Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 267
1.
PLoS One ; 19(4): e0289644, 2024.
Article En | MEDLINE | ID: mdl-38598436

Glutamate transporters play key roles in nervous physiology by modulating excitatory neurotransmitter levels, when malfunctioning, involving in a wide range of neurological and physiological disorders. However, integral transmembrane proteins including the glutamate transporters remain notoriously difficult to study, due to their localization within the cell membrane. Here we present the structural bioinformatics studies of glutamate transporters and their water-soluble variants generated through QTY-code, a protein design strategy based on systematic amino acid substitutions. These include 2 structures determined by X-ray crystallography, cryo-EM, and 6 predicted by AlphaFold2, and their predicted water-soluble QTY variants. In the native structures of glutamate transporters, transmembrane helices contain hydrophobic amino acids such as leucine (L), isoleucine (I), and phenylalanine (F). To design water-soluble variants, these hydrophobic amino acids are systematically replaced by hydrophilic amino acids, namely glutamine (Q), threonine (T) and tyrosine (Y). The QTY variants exhibited water-solubility, with four having identical isoelectric focusing points (pI) and the other four having very similar pI. We present the superposed structures of the native glutamate transporters and their water-soluble QTY variants. The superposed structures displayed remarkable similarity with RMSD 0.528Å-2.456Å, despite significant protein transmembrane sequence differences (41.1%->53.8%). Additionally, we examined the differences of hydrophobicity patches between the native glutamate transporters and their QTY variants. Upon closer inspection, we discovered multiple natural variations of L->Q, I->T, F->Y and Q->L, T->I, Y->F in these transporters. Some of these natural variations were benign and the remaining were reported in specific neurological disorders. We further investigated the characteristics of hydrophobic to hydrophilic substitutions in glutamate transporters, utilizing variant analysis and evolutionary profiling. Our structural bioinformatics studies not only provided insight into the differences between the hydrophobic helices and hydrophilic helices in the glutamate transporters, but they are also expected to stimulate further study of other water-soluble transmembrane proteins.


Amino Acid Transport System X-AG , Water , Amino Acid Transport System X-AG/genetics , Amino Acids/chemistry , Membrane Proteins , Mutation , Computational Biology , Glutamates
2.
ASN Neuro ; 15: 17590914231157974, 2023.
Article En | MEDLINE | ID: mdl-36815213

Aging is marked by complex and progressive physiological changes, including in the glutamatergic system, that lead to a decline of brain function. Increased content of senescent cells in the brain, such as glial cells, has been reported to impact cognition both in animal models and human tissue during normal aging and in the context of neurodegenerative disease. Changes in the glutamatergic synaptic activity rely on the glutamate-glutamine cycle, in which astrocytes handle glutamate taken up from synapses and provide glutamine for neurons, thus maintaining excitatory neurotransmission. However, the mechanisms of glutamate homeostasis in brain aging are still poorly understood. Herein, we showed that mouse senescent astrocytes in vitro undergo upregulation of GLT-1, GLAST, and glutamine synthetase (GS), along with the increased enzymatic activity of GS and [3H]-D-aspartate uptake. Furthermore, we observed higher levels of GS and increased [3H]-D-aspartate uptake in the hippocampus of aged mice, although the activity of GS was similar between young and old mice. Analysis of a previously available RNAseq dataset of mice at different ages revealed upregulation of GLAST and GS mRNA levels in hippocampal astrocytes during aging. Corroborating these rodent data, we showed an increased number of GS + cells, and GS and GLT-1 levels/intensity in the hippocampus of elderly humans. Our data suggest that aged astrocytes undergo molecular and functional changes that control glutamate-glutamine homeostasis upon brain aging.


Astrocytes , Neurodegenerative Diseases , Animals , Humans , Mice , Aged , Astrocytes/metabolism , Glutamine/genetics , Glutamine/metabolism , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Up-Regulation , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , D-Aspartic Acid/genetics , Glutamic Acid/metabolism , Hippocampus/metabolism
3.
Int J Med Sci ; 19(11): 1680-1694, 2022.
Article En | MEDLINE | ID: mdl-36237989

Gastric cancer is a highly malignant tumor. Gastric cancer stem cells (GCSCs) are the main causes of drug resistance, metastasis, recurrence, and poor prognosis. As a secondary metabolite of lichen, Atranorin has a variety of biological effects, such as antibacterial, anti-inflammatory, analgesic, and wound healing; however, its killing effect on GCSCs has not been reported. In this study, we constructed Atranorin complexes comprising superparamagnetic iron oxide nanoparticles (SPION) (Atranorin@SPION). In vitro and in vivo experiments confirmed that Atranorin@SPION could significantly inhibit the proliferation, invasion, angiogenesis, and tumorigenicity of CD44+/ CD24+ GCSCs, and induce oxidative stress injury, Fe2+ accumulation, and ferroptosis. Quantitative real-time reverse transcription PCR and western blotting results showed that Atranorin@SPION not only reduced the expression levels of GCSC stem cell markers and cell proliferation and division markers, but also significantly inhibited the expression levels of key molecules in the cystine/glutamate transporter (Xc-)/glutathione peroxidase 4 (GPX4) and Tet methylcytosine dioxygenase (TET) family proteins. The results of high performance liquid chromatography-mass spectrometry and Dot blotting showed that Atranorin@SPION significantly inhibited the mRNA 5­hydroxymethylcytidine modification of GCSCs. Meanwhile, the results of RNA immunoprecipitation-PCR also indicated that Atranorin@SPIONs significantly reduced the 5-hydroxymethylcytidine modification level of GPX4 and SLC7A11 mRNA 3' untranslated region in GCSCs, resulting in a decrease in their stability, shortening their half-lives and reducing translation activity. Therefore, this study revealed that Atranorin@SPIONs induced ferroptosis of GCSCs by weakening the expression of the Xc-/GPX4 axis and the 5-hydroxymethylcytidine modification of mRNAs in the pathway, thereby achieving their therapeutic effect on gastric cancer.


Dioxygenases , Ferroptosis , Stomach Neoplasms , 3' Untranslated Regions , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Amino Acid Transport System X-AG/pharmacology , Analgesics/therapeutic use , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Cystine/genetics , Cystine/metabolism , Cystine/pharmacology , Cytidine/analogs & derivatives , Dioxygenases/genetics , Dioxygenases/metabolism , Dioxygenases/pharmacology , Ferroptosis/genetics , Gene Expression Regulation, Neoplastic , Humans , Hydroxybenzoates , Magnetic Iron Oxide Nanoparticles , Neoplastic Stem Cells/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
4.
Mol Neurobiol ; 59(3): 1912-1924, 2022 Mar.
Article En | MEDLINE | ID: mdl-35032319

Exposure to xenobiotics has a significant impact in brain physiology that could be liked to an excitotoxic process induced by a massive release of the main excitatory neurotransmitter, L-glutamate. Overstimulation of extra-synaptic glutamate receptors, mainly of the N-methyl-D-aspartate subtype leads to a disturbance of intracellular calcium homeostasis that is critically involved in neuronal death. Hence, glutamate extracellular levels are tightly regulated through its uptake by glial glutamate transporters. It has been observed that glutamate regulates its own removal, both in the short-time frame via a transporter-mediated decrease in the uptake, and in the long-term through the transcriptional control of its gene expression, a process mediated by glutamate receptors that involves the Ca2+/diacylglycerol-dependent protein kinase and the transcription factor Ying Yang 1. Taking into consideration that this transcription factor is a member of the Polycomb complex and thus, part of repressive and activating chromatin remodeling factors, it might direct the interaction of DNA methyltransferases or dioxygenases of methylated cytosines to their target sequences. Here we explored the role of dynamic DNA methylation in the expression and function of glial glutamate transporters. To this end, we used the well-characterized models of primary cultures of chick cerebellar Bergmann glia cells and a human retina-derived Müller glia cell line. A time and dose-dependent increase in global DNA methylation was evident upon glutamate exposure. Under hypomethylation conditions, the glial glutamate transporter protein levels and uptake activity were increased. These results favor the notion that a dynamic DNA methylation program triggered by glutamate in glial cells modulates one of its major functions: glutamate removal.


Amino Acid Transport System X-AG , Ependymoglial Cells , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cells, Cultured , DNA Methylation/genetics , Ependymoglial Cells/metabolism , Gene Expression Regulation , Glutamic Acid/metabolism , Humans , Neuroglia/metabolism
5.
Glia ; 70(5): 858-874, 2022 05.
Article En | MEDLINE | ID: mdl-35006609

Bergmann glia (BG) predominantly use glutamate/aspartate transporters (GLAST) for glutamate uptake in the cerebellum. Recently, nitric oxide (NO) treatment has been shown to upregulate GLAST function and increase glutamate uptake in vitro. We previously discovered that neuronal nitric oxide synthase knockout (nNOS-/- ) mice displayed structural and functional neuronal abnormalities in the cerebellum during development, in addition to previously reported motor deficits. Although these developmental deficits have been identified in the nNOS-/- cerebellum, it is unknown whether BG morphology and GLAST expression are also affected in the absence of nNOS in vivo. This study is the first to characterize BG morphology and GLAST expression during development in nNOS-/- mice using immunohistochemistry and western blotting across postnatal development. Results showed that BG in nNOS-/- mice exhibited abnormal morphology and decreased GLAST expression compared with wildtype (WT) mice across postnatal development. Treating ex vivo WT cerebellar slices with the NOS inhibitor L-NAME decreased GLAST expression while treating nNOS-/- slices with the slow-release NO-donor NOC-18 increased GLAST expression when compared with their respective controls. In addition, treating primary BG isolated from WT mice with the selective nNOS inhibitor 7N decreased the membrane expression of GLAST and influx of Ca2+ /Na+ , while treating nNOS-/- BG with SNAP increased the membrane expression of GLAST and Ca2+ /Na+ influx. Moreover, the effects of SNAP on GLAST expression and Ca2+ /Na+ influx in nNOS-/- BG were significantly reduced by a PKG inhibitor. Together, these results reveal a novel role for nNOS/NO signaling in BG development, regulated by a PKG-mediated mechanism.


Amino Acid Transport System X-AG , Glutamic Acid , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid , Cerebellum/metabolism , Glutamic Acid/metabolism , Mice , Mice, Knockout , Neuroglia/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism
6.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(5): 491-496, 2022 Sep.
Article Zh | MEDLINE | ID: mdl-37088758

OBJECTIVE: To investigate the effects of glutamate aspartate transporter (GLAST)deletion on the normal auditory function of mice. METHODS: We hybridized GLAST+/- mice with C57BL/6J background and identified the genotypes of their offspring by agarose gel electrophoresis. 9-10-week-old mice were selected to detect the expression of GLAST protein in the cochlea by immunofluorescence staining and to verify the knockout results(n=3). The changes in weight from 7 days to 30 days after birth and the 30-day body length of male and female mice were compared(n=8). The auditory brainstem response(ABR) was used to detect the auditory threshold and the amplitude of wave I in 9-10-week-old male and female mice(n=5). RESULTS: Male GLAST-/- mice had shown significantly lower weight and body length compared to male GLAST+/+ and GLAST+/- mice(P<0.01), and male GLAST-/- mice showed significant differences compared to GLAST+/+ from P7 to P30 statistical time. Male GLAST-/- mice exhibited a significant reduction in weight after P15 compared to male GLAST+/- mice. In contrast, no significant differences in weight and body length were observed in female GLAST-/- mice compared with female GLAST+/+ and GLAST+/- mice. There was no difference in the hearing threshold detected by ABR between the three genotypes in both male and female mice, but the amplitude of wave I in GLAST-/- mice was significantly lower than that in male GLAST+/+ mice(P<0.01). In contrast, the amplitude of wave I in females was reduced throughout the stimulus intensity but was most significant only at high-intensity stimulation (e.g.80 dB, 90 dB) (P<0.05). CONCLUSION: GLAST knockout affects the normal growth and development of male mice, and decreases the amplitude of wave I, but do not change the threshold, suggesting that GLAST knockout may lead to synaptic pathological changes, and there are gender differences in this effect.


Excitatory Amino Acid Transporter 1 , Hearing , Animals , Female , Male , Mice , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Evoked Potentials, Auditory, Brain Stem/genetics , Excitatory Amino Acid Transporter 1/genetics , Hearing/genetics , Hearing/physiology , Mice, Inbred C57BL , Mice, Knockout , Phenotype
7.
Biochem Biophys Res Commun ; 589: 100-106, 2022 01 22.
Article En | MEDLINE | ID: mdl-34902745

Many temperate ectotherms survive winter by entering diapause - a state of developmental (or reproductive) suppression or arrest - in response to short autumnal day lengths. Day lengths are assessed by the circadian clock, the biological time-keeping system that governs biological rhythms with a period of approximately 24 h. However, clock output molecules controlling this photoperiodic response are largely unknown for many insects. To identify these molecules in Hemiptera, we performed RNAi knockdowns of several candidate genes in the bean bug Riptortus pedestris to determine whether their silencing affects photoperiodic regulation of ovarian development (reproductive diapause). Knockdown of diuretic hormone 31, short neuropeptide F, neuropeptide F, ion transport peptide, neuropeptide-like precursor 1, and choline acetyltransferase had no effect on ovarian development and were therefore ruled out as regulators of the photoperiodic response. However, knockdown of vesicular glutamate transporter promoted ovarian development under diapause-inducing short days, and this is the first report of the functional involvement of glutamate signalling in insect photoperiodism. Improved knockdown of this transporter (or receptor) and RNAi of other genes involved in glutamate signal transduction is required to verify its role as an output of the circadian clock.


Amino Acid Transport System X-AG/metabolism , Circadian Clocks/physiology , Heteroptera/physiology , Insect Proteins/metabolism , Photoperiod , Amino Acid Transport System X-AG/genetics , Animals , Circadian Clocks/genetics , Female , Gene Expression Regulation , Heteroptera/genetics , Insect Proteins/genetics , Ovary/growth & development , Ovary/metabolism , RNA Interference , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism
8.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Article En | MEDLINE | ID: mdl-34873050

Transporters cycle through large structural changes to translocate molecules across biological membranes. The temporal relationships between these changes and function, and the molecular properties setting their rates, determine transport efficiency-yet remain mostly unknown. Using single-molecule fluorescence microscopy, we compare the timing of conformational transitions and substrate uptake in the elevator-type transporter GltPh We show that the elevator-like movements of the substrate-loaded transport domain across membranes and substrate release are kinetically heterogeneous, with rates varying by orders of magnitude between individual molecules. Mutations increasing the frequency of elevator transitions and reducing substrate affinity diminish transport rate heterogeneities and boost transport efficiency. Hydrogen deuterium exchange coupled to mass spectrometry reveals destabilization of secondary structure around the substrate-binding site, suggesting that increased local dynamics leads to faster rates of global conformational changes and confers gain-of-function properties that set transport rates.


Amino Acid Transport System X-AG/metabolism , Archaeal Proteins/metabolism , Cell Membrane/metabolism , Deuterium Exchange Measurement , Amino Acid Sequence , Amino Acid Transport System X-AG/genetics , Archaeal Proteins/genetics , Biological Transport , Escherichia coli/metabolism , Fluorescence Resonance Energy Transfer , Mass Spectrometry , Mutation , Protein Binding , Single Molecule Imaging
9.
Neuropharmacology ; 192: 108602, 2021 07 01.
Article En | MEDLINE | ID: mdl-33991564

Glutamate is the major excitatory neurotransmitter in the vertebrate central nervous system. Once released, it binds to specific membrane receptors and transporters activating a wide variety of signal transduction cascades, as well as its removal from the synaptic cleft in order to avoid its extracellular accumulation and the overstimulation of extra-synaptic receptors that might result in neuronal death through a process known as excitotoxicity. Although neurodegenerative diseases are heterogenous in clinical phenotypes and genetic etiologies, a fundamental mechanism involved in neuronal degeneration is excitotoxicity. Glutamate homeostasis is critical for brain physiology and Glutamate transporters are key players in maintaining low extracellular Glutamate levels. Therefore, the characterization of Glutamate transporters has been an active area of glutamatergic research for the last 40 years. Transporter activity its regulated at different levels: transcriptional and translational control, transporter protein trafficking and membrane mobility, and through extensive post-translational modifications. The elucidation of these mechanisms has emerged as an important piece to shape our current understanding of glutamate actions in the nervous system.


Amino Acid Transport System X-AG/chemistry , Amino Acid Transport System X-AG/metabolism , Glutamic Acid/metabolism , Synaptic Transmission/physiology , Amino Acid Transport System X-AG/genetics , Animals , Excitatory Amino Acid Transporter 1/chemistry , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/chemistry , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Humans , Protein Processing, Post-Translational/physiology , Protein Structure, Secondary , Protein Structure, Tertiary
10.
Nature ; 591(7849): 327-331, 2021 03.
Article En | MEDLINE | ID: mdl-33597752

Glutamate is the most abundant excitatory neurotransmitter in the central nervous system, and its precise control is vital to maintain normal brain function and to prevent excitotoxicity1. The removal of extracellular glutamate is achieved by plasma-membrane-bound transporters, which couple glutamate transport to sodium, potassium and pH gradients using an elevator mechanism2-5. Glutamate transporters also conduct chloride ions by means of a channel-like process that is thermodynamically uncoupled from transport6-8. However, the molecular mechanisms that enable these dual-function transporters to carry out two seemingly contradictory roles are unknown. Here we report the cryo-electron microscopy structure of a glutamate transporter homologue in an open-channel state, which reveals an aqueous cavity that is formed during the glutamate transport cycle. The functional properties of this cavity, combined with molecular dynamics simulations, reveal it to be an aqueous-accessible chloride permeation pathway that is gated by two hydrophobic regions and is conserved across mammalian and archaeal glutamate transporters. Our findings provide insight into the mechanism by which glutamate transporters support their dual function, and add information that will assist in mapping the complete transport cycle shared by the solute carrier 1A transporter family.


Amino Acid Transport System X-AG/chemistry , Amino Acid Transport System X-AG/metabolism , Chloride Channels/chemistry , Chloride Channels/metabolism , Hydrophobic and Hydrophilic Interactions , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/ultrastructure , Animals , Brain/metabolism , Chloride Channels/genetics , Chloride Channels/ultrastructure , Chlorides/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Excitatory Amino Acid Transporter 1/chemistry , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 1/ultrastructure , Female , Glutamic Acid/metabolism , Humans , Models, Molecular , Mutation , Oocytes , Protein Conformation , Xenopus laevis
11.
EMBO J ; 40(1): e105415, 2021 01 04.
Article En | MEDLINE | ID: mdl-33185289

Membrane transporters mediate cellular uptake of nutrients, signaling molecules, and drugs. Their overall mechanisms are often well understood, but the structural features setting their rates are mostly unknown. Earlier single-molecule fluorescence imaging of the archaeal model glutamate transporter homologue GltPh from Pyrococcus horikoshii suggested that the slow conformational transition from the outward- to the inward-facing state, when the bound substrate is translocated from the extracellular to the cytoplasmic side of the membrane, is rate limiting to transport. Here, we provide insight into the structure of the high-energy transition state of GltPh that limits the rate of the substrate translocation process. Using bioinformatics, we identified GltPh gain-of-function mutations in the flexible helical hairpin domain HP2 and applied linear free energy relationship analysis to infer that the transition state structurally resembles the inward-facing conformation. Based on these analyses, we propose an approach to search for allosteric modulators for transporters.


Amino Acid Transport System X-AG/metabolism , Archaeal Proteins/metabolism , Biological Transport/physiology , Amino Acid Transport System X-AG/genetics , Archaea/genetics , Archaea/metabolism , Archaeal Proteins/genetics , Biological Transport/genetics , Computational Biology/methods , Gain of Function Mutation/genetics , Models, Molecular , Pyrococcus horikoshii/genetics , Pyrococcus horikoshii/metabolism , Substrate Specificity/genetics
12.
ACS Chem Neurosci ; 12(1): 163-175, 2021 01 06.
Article En | MEDLINE | ID: mdl-33315395

Excitatory amino acid transporters (EAATs) serve to maintain extracellular neurotransmitter concentrations below neurotoxic levels by transporting glutamate from the synaptic cleft into apposed glia and neurons. Although the crystal structures of the archaeal EAAT homologue from Pyrococcus horikoshii, GltPh, and the human glutamate transporter, EAAT1cryst, have been resolved, the transport mechanism of the transmembrane 3-4 (TM3-4) loop and its structural rearrangement during transport have remained poorly understood. In order to explore the spatial position and function of the TM3-4 loop in the transport cycle, we engineered a pair of cysteine residues between the TM3-4 loop and hairpin loop 2 (HP2) in cysteine-less EAAT2 (CL-EAAT2). We observed that the oxidative cross-linking reagent Cu(II)(1,10-phenanthroline)3 (CuPh) had a significant inhibitory effect on transport in the disubstituted A167C/G437C mutant, whereas dl-dithiothreitol (DTT) reversed the effect of cross-linking A167C/G437C on transport activity, as assayed by d-[3H]-aspartate uptake. Furthermore, we found that the effect of CuPh in this mutant was due to the formation of disulfide bonds in the transporter molecule. Moreover, dl-threo-ß-benzyloxyaspartic acid (TBOA) attenuated, while l-glutamate or KCl enhanced, the CuPh-mediated inhibitory effect in the A167C/G437C mutant, suggesting that the A167C and G437C cysteines were farther apart in the outward-facing configuration and closer in the inward-facing configuration. Taken together, our findings provide evidence that the TM3-4 loop and HP2 change spatial proximity during the transport cycle.


Amino Acid Transport System X-AG , Cysteine , Amino Acid Transport System X-AG/genetics , Aspartic Acid , Cysteine/genetics , Excitatory Amino Acid Transporter 2/genetics , HeLa Cells , Humans , Mutagenesis
13.
Elife ; 92020 11 06.
Article En | MEDLINE | ID: mdl-33155546

Glutamate transporters are essential players in glutamatergic neurotransmission in the brain, where they maintain extracellular glutamate below cytotoxic levels and allow for rounds of transmission. The structural bases of their function are well established, particularly within a model archaeal homolog, sodium, and aspartate symporter GltPh. However, the mechanism of gating on the cytoplasmic side of the membrane remains ambiguous. We report Cryo-EM structures of GltPh reconstituted into nanodiscs, including those structurally constrained in the cytoplasm-facing state and either apo, bound to sodium ions only, substrate, or blockers. The structures show that both substrate translocation and release involve movements of the bulky transport domain through the lipid bilayer. They further reveal a novel mode of inhibitor binding and show how solutes release is coupled to protein conformational changes. Finally, we describe how domain movements are associated with the displacement of bound lipids and significant membrane deformations, highlighting the potential regulatory role of the bilayer.


Amino Acid Transport System X-AG/chemistry , Amino Acid Transport System X-AG/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Pyrococcus horikoshii/metabolism , Amino Acid Transport System X-AG/genetics , Archaeal Proteins/genetics , Biological Transport , Cryoelectron Microscopy , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Kinetics , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Models, Molecular , Protein Domains , Pyrococcus horikoshii/chemistry , Pyrococcus horikoshii/genetics , Sodium/chemistry , Sodium/metabolism
14.
Biomed Res Int ; 2020: 2054293, 2020.
Article En | MEDLINE | ID: mdl-33195689

BACKGROUND: The aim of this study was to research the effects of glutamine synthetase (GS) and glutamate aspartate transporter (GLAST) in rat Müller cells and the effects of an adenosine A2AR antagonist (SCH 442416) on GS and GLAST in hypoxia both in vivo and in vitro. METHODS: This study used RT-PCR and Western blotting to quantify the expressions of GS and GLAST under different hypoxic conditions as well as the expressions of GS and GLAST at different drug concentrations. A cell viability assay was used to assess drug toxicity. RESULTS: mRNA and protein expression of GS and GLAST in hypoxia Group 24 h was significantly increased. mRNA and protein expressions of GS and GLAST both increased in Group 1 µM SCH 442416 compared with other groups. One micromolar SCH 442416 could upregulate GS and GLAST's activity in hypoxia both in vivo and in vitro. CONCLUSIONS: Hypoxia activates GS and GLAST in rat retinal Müller cells in a short time in vitro. (2) A2AR antagonists upregulate the activity of GS and GLAST in hypoxia both in vivo and in vitro.


Adenosine A2 Receptor Antagonists/pharmacology , Amino Acid Transport System X-AG/genetics , Glutamate-Ammonia Ligase/genetics , Hypoxia/enzymology , Hypoxia/genetics , Up-Regulation/drug effects , Amino Acid Transport System X-AG/metabolism , Animals , Cell Survival/drug effects , Disease Models, Animal , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Glutamate-Ammonia Ligase/metabolism , Pyrazoles/pharmacology , Pyrazoles/toxicity , Pyrimidines/pharmacology , Pyrimidines/toxicity , Rats, Sprague-Dawley
15.
J Mol Biol ; 432(24): 166675, 2020 12 04.
Article En | MEDLINE | ID: mdl-33058882

Neuronal hyperexcitability linked to an increase in glutamate signalling is a peculiar trait of the early stages of Alzheimer's disease (AD) and tauopathies, however, a progressive reduction in glutamate release follows in advanced stages. We recently reported that in the early phases of the neurodegenerative process, soluble, non-aggregated Tau accumulates in the nucleus and modulates the expression of disease-relevant genes directly involved in glutamatergic transmission, thus establishing a link between Tau instability and altered neurotransmission. Here we report that while the nuclear translocation of Tau in cultured cells is not impaired by its own aggregation, the nuclear amyloid inclusions of aggregated Tau abolish Tau-dependent increased expression of the glutamate transporter. Remarkably, we observed that in the prefrontal cortex (PFC) of AD patient brain, the glutamate transporter is upregulated at early stages and is downregulated at late stages. The Gene Set Enrichment Analysis indicates that the modulation of Tau-dependent gene expression along the disease progression can be extended to all protein pathways of the glutamatergic synapse. Together, this evidence links the altered glutamatergic function in the PFC during AD progression to the newly discovered function of nuclear Tau.


Alzheimer Disease/genetics , Tauopathies/genetics , Vesicular Glutamate Transport Protein 1/genetics , tau Proteins/genetics , Active Transport, Cell Nucleus/genetics , Alzheimer Disease/pathology , Amino Acid Transport System X-AG/genetics , Animals , Brain/metabolism , Embryonic Stem Cells , Gene Expression Regulation/genetics , Humans , Mice , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Synapses/genetics , Synapses/pathology , Tauopathies/pathology , tau Proteins/metabolism
16.
Respir Res ; 21(1): 208, 2020 Aug 08.
Article En | MEDLINE | ID: mdl-32771007

BACKGROUND: The carotid body (CB) plays a critical role in cyclic intermittent hypoxia (CIH)-induced chemosensitivity; however, the underlying mechanism remains uncertain. We have demonstrated the presence of multiple inotropic glutamate receptors (iGluRs) in CB, and that CIH exposure alters the level of some iGluRs in CB. This result implicates glutamatergic signaling in the CB response to hypoxia. The glutamatergic neurotransmission is not only dependent on glutamate and glutamate receptors, but is also dependent on glutamate transporters, including vesicular glutamate transporters (VGluTs) and excitatory amino acid transporters (EAATs). Here, we have further assessed the expression and distribution of VGluTs and EAATs in human and rat CB and the effect of CIH exposure on glutamate transporters expression. METHODS: The mRNA of VGluTs and EAATs in the human CB were detected by RT-PCR. The protein expression of VGluTs and EAATs in the human and rat CB were detected by Western blot. The distribution of VGluT3, EAAT2 and EAAT3 were observed by immunohistochemistry staining and immunofluorescence staining. Male Sprague-Dawley (SD) rats were exposed to CIH (FIO2 10-21%, 3 min/3 min for 8 h per day) for 2 weeks. The unpaired Student's t-test was performed. RESULTS: Here, we report on the presence of mRNAs for VGluT1-3 and EAAT1-3 in human CB, which is consistent with our previous results in rat CB. The proteins of VGluT1 and 3, EAAT2 and 3, but not VGluT2 and EAAT1, were detected with diverse levels in human and rat CB. Immunostaining showed that VGluT3, the major type of VGluTs in CB, was co-localized with tyrosine hydroxylase (TH) in type I cells. EAAT2 and EAAT3 were distributed not only in type I cells, but also in glial fibrillary acidic protein (GFAP) positive type II cells. Moreover, we found that exposure of SD rats to CIH enhanced the protein level of EAAT3 as well as TH, but attenuated the levels of VGluT3 and EAAT2 in CB. CONCLUSIONS: Our study suggests that glutamate transporters are expressed in the CB, and that glutamate transporters may contribute to glutamatergic signaling-dependent carotid chemoreflex to CIH.


Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Glutamate Plasma Membrane Transport Proteins/biosynthesis , Vesicular Glutamate Transport Proteins/biosynthesis , Amino Acid Transport System X-AG/analysis , Amino Acid Transport System X-AG/biosynthesis , Amino Acid Transport System X-AG/genetics , Animals , Carotid Body/chemistry , Chemoreceptor Cells/chemistry , Gene Expression , Glutamate Plasma Membrane Transport Proteins/analysis , Glutamate Plasma Membrane Transport Proteins/genetics , Humans , Male , Rats , Rats, Sprague-Dawley , Vesicular Glutamate Transport Proteins/analysis , Vesicular Glutamate Transport Proteins/genetics
17.
J Affect Disord ; 277: 244-252, 2020 12 01.
Article En | MEDLINE | ID: mdl-32836031

BACKGROUND: Accumulating evidence indicates that the glutamate system contributes to the pathophysiology of major depressive disorder (MDD) and suicide. We previously reported higher mRNA expression of glutamate receptors in the dorsolateral prefrontal cortex (DLPFC) of females with MDD. METHODS: In the current study, we measured the expression of mRNAs encoding glutamate transporters in the DLPFC of MDD subjects who died by suicide (MDD-S, n = 51), MDD non-suicide subjects (MDD-NS, n = 28), and individuals who did not have a history of neurological illness (CTRL, n = 32). RESULTS: Females but not males with MDD showed higher expression of EAATs and VGLUTs relative to CTRLs. VGLUT expression was significantly higher in the female MDD-S group, relative to the other groups. EAAT expression was lower in the male violent suicides. LIMITATIONS: This study has limitations common to most human studies, including medication history and demographic differences between the diagnostic groups. We mitigated the effects of confounders by including them as covariates in our analyses. CONCLUSIONS: We report sex differences in the expression of glutamate transporter genes in the DLPFC in MDD. Increased neuronal glutamate transporter expression may increase synaptic glutamate, leading to neuronal and glial loss in the DLPFC in MDD. These deficits may lower DLPFC activity, impair problem solving and impair executive function in depression, perhaps increasing vulnerability to suicidal behavior. These data add to accumulating support for the hypothesis that glutamatergic transmission is dysregulated in MDD and suicide. Glutamate transporters may be novel targets for the development of rapidly acting antidepressant therapies.


Depressive Disorder, Major , Suicide , Amino Acid Transport System X-AG/genetics , Depression , Depressive Disorder, Major/genetics , Female , Humans , Male , Prefrontal Cortex , Sex Characteristics
18.
Nat Chem Biol ; 16(9): 1006-1012, 2020 09.
Article En | MEDLINE | ID: mdl-32514183

In proteins where conformational changes are functionally important, the number of accessible states and their dynamics are often difficult to establish. Here we describe a novel 19F-NMR spectroscopy approach to probe dynamics of large membrane proteins. We labeled a glutamate transporter homolog with a 19F probe via cysteine chemistry and with a Ni2+ ion via chelation by a di-histidine motif. We used distance-dependent enhancement of the longitudinal relaxation of 19F nuclei by the paramagnetic metal to assign the observed resonances. We identified one inward- and two outward-facing states of the transporter, in which the substrate-binding site is near the extracellular and intracellular solutions, respectively. We then resolved the structure of the unanticipated second outward-facing state by cryo-EM. Finally, we showed that the rates of the conformational exchange are accessible from measurements of the metal-enhanced longitudinal relaxation of 19F nuclei.


Amino Acid Transport System X-AG/chemistry , Magnetic Resonance Spectroscopy , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Cryoelectron Microscopy , Cysteine/chemistry , Fluorine , Histidine/chemistry , Models, Molecular , Mutation , Nickel/chemistry , Protein Conformation , Protein Domains , Pyrococcus horikoshii/chemistry
19.
Mol Med Rep ; 21(4): 1809-1818, 2020 04.
Article En | MEDLINE | ID: mdl-32319622

The current study aimed to evaluate the neuroprotective effect of Ginkgo biloba extract (GbE) on the progression of acute cerebral ischemia­reperfusion injury in diabetic rats, and to determine the molecular mechanism associated with this effect. Streptozotocin (STZ) induced diabetic rats were pretreated with GbE (50, 100 and 200 mg/kg/day; intragastric) for 3 weeks. During this period, body weight changes and fasting blood glucose levels were assessed each week. Following pretreatment, rats were subjected to suture occlusion of the middle cerebral artery for 30 min, which was followed by 24 h of reperfusion. Neurological deficits were subsequently evaluated at 2 and 24 h following reperfusion. Rats were sacrificed after 24 h reperfusion, and infarct volume and S100B content were measured to evaluate the neuroprotective effect of GbE. The results of the present study demonstrated that GbE pretreatment improved neurological scores, and reduced cerebral infarct volume and S100B content. Oxidative stress markers, including glutathione (GSH) and superoxide dismutase (SOD) were increased, and malondialdehyde (MDA) contents were reduced following GbE treatment. The levels of p­Akt, p­mTOR and glutamate transporter 1 (GLT1) were observed to be increased in GbE­pretreated rats. These results indicated that GbE pretreatment may serve a protective role against cerebral ischemia­reperfusion injury in diabetic rats by inhibiting oxidative stress reaction, upregulating the expression of Akt/mTOR and promoting GLT1 expression. In conclusion, the current study revealed the protective role and molecular mechanisms of GbE in diabetic rats with cerebral ischemia­reperfusion injury, and may provide novel insight into the future clinical treatment of this condition.


Amino Acid Transport System X-AG/genetics , Diabetes Mellitus, Experimental/drug therapy , Oxidative Stress , Plant Extracts/therapeutic use , Reperfusion Injury/drug therapy , Up-Regulation , Amino Acid Transport System X-AG/metabolism , Animals , Behavior, Animal , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Fasting/blood , Ginkgo biloba , Male , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/blood , Reperfusion Injury/complications , Reperfusion Injury/physiopathology , Streptozocin , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects
20.
Nat Commun ; 11(1): 998, 2020 02 21.
Article En | MEDLINE | ID: mdl-32081874

Glutamate transporters are cation-coupled secondary active membrane transporters that clear the neurotransmitter L-glutamate from the synaptic cleft. These transporters are homotrimers, with each protomer functioning independently by an elevator-type mechanism, in which a mobile transport domain alternates between inward- and outward-oriented states. Using single-particle cryo-EM we have determined five structures of the glutamate transporter homologue GltTk, a Na+- L-aspartate symporter, embedded in lipid nanodiscs. Dependent on the substrate concentrations used, the protomers of the trimer adopt a variety of asymmetrical conformations, consistent with the independent movement. Six of the 15 resolved protomers are in a hitherto elusive state of the transport cycle in which the inward-facing transporters are loaded with Na+ ions. These structures explain how substrate-leakage is prevented - a strict requirement for coupled transport. The belt protein of the lipid nanodiscs bends around the inward oriented protomers, suggesting that membrane deformations occur during transport.


Amino Acid Transport System X-AG/chemistry , Archaeal Proteins/chemistry , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Aspartic Acid/metabolism , Binding Sites , Cryoelectron Microscopy , Lipids/chemistry , Models, Molecular , Nanostructures/chemistry , Protein Conformation , Protein Structure, Quaternary , Pyrococcus horikoshii/metabolism , Single Molecule Imaging , Symporters/chemistry , Symporters/metabolism , Thermococcus/genetics , Thermococcus/metabolism
...