Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
Add more filters

Publication year range
1.
Cell ; 141(4): 618-31, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20478254

ABSTRACT

Adaptation to different levels of illumination is central to the function of the retina. Here, we demonstrate that levels of the miR-183/96/182 cluster, miR-204, and miR-211 are regulated by different light levels in the mouse retina. Concentrations of these microRNAs were downregulated during dark adaptation and upregulated in light-adapted retinas, with rapid decay and increased transcription being responsible for the respective changes. We identified the voltage-dependent glutamate transporter Slc1a1 as one of the miR-183/96/182 targets in photoreceptor cells. We found that microRNAs in retinal neurons decay much faster than microRNAs in nonneuronal cells. The high turnover is also characteristic of microRNAs in hippocampal and cortical neurons, and neurons differentiated from ES cells in vitro. Blocking activity reduced turnover of microRNAs in neuronal cells while stimulation with glutamate accelerated it. Our results demonstrate that microRNA metabolism in neurons is higher than in most other cells types and linked to neuronal activity.


Subject(s)
MicroRNAs/metabolism , Neurons/metabolism , Animals , Dark Adaptation , Down-Regulation , Embryonic Stem Cells , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Mice , Photoreceptor Cells, Vertebrate/metabolism , Retinal Neurons/metabolism , Up-Regulation
2.
Horm Behav ; 162: 105548, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636205

ABSTRACT

Thyroid hormones are crucial for brain development and their deficiency during fetal and postnatal periods can lead to mood and cognitive disorders. We aimed to examine the consequences of thyroid hormone deficiency on anxiety-related behaviors and protein expression of hippocampal glutamate transporters in congenital hypothyroid male offspring rats. Possible beneficial effects of treadmill exercise have also been examined. Congenital hypothyroidism was induced by adding propylthiouracil (PTU) to drinking water of pregnant Wistar rats from gestational day 6 until the end of the weaning period (postnatal day 28). Next, following 4 weeks of treadmill exercise (5 days per week), anxiety-related behaviors were examined using elevated plus maze (EPM) and light/dark box tests. Thereafter, protein expression of astrocytic (GLAST and GLT-1) and neuronal (EAAC1) glutamate transporters were measured in the hippocampus by immunoblotting. Hypothyroid rats showed decreased anxiety-like behavior, as measured by longer time spent in the open arms of the EPM and in the light area of the light/dark box, compared to control rats. Hypothyroid rats had significantly higher GLAST and GLT-1 and lower EAAC1 protein levels in the hippocampus than did the euthyroid rats. Following exercise, anxiety levels decreased in the euthyroid group while protein expression of EAAC1 increased and returned to normal levels in the hypothyroid group. Our findings indicate that thyroid hormone deficiency was associated with alterations in protein expression of glutamate transporters in the hippocampus. Up-regulation of hippocampal GLAST and GLT-1 could be at least one of the mechanisms associated with the anxiolytic effects of congenital hypothyroidism.


Subject(s)
Anxiety , Congenital Hypothyroidism , Excitatory Amino Acid Transporter 2 , Hippocampus , Rats, Wistar , Animals , Male , Hippocampus/metabolism , Anxiety/metabolism , Anxiety/etiology , Rats , Female , Congenital Hypothyroidism/metabolism , Pregnancy , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 2/genetics , Thyroid Hormones/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 3/metabolism , Excitatory Amino Acid Transporter 3/genetics , Behavior, Animal/physiology , Propylthiouracil , Amino Acid Transport System X-AG/metabolism , Amino Acid Transport System X-AG/genetics , Prenatal Exposure Delayed Effects/metabolism
3.
Mol Psychiatry ; 27(3): 1515-1526, 2022 03.
Article in English | MEDLINE | ID: mdl-35058566

ABSTRACT

Obsessive-compulsive disorder (OCD) is a disabling condition that often begins in childhood. Genetic studies in OCD have pointed to SLC1A1, which encodes the neuronal glutamate transporter EAAT3, with evidence suggesting that increased expression contributes to risk. In mice, midbrain Slc1a1 expression supports repetitive behavior in response to dopaminergic agonists, aligning with neuroimaging and pharmacologic challenge studies that have implicated the dopaminergic system in OCD. These findings suggest that Slc1a1 may contribute to compulsive behavior through altered dopaminergic transmission; however, this theory has not been mechanistically tested. To examine the developmental impact of Slc1a1 overexpression on compulsive-like behaviors, we, therefore, generated a novel mouse model to perform targeted, reversible overexpression of Slc1a1 in dopaminergic neurons. Mice with life-long overexpression of Slc1a1 showed a significant increase in amphetamine (AMPH)-induced stereotypy and hyperlocomotion. Single-unit recordings demonstrated that Slc1a1 overexpression was associated with increased firing of dopaminergic neurons. Furthermore, dLight1.1 fiber photometry showed that these behavioral abnormalities were associated with increased dorsal striatum dopamine release. In contrast, no impact of overexpression was observed on anxiety-like behaviors or SKF-38393-induced grooming. Importantly, overexpression solely in adulthood failed to recapitulate these behavioral phenotypes, suggesting that overexpression during development is necessary to generate AMPH-induced phenotypes. However, doxycycline-induced reversal of Slc1a1/EAAT3 overexpression in adulthood normalized both the increased dopaminergic firing and AMPH-induced responses. These data indicate that the pathologic effects of Slc1a1/EAAT3 overexpression on dopaminergic neurotransmission and AMPH-induced stereotyped behavior are developmentally mediated, and support normalization of EAAT3 activity as a potential treatment target for basal ganglia-mediated repetitive behaviors.


Subject(s)
Excitatory Amino Acid Transporter 3 , Obsessive-Compulsive Disorder , Animals , Compulsive Behavior , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Mice , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/metabolism , Stereotyped Behavior
4.
Int J Mol Sci ; 22(9)2021 May 09.
Article in English | MEDLINE | ID: mdl-34065042

ABSTRACT

Glutathione (GSH) is the most abundant non-protein thiol, and plays crucial roles in the antioxidant defense system and the maintenance of redox homeostasis in neurons. GSH depletion in the brain is a common finding in patients with neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, and can cause neurodegeneration prior to disease onset. Excitatory amino acid carrier 1 (EAAC1), a sodium-dependent glutamate/cysteine transporter that is selectively present in neurons, plays a central role in the regulation of neuronal GSH production. The expression of EAAC1 is posttranslationally controlled by the glutamate transporter-associated protein 3-18 (GTRAP3-18) or miR-96-5p in neurons. The regulatory mechanism of neuronal GSH production mediated by EAAC1 may be a new target in therapeutic strategies for these neurodegenerative diseases. This review describes the regulatory mechanism of neuronal GSH production and its potential therapeutic application in the treatment of neurodegenerative diseases.


Subject(s)
Brain/metabolism , Glutathione/metabolism , Animals , Antioxidants/metabolism , Biomarkers , Brain/drug effects , Disease Management , Disease Susceptibility , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Gene Expression Regulation , Glutamate Plasma Membrane Transport Proteins/metabolism , Glutathione/pharmacology , Glutathione/therapeutic use , Humans , Metabolic Networks and Pathways , Microglia/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
5.
J Biol Chem ; 294(32): 12180-12190, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31235523

ABSTRACT

Plasma membrane-associated glutamate transporters play a key role in signaling by the major excitatory neurotransmitter glutamate. Uphill glutamate uptake into cells is energetically driven by coupling to co-transport of three Na+ ions. In exchange, one K+ ion is counter-transported. Currently accepted transport mechanisms assume that Na+ and K+ effects are exclusive, resulting from competition of these cations at the binding level. Here, we used electrophysiological analysis to test the effects of K+ and Na+ on neuronal glutamate transporter excitatory amino acid carrier 1 (EAAC1; the rat homologue of human excitatory amino acid transporter 3 (EAAT3)). Unexpectedly, extracellular K+ application to EAAC1 induced anion current, but only in the presence of Na+ This result could be explained with a K+/Na+ co-binding state in which the two cations simultaneously bind to the transporter. We obtained further evidence for this co-binding state, and its anion conductance, by analyzing transient currents when Na+ was exchanged for K+ and effects of the [K+]/[Na+] ratio on glutamate affinity. Interestingly, we observed the K+/Na+ co-binding state not only in EAAC1 but also in the subtypes EAAT1 and -2, which, unlike EAAC1, conducted anions in response to K+ only. We incorporated these experimental findings in a revised transport mechanism, including the K+/Na+ co-binding state and the ability of K+ to activate anion current. Overall, these results suggest that differentiation between Na+ and K+ does not occur at the binding level but is conferred by coupling of cation binding to conformational changes. These findings have implications also for other exchangers.


Subject(s)
Excitatory Amino Acid Transporter 3/metabolism , Potassium/metabolism , Sodium/metabolism , Binding, Competitive , Cations/chemistry , Excitatory Amino Acid Transporter 3/chemistry , Excitatory Amino Acid Transporter 3/genetics , Glutamic Acid/chemistry , Glutamic Acid/metabolism , HEK293 Cells , Humans , Kinetics , Patch-Clamp Techniques , Potassium/chemistry , Protein Binding , Sodium/chemistry
6.
Proc Natl Acad Sci U S A ; 114(22): 5719-5724, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28507136

ABSTRACT

Obsessive-compulsive disorder (OCD) is a chronic, disabling condition with inadequate treatment options that leave most patients with substantial residual symptoms. Structural, neurochemical, and behavioral findings point to a significant role for basal ganglia circuits and for the glutamate system in OCD. Genetic linkage and association studies in OCD point to SLC1A1, which encodes the neuronal glutamate/aspartate/cysteine transporter excitatory amino acid transporter 3 (EAAT3)/excitatory amino acid transporter 1 (EAAC1). However, no previous studies have investigated EAAT3 in basal ganglia circuits or in relation to OCD-related behavior. Here, we report a model of Slc1a1 loss based on an excisable STOP cassette that yields successful ablation of EAAT3 expression and function. Using amphetamine as a probe, we found that EAAT3 loss prevents expected increases in (i) locomotor activity, (ii) stereotypy, and (iii) immediate early gene induction in the dorsal striatum following amphetamine administration. Further, Slc1a1-STOP mice showed diminished grooming in an SKF-38393 challenge experiment, a pharmacologic model of OCD-like grooming behavior. This reduced grooming is accompanied by reduced dopamine D1 receptor binding in the dorsal striatum of Slc1a1-STOP mice. Slc1a1-STOP mice also exhibit reduced extracellular dopamine concentrations in the dorsal striatum both at baseline and following amphetamine challenge. Viral-mediated restoration of Slc1a1/EAAT3 expression in the midbrain but not in the striatum results in partial rescue of amphetamine-induced locomotion and stereotypy in Slc1a1-STOP mice, consistent with an impact of EAAT3 loss on presynaptic dopaminergic function. Collectively, these findings indicate that the most consistently associated OCD candidate gene impacts basal ganglia-dependent repetitive behaviors.


Subject(s)
Basal Ganglia/physiology , Excitatory Amino Acid Transporter 3/genetics , Motor Activity/genetics , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/physiopathology , Amphetamines/pharmacology , Animals , Cell Line , Central Nervous System Stimulants/pharmacology , Dopamine/metabolism , Glutamic Acid/metabolism , Grooming/physiology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Receptors, Dopamine D1/metabolism , Reflex, Startle/physiology
7.
J Biol Chem ; 293(37): 14200-14209, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30026234

ABSTRACT

In the brain, glutamate transporters terminate excitatory neurotransmission by removing this neurotransmitter from the synapse via cotransport with three sodium ions into the surrounding cells. Structural studies have identified the binding sites of the three sodium ions in glutamate transporters. The residue side-chains directly interact with the sodium ions at the Na1 and Na3 sites and are fully conserved from archaeal to eukaryotic glutamate transporters. The Na2 site is formed by three main-chain oxygens on the extracellular reentrant hairpin loop HP2 and one on transmembrane helix 7. A glycine residue on HP2 is located closely to the three main-chain oxygens in all glutamate transporters, except for the astroglial transporter GLT-1, which has a serine residue at that position. Unlike for WT GLT-1, substitution of the serine residue to glycine enables sustained glutamate transport also when sodium is replaced by lithium. Here, using functional and simulation studies, we studied the role of this serine/glycine switch on cation selectivity of substrate transport. Our results indicate that the side-chain oxygen of the serine residues can form a hydrogen bond with a main-chain oxygen on transmembrane helix 7. This leads to an expansion of the Na2 site such that water can participate in sodium coordination at Na2. Furthermore, we found other molecular determinants of cation selectivity on the nearby HP1 loop. We conclude that subtle changes in the composition of the two reentrant hairpin loops determine the cation specificity of acidic amino acid transport by glutamate transporters.


Subject(s)
Excitatory Amino Acid Transporter 2/metabolism , Sodium/metabolism , Binding Sites , Cations/metabolism , Excitatory Amino Acid Transporter 2/chemistry , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 3/chemistry , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Glycine/metabolism , HeLa Cells , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Serine/metabolism
8.
J Clin Pharm Ther ; 44(1): 39-48, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30315580

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Obsessive-compulsive disorder (OCD) is a chronic neuropsychiatric disorder. Selective serotonin reuptake inhibitors (SSRIs) are the first line of medication for OCD treatment; however, 40%-60% of patients with OCD do not respond to SSRIs adequately. There are growing pieces of evidence which suggest a significant role for the glutamatergic system in the genesis of OCD and its consequent treatment. In the present study, we aimed to assess the association of SLC1A1 polymorphisms (rs301430, rs2228622 and rs3780413) with OCD and its clinical characteristics, as well as the importance of these SNPs in the response of OCD patients to SSRI pharmacotherapy. METHODS: Sample study consisted of 243 OCD cases and 221 control subjects. Patients were treated 12 weeks with fluvoxamine (daily dose: 150-300 mg). Based on the reduction in obsessive and compulsive severity scores using Y-BOCS severity scale, patients were classified as responders, non-responders and refractory. A total of 239, 228 and 215 patients were genotyped for rs301430, rs2228622 and rs3780413, respectively, by the means of PCR-RFLP. RESULTS AND DISCUSSION: No association was detected between SLC1A1 SNPs and OCD, except an association between the familial form of the disease in males with rs2228622 (P = 0.033). The results of pharmacogenetic studies revealed the associations of two SLC1A1 SNPs, rs2228622 (P = 0.031) and rs3780413 (P = 0.008), with treatment response. WHAT IS NEW AND CONCLUSION: Results of the current study suggest a role for the glutamate transporter in OCD treatment response with SSRIs which should encourage researchers to further investigate the importance of glutamate transporter in OCD pharmacogenetics.


Subject(s)
Excitatory Amino Acid Transporter 3/genetics , Fluvoxamine/therapeutic use , Obsessive-Compulsive Disorder/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Female , Fluvoxamine/administration & dosage , Genetic Predisposition to Disease , Genotype , Humans , Iran , Male , Middle Aged , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/physiopathology , Pharmacogenomic Testing , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Polymorphism, Single Nucleotide , Selective Serotonin Reuptake Inhibitors/administration & dosage , Severity of Illness Index , Sex Factors , Treatment Outcome , Young Adult
9.
Turk J Med Sci ; 49(2): 531-537, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30862152

ABSTRACT

Background/aim: This study aimed to comparatively analyze the expression levels of the SLC1A1 gene in renal specimens from tumors and adjacent healthy kidney tissues of patients with clear cell renal cell carcinoma (ccRCC). Materials and methods: Nineteen patients diagnosed with ccRCC were included in the study. The expression levels of the SLC1A1 and GAPDH genes were measured in tumor and formalin-fixed paraffin-embedded (FFPE) tissue specimens from the adjacent healthy kidney of each subject. Via the GEPIA database, the distribution of SLC1A1 gene expressions in ccRCC and healthy kidney tissues was obtained. The relative expression of SLC1A1 was evaluated for the association with the clinical parameters of the patients. Results: The expression of the SLC1A1 gene was significantly higher in males than females (P = 0.029). Also, there were statistically significant associations between stages II­IV and Fuhrman grades 2­4 with respect to SLC1A1 gene expression (P < 0.001 for both). Moreover, low levels of red blood cell and hemoglobin counts were significantly associated with the SLC1A1 expression (P < 0.001 and P = 0.005, respectively). The expression of the SLC1A1 gene in tumor tissues increased approximately 3 times compared with normal kidney tissues (P < 0.05). According to the GEPIA database, SLC1A1 gene expression is significantly higher in ccRCC patients than healthy persons (P = 0.01). Conclusion: The change in the expression of SLC1A1 may be crucial for ccRCC pathophysiology.


Subject(s)
Carcinoma, Renal Cell/genetics , Excitatory Amino Acid Transporter 3/genetics , Kidney Neoplasms/genetics , Adult , Analysis of Variance , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/pathology , Male , Middle Aged
10.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 317-323, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28962897

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system, characterized by chronic inflammation, demyelination and scarring as well as a broad spectrum of signs and symptoms. MicroRNA plays pivotal roles in cellular and developmental processes by regulating gene expression at the post-transcriptional level. Increasing evidence suggests the involvement of microRNAs in the pathogenesis of neurodegenerative diseases, including MS. We have already found that the expression of a specific miRNA, hsa-mir-26a-5p (miR-26a), changed during INF-ß treatment in responder Relapsing-Remitting MS patients. Functional annotations of mir-26a targets revealed that a number of genes were implicated in Glutamate Receptor Signaling pathway, which is notoriously altered in neurodegenerative diseases as MS. In this study, the different potential targets were subjected to a validation test based on luciferase reporter constructs transfected in an oligodendroglial cell line. In this functional screening, miR-26a was able to interact with SLC1A1 3' UTR suppressing the reporter activity. Transfection of a miR-26a mimic was then shown to decrease the endogenous SLC1A1 mRNA. Afterward, we have evaluated in blood platelets from interferon-ß treated Multiple Sclerosis patients the expression of miR-26a and SLC1A1, finding not only their converse expression, but also a responsiveness to interferon-ß therapy. Overall, these data suggest that mir-26a and SLC1A1 may play a role in the MS pathogenesis, and may be potential targets for the development of new biomarkers and/or therapeutic tools.


Subject(s)
Excitatory Amino Acid Transporter 3/genetics , MicroRNAs/physiology , Multiple Sclerosis/genetics , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Signal Transduction/drug effects , Signal Transduction/genetics
11.
Alcohol Clin Exp Res ; 42(8): 1444-1453, 2018 08.
Article in English | MEDLINE | ID: mdl-29786868

ABSTRACT

BACKGROUND: Alcohol use disorders (AUDs) are influenced by complex interactions between the genetics of the individual and their environment. We have previously identified hundreds of polygenic genetic variants between the selectively bred high- and low-alcohol drinking (HAD and LAD) rat lines. Here, we report allele-specific expression (ASE) differences, between the HAD2 and LAD2 rat lines. METHODS: The HAD2 and LAD2 rats, which have been sequenced, were reciprocally crossed to generate 10 litters of F1 progeny. For 5 of these litters, the sire was HAD2, and for the other 5 litters, the sire was a LAD2. From these 10 litters, 2 males and 2 females were picked from each F1 litter (N = 40 total). The F1 pups were divided, balancing for sex and direction of cross, into an alcohol (15%) versus a water control group. Alcohol drinking started in the middle of adolescence (~postnatal day 35) and lasted 9 weeks. At the end of these treatments, rats were euthanized, the nucleus accumbens was dissected, and RNA was processed for RNA-sequencing and ASE analyses. RESULTS: Analyses revealed that adolescent ethanol (EtOH) drinking, individual EtOH drinking levels, parentage, and sex-of-animal affected ASEs of about 300 genes. The identified genes included those associated with EtOH metabolism (e.g., Aldh2); neuromodulatory function (e.g., Cckbr, Slc6a7, and Slc1a1); ion channel activity (e.g., Kcnc3); and other synaptic and epigenetic functions. CONCLUSIONS: These data indicate that EtOH drinking differentially amplified paternal versus maternal allelic contribution to the transcriptome. We hypothesize that this was due, at least in part, to EtOH-induced changes in cis-regulation of polymorphisms previously identified between the HAD2 and LAD2 rat lines. This report highlights the complexity of gene-by-environment interactions mediating a genetic predisposition for, and/or the active development of, AUDs.


Subject(s)
Alcohol Drinking/genetics , Alcoholism/genetics , Alleles , Gene Expression Regulation/drug effects , Genotype , Sex Factors , Alcohol Drinking/physiopathology , Alcoholism/physiopathology , Aldehyde Dehydrogenase, Mitochondrial/genetics , Amino Acid Transport Systems, Neutral/genetics , Animals , Breeding/methods , Crosses, Genetic , Ethanol/metabolism , Excitatory Amino Acid Transporter 3/genetics , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene-Environment Interaction , Male , Rats , Shaw Potassium Channels/genetics
12.
J Biol Chem ; 291(22): 11852-64, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27044739

ABSTRACT

Glutamate transporters actively take up glutamate into the cell, driven by the co-transport of sodium ions down their transmembrane concentration gradient. It was proposed that glutamate binds to its binding site and is subsequently transported across the membrane in the negatively charged form. With the glutamate binding site being located partially within the membrane domain, the possibility has to be considered that glutamate binding is dependent on the transmembrane potential and, thus, is electrogenic. Experiments presented in this report test this possibility. Rapid application of glutamate to the wild-type glutamate transporter subtype EAAC1 (excitatory amino acid carrier 1) through photo-release from caged glutamate generated a transient inward current, as expected for the electrogenic inward movement of co-transported Na(+) In contrast, glutamate application to a transporter with the mutation A334E induced transient outward current, consistent with movement of negatively charged glutamate into its binding site within the dielectric of the membrane. These results are in agreement with electrostatic calculations, predicting a valence for glutamate binding of -0.27. Control experiments further validate and rule out other possible explanations for the transient outward current. Electrogenic glutamate binding can be isolated in the mutant glutamate transporter because reactions, such as glutamate translocation and/or Na(+) binding to the glutamate-bound state, are inhibited by the A334E substitution. Electrogenic glutamate binding has to be considered together with other voltage-dependent partial reactions to cooperatively determine the voltage dependence of steady-state glutamate uptake and glutamate buffering at the synapse.


Subject(s)
Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Sodium/metabolism , Binding Sites , Biological Transport , Electrophysiology , Excitatory Amino Acid Transporter 3/chemistry , Excitatory Amino Acid Transporter 3/genetics , Humans , Kinetics , Membrane Potentials , Molecular Dynamics Simulation , Mutation/genetics , Patch-Clamp Techniques , Protein Conformation , Substrate Specificity
13.
Immunology ; 150(2): 162-171, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27649685

ABSTRACT

Although type 1 diabetes (T1D) is a T-cell-mediated disease in the effector stage, the mechanism behind the initial beta cell assault is less understood. Metabolomic differences, including elevated levels of glutamic acid, have been observed in patients with T1D before disease onset, as well as in pre-diabetic non-obese diabetic (NOD) mice. Increased levels of glutamic acid damage both neurons and beta cells, implying that this could contribute to the initial events of T1D pathogenesis. We investigated the underlying genetic factors and consequences of the increased levels of glutamic acid in NOD mice. Serum glutamic acid levels from a (NOD×B6)F2 cohort (n = 182) were measured. By genome-wide and Idd region targeted microsatellite mapping, genetic association was detected for six regions including Idd2, Idd4 and Idd22. In silico analysis of potential enzymes and transporters located in and around the mapped regions that are involved in glutamic acid metabolism consisted of alanine aminotransferase, glutamic-oxaloacetic transaminase, aldehyde dehydrogenase 18 family, alutamyl-prolyl-tRNA synthetase, glutamic acid transporters GLAST and EAAC1. Increased EAAC1 protein expression was observed in lysates from livers of NOD mice compared with B6 mice. Functional consequence of the elevated glutamic acid level in NOD mice was tested by culturing NOD. Rag2-/- Langerhans' islets with glutamic acid. Induction of apoptosis of the islets was detected upon glutamic acid challenge using TUNEL assay. Our results support the notion that a dysregulated metabolome could contribute to the initiation of T1D. We suggest that targeting of the increased glutamic acid in pre-diabetic patients could be used as a potential therapy.


Subject(s)
Apoptosis/genetics , Diabetes Mellitus, Type 1/genetics , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Islets of Langerhans/physiology , Alanine Transaminase/genetics , Animals , Aspartate Aminotransferases/genetics , DNA-Binding Proteins/genetics , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 3/genetics , Humans , Metabolome/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Microsatellite Repeats/genetics , Polymorphism, Single Nucleotide
14.
Hum Genet ; 136(6): 693-703, 2017 06.
Article in English | MEDLINE | ID: mdl-28324217

ABSTRACT

Hot water epilepsy is sensory epilepsy, wherein seizures are triggered by an unusual stimulus: contact with hot water. Although genetic factors contribute to the etiology of hot water epilepsy, molecular underpinnings of the disorder remain largely unknown. We aimed to identify the molecular genetic basis of the disorder by studying families with two or more of their members affected with hot water epilepsy. Using a combination of genome-wide linkage mapping and whole exome sequencing, a missense variant was identified in SLC1A1 in a three-generation family. Further, we examined SLC1A1in probands of 98 apparently unrelated HWE families with positive histories of seizures provoked by contact with hot water. In doing so, we found three rare variants, p.Asp174Asn, p.Val251Ile and p.Ile304Met in the gene. SLC1A1 is a neuronal glutamate transporter which limits excitotoxicity and its loss-of-function leads to age-dependent neurodegeneration. We examined functional attributes of the variants in cultured mammalian cells. All three non-synonymous variants affected glutamate uptake, exhibited altered glutamate kinetics and anion conductance properties of SLC1A1. These observations provide insights into the molecular basis of hot water epilepsy and show the role of SLC1A1 variants in this intriguing neurobehavioral disorder.


Subject(s)
Epilepsy/genetics , Excitatory Amino Acid Transporter 3/genetics , Hot Temperature , Amino Acid Sequence , Animals , Epilepsy/etiology , Excitatory Amino Acid Transporter 3/chemistry , Exome , Female , Genetic Linkage , Humans , Male , Patch-Clamp Techniques , Pedigree , Sequence Homology, Amino Acid , Water
15.
Amino Acids ; 49(9): 1587-1599, 2017 09.
Article in English | MEDLINE | ID: mdl-28623466

ABSTRACT

In-feed antibiotics have been commonly used to promote the growth performance of piglets. The antibiotics can increase protein utilization, but the underlying mechanism is largely unknown. The present study investigated the effects of in-feed antibiotics on intestinal AA transporters and receptors to test the hypothesis that the alteration of circulating AA profiles may be concomitant with the change of intestinal AA transporters and receptors. Sixteen litters of piglets at day 7 started to receive creep feed with (Antibiotic) or without (Control) antibiotic. Piglets were weaned at day 23 after birth, and fed the same diets until day 42. In-feed antibiotics did not affect the BW of 23-day-old (P = 0.248), or 42-day-old piglets (P = 0.089), but increased the weight gain to feed ratio from day 23 to 42 (P = 0.020). At day 42 after birth, antibiotic treatment increased the concentrations of most AAs in serum (P < 0.05), and decreased the concentrations of most AAs in jejunal and ileal digesta. Antibiotics upregulated (P < 0.05) the mRNA expression levels for jejunal AAs transporters (CAT1, EAAC1, ASCT2, y+LAT1), peptide transporters (PepT1), and Na+-K+-ATPase (ATP1A1), and ileal AA transporters (ASCT2, y+LAT1, b0,+AT, and B0AT1), and ATP1A1. The antibiotics also upregulated the mRNA expression of jejunal AAs receptors T1R3 and CaSR, and ileal T1R3. Protein expression levels for jejunal AA transporters (EAAC1, b0,+AT, and ASCT2) and PepT1 were also upregulated. Correlation analysis revealed that the alterations of AA profiles in serum after the in-feed antibiotics were correlated with the upregulations of mRNA expression levels for key AA transporters and receptors in the small intestine. In conclusion, the in-feed antibiotics increased serum level of most AAs and decreased most AAs in the small intestine. These changes correlated with the upregulations of mRNA expression levels for key AA transporters and receptors in the small intestine. The findings provide further insights into the mechanism of in-feed antibiotics, which may provide new framework for designing alternatives to antibiotics in animal feed in the future.


Subject(s)
Amino Acids/blood , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation/drug effects , Weight Gain/drug effects , Amino Acid Transport System ASC/agonists , Amino Acid Transport System ASC/genetics , Amino Acid Transport System ASC/metabolism , Animal Feed/analysis , Animals , Animals, Newborn , Biological Transport/drug effects , Excitatory Amino Acid Transporter 3/agonists , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Kitasamycin/pharmacology , Large Neutral Amino Acid-Transporter 1/genetics , Large Neutral Amino Acid-Transporter 1/metabolism , Oxytetracycline/pharmacology , Peptide Transporter 1/agonists , Peptide Transporter 1/genetics , Peptide Transporter 1/metabolism , Quinoxalines/pharmacology , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Swine , TRPV Cation Channels/agonists , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Weaning
16.
Biol Res ; 50(1): 29, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28927446

ABSTRACT

BACKGROUND: Obsessive-compulsive disorder (OCD) is a severe neuropsychiatric condition affecting 1-3% of the worldwide population. OCD has a strong genetic component, and the SLC1A1 gene that encodes neuronal glutamate transporter EAAT3 is a strong candidate for this disorder. To evaluate the impact of reduced EAAT3 expression in vivo, we studied male EAAT3 heterozygous and wild-type littermate mice using a battery of behavioral paradigms relevant to anxiety (open field test, elevated plus maze) and compulsivity (marble burying), as well as locomotor activity induced by amphetamine. Using high-performance liquid chromatography, we also determined tissue neurotransmitter levels in cortex, striatum and thalamus-brain areas that are relevant to OCD. RESULTS: Compared to wild-type littermates, EAAT3 heterozygous male mice have unaltered baseline anxiety-like, compulsive-like behavior and locomotor activity. Administration of acute amphetamine (5 mg/kg intraperitoneally) increased locomotion with no differences across genotypes. Tissue levels of glutamate, GABA, dopamine and serotonin did not vary between EAAT3 heterozygous and wild-type mice. CONCLUSIONS: Our results indicate that reduced EAAT3 expression does not impact neurotransmitter content in the corticostriatal circuit nor alter anxiety or compulsive-like behaviors.


Subject(s)
Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Obsessive-Compulsive Disorder/metabolism , Animals , Disease Models, Animal , Excitatory Amino Acid Transporter 3/genetics , Genotype , Glutamic Acid/genetics , Heterozygote , Male , Mice , Obsessive-Compulsive Disorder/genetics
17.
Hum Mol Genet ; 23(6): 1669-76, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24163246

ABSTRACT

Large and rare copy number variants (CNVs) at several loci have been shown to increase risk for schizophrenia. Aiming to discover novel susceptibility CNV loci, we analyzed 6882 cases and 11 255 controls genotyped on Illumina arrays, most of which have not been used for this purpose before. We identified genes enriched for rare exonic CNVs among cases, and then attempted to replicate the findings in additional 14 568 cases and 15 274 controls. In a combined analysis of all samples, 12 distinct loci were enriched among cases with nominal levels of significance (P < 0.05); however, none would survive correction for multiple testing. These loci include recurrent deletions at 16p12.1, a locus previously associated with neurodevelopmental disorders (P = 0.0084 in the discovery sample and P = 0.023 in the replication sample). Other plausible candidates include non-recurrent deletions at the glutamate transporter gene SLC1A1, a CNV locus recently suggested to be involved in schizophrenia through linkage analysis, and duplications at 1p36.33 and CGNL1. A burden analysis of large (>500 kb), rare CNVs showed a 1.2% excess in cases after excluding known schizophrenia-associated loci, suggesting that additional susceptibility loci exist. However, even larger samples are required for their discovery.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 1 , Cytoskeletal Proteins/genetics , Excitatory Amino Acid Transporter 3/genetics , Gene Duplication , Schizophrenia/genetics , DNA Copy Number Variations , Female , Gene Deletion , Gene Dosage , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Humans , Male
18.
J Membr Biol ; 249(3): 239-49, 2016 06.
Article in English | MEDLINE | ID: mdl-26690923

ABSTRACT

Excitatory amino acid transporters EAAT1 (SLC1A3), EAAT2 (SLC1A2), EAAT3 (SLC1A1), and EAAT4 (SLC1A6) serve to clear L-glutamate from the synaptic cleft and are thus important for the limitation of neuronal excitation. EAAT3 has previously been shown to form complexes with caveolin-1, a major component of caveolae, which participate in the regulation of transport proteins. The present study explored the impact of caveolin-1 on electrogenic transport by excitatory amino acid transporter isoforms EAAT1-4. To this end cRNA encoding EAAT1, EAAT2, EAAT3, or EAAT4 was injected into Xenopus oocytes without or with additional injection of cRNA encoding caveolin-1. The L-glutamate (2 mM)-induced inward current (I Glu) was taken as a measure of glutamate transport. As a result, I Glu was observed in EAAT1-, EAAT2-, EAAT3-, or EAAT4-expressing oocytes but not in water-injected oocytes, and was significantly decreased by coexpression of caveolin-1. Caveolin-1 decreased significantly the maximal transport rate. Treatment of EAATs-expressing oocytes with brefeldin A (5 µM) was followed by a decrease in conductance, which was similar in oocytes expressing EAAT together with caveolin-1 as in oocytes expressing EAAT1-4 alone. Thus, caveolin-1 apparently does not accelerate transporter protein retrieval from the cell membrane. In conclusion, caveolin-1 is a powerful negative regulator of the excitatory glutamate transporters EAAT1, EAAT2, EAAT3, and EAAT4.


Subject(s)
Caveolin 1/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Excitatory Amino Acid Transporter 4/metabolism , Animals , Biological Transport , Caveolin 1/genetics , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 4/genetics , Gene Expression , Glutamic Acid/metabolism , Humans , Oocytes/metabolism , Xenopus laevis
19.
Article in English | MEDLINE | ID: mdl-27507301

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small regulatory molecules that cause translational repression by base pairing with target mRNAs. Cumulative evidence suggests that changes in miRNA expression may in part underlie the pathophysiology and treatment of neuropsychiatric disorders, including major depressive disorder (MDD). METHODS: A miRNA expression assay that can simultaneously detect 423 rat miRNAs (miRBase v.17) was used to profile the prefrontal cortex (PFC) of a genetic rat model of MDD (the Flinders Sensitive Line [FSL]) and the controls, the Flinders Resistant Line (FRL). Gene expression data from the PFC of FSL/FRL animals (GEO accession no. GSE20388) were used to guide mRNA target selection. Luciferase reporter assays were used to verify miRNA targets in vitro. RESULTS: We identified 23 miRNAs that were downregulated in the PFC of the FSL model compared with controls. Interestingly, one of the identified miRNAs (miR-101b) is highly conserved between rat and human and was recently found to be downregulated in the PFC of depressed suicide subjects. Using a combination of in silico and in vitro analyses, we found that miR-101b targets the neuronal glutamate transporter SLC1A1 (also known as EAAC1 or EAAT3). Accordingly, both mRNA and protein levels of SLC1A1 were found to be upregulated in the PFC of the FSL model. CONCLUSIONS: Besides providing a list of novel miRNAs associated with depression-like states, this preclinical study replicated the human association of miR-101 with depression. In addition, since one of the targets of miR-101b appears to be a glutamate transporter, our preclinical data support the hypothesis of a glutamatergic dysregulation being implicated in the etiology of depression.


Subject(s)
Depressive Disorder, Major/genetics , Excitatory Amino Acid Transporter 3/genetics , Glutamic Acid/metabolism , MicroRNAs/genetics , Prefrontal Cortex/metabolism , Animals , Behavior, Animal , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/psychology , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Transporter 3/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks , Genetic Predisposition to Disease , Male , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Prefrontal Cortex/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred Strains , Signal Transduction
20.
Epilepsia ; 57(6): 984-93, 2016 06.
Article in English | MEDLINE | ID: mdl-27153812

ABSTRACT

OBJECTIVE: Posttraumatic seizures (PTS) commonly occur following severe traumatic brain injury (sTBI). Risk factors for PTS have been identified, but variability in who develops PTS remains. Excitotoxicity may influence epileptogenesis following sTBI. Glutamate transporters manage glutamate levels and excitatory neurotransmission, and they have been associated with both epilepsy and TBI. Therefore, we aimed to determine if genetic variation in neuronal glutamate transporter genes is associated with accelerated epileptogenesis and increased PTS risk after sTBI. METHODS: Individuals (N = 253) 18-75 years of age with sTBI were assessed for genetic relationships with PTS. Single nucleotide polymorphisms (SNPs) within SLC1A1 and SLC1A6 were assayed. Kaplan-Meier estimates and log-rank statistics were used to compare seizure rates from injury to 3 years postinjury for SNPs by genotype. Hazard ratios (HRs) were estimated using Cox proportional hazards regression for SNPs significant in Kaplan-Meier analyses adjusting for known PTS risk factors. RESULTS: Thirty-two tagging SNPs were examined (SLC1A1: n = 28, SLC1A6: n = 4). Forty-nine subjects (19.37%) had PTS. Of these, 18 (36.7%) seized within 7 days, and 31 (63.3%) seized between 8 days and 3 years post-TBI. With correction for multiple comparisons, genotypes at SNP rs10974620 (SLC1A1) were significantly associated with time to first seizure across the full 3-year follow-up (seizure rates: 77.1% minor allele homozygotes, 24.8% heterozygotes, 16.6% major allele homozygotes; p = 0.001). When seizure follow-up began day 2 postinjury, genotypes at SNP rs7858819 (SLC1A1) were significantly associated with PTS risk (seizure rates: 52.7% minor allele homozygotes, 11.8% heterozygotes, 21.1% major allele homozygotes; p = 0.002). After adjusting for covariates, we found that rs10974620 remained significant (p = 0.017, minor allele versus major allele homozygotes HR 3.4, 95% confidence interval [CI] 1.3-9.3). rs7858819 also remained significant in adjusted models (p = 0.023, minor allele versus major allele homozygotes HR 3.4, 95%CI 1.1-10.5). SIGNIFICANCE: Variations within SLC1A1 are associated with risk of epileptogenesis following sTBI. Future studies need to confirm findings, but variation within neuronal glutamate transporter genes may represent a possible pharmaceutical target for PTS prevention and treatment.


Subject(s)
Epilepsy, Post-Traumatic/genetics , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 4/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , Adolescent , Adult , Aged , Female , Follow-Up Studies , Genetic Association Studies , Genotype , Glasgow Coma Scale , Humans , Male , Middle Aged , Outcome Assessment, Health Care , Proportional Hazards Models , Statistics, Nonparametric , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL