluted using SDS-PAGE sample buffer. Analysis was performed on aliquots taken: (a) prior to incubation with beads (as total lysate) and (b) of the bead elute (as the biotinylated membrane fraction). Then, immunoblot analysis was carried out as described above. Analysis was performed on aliquots taken: (a) prior to incubation with beads (as total lysate) and (b) of the bead elute (as the biotinylated membrane fraction). Then, Western blot analysis was carried out as described above. For the biotinylated membrane fraction, after Western blot analysis, the membrane was stained with Coomassie Brilliant Blue (CBB) as a protein-loading control.

Time-controlled transcardiac perfusion cross-linking and immunoprecipitation

The time-controlled transcardiac perfusion cross-linking (tcTPC) experiments were performed as described previously [40]. Mice were anesthetized and perfused with saline at 25 ml/min for 2 min to purge the blood vessels. The perfusate was switched to fixative solution (4% formaldehyde in PBS (-)) at 25 ml/min and cross-linking was carried out for 6 min. After perfusion, brains were rapidly removed from the skull, postfixed in tcTPC reagent and immediately frozen by immersion in liquid nitrogen. The perfusion and postfixing procedures were completed within 15 min. Mouse brains were homogenized on ice using a homogenizer (Iuchi, Osaka, Japan), in 5 ml of homogenization buffer (50 mM NH4Cl, 40 mM Tris–HCl, pH 8.0) supplemented with 1× complete protease inhibitor cocktail (Roche Applied Science) per brain. The same amount of extraction buffer (20 mM NaCl, 20 mM Tris–HCl, pH 8.0, 1% NP-40, 1% deoxycholate) was added, followed by incubation at 4°C for 30 min with rotation. Insoluble cellular debris was removed by centrifugation (3,000 rpm, 10 min), and the supernatants were then used as a brain extract. Brain extracts were pre-cleared with 30 μl of protein G-Sepharose (Thermo Fisher Scientific, Inc, Waltham, MA, USA) for 1 h at 4°C. Cleared lysates were first incubated with an anti-SERT antibody (made by two of the authors, TT and SY) at 4°C for 3 h, and then with 20 μl of protein G-Sepharose for 1 h at RT. The complex-bound resin was washed five times with IP buffer (25 mM Tris–HCl, 150 mM NaCl; pH 7.2). Immunoprecipitated complexes were boiled in 2× SDS-PAGE sample buffer for 5 min to elute bound proteins. Western blot analysis was carried out as described above.

Post-mortem brain tissues

The ethics committee of the Hamamatsu University School of Medicine approved this study. The Autism Tissue Program (Princeton, NJ, USA) [41], the National Institute of Child Health and Human Development’s Brain and Tissue Bank for Developmental Disorders (Baltimore, MD, USA) [42] and the Harvard Brain Tissue Resource Center (Belmont, MD, USA) [43] provided frozen post-mortem brain tissues from dorsal raphe regions (n = 11 control and n = 7 autism).

Lymphocyte samples

The participants in this study were 30 male subjects with autism spectrum disorder (ASD) and 30 healthy male controls. All participants were Japanese. They were born and lived in restricted areas of central Japan, including Aichi, Gifu and Shizuoka prefectures. Based on interviews and available information, including hospital records, diagnoses of ASD were made by an experienced child psychiatrist (TS) based on the DSM-IV-TR criteria. The Autism Diagnostic Interview-Revised (ADI-R) [44] was also conducted by two of the authors (KJT and KM), both of whom have established reliability for diagnosing autism with the Japanese version of the ADI-R. The ADI-R is a semi-structured interview conducted with a parent, usually the mother, and is used to confirm the diagnosis and also to evaluate the core symptoms of ASD. The ADI-R domain A score quantifies impairment in social interaction, the domain BV score quantifies impairment in communication, and the domain C score quantifies restricted, repetitive and stereotyped patterns of behavior and interests. The ADI-R domain D corresponds to the age of onset criterion for autistic disorder. The manual for the Wechsler Intelligence Scale for Children, Third Edition [45], was used to evaluate the intelligence quotient (IQ) of all the participants. Co-morbid psychiatric illnesses were excluded by means of the Structured Clinical Interview for DSM-IV (SCID). Participants were excluded from the study if they had any symptoms of inflammation, a diagnosis of fragile X syndrome, epileptic seizures, obsessive-compulsive disorder, affective disorders or any additional psychiatric or neurological diagnoses. None of the participants had ever received psychoactive medications before this study. Healthy control subjects were recruited locally by advertisement. All control subjects underwent a comprehensive assessment of their medical history to eliminate individuals with any neurological or other medical disorders. SCIDs were also conducted to identify any personal or family history of past or present mental illness. None of the comparison subjects initially recruited was found to fulfill any of these exclusion criteria.
This study was approved by the ethics committee of the Hamamatsu University School of Medicine. All participants as well as their guardians were given a complete description of the study, and provided written informed consent before enrollment. Whole-blood samples were collected by venipuncture from all participants. Lymphocytes were isolated from blood samples by means of the Ficoll-Paque gradient method (purity 80%) within 2 h after sampling.

Quantitative real-time reverse-transcription-polymerase chain reaction

Total RNA was isolated from the dorsal raphe regions of post-mortem brains and lymphocytes using TRIZOL reagent (Invitrogen). The RNA samples were further purified using the RNeasy Micro Kit (QIAGEN, Hilden, Germany). First-strand cDNA was synthesized from the RNA samples using the SuperScript III First-Strand Synthesis System (Invitrogen). Quantitative real-time reverse-transcription polymerase chain reaction (qRT-PCR) analysis was performed using the TaqMan method in the ABI StepOnePlus TM Real-Time PCR System (Applied Biosystems, Foster City, CA, USA). TaqMan assay IDs of the genes are as follows: SLC6A4, Hs00984349_m1 and NSF, Hs00938040_m1. Actin, beta (ACTB; Hs99999903_m1) was used as the endogenous reference. Relative quantification of NSF and SERT expression levels in post-mortem brains was performed using the delta-delta CT method [46], with the constitutively expressed gene ACTB as an internal control. Standard curves were constructed for NSF, SERT and ACTB primers to validate the application of the delta-delta CT method. Relative quantification of NSF and SERT expression levels in lymphocytes was performed using the relative standard curve method, with the constitutively expressed gene ACTB as an internal control.

Statistical analysis

The data were analyzed using a two-tailed unpaired t-test after it had been confirmed that there were no statistically significant differences in variance as assessed by the F test. One-way analysis of variance (ANOVA) followed by Tukey’s correction was used for multiple comparisons. One-way repeated-measures ANOVA with Tukey’s post hoc test was used for analysis of data from the uptake assay. The Mann–Whitney U test was used to evaluate differences in age, post-mortem interval (PMI) and IQs between the autism and control groups, and gene expression levels in the post-mortem brains and lymphocytes between these groups. Fisher’s exact test was used to evaluate differences in race and gender between the autism and control groups. Evaluation of the relationships between NSF expression level and clinical variables and symptom profiles was performed using Spearman’s rank correlation coefficient. P values of less than 0.05 were considered to indicate statistical significance. All statistical analyses were performed using statistical analysis software (SPSS, version 12.0 J, IBM, Armonk, NY, USA).

Results

Identification of N-ethylmaleimide-sensitive factor as a novel serotonin transporter-binding protein

To identify novel binding proteins for SERT, we conducted pull-down experiments using GST-N-SERT or GST-C-SERT with and without (as a negative control) mouse brain lysates. After SDS-PAGE and silver staining of the gels, at least ten specific bands were observed in the lane containing proteins eluted from GST-N-SERT beads incubated with brain lysates, and at least three bands were observed in the lane containing proteins eluted from GST-C-SERT beads incubated with brain lysates (Figure 1A). The protein bands were excised from the gel and subjected to in-gel trypsin digestion. The tryptic peptide mixtures were analyzed by mass spectrometry. Excluding proteins that bound to both termini of SERT, we identified seven N-terminal-specific binding proteins, but no C-terminal-specific binding proteins (Table 1). One of the N-terminal specific bands, migrating at around 70 kDa, N-4 (Figure 1A), was identified as NSF, which regulates membrane fusion events [47, 48], based on 24 independent MS spectra (Figure 1B and Table 1). We focused on the interaction between NSF and SERT in the present study for the following reasons. First, we identified NSF as having the highest reliability score (Table 1). Second, NSF interacts with neurotransmitter receptors, such as AMPA, β2 adrenergic and GABAA receptors, and it regulates the membrane trafficking and synaptic stabilization of these receptors [4957]. Finally, in the photoreceptor synapse, the NSF and Arrestin 1 interaction regulates expression of vesicular glutamate transporter 1 and excitatory amino acid transporter 5 in the photoreceptor synapse [58]. These findings suggest that NSF may interact with neurotransmitter transporters and regulates these functions in the central nervous system (CNS). To verify the interaction of NSF with SERT, we conducted Western blot analysis. GST, GST-N-SERT and GST-C-SERT were incubated with mouse brain extracts. As shown in Figure 1C, NSF bound the N-terminal region of SERT specifically. In support of previous studies, N-terminal-specific binding of syntaxin-1A was confirmed [2123] (Additional file 1: Figure S1).
Table 1
Identification of GST-N-SERT and GST-C-SERT pulled-down proteins from mouse brain extracts
Spot number
Gene name
Protein name
MW (Da)
Number
Sequence coverage
Score
Accession number
N-terminal specific
Cellular and molecular events
N-1
Synj1
Synaptojanin 1
172,509
23
14%
785
Q8CHC4
Endocytosis
N-2
Cand1
Cullin-associated NEDD8-dissociated protein 1
136,245
14
10%
526
Q6ZQ38
SCF complex assembly
N-3
Aco2
Aconitate hydratase, mitochondrial
85,410
14
19%
534
Q99KI0
  
N-4
Nsf
Vesicle-fusing ATPase (NSF)
82,561
24
27%
1,010
P46460
 
N-5
Atp6v1a
V-type proton ATPase catalytic subunit A
68,283
13
21%
466
P50516
Hydrolysis
N-6
Crmp1
Dihydropyrimidinase-related protein 1
62,129
12
20%
441
P97427
Axon guidance and cell migration
N-7
Cct2
T-complex protein 1 subunit beta
57,441
11
19%
202
P80314
Molecular chaperone
N-8
Fscn1
Fascin
54,474
14
25%
174
Q61553
Actin filament binding
N-9
Eno1
Alpha-enolase
47,111
16
24%
703
P17182
  
N-10
Cnp
2′,3′-cyclic-nucleotide 3′-phosphodiesterase
47,094
22
40%
341
P16330
  
C-1
Aco2
Aconitate hydratase, mitochondrial
85,410
8
9%
287
Q99KI0
  
C-2
Eno1
Alpha-enolase
47,111
8
20%
384
P17182
  
C-3
Cnp
2′,3′-cyclic-nucleotide 3′-phosphodiesterase
47,094
18
32%
225
P16330
  
C-SERT, C-terminal domain of the serotonin transporter; GST, glutathione S-transferase; MW, molecular weight; N-SERT, N-terminal domain of the serotonin transporter.

Co-localization of serotonin transporter and N-ethylmaleimide-sensitive factor in HEK293-hSERT cells

The subcellular localization of SERT and NSF was examined using immunofluorescence confocal microscopy. NSF is expressed endogenously in HEK293 cells. We established a stable human SERT-expressing cell line, HEK293-hSERT, using HEK293 cells as described in the Methods section. It was confirmed that SERT was transported to the plasma membrane in this cell line by double staining using antibodies to SERT and cadherin, a membrane marker (see Additional file 2: Figure S2). HEK293-hSERT cells were double labeled with antibodies to NSF and SERT, and it was revealed that NSF co-localized with SERT in the plasma membrane (Figure 2A,B,C) and intracellular particles (Figure 2D,E,F).

Effect of N-ethylmaleimide-sensitive factor knockdown on serotonin transporter function and cellular localization

We used RNA interference to knock down endogenous NSF expression. We confirmed that the efficacy of siRNA transfection into HEK293-hSERT cells was >90% (see Additional file 3: Figure S3). As shown in Figure 3A,B, it was confirmed that both of the siRNAs (siRNA-1 and -2) targeting NSF suppressed endogenous NSF protein levels by approximately 60% (P < 0.001, one-way ANOVA with Tukey’s post hoc test, n = 3 each). Importantly, whole-cell SERT protein levels were not changed significantly by the siRNAs targeting NSF (F(2,14) = 1.057; P = 0.374, one-way ANOVA, n = 5 to 6 each) (Figure 3C,D). To investigate the effect of NSF on SERT uptake function, we conducted a fluorescence-based uptake assay in HEK293-hSERT cells. As shown in Figure 4, both NSF siRNAs decreased fluorescence uptake (siRNA-1; P = 0.005 and siRNA-2; P < 0.001, one-way repeated measures ANOVA with Tukey’s post hoc test, n = 8 each). Fluoxetine completely inhibited uptake (Figure 4), including nonspecific uptake.
Next, we conducted biotinylation experiments in HEK293-hSERT cells using sulfo-NHS-SS-biotin. This compound, which binds to lysine and arginine residues in proteins, is cell impermeant and labels cell-surface proteins. Cells transfected with the siRNA of NSF (siRNA-2) or a negative control were incubated with sulfo-NHS-SS-biotin, followed by isolation of labeled proteins with avidin beads and analysis by Western blotting using anti-SERT antibodies. For the biotinylated membrane fraction, after Western blot analysis, the membrane was stained with CBB as a protein-loading control (Additional file 4: Figure S4). As shown in Figure 5A,B, the level of SERT protein at the cell membrane was decreased by an average of 50% (t = 5.399; df = 16; P < 0.001, two-tailed unpaired t-test, n = 9) following NSF knockdown, despite no change in the total levels of SERT protein (t = -1.565; df = 10; P = 0.149, two-tailed unpaired t-test, n = 6). Finally, we examined the distribution of SERT in HEK293-hSERT cells when NSF was suppressed. In support of the results of the experiment using sulfo-NHS-SS-biotin, the membrane expression of SERT was decreased by NSF knockdown in HEK293-hSERT cells (Figure 5C).

Association between serotonin transporter and N-ethylmaleimide-sensitive factor in vivo

To determine the physiological significance of our findings in vivo, we examined: (a) the interaction between SERT and NSF in the mouse brain by immunoprecipitation and Western blotting and (b) the cellular distributions of NSF and SERT in cultured mouse raphe neurons by immunocytochemistry and microscopy.
Schmitt-Ulms and colleagues have established a method that covalently conserves protein interactions through tcTPC [40]. This method enables the preservation of protein–protein interactions that occur under physiological conditions. We investigated the interaction of SERT with NSF in the mouse brain using this tcTPC method. First, we examined the accuracy of the method. Total protein from non-tcTPC- or tcTPC-treated mouse brains was analyzed by immunoblotting, and we confirmed that SERT-containing cross-linked complexes were retained by this method (see Additional file 5: Figure S5A). Second, we checked whether the complexes were precipitated by anti-SERT antibodies and confirmed that SERT-containing cross-linked complexes were precipitated in a dose-dependent manner using this antibody (see Additional file 5: Figure S5B). Then, finally, we investigated the binding of SERT to NSF. As shown in Figure 6A, NSF co-immunoprecipitated with SERT from tcTPC-treated brain cells indicating that NSF interacts with SERT in the mouse brain under physiological conditions. Next, the cellular distributions of NSF and SERT in cultured mouse raphe neurons were examined. About 10% of all cultured cells were 5-HT-positive neurons in support of a previous report (data not shown) [36]. NSF was ubiquitously expressed in all cultured cells (data not shown). As shown in Figure 6B, triple immunocytochemical staining for SERT, NSF and 5-HT revealed that NSF co-localizes with SERT in the cell body and fibers of cultured serotonergic neurons.

SLC6A4 and N-ethylmaleimide-sensitive factor expression in the raphe region of post-mortem brains from autism patients

The demographic characteristics of subjects (seven with autism and eleven control subjects) are described in Tables 2 and 3. There were no significant differences in age (P = 1.000, Mann–Whitney U test), race (P = 0.305, Fisher’s exact test), gender (P = 0.596, Fisher’s exact test) and PMI (P = 0.513, Mann–Whitney U test) between the autism and control groups (Table 3). Although changes in SERT function and expression have been implicated in autism, mRNA expression of the SLC6A4 gene that encodes SERT in the brains of autistic individuals has never been reported. Therefore, first, we measured SLC6A4 expression in the raphe region of post-mortem brains from autistic individuals and controls using qRT-PCR. SLC6A4 expression was normalized to the expression levels of an internal control (ACTB). As shown in Figure 7A, there are wide individual differences in the expression level of SLC6A4 among the subjects, and the level did not differ significantly between subjects with autism and controls (P = 0.928, Mann–Whitney U test). Then, we measured NSF expression in the same way. NSF expression was normalized to the expression of ACTB. We found that the NSF expression level in autism patients tended to be lower than that in controls; however, this trend was not statistically significant (P = 0.069, Mann–Whitney U test) (Figure 7B).
Table 2
Information for post-mortem brain tissues
Sample ID
Diagnosis
Age (years)
Gender
Post-mortem interval (hours)
Race
Cause of death
1065
Control
15
M
12
Caucasian
Multiple injuries
1297
Control
15
M
16
African-American
Multiple injuries
1407
Control
9
F
20
African-American
Asthma
1541
Control
20
F
19
Caucasian
Head injuries
1708
Control
8
F
20
African-American
Asphyxia, multiple injuries
1790
Control
14
M
18
Caucasian
Multiple injuries
1793
Control
12
M
19
African-American
Drowning
1860
Control
8
M
5
Caucasian
Cardiac arrhythmia
4543
Control
29
M
13
Caucasian
Multiple injuries
4638
Control
15
F
5
Caucasian
Chest injuries
4722
Control
14
M
16
Caucasian
Multiple injuries
797
Autism
9
M
13
Caucasian
Drowning
1638
Autism
20
F
50
Caucasian
Seizure
4231
Autism
8
M
12
African-American
Drowning
4721
Autism
8
M
16
African-American
Drowning
4899
Autism
14
M
9
Caucasian
Drowning
5000
Autism
27
M
8.3
NA
NA
6294
Autism
16
M
NA
NA
NA
F, female; M, male; NA, not available.
Table 3
Demographic data associated with raphe brain-tissue samples
 
Control (n = 11)
Autism (n = 7)
P value
Age (years) (range)
14.45 (8–29)
14.57 (8–27)
NSa
Race, n (%)
Caucasian 7 (63.6), African-American 4 (36.4)
Caucasian 3 (42.9), African-American 2 (28.6), NA 2 (28.6)
NSb
Gender, n (%)
Male 7 (63.6), Female 4 (36.4)
Male 6 (85.7), Female 1 (14.3)
NSb
Post-mortem interval (hours) (range)
14.82 (5–20)
18.05 (8.3-50)
NSa
aDerived from Mann–Whitney U test, bDerived from Fisher’s exact test.
NA, not available; NS, not significant.

SLC6A4 and N-ethylmaleimide-sensitive factor expression in lymphocytes from patients with autism spectrum disorders

NSF is expressed ubiquitously in all normal human tissues including lymphocytes [59]. Lymphocytes also carry SERT [60]. Thus, we measured expressions of these genes in lymphocytes from individuals with ASD and age- and sex-matched controls by qRT-PCR. The demographic characteristics of the subjects (30 with ASD and 30 control subjects) are described in Table 4. There were no significant differences in age (P = 0.928, Mann–Whitney U test) or IQs (verbal IQ, P = 0.098, Mann–Whitney U test; performance IQ, P = 0.076, Mann–Whitney U test; full-scale IQ, P = 0.554, Mann–Whitney U test) between the ASD and control groups (Table 4). As shown in Figure 8A, the expression level of SLC6A4 did not differ significantly between subjects with ASD and controls (P = 0.518, Mann–Whitney U test). On the other hand, we found that the NSF expression level in ASD patients were significantly lower than that in controls (P = 0.0011, Mann–Whitney U test) (Figure 8B). Moreover, there was a significantly negative correlation between NSF expression and ADI-R Domain A score, which quantified impairment in social interaction, in individuals with ASD (r s = 0.131, P = 0.0498, Spearman’s rank correlation coefficient test) (Figure 8C). There were no significant correlations between NSF expression levels and levels of SLC6A4 and any other symptom profile or clinical variables (data not shown).
Table 4
Demographic data associated with lymphocyte samples
 
Control (N = 30)b
Autism (N = 30)b
P value
Age (years)
11.1 ± 2.3 (6–16)
11.6 ± 2.7 (7–16)
NSa
ADI-R
   
  Domain A score
 
20.0 ± 5.3 (10–30)
 
  Domain BV score
 
14.3 ± 4.0 (8–23)
 
  Domain C score
 
8.5 ± 3.4 (3–9)
 
  Domain D score
 
3.1 ± 1.1 (1–5)
 
WISC-III
   
  Verbal IQ
99.1 ± 10.3 (77–120)
90.4 ± 28.7 (44–153)
NSa
  Performance IQ
97.0 ± 10.2 (76–114)
89.8 ± 22.9 (47–131)
NSa
  Full-scale IQ
97.8 ± 9.5 (82–115)
89.0 ± 26.9 (42–140)
NSa
aDerived from Mann–Whitney U test; bvalues are expressed as mean ± standard deviation (range).
ADI-R, Autism Diagnostic Interview-Revised; IQ, intelligence quotient; NS, not significant; WISC-III, the third edition of the Wechsler Intelligence Scale for Children.

Discussion

In this study, NSF was identified as a novel SERT-binding protein interacting with the N-terminal region of SERT. NSF knockdown resulted in decreased membrane expression of SERT and decreased uptake of substrate. These results clearly show that NSF modulates SERT membrane trafficking, which is consistent with its uptake function. An immunoprecipitation assay using mouse brain and immunocytochemistry of cultured mouse raphe neurons clearly indicated that SERT–NSF complexes were formed under physiological conditions in vivo. In addition, a study of post-mortem brains revealed that the SLC6A4 expression level was not affected in subjects with autism, but the NSF expression level in the raphe region tended to be decreased; however, this potential trend is not statistically significant. In lymphocytes, the SLC6A4 expression level was also unchanged, but the NSF expression level was significantly decreased in subjects with ASD and correlated with the severity of clinical symptoms.

N-ethylmaleimide-sensitive factor functions and protein binding

NSF is a homohexameric ATPase [61, 62], which is an essential component of the protein machinery responsible for various membrane fusion events, including intercisternal Golgi protein transport and the exocytosis of synaptic vesicles [63]. NSF binds to soluble NSF attachment protein–receptor (SNARE) complexes and mediates the recycling of spent SNARE complexes for subsequent rounds of membrane fusion [63, 64]. While this is a major function of NSF, it also interacts with receptor proteins, such as AMPA, β2 adrenergic and GABAA receptors, and is thought to affect their trafficking patterns or recycling [4957]. Additionally, an interaction between NSF and arrestin 1 regulates the expression of vesicular glutamate transporter 1 and excitatory amino acid transporter 5 in the photoreceptor synapse [58]. In the present study, we found, for the first time, that NSF binds the neurotransmitter transporter SERT and regulates its function in the CNS.

Serotonin transporter forms complexes with N-ethylmaleimide-sensitive factor in vivo

Several putative SERT-binding proteins have been reported [2132]. However, almost all of these were identified using the yeast two-hybrid system and little is known regarding whether any of these proteins bind to SERT and regulate its function in the mammalian brain. Also, little is known about the involvement of these proteins in autism [65, 66]. Therefore, in this study, we used a pull-down system together with mouse brain tissue to identify novel SERT-binding proteins. Moreover, we used the tcTPC method, which is an innovative tool for studying proteins in living tissues [40]. This method enabled us to preserve protein–protein interactions occurring under physiological conditions. This cross-linking also preserves membrane protein assemblies, which are degraded by solubilizing detergents. For instance, whereas most detergents cause rapid disintegration of the γ-secretase complex, three of four known components of the complex were purified and identified from harsh detergents and a high salt concentration by tcTPC [40]. Because NSF was not co-immunoprecipitated with SERT from non-tcTPC-treated brains (Figure 6A), it is likely that SERT–NSF complexes are sensitive to solubilizing detergents. The discovery of complexes including NSF and SERT, which form in the mammalian brain under physiological conditions, in the present study, is important from the viewpoint of their potential involvement in the pathophysiology of disorders such as autism. It is not yet clear whether NSF binds SERT directly or indirectly. In addition, the band for the SERT–NSF complex was smeared, suggesting that multiple types of SERT–NSF complexes exist. It is possible that SERT interacts with NSF through other proteins. Indeed, it is possible that GABAA receptors interact with NSF via GABAA receptor-associated protein, and regulate its intracellular distribution and recycling [56, 67]. Detailed analyses of these SERT–NSF complexes are needed.

Serotonin transporter and N-ethylmaleimide-sensitive factor expressions in autism

Recently, Nakamura and colleagues reported that the levels of SERT based on its radioligand binding were significantly lower throughout the brain in autistic individuals compared with controls [17]. On the other hand, Azmitia and colleagues reported increased immunoreactivity to a SERT antibody of serotonin axons in the post-mortem cortex of autism patients [18]. Our results show that, at least, SLC6A4 mRNA expression is normal in the raphe region of post-mortem brains from subjects with autism. Our findings and previous results lead us to two suggestions. First, although the transcription of SLC6A4 is normal in subjects with autism, the level of SERT protein at the pre-synaptic membrane is decreased because of an impairment of the trafficking system. Second, SERT protein that is not delivered to the pre-synaptic membrane accumulates in axon fibers in the brains of subjects with autism. In lymphocytes, we found that SLC6A4 expression was not changed in subjects with ASD. In contrast with our finding, Hu et al. previously reported that there was a significant decrease in the expression in the more severely affected twin for autistic twin pairs studied using lymphoblastoid cell lines [68]. This study used lymphoblastoid cell lines, not lymphocytes, from only three sets of discordant twins, and SLC6A4 expression was not compared with normal controls [68]. These differences may be the cause of the discrepancies between the present study and that report.
We found that the NSF expression levels tended to decrease in the raphe region of post-mortem brains from subjects with autism; however, this trend was not statistically significant (n = 11 control and n = 7 autism). Further studies with larger numbers of post-mortem brains are needed to clarify NSF expression status in the brain of autism patients. In lymphocytes, we found, for the first time, that NSF expression was significantly lower in subjects with ASD and lower NSF expression correlated with the severity of impairments in social interaction. Our findings suggest that peripheral NSF mRNA levels may serve as a reliable peripheral biological marker of ASD.
Sullivan et al. reported that the expression levels of a number of biologically relevant genes are statistically similar between lymphocytes and CNS tissues including the brain, and suggested that the cautious and thoughtful use of lymphocytic gene expression may be a useful surrogate for gene expression in the CNS when it has been determined that the gene is expressed in both [69]. In support of previous findings [59, 60], the expressions of SLC6A4 and NSF were detected in both tissues, and it is likely that levels of SLC6A4 and NSF in the peripheral lymphocytes may reflect the levels in post-mortem brains, although further study is needed.

The serotonin transporter–N-ethylmaleimide-sensitive factor binding and implications for pathophysiology in autism

Sanyal and Krishnan reported a lethal mutation in the Drosophila homolog of NSF [70]. Intriguingly, mutant adult survivors show abnormal seizure-like paralytic behavior [70]. Additionally, Matveeva and colleagues reported that decreased production of NSF is associated with epilepsy in rats [71]. Importantly, a high rate of co-occurrence of autism and epilepsy has been described [7276]. Approximately 30% of children with autism have epilepsy and 30% of children with epilepsy have autism [77]. Interestingly, an abnormal status for SERT has been reported in epileptic patients as follows. Autoradiography experiments have revealed that the temporal neocortex surrounding the epileptic focus of patients with mesial temporal lobe epilepsy presents diminished SERT binding in all cortical layers [78]. A significant decrease was found in the SERT density in the platelet membranes from epileptic patients having undergone an epileptic seizure [79, 80]. Additionally, it has been shown that epileptic patients who had been treated with inhibitors of serotonin reuptake, such as fluoxetine and citalopram, in addition to their ongoing antiepileptic therapy displayed remarkable clinical improvements [81, 82]. This indirect evidence implies the relationship between SERT and NSF in neurological disorders, such as autism. Further investigations of the status of SERT–NSF binding in the brain of autism patients would be useful for understanding the mechanisms that underlie autism. In addition, an animal model, such as an NSF conditional knockout mouse, would be a useful tool for understanding the mechanisms that underlie ASD.
As mentioned above, NSF interacts with neurotransmitter receptors such as AMPA, β2 adrenergic and GABAA receptors, and regulates the membrane trafficking and recycling of these receptors [4957]. An abnormal status of many of these receptors has been reported in autism. Binding of GABAAα5 and its radioligand was significantly lower throughout the brains of participants with ASDs compared with controls [83]. The mRNA levels of AMPA receptor were significantly increased in the post-mortem cerebellum of autistic individuals, while the receptor density was slightly decreased in people with autism [84]. It is possible that NSF may contribute to the pathophysiology of autism through these known interactions with relevant molecules.

Conclusions

This study showed that dysfunctional trafficking of SERT mediated by NSF may be linked with the pathophysiology of autism. The identification of SERT-binding proteins provides new opportunities not only to dissect the accessory components involved in SERT function and regulation, but also to elucidate the pathophysiology of psychiatric disorders or developmental disorders, such as autism. Future studies should examine the pathophysiological implications of SERT–NSF interactions for autism.

Acknowledgements

We thank Dr Jane Pickett, Director of Brain Resources and Data, Autism Tissue Program, for facilitating brain tissue collection. Human tissue was obtained from the National Institute of Child Health and Development Brain and Tissue Bank for Developmental Disorders at the University of Maryland, Baltimore, MD. Tissue samples were also provided by the Harvard Brain Tissue Resource Center. Written informed consent was obtained from the participants or their families for publication of this manuscript and accompanying figures. We would like to thank all the participants and families for their time and effort. We thank Heinrich Betz for the generous supply of full-length rat SERT cDNA. We also thank Masako Suzuki (Research Equipment Center, Hamamatsu University School of Medicine) for the technical support of the mass spectrometry analysis. This study was supported by a Grant-in-Aid for Scientific Research (C) from the Ministry of Education, Culture, Sports, Science and Technology of Japan to KI and a Grant-in-Aid for Scientific Research on Innovative Areas from the Ministry of Education, Culture, Sports, Science and Technology of Japan to HM. A part of this study is the result of integrated research on neuropsychiatric disorders carried out under the Strategic Research Program for Brain Sciences by the Ministry of Education, Culture, Sports, Science and Technology of Japan.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HM and TK co-designed the study. KI and HM collected blood samples; collected, analyzed and interpreted the data and prepared the manuscript. TT and SY produced the SERT antibody. KO, HT, KY and SM collected, analyzed and interpreted the data. KN recruited participants, collected blood samples and obtained post-mortem brain samples. KJT and KM collected blood samples and undertook clinical evaluations. MT recruited participants. TS recruited participants and diagnosed ASD. TK collected, analyzed and interpreted the data and prepared the manuscript. NM analyzed and interpreted the data, and prepared the manuscript. All authors read and approved the final manuscript.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Anderson GM, Freedman DX, Cohen DJ, Volkmar FR, Hoder EL, McPhedran P, Minderaa RB, Hansen CR, Young JG: Whole blood serotonin in autistic and normal subjects. J Child Psychol Psychiatry. 1987, 28: 885-900. 10.1111/j.1469-7610.1987.tb00677.x.CrossRefPubMed Anderson GM, Freedman DX, Cohen DJ, Volkmar FR, Hoder EL, McPhedran P, Minderaa RB, Hansen CR, Young JG: Whole blood serotonin in autistic and normal subjects. J Child Psychol Psychiatry. 1987, 28: 885-900. 10.1111/j.1469-7610.1987.tb00677.x.CrossRefPubMed
2.
Zurück zum Zitat Ciaranello RD: Hyperserotonemia and early infantile autism. N Engl J Med. 1982, 307: 181-183. 10.1056/NEJM198207153070310.CrossRefPubMed Ciaranello RD: Hyperserotonemia and early infantile autism. N Engl J Med. 1982, 307: 181-183. 10.1056/NEJM198207153070310.CrossRefPubMed
3.
Zurück zum Zitat Cook EH, Leventhal BL, Freedman DX: Serotonin and measured intelligence. J Autism Dev Disord. 1988, 18: 553-559. 10.1007/BF02211873.CrossRefPubMed Cook EH, Leventhal BL, Freedman DX: Serotonin and measured intelligence. J Autism Dev Disord. 1988, 18: 553-559. 10.1007/BF02211873.CrossRefPubMed
4.
Zurück zum Zitat Hanley HG, Stahl SM, Freedman DX: Hyperserotonemia and amine metabolites in autistic and retarded children. Arch Gen Psychiatry. 1977, 34: 521-531. 10.1001/archpsyc.1977.01770170031002.CrossRefPubMed Hanley HG, Stahl SM, Freedman DX: Hyperserotonemia and amine metabolites in autistic and retarded children. Arch Gen Psychiatry. 1977, 34: 521-531. 10.1001/archpsyc.1977.01770170031002.CrossRefPubMed
5.
Zurück zum Zitat Schain RJ, Freedman DX: Studies on 5-hydroxyindole metabolism in autistic and other mentally retarded children. J Pediatr. 1961, 58: 315-320. 10.1016/S0022-3476(61)80261-8.CrossRefPubMed Schain RJ, Freedman DX: Studies on 5-hydroxyindole metabolism in autistic and other mentally retarded children. J Pediatr. 1961, 58: 315-320. 10.1016/S0022-3476(61)80261-8.CrossRefPubMed
6.
Zurück zum Zitat Abramson RK, Wright HH, Carpenter R, Brennan W, Lumpuy O, Cole E, Young SR: Elevated blood serotonin in autistic probands and their first-degree relatives. J Autism Dev Disord. 1989, 19: 397-407. 10.1007/BF02212938.CrossRefPubMed Abramson RK, Wright HH, Carpenter R, Brennan W, Lumpuy O, Cole E, Young SR: Elevated blood serotonin in autistic probands and their first-degree relatives. J Autism Dev Disord. 1989, 19: 397-407. 10.1007/BF02212938.CrossRefPubMed
7.
Zurück zum Zitat Cook EH, Leventhal BL, Heller W, Metz J, Wainwright M, Freedman DX: Autistic children and their first-degree relatives: relationships between serotonin and norepinephrine levels and intelligence. J Neuropsychiatry Clin Neurosci. 1990, 2: 268-274.CrossRefPubMed Cook EH, Leventhal BL, Heller W, Metz J, Wainwright M, Freedman DX: Autistic children and their first-degree relatives: relationships between serotonin and norepinephrine levels and intelligence. J Neuropsychiatry Clin Neurosci. 1990, 2: 268-274.CrossRefPubMed
8.
Zurück zum Zitat Cross S, Kim SJ, Weiss LA, Delahanty RJ, Sutcliffe JS, Leventhal BL, Cook EH, Veenstra-Vanderweele J: Molecular genetics of the platelet serotonin system in first-degree relatives of patients with autism. Neuropsychopharmacology. 2008, 33: 353-360. 10.1038/sj.npp.1301406.PubMedCentralCrossRefPubMed Cross S, Kim SJ, Weiss LA, Delahanty RJ, Sutcliffe JS, Leventhal BL, Cook EH, Veenstra-Vanderweele J: Molecular genetics of the platelet serotonin system in first-degree relatives of patients with autism. Neuropsychopharmacology. 2008, 33: 353-360. 10.1038/sj.npp.1301406.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat McDougle CJ, Naylor ST, Cohen DJ, Aghajanian GK, Heninger GR, Price LH: Effects of tryptophan depletion in drug-free adults with autistic disorder. Arch Gen Psychiatry. 1996, 53: 993-1000. 10.1001/archpsyc.1996.01830110029004.CrossRefPubMed McDougle CJ, Naylor ST, Cohen DJ, Aghajanian GK, Heninger GR, Price LH: Effects of tryptophan depletion in drug-free adults with autistic disorder. Arch Gen Psychiatry. 1996, 53: 993-1000. 10.1001/archpsyc.1996.01830110029004.CrossRefPubMed
10.
Zurück zum Zitat Cook EH, Courchesne R, Lord C, Cox NJ, Yan S, Lincoln A, Haas R, Courchesne E, Leventhal BL: Evidence of linkage between the serotonin transporter and autistic disorder. Mol Psychiatry. 1997, 2: 247-250. 10.1038/sj.mp.4000266.CrossRefPubMed Cook EH, Courchesne R, Lord C, Cox NJ, Yan S, Lincoln A, Haas R, Courchesne E, Leventhal BL: Evidence of linkage between the serotonin transporter and autistic disorder. Mol Psychiatry. 1997, 2: 247-250. 10.1038/sj.mp.4000266.CrossRefPubMed
11.
Zurück zum Zitat Conroy J, Meally E, Kearney G, Fitzgerald M, Gill M, Gallagher L: Serotonin transporter gene and autism: a haplotype analysis in an Irish autistic population. Mol Psychiatry. 2004, 9: 587-593. 10.1038/sj.mp.4001459.CrossRefPubMed Conroy J, Meally E, Kearney G, Fitzgerald M, Gill M, Gallagher L: Serotonin transporter gene and autism: a haplotype analysis in an Irish autistic population. Mol Psychiatry. 2004, 9: 587-593. 10.1038/sj.mp.4001459.CrossRefPubMed
12.
Zurück zum Zitat Kim SJ, Cox N, Courchesne R, Lord C, Corsello C, Akshoomoff N, Guter S, Leventhal BL, Courchesne E, Cook EH: Transmission disequilibrium mapping at the serotonin transporter gene (SLC6A4) region in autistic disorder. Mol Psychiatry. 2002, 7: 278-288. 10.1038/sj.mp.4001033.CrossRefPubMed Kim SJ, Cox N, Courchesne R, Lord C, Corsello C, Akshoomoff N, Guter S, Leventhal BL, Courchesne E, Cook EH: Transmission disequilibrium mapping at the serotonin transporter gene (SLC6A4) region in autistic disorder. Mol Psychiatry. 2002, 7: 278-288. 10.1038/sj.mp.4001033.CrossRefPubMed
13.
Zurück zum Zitat Maestrini E, Lai C, Marlow A, Matthews N, Wallace S, Bailey A, Cook EH, Weeks DE, Monaco AP, International Molecular Genetic Study of Autism Consortium: Serotonin transporter (5-HTT) and γ-aminobutyric acid receptor subunit β3 (GABRB3) gene polymorphisms are not associated with autism in the IMGSA families. Am J Med Genet. 1999, 88: 492-496. 10.1002/(SICI)1096-8628(19991015)88:5<492::AID-AJMG11>3.0.CO;2-X.CrossRefPubMed Maestrini E, Lai C, Marlow A, Matthews N, Wallace S, Bailey A, Cook EH, Weeks DE, Monaco AP, International Molecular Genetic Study of Autism Consortium: Serotonin transporter (5-HTT) and γ-aminobutyric acid receptor subunit β3 (GABRB3) gene polymorphisms are not associated with autism in the IMGSA families. Am J Med Genet. 1999, 88: 492-496. 10.1002/(SICI)1096-8628(19991015)88:5<492::AID-AJMG11>3.0.CO;2-X.CrossRefPubMed
14.
Zurück zum Zitat Ramoz N, Reichert JG, Corwin TE, Smith CJ, Silverman JM, Hollander E, Buxbaum JD: Lack of evidence for association of the serotonin transporter gene SLC6A4 with autism. Biol Psychiatry. 2006, 60: 186-191. 10.1016/j.biopsych.2006.01.009.CrossRefPubMed Ramoz N, Reichert JG, Corwin TE, Smith CJ, Silverman JM, Hollander E, Buxbaum JD: Lack of evidence for association of the serotonin transporter gene SLC6A4 with autism. Biol Psychiatry. 2006, 60: 186-191. 10.1016/j.biopsych.2006.01.009.CrossRefPubMed
15.
Zurück zum Zitat Sutcliffe JS, Delahanty RJ, Prasad HC, McCauley JL, Han Q, Jiang L, Li C, Folstein SE, Blakely RD: Allelic heterogeneity at the serotonin transporter locus (SLC6A4) confers susceptibility to autism and rigid-compulsive behaviors. Am J Hum Genet. 2005, 77: 265-279. 10.1086/432648.PubMedCentralCrossRefPubMed Sutcliffe JS, Delahanty RJ, Prasad HC, McCauley JL, Han Q, Jiang L, Li C, Folstein SE, Blakely RD: Allelic heterogeneity at the serotonin transporter locus (SLC6A4) confers susceptibility to autism and rigid-compulsive behaviors. Am J Hum Genet. 2005, 77: 265-279. 10.1086/432648.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Makkonen I, Riikonen R, Kokki H, Airaksinen MM, Kuikka JT: Serotonin and dopamine transporter binding in children with autism determined by SPECT. Dev Med Child Neurol. 2008, 50: 593-597. 10.1111/j.1469-8749.2008.03027.x.CrossRefPubMed Makkonen I, Riikonen R, Kokki H, Airaksinen MM, Kuikka JT: Serotonin and dopamine transporter binding in children with autism determined by SPECT. Dev Med Child Neurol. 2008, 50: 593-597. 10.1111/j.1469-8749.2008.03027.x.CrossRefPubMed
17.
Zurück zum Zitat Nakamura K, Sekine Y, Ouchi Y, Tsujii M, Yoshikawa E, Futatsubashi M, Tsuchiya KJ, Sugihara G, Iwata Y, Suzuki K, Matsuzaki H, Suda S, Sugiyama T, Takei N, Mori N: Brain serotonin and dopamine transporter bindings in adults with high-functioning autism. Arch Gen Psychiatry. 2010, 67: 59-68. 10.1001/archgenpsychiatry.2009.137.CrossRefPubMed Nakamura K, Sekine Y, Ouchi Y, Tsujii M, Yoshikawa E, Futatsubashi M, Tsuchiya KJ, Sugihara G, Iwata Y, Suzuki K, Matsuzaki H, Suda S, Sugiyama T, Takei N, Mori N: Brain serotonin and dopamine transporter bindings in adults with high-functioning autism. Arch Gen Psychiatry. 2010, 67: 59-68. 10.1001/archgenpsychiatry.2009.137.CrossRefPubMed
18.
Zurück zum Zitat Azmitia EC, Singh JS, Whitaker-Azmitia PM: Increased serotonin axons (immunoreactive to 5-HT transporter) in postmortem brains from young autism donors. Neuropharmacology. 2011, 60: 1347-1354. 10.1016/j.neuropharm.2011.02.002.CrossRefPubMed Azmitia EC, Singh JS, Whitaker-Azmitia PM: Increased serotonin axons (immunoreactive to 5-HT transporter) in postmortem brains from young autism donors. Neuropharmacology. 2011, 60: 1347-1354. 10.1016/j.neuropharm.2011.02.002.CrossRefPubMed
19.
Zurück zum Zitat Blakely RD, Bauman AL: Biogenic amine transporters: regulation in flux. Curr Opin Neurobiol. 2000, 10: 328-336. 10.1016/S0959-4388(00)00088-X.CrossRefPubMed Blakely RD, Bauman AL: Biogenic amine transporters: regulation in flux. Curr Opin Neurobiol. 2000, 10: 328-336. 10.1016/S0959-4388(00)00088-X.CrossRefPubMed
20.
Zurück zum Zitat Robinson MB: Regulated trafficking of neurotransmitter transporters: common notes but different melodies. J Neurochem. 2002, 80: 1-11.CrossRefPubMed Robinson MB: Regulated trafficking of neurotransmitter transporters: common notes but different melodies. J Neurochem. 2002, 80: 1-11.CrossRefPubMed
21.
Zurück zum Zitat Ciccone MA, Timmons M, Phillips A, Quick MW: Calcium/calmodulin-dependent kinase II regulates the interaction between the serotonin transporter and syntaxin 1A. Neuropharmacology. 2008, 55: 763-770. 10.1016/j.neuropharm.2008.06.018.PubMedCentralCrossRefPubMed Ciccone MA, Timmons M, Phillips A, Quick MW: Calcium/calmodulin-dependent kinase II regulates the interaction between the serotonin transporter and syntaxin 1A. Neuropharmacology. 2008, 55: 763-770. 10.1016/j.neuropharm.2008.06.018.PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Quick MW: Role of syntaxin 1A on serotonin transporter expression in developing thalamocortical neurons. Int J Dev Neurosci. 2002, 20: 219-224. 10.1016/S0736-5748(02)00021-7.CrossRefPubMed Quick MW: Role of syntaxin 1A on serotonin transporter expression in developing thalamocortical neurons. Int J Dev Neurosci. 2002, 20: 219-224. 10.1016/S0736-5748(02)00021-7.CrossRefPubMed
23.
Zurück zum Zitat Quick MW: Regulating the conducting states of a mammalian serotonin transporter. Neuron. 2003, 40: 537-549. 10.1016/S0896-6273(03)00605-6.CrossRefPubMed Quick MW: Regulating the conducting states of a mammalian serotonin transporter. Neuron. 2003, 40: 537-549. 10.1016/S0896-6273(03)00605-6.CrossRefPubMed
24.
Zurück zum Zitat Muller HK, Wiborg O, Haase J: Subcellular redistribution of the serotonin transporter by secretory carrier membrane protein 2. J Biol Chem. 2006, 281: 28901-28909. 10.1074/jbc.M602848200.CrossRefPubMed Muller HK, Wiborg O, Haase J: Subcellular redistribution of the serotonin transporter by secretory carrier membrane protein 2. J Biol Chem. 2006, 281: 28901-28909. 10.1074/jbc.M602848200.CrossRefPubMed
25.
Zurück zum Zitat Jess U, El Far O, Kirsch J, Betz H: Interaction of the C-terminal region of the rat serotonin transporter with MacMARCKS modulates 5-HT uptake regulation by protein kinase C. Biochem Biophys Res Commun. 2002, 294: 272-279. 10.1016/S0006-291X(02)00460-6.CrossRefPubMed Jess U, El Far O, Kirsch J, Betz H: Interaction of the C-terminal region of the rat serotonin transporter with MacMARCKS modulates 5-HT uptake regulation by protein kinase C. Biochem Biophys Res Commun. 2002, 294: 272-279. 10.1016/S0006-291X(02)00460-6.CrossRefPubMed
26.
Zurück zum Zitat Carneiro AM, Cook EH, Murphy DL, Blakely RD: Interactions between integrin αIIbβ3 and the serotonin transporter regulate serotonin transport and platelet aggregation in mice and humans. J Clin Invest. 2008, 118: 1544-1552. 10.1172/JCI33374.PubMedCentralCrossRefPubMed Carneiro AM, Cook EH, Murphy DL, Blakely RD: Interactions between integrin αIIbβ3 and the serotonin transporter regulate serotonin transport and platelet aggregation in mice and humans. J Clin Invest. 2008, 118: 1544-1552. 10.1172/JCI33374.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Chanrion B, Mannoury la Cour C, Bertaso F, Lerner-Natoli M, Freissmuth M, Millan MJ, Bockaert J, Marin P: Physical interaction between the serotonin transporter and neuronal nitric oxide synthase underlies reciprocal modulation of their activity. Proc Natl Acad Sci USA. 2007, 104: 8119-8124. 10.1073/pnas.0610964104.PubMedCentralCrossRefPubMed Chanrion B, Mannoury la Cour C, Bertaso F, Lerner-Natoli M, Freissmuth M, Millan MJ, Bockaert J, Marin P: Physical interaction between the serotonin transporter and neuronal nitric oxide synthase underlies reciprocal modulation of their activity. Proc Natl Acad Sci USA. 2007, 104: 8119-8124. 10.1073/pnas.0610964104.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Carneiro AM, Ingram SL, Beaulieu JM, Sweeney A, Amara SG, Thomas SM, Caron MG, Torres GE: The multiple LIM domain-containing adaptor protein Hic-5 synaptically colocalizes and interacts with the dopamine transporter. J Neurosci. 2002, 22: 7045-7054.PubMed Carneiro AM, Ingram SL, Beaulieu JM, Sweeney A, Amara SG, Thomas SM, Caron MG, Torres GE: The multiple LIM domain-containing adaptor protein Hic-5 synaptically colocalizes and interacts with the dopamine transporter. J Neurosci. 2002, 22: 7045-7054.PubMed
29.
Zurück zum Zitat Carneiro AM, Blakely RD: Serotonin-, protein kinase C-, and Hic-5-associated redistribution of the platelet serotonin transporter. J Biol Chem. 2006, 281: 24769-24780. 10.1074/jbc.M603877200.CrossRefPubMed Carneiro AM, Blakely RD: Serotonin-, protein kinase C-, and Hic-5-associated redistribution of the platelet serotonin transporter. J Biol Chem. 2006, 281: 24769-24780. 10.1074/jbc.M603877200.CrossRefPubMed
30.
Zurück zum Zitat Bauman AL, Apparsundaram S, Ramamoorthy S, Wadzinski BE, Vaughan RA, Blakely RD: Cocaine and antidepressant-sensitive biogenic amine transporters exist in regulated complexes with protein phosphatase 2A. J Neurosci. 2000, 20: 7571-7578.PubMed Bauman AL, Apparsundaram S, Ramamoorthy S, Wadzinski BE, Vaughan RA, Blakely RD: Cocaine and antidepressant-sensitive biogenic amine transporters exist in regulated complexes with protein phosphatase 2A. J Neurosci. 2000, 20: 7571-7578.PubMed
31.
Zurück zum Zitat Wersinger C, Sidhu A: Partial regulation of serotonin transporter function by gamma-synuclein. Neurosci Lett. 2009, 453: 157-161. 10.1016/j.neulet.2009.02.033.PubMedCentralCrossRefPubMed Wersinger C, Sidhu A: Partial regulation of serotonin transporter function by gamma-synuclein. Neurosci Lett. 2009, 453: 157-161. 10.1016/j.neulet.2009.02.033.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Wersinger C, Rusnak M, Sidhu A: Modulation of the trafficking of the human serotonin transporter by human alpha-synuclein. Eur J Neurosci. 2006, 24: 55-64. 10.1111/j.1460-9568.2006.04900.x.CrossRefPubMed Wersinger C, Rusnak M, Sidhu A: Modulation of the trafficking of the human serotonin transporter by human alpha-synuclein. Eur J Neurosci. 2006, 24: 55-64. 10.1111/j.1460-9568.2006.04900.x.CrossRefPubMed
33.
Zurück zum Zitat Sur C, Betz H, Schloss P: A single serine residue controls the cation dependence of substrate transport by the rat serotonin transporter. Proc Natl Acad Sci USA. 1997, 94: 7639-7644. 10.1073/pnas.94.14.7639.PubMedCentralCrossRefPubMed Sur C, Betz H, Schloss P: A single serine residue controls the cation dependence of substrate transport by the rat serotonin transporter. Proc Natl Acad Sci USA. 1997, 94: 7639-7644. 10.1073/pnas.94.14.7639.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Iwata K, Izumo N, Matsuzaki H, Manabe T, Ishibashi Y, Ichitani Y, Yamada K, Thanseem I, Anitha A, Vasu MM, Shimmura C, Wakuda T, Kameno Y, Takahashi T, Iwata Y, Suzuki K, Nakamura K, Mori N: Vldlr overexpression causes hyperactivity in rats. Mol Autism. 2012, 3: 11-10.1186/2040-2392-3-11.PubMedCentralCrossRefPubMed Iwata K, Izumo N, Matsuzaki H, Manabe T, Ishibashi Y, Ichitani Y, Yamada K, Thanseem I, Anitha A, Vasu MM, Shimmura C, Wakuda T, Kameno Y, Takahashi T, Iwata Y, Suzuki K, Nakamura K, Mori N: Vldlr overexpression causes hyperactivity in rats. Mol Autism. 2012, 3: 11-10.1186/2040-2392-3-11.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Czesak M, Burns AM, Lenicov FR, Albert PR: Characterization of rat rostral raphe primary cultures: multiplex quantification of serotonergic markers. J Neurosci Methods. 2007, 164: 59-67. 10.1016/j.jneumeth.2007.04.002.CrossRefPubMed Czesak M, Burns AM, Lenicov FR, Albert PR: Characterization of rat rostral raphe primary cultures: multiplex quantification of serotonergic markers. J Neurosci Methods. 2007, 164: 59-67. 10.1016/j.jneumeth.2007.04.002.CrossRefPubMed
37.
Zurück zum Zitat Lautenschlager M, Holtje M, von Jagow B, Veh RW, Harms C, Bergk A, Dirnagl U, Ahnert-Hilger G, Hortnagl H: Serotonin uptake and release mechanisms in developing cultures of rat embryonic raphe neurons: age- and region-specific differences. Neuroscience. 2000, 99: 519-527. 10.1016/S0306-4522(00)00222-0.CrossRefPubMed Lautenschlager M, Holtje M, von Jagow B, Veh RW, Harms C, Bergk A, Dirnagl U, Ahnert-Hilger G, Hortnagl H: Serotonin uptake and release mechanisms in developing cultures of rat embryonic raphe neurons: age- and region-specific differences. Neuroscience. 2000, 99: 519-527. 10.1016/S0306-4522(00)00222-0.CrossRefPubMed
38.
Zurück zum Zitat Jorgensen S, Nielsen EO, Peters D, Dyhring T: Validation of a fluorescence-based high-throughput assay for the measurement of neurotransmitter transporter uptake activity. J Neurosci Methods. 2008, 169: 168-176. 10.1016/j.jneumeth.2007.12.004.CrossRefPubMed Jorgensen S, Nielsen EO, Peters D, Dyhring T: Validation of a fluorescence-based high-throughput assay for the measurement of neurotransmitter transporter uptake activity. J Neurosci Methods. 2008, 169: 168-176. 10.1016/j.jneumeth.2007.12.004.CrossRefPubMed
39.
Zurück zum Zitat Tsuruda PR, Yung J, Martin WJ, Chang R, Mai N, Smith JA: Influence of ligand binding kinetics on functional inhibition of human recombinant serotonin and norepinephrine transporters. J Pharmacol Toxicol Methods. 2010, 61: 192-204. 10.1016/j.vascn.2009.12.003.CrossRefPubMed Tsuruda PR, Yung J, Martin WJ, Chang R, Mai N, Smith JA: Influence of ligand binding kinetics on functional inhibition of human recombinant serotonin and norepinephrine transporters. J Pharmacol Toxicol Methods. 2010, 61: 192-204. 10.1016/j.vascn.2009.12.003.CrossRefPubMed
40.
Zurück zum Zitat Schmitt-Ulms G, Hansen K, Liu J, Cowdrey C, Yang J, DeArmond SJ, Cohen FE, Prusiner SB, Baldwin MA: Time-controlled transcardiac perfusion cross-linking for the study of protein interactions in complex tissues. Nat Biotechnol. 2004, 22: 724-731. 10.1038/nbt969.CrossRefPubMed Schmitt-Ulms G, Hansen K, Liu J, Cowdrey C, Yang J, DeArmond SJ, Cohen FE, Prusiner SB, Baldwin MA: Time-controlled transcardiac perfusion cross-linking for the study of protein interactions in complex tissues. Nat Biotechnol. 2004, 22: 724-731. 10.1038/nbt969.CrossRefPubMed
44.
Zurück zum Zitat Lord C, Rutter M, Le Couteur A: Autism Diagnostic Interview – Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994, 24: 659-685. 10.1007/BF02172145.CrossRefPubMed Lord C, Rutter M, Le Couteur A: Autism Diagnostic Interview – Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994, 24: 659-685. 10.1007/BF02172145.CrossRefPubMed
45.
Zurück zum Zitat Wechsler D: Wechsler Intelligence Scale for Children Third Edition manual. 1991, New York, NY: The Psychological Corporation Wechsler D: Wechsler Intelligence Scale for Children Third Edition manual. 1991, New York, NY: The Psychological Corporation
46.
Zurück zum Zitat Bookout AL, Mangelsdorf DJ: Quantitative real-time PCR protocol for analysis of nuclear receptor signaling pathways. Nucl Recept Signal. 2003, 1: e012-PubMedCentralCrossRefPubMed Bookout AL, Mangelsdorf DJ: Quantitative real-time PCR protocol for analysis of nuclear receptor signaling pathways. Nucl Recept Signal. 2003, 1: e012-PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Zhao CX, Slevin JT, Whiteheart SW: Cellular functions of NSF: not just SNAPs and SNAREs. Febs Letters. 2007, 581: 2140-2149. 10.1016/j.febslet.2007.03.032.PubMedCentralCrossRefPubMed Zhao CX, Slevin JT, Whiteheart SW: Cellular functions of NSF: not just SNAPs and SNAREs. Febs Letters. 2007, 581: 2140-2149. 10.1016/j.febslet.2007.03.032.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Zhao C, Smith EC, Whiteheart SW: Requirements for the catalytic cycle of the N-ethylmaleimide-Sensitive Factor (NSF). Biochim Biophys Acta. 2012, 1823: 159-171. 10.1016/j.bbamcr.2011.06.003.PubMedCentralCrossRefPubMed Zhao C, Smith EC, Whiteheart SW: Requirements for the catalytic cycle of the N-ethylmaleimide-Sensitive Factor (NSF). Biochim Biophys Acta. 2012, 1823: 159-171. 10.1016/j.bbamcr.2011.06.003.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Song I, Kamboj S, Xia J, Dong H, Liao D, Huganir RL: Interaction of the N-ethylmaleimide-sensitive factor with AMPA receptors. Neuron. 1998, 21: 393-400. 10.1016/S0896-6273(00)80548-6.CrossRefPubMed Song I, Kamboj S, Xia J, Dong H, Liao D, Huganir RL: Interaction of the N-ethylmaleimide-sensitive factor with AMPA receptors. Neuron. 1998, 21: 393-400. 10.1016/S0896-6273(00)80548-6.CrossRefPubMed
50.
Zurück zum Zitat Osten P, Srivastava S, Inman GJ, Vilim FS, Khatri L, Lee LM, States BA, Einheber S, Milner TA, Hanson PI, Ziff EB: The AMPA receptor GluR2 C terminus can mediate a reversible, ATP-dependent interaction with NSF and alpha- and beta-SNAPs. Neuron. 1998, 21: 99-110. 10.1016/S0896-6273(00)80518-8.CrossRefPubMed Osten P, Srivastava S, Inman GJ, Vilim FS, Khatri L, Lee LM, States BA, Einheber S, Milner TA, Hanson PI, Ziff EB: The AMPA receptor GluR2 C terminus can mediate a reversible, ATP-dependent interaction with NSF and alpha- and beta-SNAPs. Neuron. 1998, 21: 99-110. 10.1016/S0896-6273(00)80518-8.CrossRefPubMed
51.
Zurück zum Zitat Nishimune A, Isaac JT, Molnar E, Noel J, Nash SR, Tagaya M, Collingridge GL, Nakanishi S, Henley JM: NSF binding to GluR2 regulates synaptic transmission. Neuron. 1998, 21: 87-97. 10.1016/S0896-6273(00)80517-6.CrossRefPubMed Nishimune A, Isaac JT, Molnar E, Noel J, Nash SR, Tagaya M, Collingridge GL, Nakanishi S, Henley JM: NSF binding to GluR2 regulates synaptic transmission. Neuron. 1998, 21: 87-97. 10.1016/S0896-6273(00)80517-6.CrossRefPubMed
52.
Zurück zum Zitat Hanley JG, Khatri L, Hanson PI, Ziff EB: NSF ATPase and alpha-/beta-SNAPs disassemble the AMPA receptor-PICK1 complex. Neuron. 2002, 34: 53-67. 10.1016/S0896-6273(02)00638-4.CrossRefPubMed Hanley JG, Khatri L, Hanson PI, Ziff EB: NSF ATPase and alpha-/beta-SNAPs disassemble the AMPA receptor-PICK1 complex. Neuron. 2002, 34: 53-67. 10.1016/S0896-6273(02)00638-4.CrossRefPubMed
53.
Zurück zum Zitat Evers DM, Matta JA, Hoe HS, Zarkowsky D, Lee SH, Isaac JT, Pak DT: Plk2 attachment to NSF induces homeostatic removal of GluA2 during chronic overexcitation. Nat Neurosci. 2010, 13: 1199-1207. 10.1038/nn.2624.PubMedCentralCrossRefPubMed Evers DM, Matta JA, Hoe HS, Zarkowsky D, Lee SH, Isaac JT, Pak DT: Plk2 attachment to NSF induces homeostatic removal of GluA2 during chronic overexcitation. Nat Neurosci. 2010, 13: 1199-1207. 10.1038/nn.2624.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Cong M, Perry SJ, Hu LA, Hanson PI, Claing A, Lefkowitz RJ: Binding of the beta2 adrenergic receptor to N-ethylmaleimide-sensitive factor regulates receptor recycling. J Biol Chem. 2001, 276: 45145-45152. 10.1074/jbc.M106087200.CrossRefPubMed Cong M, Perry SJ, Hu LA, Hanson PI, Claing A, Lefkowitz RJ: Binding of the beta2 adrenergic receptor to N-ethylmaleimide-sensitive factor regulates receptor recycling. J Biol Chem. 2001, 276: 45145-45152. 10.1074/jbc.M106087200.CrossRefPubMed
55.
Zurück zum Zitat Chou WH, Wang D, McMahon T, Qi ZH, Song M, Zhang C, Shokat KM, Messing RO: GABAA receptor trafficking is regulated by protein kinase C(epsilon) and the N-ethylmaleimide-sensitive factor. J Neurosci. 2010, 30: 13955-13965. 10.1523/JNEUROSCI.0270-10.2010.PubMedCentralCrossRefPubMed Chou WH, Wang D, McMahon T, Qi ZH, Song M, Zhang C, Shokat KM, Messing RO: GABAA receptor trafficking is regulated by protein kinase C(epsilon) and the N-ethylmaleimide-sensitive factor. J Neurosci. 2010, 30: 13955-13965. 10.1523/JNEUROSCI.0270-10.2010.PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Kittler JT, Rostaing P, Schiavo G, Fritschy JM, Olsen R, Triller A, Moss SJ: The subcellular distribution of GABARAP and its ability to interact with NSF suggest a role for this protein in the intracellular transport of GABAA receptors. Mol Cell Neurosci. 2001, 18: 13-25. 10.1006/mcne.2001.1005.CrossRefPubMed Kittler JT, Rostaing P, Schiavo G, Fritschy JM, Olsen R, Triller A, Moss SJ: The subcellular distribution of GABARAP and its ability to interact with NSF suggest a role for this protein in the intracellular transport of GABAA receptors. Mol Cell Neurosci. 2001, 18: 13-25. 10.1006/mcne.2001.1005.CrossRefPubMed
57.
Zurück zum Zitat Leil TA, Chen ZW, Chang CS, Olsen RW: GABAA receptor-associated protein traffics GABAA receptors to the plasma membrane in neurons. J Neurosci. 2004, 24: 11429-11438. 10.1523/JNEUROSCI.3355-04.2004.CrossRefPubMed Leil TA, Chen ZW, Chang CS, Olsen RW: GABAA receptor-associated protein traffics GABAA receptors to the plasma membrane in neurons. J Neurosci. 2004, 24: 11429-11438. 10.1523/JNEUROSCI.3355-04.2004.CrossRefPubMed
58.
Zurück zum Zitat Huang SP, Brown BM, Craft CM: Visual Arrestin 1 acts as a modulator for N-ethylmaleimide-sensitive factor in the photoreceptor synapse. J Neurosci. 2010, 30: 9381-9391. 10.1523/JNEUROSCI.1207-10.2010.PubMedCentralCrossRefPubMed Huang SP, Brown BM, Craft CM: Visual Arrestin 1 acts as a modulator for N-ethylmaleimide-sensitive factor in the photoreceptor synapse. J Neurosci. 2010, 30: 9381-9391. 10.1523/JNEUROSCI.1207-10.2010.PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Jahn R, Scheller RH: SNAREs – engines for membrane fusion. Nat Rev Mol Cell Biol. 2006, 7: 631-643. 10.1038/nrm2002.CrossRefPubMed Jahn R, Scheller RH: SNAREs – engines for membrane fusion. Nat Rev Mol Cell Biol. 2006, 7: 631-643. 10.1038/nrm2002.CrossRefPubMed
60.
Zurück zum Zitat Faraj BA, Olkowski ZL, Jackson RT: Expression of a high-affinity serotonin transporter in human lymphocytes. Int J Immunopharmacol. 1994, 16: 561-567. 10.1016/0192-0561(94)90107-4.CrossRefPubMed Faraj BA, Olkowski ZL, Jackson RT: Expression of a high-affinity serotonin transporter in human lymphocytes. Int J Immunopharmacol. 1994, 16: 561-567. 10.1016/0192-0561(94)90107-4.CrossRefPubMed
61.
Zurück zum Zitat Fleming KG, Hohl TM, Yu RC, Muller SA, Wolpensinger B, Engel A, Engelhardt H, Brunger AT, Sollner TH, Hanson PI: A revised model for the oligomeric state of the N-ethylmaleimide-sensitive fusion protein, NSF. J Biol Chem. 1998, 273: 15675-15681. 10.1074/jbc.273.25.15675.CrossRefPubMed Fleming KG, Hohl TM, Yu RC, Muller SA, Wolpensinger B, Engel A, Engelhardt H, Brunger AT, Sollner TH, Hanson PI: A revised model for the oligomeric state of the N-ethylmaleimide-sensitive fusion protein, NSF. J Biol Chem. 1998, 273: 15675-15681. 10.1074/jbc.273.25.15675.CrossRefPubMed
62.
Zurück zum Zitat Hanson PI, Roth R, Morisaki H, Jahn R, Heuser JE: Structure and conformational changes in NSF and its membrane receptor complexes visualized by quick-freeze/deep-etch electron microscopy. Cell. 1997, 90: 523-535. 10.1016/S0092-8674(00)80512-7.CrossRefPubMed Hanson PI, Roth R, Morisaki H, Jahn R, Heuser JE: Structure and conformational changes in NSF and its membrane receptor complexes visualized by quick-freeze/deep-etch electron microscopy. Cell. 1997, 90: 523-535. 10.1016/S0092-8674(00)80512-7.CrossRefPubMed
63.
Zurück zum Zitat Rothman JE: Mechanisms of intracellular protein transport. Nature. 1994, 372: 55-63. 10.1038/372055a0.CrossRefPubMed Rothman JE: Mechanisms of intracellular protein transport. Nature. 1994, 372: 55-63. 10.1038/372055a0.CrossRefPubMed
64.
Zurück zum Zitat Hay JC, Scheller RH: SNAREs and NSF in targeted membrane fusion. Curr Opin Cell Biol. 1997, 9: 505-512. 10.1016/S0955-0674(97)80026-9.CrossRefPubMed Hay JC, Scheller RH: SNAREs and NSF in targeted membrane fusion. Curr Opin Cell Biol. 1997, 9: 505-512. 10.1016/S0955-0674(97)80026-9.CrossRefPubMed
65.
Zurück zum Zitat Nakamura K, Anitha A, Yamada K, Tsujii M, Iwayama Y, Hattori E, Toyota T, Suda S, Takei N, Iwata Y, Suzuki K, Matsuzaki H, Kawai M, Sekine Y, Tsuchiya KJ, Sugihara G, Ouchi Y, Sugiyama T, Yoshikawa T, Mori N: Genetic and expression analyses reveal elevated expression of syntaxin 1A (STX1A) in high functioning autism. Int J Neuropsychopharmacol. 2008, 11: 1073-1084. 10.1017/S1461145708009036.CrossRefPubMed Nakamura K, Anitha A, Yamada K, Tsujii M, Iwayama Y, Hattori E, Toyota T, Suda S, Takei N, Iwata Y, Suzuki K, Matsuzaki H, Kawai M, Sekine Y, Tsuchiya KJ, Sugihara G, Ouchi Y, Sugiyama T, Yoshikawa T, Mori N: Genetic and expression analyses reveal elevated expression of syntaxin 1A (STX1A) in high functioning autism. Int J Neuropsychopharmacol. 2008, 11: 1073-1084. 10.1017/S1461145708009036.CrossRefPubMed
66.
Zurück zum Zitat Nakamura K, Iwata Y, Anitha A, Miyachi T, Toyota T, Yamada S, Tsujii M, Tsuchiya KJ, Iwayama Y, Yamada K, Hattori E, Matsuzaki H, Matsumoto K, Suzuki K, Suda S, Takebayashi K, Takei N, Ichikawa H, Sugiyama T, Yoshikawa T, Mori N: Replication study of Japanese cohorts supports the role of STX1A in autism susceptibility. Prog Neuropsychopharmacol Biol Psychiatry. 2011, 35: 454-458. 10.1016/j.pnpbp.2010.11.033.CrossRefPubMed Nakamura K, Iwata Y, Anitha A, Miyachi T, Toyota T, Yamada S, Tsujii M, Tsuchiya KJ, Iwayama Y, Yamada K, Hattori E, Matsuzaki H, Matsumoto K, Suzuki K, Suda S, Takebayashi K, Takei N, Ichikawa H, Sugiyama T, Yoshikawa T, Mori N: Replication study of Japanese cohorts supports the role of STX1A in autism susceptibility. Prog Neuropsychopharmacol Biol Psychiatry. 2011, 35: 454-458. 10.1016/j.pnpbp.2010.11.033.CrossRefPubMed
67.
Zurück zum Zitat Kneussel M: Dynamic regulation of GABAA receptors at synaptic sites. Brain Res Rev. 2002, 39: 74-83. 10.1016/S0165-0173(02)00159-5.CrossRefPubMed Kneussel M: Dynamic regulation of GABAA receptors at synaptic sites. Brain Res Rev. 2002, 39: 74-83. 10.1016/S0165-0173(02)00159-5.CrossRefPubMed
68.
Zurück zum Zitat Hu VW, Frank BC, Heine S, Lee NH, Quackenbush J: Gene expression profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity of autism reveals differential regulation of neurologically relevant genes. BMC Genomics. 2006, 7: 118-10.1186/1471-2164-7-118.PubMedCentralCrossRefPubMed Hu VW, Frank BC, Heine S, Lee NH, Quackenbush J: Gene expression profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity of autism reveals differential regulation of neurologically relevant genes. BMC Genomics. 2006, 7: 118-10.1186/1471-2164-7-118.PubMedCentralCrossRefPubMed
69.
Zurück zum Zitat Sullivan PF, Fan C, Perou CM: Evaluating the comparability of gene expression in blood and brain. Am J Med Genet B Neuropsychiatr Genet. 2006, 141B: 261-268. 10.1002/ajmg.b.30272.CrossRefPubMed Sullivan PF, Fan C, Perou CM: Evaluating the comparability of gene expression in blood and brain. Am J Med Genet B Neuropsychiatr Genet. 2006, 141B: 261-268. 10.1002/ajmg.b.30272.CrossRefPubMed
70.
Zurück zum Zitat Sanyal S, Krishnan KS: Lethal comatose mutation in Drosophila reveals possible role for NSF in neurogenesis. Neuroreport. 2001, 12: 1363-1366. 10.1097/00001756-200105250-00015.CrossRefPubMed Sanyal S, Krishnan KS: Lethal comatose mutation in Drosophila reveals possible role for NSF in neurogenesis. Neuroreport. 2001, 12: 1363-1366. 10.1097/00001756-200105250-00015.CrossRefPubMed
71.
Zurück zum Zitat Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT: Asymmetric accumulation of hippocampal 7S SNARE complexes occurs regardless of kindling paradigm. Epilepsy Res. 2007, 73: 266-274. 10.1016/j.eplepsyres.2006.11.003.PubMedCentralCrossRefPubMed Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT: Asymmetric accumulation of hippocampal 7S SNARE complexes occurs regardless of kindling paradigm. Epilepsy Res. 2007, 73: 266-274. 10.1016/j.eplepsyres.2006.11.003.PubMedCentralCrossRefPubMed
72.
Zurück zum Zitat Brooks-Kayal A: Epilepsy and autism spectrum disorders: are there common developmental mechanisms?. Brain Dev. 2010, 32: 731-738. 10.1016/j.braindev.2010.04.010.CrossRefPubMed Brooks-Kayal A: Epilepsy and autism spectrum disorders: are there common developmental mechanisms?. Brain Dev. 2010, 32: 731-738. 10.1016/j.braindev.2010.04.010.CrossRefPubMed
73.
74.
Zurück zum Zitat Spence SJ, Schneider MT: The role of epilepsy and epileptiform EEGs in autism spectrum disorders. Pediatr Res. 2009, 65: 599-606. 10.1203/PDR.0b013e31819e7168.PubMedCentralCrossRefPubMed Spence SJ, Schneider MT: The role of epilepsy and epileptiform EEGs in autism spectrum disorders. Pediatr Res. 2009, 65: 599-606. 10.1203/PDR.0b013e31819e7168.PubMedCentralCrossRefPubMed
75.
Zurück zum Zitat Tuchman R, Alessandri M, Cuccaro M: Autism spectrum disorders and epilepsy: moving towards a comprehensive approach to treatment. Brain Dev. 2010, 32: 719-730. 10.1016/j.braindev.2010.05.007.CrossRefPubMed Tuchman R, Alessandri M, Cuccaro M: Autism spectrum disorders and epilepsy: moving towards a comprehensive approach to treatment. Brain Dev. 2010, 32: 719-730. 10.1016/j.braindev.2010.05.007.CrossRefPubMed
76.
Zurück zum Zitat Deonna T, Roulet E: Autistic spectrum disorder: evaluating a possible contributing or causal role of epilepsy. Epilepsia. 2006, 47 (Suppl 2): 79-82.CrossRefPubMed Deonna T, Roulet E: Autistic spectrum disorder: evaluating a possible contributing or causal role of epilepsy. Epilepsia. 2006, 47 (Suppl 2): 79-82.CrossRefPubMed
77.
78.
Zurück zum Zitat Rocha L, Lorigados-Pedre L, Orozco-Suarez S, Morales-Chacon L, Alonso-Vanegas M, Garcia-Maeso I, Villeda-Hernandez J, Osorio-Rico L, Estupinan B, Quintana C: Autoradiography reveals selective changes in serotonin binding in neocortex of patients with temporal lobe epilepsy. Prog Neuropsychopharmacol Biol Psychiatry. 2007, 31: 1208-1218. 10.1016/j.pnpbp.2007.04.014.CrossRefPubMed Rocha L, Lorigados-Pedre L, Orozco-Suarez S, Morales-Chacon L, Alonso-Vanegas M, Garcia-Maeso I, Villeda-Hernandez J, Osorio-Rico L, Estupinan B, Quintana C: Autoradiography reveals selective changes in serotonin binding in neocortex of patients with temporal lobe epilepsy. Prog Neuropsychopharmacol Biol Psychiatry. 2007, 31: 1208-1218. 10.1016/j.pnpbp.2007.04.014.CrossRefPubMed
79.
Zurück zum Zitat Cupello A, Audenino D, Scarrone S, Fornaro M, Gatta E, Fornaro P, Albano C: Epileptic seizures but not pseudoseizures are associated with decreased density of the serotonin transporter in blood platelet membranes. Neurochem Res. 2008, 33: 2263-2268. 10.1007/s11064-008-9708-7.CrossRefPubMed Cupello A, Audenino D, Scarrone S, Fornaro M, Gatta E, Fornaro P, Albano C: Epileptic seizures but not pseudoseizures are associated with decreased density of the serotonin transporter in blood platelet membranes. Neurochem Res. 2008, 33: 2263-2268. 10.1007/s11064-008-9708-7.CrossRefPubMed
80.
Zurück zum Zitat Cupello A, Favale E, Audenino D, Scarrone S, Gastaldi S, Albano C: Decrease of serotonin transporters in blood platelets after epileptic seizures. Neurochem Res. 2005, 30: 425-428. 10.1007/s11064-005-2676-2.CrossRefPubMed Cupello A, Favale E, Audenino D, Scarrone S, Gastaldi S, Albano C: Decrease of serotonin transporters in blood platelets after epileptic seizures. Neurochem Res. 2005, 30: 425-428. 10.1007/s11064-005-2676-2.CrossRefPubMed
81.
Zurück zum Zitat Favale E, Audenino D, Cocito L, Albano C: The anticonvulsant effect of citalopram as an indirect evidence of serotonergic impairment in human epileptogenesis. Seizure. 2003, 12: 316-318. 10.1016/S1059-1311(02)00315-1.CrossRefPubMed Favale E, Audenino D, Cocito L, Albano C: The anticonvulsant effect of citalopram as an indirect evidence of serotonergic impairment in human epileptogenesis. Seizure. 2003, 12: 316-318. 10.1016/S1059-1311(02)00315-1.CrossRefPubMed
82.
Zurück zum Zitat Favale E, Rubino V, Mainardi P, Lunardi G, Albano C: Anticonvulsant effect of fluoxetine in humans. Neurology. 1995, 45: 1926-1927. 10.1212/WNL.45.10.1926.CrossRefPubMed Favale E, Rubino V, Mainardi P, Lunardi G, Albano C: Anticonvulsant effect of fluoxetine in humans. Neurology. 1995, 45: 1926-1927. 10.1212/WNL.45.10.1926.CrossRefPubMed
83.
Zurück zum Zitat Mendez MA, Horder J, Myers J, Coghlan S, Stokes P, Erritzoe D, Howes O, Lingford-Hughes A, Murphy D, Nutt D: The brain GABA-benzodiazepine receptor alpha-5 subtype in autism spectrum disorder: A pilot [11C]Ro15-4513 positron emission tomography study. Neuropharmacology. 2012, 68: 195-201.PubMedCentralCrossRefPubMed Mendez MA, Horder J, Myers J, Coghlan S, Stokes P, Erritzoe D, Howes O, Lingford-Hughes A, Murphy D, Nutt D: The brain GABA-benzodiazepine receptor alpha-5 subtype in autism spectrum disorder: A pilot [11C]Ro15-4513 positron emission tomography study. Neuropharmacology. 2012, 68: 195-201.PubMedCentralCrossRefPubMed
84.
Zurück zum Zitat Purcell AE, Jeon OH, Zimmerman AW, Blue ME, Pevsner J: Postmortem brain abnormalities of the glutamate neurotransmitter system in autism. Neurology. 2001, 57: 1618-1628. 10.1212/WNL.57.9.1618.CrossRefPubMed Purcell AE, Jeon OH, Zimmerman AW, Blue ME, Pevsner J: Postmortem brain abnormalities of the glutamate neurotransmitter system in autism. Neurology. 2001, 57: 1618-1628. 10.1212/WNL.57.9.1618.CrossRefPubMed
Metadaten
Titel
N-ethylmaleimide-sensitive factor interacts with the serotonin transporter and modulates its trafficking: implications for pathophysiology in autism
verfasst von
Keiko Iwata
Hideo Matsuzaki
Taro Tachibana
Koji Ohno
Saori Yoshimura
Hironori Takamura
Kohei Yamada
Shinsuke Matsuzaki
Kazuhiko Nakamura
Kenji J Tsuchiya
Kaori Matsumoto
Masatsugu Tsujii
Toshirou Sugiyama
Taiichi Katayama
Norio Mori
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Molecular Autism / Ausgabe 1/2014
Elektronische ISSN: 2040-2392
DOI
https://doi.org/10.1186/2040-2392-5-33

Weitere Artikel der Ausgabe 1/2014

Molecular Autism 1/2014 Zur Ausgabe