881 results on '"Excitatory Amino Acid Transporter 1"'
Search Results
2. Genetic interactions regulate hypoxia tolerance conferred by activating Notch in excitatory amino acid transporter 1-positive glial cells in Drosophila melanogaster
- Author
-
Zhou, Dan, Stobdan, Tsering, Visk, DeeAnn, Xue, Jin, and Haddad, Gabriel G
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Stroke ,Human Genome ,Biotechnology ,1.1 Normal biological development and functioning ,Aetiology ,Underpinning research ,2.1 Biological and endogenous factors ,Animals ,Drosophila Proteins ,Drosophila melanogaster ,Excitatory Amino Acid Transporter 1 ,Hypoxia ,Neuroglia ,Phenotype ,Receptors ,Notch ,Notch signaling ,genetic interactions ,eaat1-posive glia ,Drosophila melanogaster ,Biochemistry and cell biology ,Statistics - Abstract
Hypoxia is a critical pathological element in many human diseases, including ischemic stroke, myocardial infarction, and solid tumors. Of particular significance and interest of ours are the cellular and molecular mechanisms that underlie susceptibility or tolerance to low O2. Previous studies have demonstrated that Notch signaling pathway regulates hypoxia tolerance in both Drosophila melanogaster and humans. However, the mechanisms mediating Notch-conferred hypoxia tolerance are largely unknown. In this study, we delineate the evolutionarily conserved mechanisms underlying this hypoxia tolerant phenotype. We determined the role of a group of conserved genes that were obtained from a comparative genomic analysis of hypoxia-tolerant D.melanogaster populations and human highlanders living at the high-altitude regions of the world (Tibetans, Ethiopians, and Andeans). We developed a novel dual-UAS/Gal4 system that allows us to activate Notch signaling in the Eaat1-positive glial cells, which remarkably enhances hypoxia tolerance in D.melanogaster, and, simultaneously, knock down a candidate gene in the same set of glial cells. Using this system, we discovered that the interactions between Notch signaling and bnl (fibroblast growth factor), croc (forkhead transcription factor C), or Mkk4 (mitogen-activated protein kinase kinase 4) are important for hypoxia tolerance, at least in part, through regulating neuronal development and survival under hypoxic conditions. Becausethese genetic mechanisms are evolutionarily conserved, this group of genes may serve as novel targets for developing therapeutic strategies and have a strong potential to be translated to humans to treat/prevent hypoxia-related diseases.
- Published
- 2021
3. New Spinal Cord Injury Study Results Reported from University of Puerto Rico (The sexually dimorphic expression of glutamate transporters and their implication in pain after spinal cord injury).
- Abstract
A new study from the University of Puerto Rico explores the cellular-molecular mechanisms behind pain after spinal cord injury. The researchers found that plasticity within the injury site leads to abnormal sensory transmission and the formation of new connections, resulting in central neuropathic pain. The study focused on the role of glutamate transporters and their potential as a therapeutic target. The researchers discovered a sexually dimorphic expression of glutamate transporters in the spinal cord, with female rats showing a higher level of expression. This finding provides a potential opportunity for treating chronic pain in a sex-specific manner. [Extracted from the article]
- Published
- 2024
4. Novel Genetic Variants Associated With Increased Vertebral Volumetric BMD, Reduced Vertebral Fracture Risk, and Increased Expression of SLC1A3 and EPHB2
- Author
-
Nielson, Carrie M, Liu, Ching‐Ti, Smith, Albert V, Ackert‐Bicknell, Cheryl L, Reppe, Sjur, Jakobsdottir, Johanna, Wassel, Christina, Register, Thomas C, Oei, Ling, Alonso, Nerea, Oei, Edwin H, Parimi, Neeta, Samelson, Elizabeth J, Nalls, Mike A, Zmuda, Joseph, Lang, Thomas, Bouxsein, Mary, Latourelle, Jeanne, Claussnitzer, Melina, Siggeirsdottir, Kristin, Srikanth, Priya, Lorentzen, Erik, Vandenput, Liesbeth, Langefeld, Carl, Raffield, Laura, Terry, Greg, Cox, Amanda J, Allison, Matthew A, Criqui, Michael H, Bowden, Don, Ikram, M Arfan, Mellström, Dan, Karlsson, Magnus K, Carr, John, Budoff, Matthew, Phillips, Caroline, Cupples, L Adrienne, Chou, Wen‐Chi, Myers, Richard H, Ralston, Stuart H, Gautvik, Kaare M, Cawthon, Peggy M, Cummings, Steven, Karasik, David, Rivadeneira, Fernando, Gudnason, Vilmundur, Orwoll, Eric S, Harris, Tamara B, Ohlsson, Claes, Kiel, Douglas P, and Hsu, Yi‐Hsiang
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Genetics ,Human Genome ,Aging ,Osteoporosis ,Biomedical Imaging ,Aetiology ,2.1 Biological and endogenous factors ,Musculoskeletal ,Animals ,Biopsy ,Bone Density ,Cancellous Bone ,Excitatory Amino Acid Transporter 1 ,Gene Expression Regulation ,Genetic Association Studies ,Genetic Predisposition to Disease ,Humans ,Linkage Disequilibrium ,Lumbar Vertebrae ,Mice ,Molecular Sequence Annotation ,Organ Size ,Osteoblasts ,Polymorphism ,Single Nucleotide ,Quantitative Trait Loci ,Receptor ,EphB2 ,Risk Factors ,Spinal Fractures ,Spine ,BONE QCT ,CT ,ANALYSIS ,QUANTITATION OF BONE ,OSTEOPOROSIS ,DISEASES AND DISORDERS OF ,RELATED TO BONE ,GENERAL POPULATION STUDIES ,EPIDEMIOLOGY ,HUMAN ASSOCIATION STUDIES ,GENETIC RESEARCH ,FRACTURE RISK ASSESSMENT ,ANALYSIS/QUANTITATION OF BONE ,BONE QCT/μCT ,DISEASES AND DISORDERS OF/RELATED TO BONE ,EPIDEMIOLOGY ,HUMAN ASSOCIATION STUDIES ,Engineering ,Medical and Health Sciences ,Anatomy & Morphology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Genome-wide association studies (GWASs) have revealed numerous loci for areal bone mineral density (aBMD). We completed the first GWAS meta-analysis (n = 15,275) of lumbar spine volumetric BMD (vBMD) measured by quantitative computed tomography (QCT), allowing for examination of the trabecular bone compartment. SNPs that were significantly associated with vBMD were also examined in two GWAS meta-analyses to determine associations with morphometric vertebral fracture (n = 21,701) and clinical vertebral fracture (n = 5893). Expression quantitative trait locus (eQTL) analyses of iliac crest biopsies were performed in 84 postmenopausal women, and murine osteoblast expression of genes implicated by eQTL or by proximity to vBMD-associated SNPs was examined. We identified significant vBMD associations with five loci, including: 1p36.12, containing WNT4 and ZBTB40; 8q24, containing TNFRSF11B; and 13q14, containing AKAP11 and TNFSF11. Two loci (5p13 and 1p36.12) also contained associations with radiographic and clinical vertebral fracture, respectively. In 5p13, rs2468531 (minor allele frequency [MAF] = 3%) was associated with higher vBMD (β = 0.22, p = 1.9 × 10-8 ) and decreased risk of radiographic vertebral fracture (odds ratio [OR] = 0.75; false discovery rate [FDR] p = 0.01). In 1p36.12, rs12742784 (MAF = 21%) was associated with higher vBMD (β = 0.09, p = 1.2 × 10-10 ) and decreased risk of clinical vertebral fracture (OR = 0.82; FDR p = 7.4 × 10-4 ). Both SNPs are noncoding and were associated with increased mRNA expression levels in human bone biopsies: rs2468531 with SLC1A3 (β = 0.28, FDR p = 0.01, involved in glutamate signaling and osteogenic response to mechanical loading) and rs12742784 with EPHB2 (β = 0.12, FDR p = 1.7 × 10-3 , functions in bone-related ephrin signaling). Both genes are expressed in murine osteoblasts. This is the first study to link SLC1A3 and EPHB2 to clinically relevant vertebral osteoporosis phenotypes. These results may help elucidate vertebral bone biology and novel approaches to reducing vertebral fracture incidence. © 2016 American Society for Bone and Mineral Research.
- Published
- 2016
5. Reduced excitatory amino acid transporter 1 and metabotropic glutamate receptor 5 expression in the cerebellum of fragile X mental retardation gene 1 premutation carriers with fragile X-associated tremor/ataxia syndrome
- Author
-
Pretto, Dalyir I, Kumar, Madhur, Cao, Zhengyu, Cunningham, Christopher L, Durbin-Johnson, Blythe, Qi, Lihong, Berman, Robert, Noctor, Stephen C, Hagerman, Randi J, Pessah, Isaac N, and Tassone, Flora
- Subjects
Biological Psychology ,Psychology ,Neurosciences ,Genetics ,Fragile X Syndrome ,Mental Health ,Pediatric ,Neurodegenerative ,Rare Diseases ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Mental health ,Aged ,Aged ,80 and over ,Aging ,Ataxia ,Brain ,Cerebellum ,Cognitive Dysfunction ,Excitatory Amino Acid Transporter 1 ,Fragile X Mental Retardation Protein ,Gene Expression ,Heterozygote ,Humans ,Male ,Mutation ,RNA ,Messenger ,Receptor ,Metabotropic Glutamate 5 ,Syndrome ,Tremor ,FMR1 ,FMRP ,Premutation ,Fragile X tremor/ataxia syndrome ,FXTAS ,Glu transporters ,EAAT1 ,EAAT2 ,mGluR5 ,Clinical Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
A premutation (PM) expansion (55-200 CGG) in the fragile X mental retardation gene 1 causes elevated messenger RNA and reduced fragile X mental retardation gene 1 protein. Young PM carriers can develop characteristic physical features and mild cognitive disabilities. In addition, individuals with PM, particularly male carriers, are at high risk to develop fragile X-associated tremor/ataxia syndrome (FXTAS) with aging. Human postmortem FXTAS brains show extensive white matter disease in the cerebellum and the presence of intranuclear inclusions throughout the brain, although their etiologic significance is unknown. In the current work, expression levels of the metabotropic glutamate (Glu) receptor 5 and the Glu transporter excitatory amino acid transporter 1, examined by reverse transcription polymerase chain reaction and western blot analyses, were found to be reduced in the postmortem cerebellum of PM carriers with FXTAS compared with age matched controls, with higher CGG repeat number having greater reductions in both proteins. These data suggests a dysregulation of Glu signaling in PM carriers, which would likely contribute to the development and severity of FXTAS.
- Published
- 2014
6. Finding disease candidate genes by liquid association
- Author
-
Li, Ker-Chau, Palotie, Aarno, Yuan, Shinsheng, Bronnikov, Denis, Chen, Daniel, Wei, Xuelian, Choi, Oi-Wa, Saarela, Janna, and Peltonen, Leena
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Computational Biology ,Excitatory Amino Acid Transporter 1 ,Finland ,Gene Expression ,Genetic Predisposition to Disease ,Genetic Testing ,Genomics ,Genotype ,Humans ,Multiple Sclerosis ,Protein Kinase C-alpha ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
A novel approach to finding candidate genes by using gene expression data through liquid association is developed and used to identify multiple sclerosis susceptibility candidate genes.
- Published
- 2007
7. Fibroblasts enable penile erection.
- Author
-
Ryu JK and Koh GY
- Subjects
- Humans, Male, Mice, Animals, Erectile Dysfunction therapy, Excitatory Amino Acid Transporter 1, Fibroblasts metabolism, Fibroblasts physiology, Penile Erection
- Abstract
Perivascular fibroblasts may underlie erectile dysfunction.
- Published
- 2024
- Full Text
- View/download PDF
8. The episodic ataxias.
- Author
-
Graves TD, Snell HD, Khodakhah K, Griggs RC, and Jen JC
- Subjects
- Humans, Fibroblast Growth Factors genetics, Kv1.1 Potassium Channel genetics, Calcium Channels genetics, Excitatory Amino Acid Transporter 1, Ataxia genetics, Ataxia diagnosis, Mutation genetics
- Abstract
The primary episodic ataxias (EAs) are a group of autosomal-dominant disorders characterized by transient recurrent incoordination and truncal instability, often triggered by physical exertion or emotional stress and variably associated with progressive baseline ataxia. There are now nine designated subtypes EA1-9 (OMIM) and late onset cerebellar ataxia with episodic features as newly designated SCA27B, based largely on genetic loci. Mutations have been identified in multiple individuals and families in 4 of the 9 EA subtypes, mostly with the onset before adulthood. This chapter focuses on the clinical assessment and management of EA, genetic diagnosis, and neurophysiologic consequences of the causative mutations in the best characterized EA syndromes: EA1 caused by mutations in KCNA1 encoding a neuronal voltage-gated potassium channel, EA2 caused by mutations in CACNA1A encoding a neuronal voltage-gated calcium channel, EA6 caused by mutations in SLC1A3 encoding a glutamate transporter that is also an anion channel, and SCA27B with late onset episodic ataxia caused by an intronic trinucleotide repeat in FGF14 encoding fibroblast growth factor 14 important in regulating the distribution of voltage-gated sodium channels in the cerebellar Purkinje and granule cells. The study of EA has illuminated previously unrecognized but important roles of ion channels and transporters in brain function with shared mechanisms underlying cerebellar ataxia, migraine, and epilepsy., (Copyright © 2024 Elsevier B.V. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
9. Glutamate–aspartate transporter 1 attenuates oxygen–glucose deprivation‐induced injury by promoting glutamate metabolism in primary cortical neurons
- Author
-
Yun‐Zhi Zhao, Jun Wei, Ke‐Xin Song, Chen Zhou, and Zhen Chai
- Subjects
Excitatory Amino Acid Transporter 1 ,Neurons ,Oxygen ,Glucose ,Physiology ,Clinical Biochemistry ,Animals ,Glutamic Acid ,Sciuridae ,Cell Biology ,Cells, Cultured ,Ischemic Stroke ,Rats - Abstract
Ischemic stroke is a common cerebral disease. However, the treatment for the disease is limited. Daurian ground squirrel (GS; Spermophilus dauricus), a hibernating mammalian species, is highly tolerant to ischemia. In the present study, GS neurons in a non-hibernating state were found to be more resistant to oxygen-glucose deprivation (OGD), an ischemic model in vitro. We leveraged the differences in the endurance capacity of GS and rats to investigate the mechanisms of resistance to ischemia in GS neurons. We first identified glutamate-aspartate transporter 1 (GLAST) as a cytoprotective factor that contributed to tolerance against OGD injury of GS neurons. The expression of GLAST in GS neurons was much higher than that in rat neurons. Overexpression of GLAST rescued viability in rat neurons, and GS neurons exhibited decreased viability following GLAST knockdown under OGD conditions. Mechanistically, more glutamate was transported into neurons after GLAST overexpression and served as substrates for ATP production. Furthermore, eukaryotic transcription initiation factor 4E binding protein 1 was downregulated by GLAST to rescue neuronal viability. Our findings not only revealed an important molecular mechanism underlying the survival of hibernating mammals but also suggested that neuronal GLAST may be a potential target for ischemic stroke therapy.
- Published
- 2022
- Full Text
- View/download PDF
10. EAAT1-dependent
- Author
-
Dinorah, Hernández-Melchor, Leticia, Ramírez-Martínez, Luis, Cid, Cecilia, Palafox-Gómez, Esther, López-Bayghen, and Arturo, Ortega
- Subjects
Excitatory Amino Acid Transporter 1 ,Gene Expression Regulation ,Glutamic Acid ,Neuroglia ,Cells, Cultured - Abstract
EAAT1/GLAST down-regulates its expression and function at the transcriptional level by activating a signaling pathway that includes PI3K, PKC and NF-κB, favoring the notion of an activity-dependent fine-tuning of glutamate recycling and its synaptic transactions through glial cells.
- Published
- 2022
11. ACSA‐2 and GLAST classify subpopulations of multipotent and glial‐restricted cerebellar precursors
- Author
-
Elena Parmigiani, Valentina Cerrato, Melanie Jungblut, Annalisa Buffo, Stefan Tomiuk, Christina Geraldine Kantzer, Andreas Bosio, and Michail Knauel
- Subjects
Male ,0301 basic medicine ,Cerebellum ,RRID:AB_10013382 ,ACSA-2 ,Bergmann glia ,RRID:AB_10000325 ,RRID:AB_10000343 ,RRID:AB_1036062 ,RRID:AB_10829302 ,RRID:AB_10829314 ,RRID:AB_1566510 ,RRID:AB_2113602 ,RRID:AB_221569 ,RRID:AB_2299035 ,RRID:AB_2307313 ,RRID:AB_244365 ,RRID:AB_244373 ,RRID:AB_2651192 ,RRID:AB_2655070 ,RRID:AB_2655072 ,RRID:AB_2655591 ,RRID:AB_2660782 ,RRID:AB_2660783 ,RRID:AB_2727361 ,RRID:AB_2727421 ,RRID:AB_2727423 ,RRID:AB_442102 ,RRID:AB_477499 ,RRID:AB_641021 ,RRID:AB_871621 ,RRID:AB_871623 ,RRID:AB_90949 ,RRID:AB_91789 ,RRID:ISMR_CARD:290 ,RRID:SCR_001905 ,RRID:SCR_001915 ,RRID:SCR_002798 ,RRID:SCR_003070 ,RRID:SCR_005012 ,astrocytes ,cerebellum ,prospective white matter ,ACSA‐2 ,Cell ,Mice ,0302 clinical medicine ,Research Articles ,education.field_of_study ,Cell biology ,Excitatory Amino Acid Transporter 1 ,medicine.anatomical_structure ,Antigens, Surface ,Female ,Neuroglia ,Research Article ,Astrocyte ,Population ,Mice, Transgenic ,Granular layer ,Biology ,White matter ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,medicine ,Animals ,Progenitor cell ,education ,Progenitor ,Immunomagnetic Separation ,Sequence Analysis, RNA ,Multipotent Stem Cells ,Mice, Inbred C57BL ,030104 developmental biology ,Animals, Newborn ,nervous system ,030217 neurology & neurosurgery - Abstract
The formation of the cerebellum is highly coordinated to obtain its characteristic morphology and all cerebellar cell types. During mouse postnatal development, cerebellar progenitors with astroglial‐like characteristics generate mainly astrocytes and oligodendrocytes. However, a subset of astroglial‐like progenitors found in the prospective white matter (PWM) produces astroglia and interneurons. Characterizing these cerebellar astroglia‐like progenitors and distinguishing their developmental fates is still elusive. Here, we reveal that astrocyte cell surface antigen‐2 (ACSA‐2), lately identified as ATPase, Na+/K+ transporting, beta 2 polypeptide, is expressed by glial precursors throughout postnatal cerebellar development. In contrast to common astrocyte markers, ACSA‐2 appears on PWM cells but is absent on Bergmann glia (BG) precursors. In the adult cerebellum, ACSA‐2 is broadly expressed extending to velate astrocytes in the granular layer, white matter astrocytes, and to a lesser extent to BG. Cell transplantation and transcriptomic analysis revealed that marker staining discriminates two postnatal progenitor pools. One subset is defined by the co‐expression of ACSA‐2 and GLAST and the expression of markers typical of parenchymal astrocytes. These are PWM precursors that are exclusively gliogenic. They produce predominantly white matter and granular layer astrocytes. Another subset is constituted by GLAST positive/ACSA‐2 negative precursors that express neurogenic and BG‐like progenitor genes. This population displays multipotency and gives rise to interneurons besides all glial types, including BG. In conclusion, this work reports about ACSA‐2, a marker that in combination with GLAST enables for the discrimination and isolation of multipotent and glia‐committed progenitors, which generate different types of cerebellar astrocytes., Glial cells of the cerebellum are still insufficiently classified. In this study we describe the subclassification of GLAST+ precursors in the prospective white matter using immunophenotyping, transcriptomics, and transplantation assays. A subclass of GLAST+ cells, which co‐express astrocyte cell surface antigen‐2 (ACSA‐2), generates only parenchymal astrocytes—as revealed by protein expression, their genetic signature and transplantation. The absence of the astrocyte marker (ACSA‐2) reveals GLAST+ cells to maintain a multipotent potential, giving rise to interneurons, glia, and Bergmann glia.
- Published
- 2021
- Full Text
- View/download PDF
12. IκB kinase inhibition remodeled connexins, pannexin‐1, and excitatory amino‐acid transporters expressions to promote neuroprotection of galantamine and morphine
- Author
-
Shimaa Magdy, Nivin Sharawy, Nancy F Samir, Haitham S. Mohammed, Basma Emad Aboulhoda, Maha Gamal, and Laila A. Rashed
- Subjects
Lipopolysaccharides ,Male ,0301 basic medicine ,Physiology ,Clinical Biochemistry ,Anti-Inflammatory Agents ,Glutamic Acid ,Nerve Tissue Proteins ,IκB kinase ,Neuroprotection ,Connexins ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Glutamate homeostasis ,Nitriles ,Animals ,Glutamate reuptake ,Sulfones ,Neurons ,Morphine ,Galantamine ,Chemistry ,Glutamate receptor ,Brain ,Neurodegenerative Diseases ,Transporter ,Cell Biology ,Pannexin ,I-kappa B Kinase ,Cell biology ,Excitatory Amino Acid Transporter 1 ,Disease Models, Animal ,Neuroprotective Agents ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,030220 oncology & carcinogenesis ,Excitatory postsynaptic potential ,Neuroglia ,Signal Transduction - Abstract
Inflammatory pathway and disruption in glutamate homeostasis join at the level of the glia, resulting in various neurological disorders. In vitro studies have provided evidence that membrane proteins connexions (Cxs) are involved in glutamate release, meanwhile, excitatory amino-acid transporters (EAATs) are crucial for glutamate reuptake (clearance). Moreover, pannexin-1 (Panx-1) activation is more detrimental to neurons. Their expression patterns during inflammation and the impacts of IκB kinase (IKK) inhibition, morphine, and galantamine on the inflammatory-associated glutamate imbalance remain elusive. To investigate this, rats were injected with saline or lipopolysaccharide. Thereafter, vehicles, morphine, galantamine, and BAY-117082 were administered in different groups of animals. Subsequently, electroencephalography, enzyme-linked immunosorbent assay, western blot, and histopathological examinations were carried out and various indicators of inflammation and glutamate level were determined. Parallel analysis of Cxs, Panx-1, and EAAts in the brain was performed. Our findings strengthen the concept that unregulated expressions of Cxs, Panx-1, and EAATs contribute to glutamate accumulation and neuronal cell loss. Nuclear factor-kB (NF-κB) pathway can significantly contribute to glutamate homeostasis via modulating Cxs, Panx-1, and EAATs expressions. BAY-117082, via inhibition of IkK, promoted the anti-inflammatory effects of morphine as well as galantamine. We concluded that NF-κB is an important component of reshaping the expressions of Cxs, panx-1, and EAATs and the development of glutamate-induced neuronal degeneration.
- Published
- 2021
- Full Text
- View/download PDF
13. Hashimoto's Thyroiditis Induces Hippocampus-Dependent Cognitive Alterations by Impairing Astrocytes in Euthyroid Mice
- Author
-
Yan Sun, Yong-Xia Xu, Tiantian Liu, Hao Yang, Nan Wang, Qin Xia, Defa Zhu, Fen Wang, and Yaojun Cai
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Long-Term Potentiation ,Hippocampus ,Morris water navigation task ,030209 endocrinology & metabolism ,Hashimoto Disease ,Hippocampal formation ,Biology ,Receptors, N-Methyl-D-Aspartate ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Endocrinology ,Memory ,Mice, Inbred NOD ,Morris Water Maze Test ,Internal medicine ,LTP induction ,medicine ,Animals ,Cognitive Dysfunction ,Euthyroid ,CA1 Region, Hippocampal ,Behavior, Animal ,musculoskeletal, neural, and ocular physiology ,Glutamate Synthase ,Glutamate receptor ,Excitatory Postsynaptic Potentials ,Long-term potentiation ,Excitatory Amino Acid Transporter 1 ,Disease Models, Animal ,Excitatory Amino Acid Transporter 2 ,nervous system ,Astrocytes ,030220 oncology & carcinogenesis ,Synaptic plasticity ,Female - Abstract
Background: Although studies have reported an increased risk for cognitive disorders in Hashimoto's thyroiditis (HT) patients, even in the euthyroid state, the mechanisms involved remain unclear. The hippocampus is a classic brain region associated with cognitive function, among which the formation of long-term potentiation (LTP) in the Schaffer collateral-CA1 pathway plays an important role in the process of learning and memory. Therefore, this study established a euthyroid HT model in mice and investigated whether and how HT itself has the ability to trigger LTP alterations accompanied by learning and memory abnormality. Methods: An experimental euthyroid HT model was established in NOD mice through immunization with porcine thyroglobulin (Tg). Morris water maze was measured to determine mice spatial learning and memory. We investigated the effect of HT on synaptic transmission and high-frequency stimulation-induced LTP in the Schaffer collateral-CA1 synapse of mice hippocampus in vivo. Then, animals were sacrificed for thyroid-related parameter measure as well as detection of cellular and molecular events associated with the induction of LTP. Results: HT mice showed intrathyroidal lymphocyte infiltration and rising serum thyroid autoantibody levels accompanied by normal thyroid function. The HT mice had poorer performance in Morris water maze than controls. These alterations were mirrored by abnormalities in synaptic plasticity in the Schaffer collateral-CA1 synapses of the hippocampus in vivo. The integrity of the synaptic structure is the premise for the production of LTP. As detected by transmission electron microscopy, the ultrastructure of synapse and astrocyte in the hippocampus were impaired in euthyroid HT mice. Additionally, Western blot and real-time polymerase chain reaction analyses confirmed that in HT mice, GS, GLAST, and GLT-1, key elements in glutamate-glutamine circulation located in astrocyte, were downregulated, accompanied by elevated levels of glutamate in the hippocampus, which impaired the material basis for LTP induction. NMDR2B expression in the hippocampus was also downregulated. Conclusion: HT can induce damage of LTP in the hippocampal Schaffer collateral-CA1 pathway in the euthyroid state, and this can be attributed, at least partly, to astrocytes impairment, which may underlie the deleterious effects of HT itself on hippocampal-dependent learning and memory function.
- Published
- 2021
- Full Text
- View/download PDF
14. Glutamate transporters have a chloride channel with two hydrophobic gates
- Author
-
Alastair G. Stewart, Emad Tajkhorshid, Rosemary J. Cater, Renae M. Ryan, Josep Font, Qianyi Wu, Ichia Chen, Shashank Pant, Robert J. Vandenberg, and Meghna Sobti
- Subjects
Models, Molecular ,Protein Conformation ,Sodium ,Amino Acid Transport System X-AG ,chemistry.chemical_element ,Glutamic Acid ,Crystallography, X-Ray ,Chloride ,Article ,03 medical and health sciences ,Xenopus laevis ,0302 clinical medicine ,Chlorides ,Chloride Channels ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Aqueous solution ,Chemistry ,Molecular biophysics ,Cryoelectron Microscopy ,Glutamate receptor ,Brain ,Transporter ,Solute carrier family ,Excitatory Amino Acid Transporter 1 ,Mutation ,Chloride channel ,Biophysics ,Oocytes ,Female ,Hydrophobic and Hydrophilic Interactions ,030217 neurology & neurosurgery ,medicine.drug - Abstract
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. Glutamate transporters conduct chloride ions through an aqueous channel with hydrophobic gates that forms during the glutamate transport cycle.
- Published
- 2021
15. Research on the Relationship Between Schizophrenia and Excitatory Amino Acid Transporter 1 Gene Based on Nanogold Amplification Technology
- Author
-
Wen Xie, Shuang Zheng, Longcai Fei, and Nannan Zhu
- Subjects
Candidate gene ,Materials science ,Population ,Central nervous system ,Biomedical Engineering ,Bioengineering ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Humans ,SNP ,Genetic Predisposition to Disease ,General Materials Science ,Allele ,education ,Gene ,030304 developmental biology ,Genetics ,0303 health sciences ,education.field_of_study ,Brain ,General Chemistry ,Condensed Matter Physics ,Excitatory Amino Acid Transporter 1 ,medicine.anatomical_structure ,chemistry ,Schizophrenia ,030217 neurology & neurosurgery ,DNA ,Cysteine - Abstract
Schizophrenia is one of the most common central nervous system diseases, which is caused by abnormal discharge of neurons in the brain. Its occurrence and development are affected by both genetic and environmental factors. The variation of gene level can affect the development of schizophrenia and the treatment of prognosis by affecting the susceptibility, clinical phenotype and drug response. At present, the research results of susceptibility genes screened by candidate gene association research are not consistent. The method of gene recognition on DNA was studied by QCM and nano gold composite. By using this method, the enantioselective recognition of cysteine on cyclodextrin self-assembled membrane was studied. In this study, EAAT1 gene, which is highly expressed in astrocytes, was used as a candidate gene to analyze the relationship between polymorphism and schizophrenia. The experimental results show that the introduction of nano gold can significantly improve the sensing signal, detection sensitivity and gene differentiation. In addition, this study suggested that EAAT1 gene might be a susceptibility gene of schizophrenia in the population. The results showed that a common SNP allele rs1030239-g was the risk factor (83.8% vs. 79.2%,P= 0.00067, or = 1.35, 95% CI = 1.08-1.69). The results showed that A-T-G increased the risk of schizophrenia.
- Published
- 2021
- Full Text
- View/download PDF
16. Ataxia-linked SLC1A3 mutations alter EAAT1 chloride channel activity and glial regulation of CNS function
- Author
-
Alastair Garner, Jin Xin Zhu, Qianyi Wu, Emad Tajkhorshid, Don J. Van Meyel, Shashank Pant, Renae M. Ryan, Tomoko Ohyama, Eunjoo Cho, and Azman Akhter
- Subjects
Xenopus ,Biophysics ,Glutamic Acid ,Neurotransmission ,Channelopathy ,Chloride Channels ,medicine ,Animals ,Humans ,Chloride channel activity ,Mammals ,Episodic ataxia ,biology ,Chemistry ,Glutamate receptor ,Neurotoxicity ,General Medicine ,medicine.disease ,biology.organism_classification ,Cell biology ,Excitatory Amino Acid Transporter 1 ,Drosophila melanogaster ,Mutation ,Excitatory postsynaptic potential ,Ataxia ,Neuroglia - Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). Excitatory Amino Acid Transporters (EAATs) regulate extracellular glutamate by transporting it into cells, mostly glia, to terminate neurotransmission and to avoid neurotoxicity. EAATs are also chloride (Cl−) channels, but the physiological role of Cl− conductance through EAATs is poorly understood. Mutations of human EAAT1 (hEAAT1) have been identified in patients with episodic ataxia type 6 (EA6). One mutation showed increased Cl− channel activity and decreased glutamate transport, but the relative contributions of each function of hEAAT1 to mechanisms underlying the pathology of EA6 remain unclear. Here we investigated the effects of five additional EA6-related mutations on hEAAT1 function in Xenopus laevis oocytes, and on CNS function in a Drosophila melanogaster model of locomotor behavior. Our results indicate that mutations with decreased hEAAT1 Cl− channel activity and functional glutamate transport can also contribute to the pathology of EA6, highlighting the importance of Cl− homeostasis in glial cells for proper CNS function. We also identified a novel mechanism involving an ectopic sodium (Na+) leak conductance in glial cells. Together, these results strongly support the idea that EA6 is primarily an ion channelopathy of CNS glia.
- Published
- 2022
- Full Text
- View/download PDF
17. Effect of early weaning on the expression of excitatory amino acid transporter 1 in the jejunum and ileum of piglets.
- Author
-
XIU‑YING ZHANG, QIU‑JU WANG, YI‑ZHE CUI, and JING SU
- Subjects
- *
ANIMAL weaning , *PIGLETS , *AMINO acids , *JEJUNUM , *ILEUM - Abstract
The present study aimed to compare the expression levels of excitatory amino acid transporters (EAATs) and growth status of piglets weaned at 10‑20 days after birth with suckling piglets. A total of 40 hybrid piglets (Landrace x Large White x Duroc) born to 40 different sows, with similar body weight were selected for the present study. They were randomly divided into two groups (n=20 per group): Control group (suckling piglets) and experimental group (weaned piglets, reared in isolation). The experiment lasted for 10 days. At the end of the experiment, 12 piglets were randomly selected from each group and the jejunum and the ileum were collected in order to determine excitatory amino acid carrier 1 (EAAC1) expression levels and free amino acid content. The present study determined that early weaning significantly reduced EAAC1 gene and protein (57 and 73 kDa) expression levels and glutamate transporter associate protein 3‑18 (GTRAP3‑18; 50 kDa) in the jejunum and the ileum compared with the suckling group (P<0.05). Weaning led to an increased content of free glutamic acid (Glu) and total amino acids in the jejunum; however, content of free Glu and total amino acids in the ileum was significantly reduced (P<0.05). Early weaning reduced the expression of EAAC1 and GTRAP3‑18, which was possibly due to the amino acid absorption and transport disorder in the small intestine due to the Glu deficiency. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
18. Neuroprotective Potential of L-Glutamate Transporters in Human Induced Pluripotent Stem Cell-Derived Neural Cells against Excitotoxicity.
- Author
-
Takahashi K, Ishibashi Y, Chujo K, Suzuki I, and Sato K
- Subjects
- Humans, Neurons, Amino Acid Transport System X-AG, Biological Transport, Excitatory Amino Acid Transporter 1, Glutamic Acid toxicity, Induced Pluripotent Stem Cells
- Abstract
Human induced pluripotent stem cell (hiPSC)-derived neural cells have started to be used in safety/toxicity tests at the preclinical stage of drug development. As previously reported, hiPSC-derived neurons exhibit greater tolerance to excitotoxicity than those of primary cultures of rodent neurons; however, the underlying mechanisms remain unknown. We here investigated the functions of L-glutamate (L-Glu) transporters, the most important machinery to maintain low extracellular L-Glu concentrations, in hiPSC-derived neural cells. We also clarified the contribution of respective L-Glu transporter subtypes. At 63 days in vitro (DIV), we detected neuronal circuit functions in hiPSC-derived neural cells by a microelectrode array system (MEA). At 63 DIV, exposure to 100 μM L-Glu for 24 h did not affect the viability of neural cells. 100 µM L-Glu in the medium decreased to almost 0 μM in 60 min. Pharmacological inhibition of excitatory amino acid transporter 1 (EAAT1) and EAAT2 suppressed almost 100% of L-Glu decrease. In the presence of this inhibitor, 100 μM L-Glu dramatically decreased cell viability. These results suggest that in hiPSC-derived neural cells, EAAT1 and EAAT2 are the predominant L-Glu transporters, and their uptake potentials are the reasons for the tolerance of hiPSC-derived neurons to excitotoxicity.
- Published
- 2023
- Full Text
- View/download PDF
19. Brain Tumor Stem Cell Dependence on Glutaminase Reveals a Metabolic Vulnerability through the Amino Acid Deprivation Response Pathway
- Author
-
Samuel Weiss, H. Artee Luchman, Trevor J. Pugh, Ian Restall, Laura M. Richards, and Orsolya Cseh
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Citric Acid Cycle ,Population ,Benzeneacetamides ,Glutamic Acid ,03 medical and health sciences ,0302 clinical medicine ,Glutaminase ,Cancer stem cell ,Cell Line, Tumor ,Thiadiazoles ,Humans ,Amino Acids ,education ,Cell Proliferation ,education.field_of_study ,Glutaminolysis ,Brain Neoplasms ,Cell growth ,Chemistry ,Cell biology ,Excitatory Amino Acid Transporter 1 ,Glutamine ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,Oncology ,Astrocytes ,030220 oncology & carcinogenesis ,Cancer cell ,Neoplastic Stem Cells ,Stem cell ,Glioblastoma ,Signal Transduction - Abstract
Cancer cells can metabolize glutamine to replenish TCA cycle intermediates, leading to a dependence on glutaminolysis for cell survival. However, a mechanistic understanding of the role that glutamine metabolism has on the survival of glioblastoma (GBM) brain tumor stem cells (BTSC) has not yet been elucidated. Here, we report that across a panel of 19 GBM BTSC lines, inhibition of glutaminase (GLS) showed a variable response from complete blockade of cell growth to absolute resistance. Surprisingly, BTSC sensitivity to GLS inhibition was a result of reduced intracellular glutamate triggering the amino acid deprivation response (AADR) and not due to the contribution of glutaminolysis to the TCA cycle. Moreover, BTSC sensitivity to GLS inhibition negatively correlated with expression of the astrocytic glutamate transporters EAAT1 and EAAT2. Blocking glutamate transport in BTSCs with high EAAT1/EAAT2 expression rendered cells susceptible to GLS inhibition, triggering the AADR and limiting cell growth. These findings uncover a unique metabolic vulnerability in BTSCs and support the therapeutic targeting of upstream activators and downstream effectors of the AADR pathway in GBM. Moreover, they demonstrate that gene expression patterns reflecting the cellular hierarchy of the tissue of origin can alter the metabolic requirements of the cancer stem cell population. Significance: Glioblastoma brain tumor stem cells with low astrocytic glutamate transporter expression are dependent on GLS to maintain intracellular glutamate to prevent the amino acid deprivation response and cell death.
- Published
- 2020
- Full Text
- View/download PDF
20. Conventional immunomarkers stain a fraction of astrocytes in vitro : A comparison of rat cortical and spinal cord astrocytes in naïve and stimulated cultures
- Author
-
Jessica L Funnell, Manoj K. Gottipati, Kathy Lin, Ryan J. Gilbert, Bailey Balouch, Alexis M. Ziemba, and Devan L Puhl
- Subjects
0301 basic medicine ,Primary Cell Culture ,Central nervous system ,Rats, Sprague-Dawley ,Transforming Growth Factor beta1 ,Cerebellar Cortex ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Transforming Growth Factor beta3 ,0302 clinical medicine ,Glutamate-Ammonia Ligase ,Glial Fibrillary Acidic Protein ,medicine ,Glutamate aspartate transporter ,Animals ,Oxidoreductases Acting on CH-NH Group Donors ,Glial fibrillary acidic protein ,biology ,Microglia ,Glutamate receptor ,Brain ,SOX9 Transcription Factor ,medicine.disease ,Molecular biology ,Oligodendrocyte ,Rats ,Astrogliosis ,Excitatory Amino Acid Transporter 1 ,Oligodendroglia ,030104 developmental biology ,medicine.anatomical_structure ,Animals, Newborn ,Excitatory Amino Acid Transporter 2 ,Spinal Cord ,Astrocytes ,biology.protein ,Neuroglia ,030217 neurology & neurosurgery ,Astrocyte - Abstract
Astrocytes are responsible for a wide variety of essential functions throughout the central nervous system. The protein markers glial fibrillary acidic protein (GFAP), glutamate aspartate transporter (GLAST), glutamate transporter-1 (GLT-1), glutamine synthetase (GS), 10-formyltetrahydrofolate dehydrogenase (ALDH1L1), and the transcription factor SOX9 are routinely used to label astrocytes in primary rodent cultures. However, GLAST, GLT-1, GS, and SOX9 are also produced by microglia and oligodendrocytes and GFAP, GLAST, GLT-1, and GS production levels are affected by astrocyte phenotypic changes associated with reactive astrogliosis. No group has performed a comprehensive immunocytochemical evaluation to quantify the percentage of cells labeled by these markers in vitro, nor compared changes in staining between cortex- and spinal cord-derived cells in naïve and stimulated cultures. Here, we quantified the percentage of cells positively stained for these six markers in astrocyte, microglia, and oligodendrocyte cultures isolated from neonatal rat cortices and spinal cords. Additionally, we incubated the astrocytes with transforming growth factor (TGF)-β1 or TGF-β3 to determine if the labeling of these markers is altered by these stimuli. We found that only SOX9 in cortical cultures and ALDH1L1 in spinal cord cultures labeled more than 75% of the cells in naïve and stimulated astrocyte cultures and stained less than 5% of the cells in microglia and oligodendrocyte cultures. Furthermore, significantly more cortical than spinal cord astrocytes stained for GFAP, GLAST, and ALDH1L1 in naïve cultures, whereas significantly more spinal cord than cortical astrocytes stained for GLAST and GS in TGF-β1-treated cultures. These findings are important as variability in marker staining may lead to misinterpretation of the astrocyte response in cocultures, migration assays, or engineered disease models.
- Published
- 2020
- Full Text
- View/download PDF
21. Astrocyte-specific deletion of the transcription factor Yin Yang 1 in murine substantia nigra mitigates manganese-induced dopaminergic neurotoxicity
- Author
-
Eun Sook Lee, Julia Bornhorst, Tania Schwerdtle, Asha Rizor, Deok Soo Son, Michael Aschner, Ivan Nyarko-Danquah, Edward Alain B. Pajarillo, Getinet Adinew, Michael Stiboller, and James Johnson
- Subjects
Male ,0301 basic medicine ,Tyrosine 3-Monooxygenase ,Down-Regulation ,Substantia nigra ,Striatum ,Biochemistry ,Histone Deacetylases ,Mice ,03 medical and health sciences ,Chlorides ,Conditional gene knockout ,medicine ,Animals ,RNA, Messenger ,Molecular Biology ,YY1 Transcription Factor ,Mice, Knockout ,030102 biochemistry & molecular biology ,Tyrosine hydroxylase ,Chemistry ,Manganese Poisoning ,Dopaminergic ,Glutamate receptor ,Neurotoxicity ,Molecular Bases of Disease ,Cell Biology ,medicine.disease ,Molecular biology ,Excitatory Amino Acid Transporter 1 ,Substantia Nigra ,030104 developmental biology ,medicine.anatomical_structure ,Excitatory Amino Acid Transporter 2 ,Manganese Compounds ,Astrocytes ,embryonic structures ,Female ,Locomotion ,Astrocyte - Abstract
Manganese (Mn)-induced neurotoxicity resembles Parkinson's disease (PD), but the mechanisms underpinning its effects remain unknown. Mn dysregulates astrocytic glutamate transporters, GLT-1 and GLAST, and dopaminergic function, including tyrosine hydroxylase (TH). Our previous in vitro studies have shown that Mn repressed GLAST and GLT-1 via activation of transcription factor Yin Yang 1 (YY1). Here, we investigated if in vivo astrocytic YY1 deletion mitigates Mn-induced dopaminergic neurotoxicity, attenuating Mn-induced reduction in GLAST/GLT-1 expression in murine substantia nigra (SN). AAV5-GFAP-Cre-GFP particles were infused into the SN of 8-week–old YY1(flox/flox) mice to generate a region-specific astrocytic YY1 conditional knockout (cKO) mouse model. 3 weeks after adeno-associated viral (AAV) infusion, mice were exposed to 330 μg of Mn (MnCl(2) 30 mg/kg, intranasal instillation, daily) for 3 weeks. After Mn exposure, motor functions were determined in open-field and rotarod tests, followed by Western blotting, quantitative PCR, and immunohistochemistry to assess YY1, TH, GLAST, and GLT-1 levels. Infusion of AAV5-GFAP-Cre-GFP vectors into the SN resulted in region-specific astrocytic YY1 deletion and attenuation of Mn-induced impairment of motor functions, reduction of TH-expressing cells in SN, and TH mRNA/protein levels in midbrain/striatum. Astrocytic YY1 deletion also attenuated the Mn-induced decrease in GLAST/GLT-1 mRNA/protein levels in midbrain. Moreover, YY1 deletion abrogated its interaction with histone deacetylases in astrocytes. These results indicate that astrocytic YY1 plays a critical role in Mn-induced neurotoxicity in vivo, at least in part, by reducing astrocytic GLAST/GLT-1. Thus, YY1 might be a potential target for treatment of Mn toxicity and other neurological disorders associated with dysregulation of GLAST/GLT-1.
- Published
- 2020
- Full Text
- View/download PDF
22. Potential Mechanism of Cellular Uptake of the Excitotoxin Quinolinic Acid in Primary Human Neurons
- Author
-
Hayden Alicajic, David V. Pow, Gilles J. Guillemin, Joseph A. Nicolazzo, Bruce J. Brew, Bat-Erdene Jugder, Nady Braidy, and Jason R. Smith
- Subjects
Models, Molecular ,0301 basic medicine ,Time Factors ,Kynurenine pathway ,Neurotoxins ,Neuroscience (miscellaneous) ,Excitotoxicity ,medicine.disease_cause ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Fetus ,0302 clinical medicine ,Lysosomal-Associated Membrane Protein 2 ,medicine ,Humans ,Cells, Cultured ,Neurons ,chemistry.chemical_classification ,LAMP2 ,Chemistry ,Glutamate receptor ,Transporter ,Quinolinic Acid ,Endocytosis ,Amino acid ,Excitatory Amino Acid Transporter 1 ,Kinetics ,Excitatory Amino Acid Transporter 3 ,030104 developmental biology ,Neurology ,Excitatory postsynaptic potential ,Biophysics ,030217 neurology & neurosurgery ,Quinolinic acid - Abstract
In Alzheimer’s disease (AD), excessive amounts of quinolinic acid (QUIN) accumulate within the brain parenchyma and dystrophic neurons. QUIN also regulates glutamate uptake into neurons, which may be due to modulation of Na+-dependent excitatory amino acid transporters (EAATs). To determine the biological relationships between QUIN and glutamate dysfunction, we first quantified the functionality and kinetics of [3H]QUIN uptake in primary human neurons using liquid scintillation. We then measured changes in the protein expression of the glutamate transporter EAAT3 and EAAT1b in primary neurons treated with QUIN and the EAAT inhibitor l-trans-pyrrolidine-2,4-dicarboxylic acid (2,4-PDC) using western blotting and immunohistochemistry. Immunohistochemistry was further used to elucidate intracellular transport of exogenous QUIN and the lysosomal-associated membrane protein 2 (LAMP2). Structural insights into the binding between QUIN and EAAT3 were further investigated using molecular docking techniques. We report significant temperature-dependent high-affinity transport leading to neuronal uptake of [3H]QUIN with a Km of 42.2 μM, and a Vmax of 9.492 pmol/2 min/mg protein, comparable with the uptake of glutamate. We also found that QUIN increases expression of the EAAT3 monomer while decreasing the functional trimer. QUIN uptake into primary neurons was shown to involve EAAT3 as uptake was significantly attenuated following EAAT inhibition. We also demonstrated that QUIN increases the expression of aberrant EAAT1b protein in neurons further implicating QUIN-induced glutamate dysfunction. Furthermore, we demonstrated that QUIN is metabolised exclusively in lysosomes. The involvement of EAAT3 as a modulator for QUIN uptake was further confirmed using molecular docking. This study is the first to characterise a mechanism for QUIN uptake into primary human neurons involving EAAT3, opening potential targets to attenuate QUIN-induced excitotoxicity in neuroinflammatory diseases.
- Published
- 2020
- Full Text
- View/download PDF
23. EAAT1 variants associated with glaucoma
- Author
-
Michiko Yanagisawa, Kazuhiko Namekata, Tomomi Aida, Sayaka Katou, Takuya Takeda, Takayuki Harada, Nobuo Fuse, null the Glaucoma Gene Research Group, and Kohichi Tanaka
- Subjects
Retinal Ganglion Cells ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,genetic structures ,Mutation, Missense ,Biophysics ,Gene Expression ,Glaucoma ,Biochemistry ,Retinal ganglion ,Cell Line ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Risk Factors ,medicine ,Animals ,Humans ,Missense mutation ,Amino Acid Sequence ,Low Tension Glaucoma ,Molecular Biology ,Allele frequency ,Gene ,Alleles ,Intraocular Pressure ,Silent Mutation ,Mice, Knockout ,Retina ,Sequence Homology, Amino Acid ,business.industry ,Optic Nerve ,Cell Biology ,medicine.disease ,eye diseases ,Excitatory Amino Acid Transporter 1 ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,030220 oncology & carcinogenesis ,Knockout mouse ,sense organs ,business ,Sequence Alignment ,Glaucoma, Open-Angle - Abstract
Glaucoma is one of the leading causes of blindness characterized by progressive loss of retinal ganglion cells (RGCs) and their axons. We reported that glutamate/aspartate transporter (GLAST) knockout mice showed progressive RGC loss and optic nerve degeneration that are similar to glaucoma. To explore the possibility that rare variants in the EAAT1 gene (the human homolog of GLAST) cause susceptibility to glaucoma, we performed targeted sequencing of EAAT1 in 440 patients with glaucoma and 450 control subjects. We identified 8 rare variants in 20 out of 440 patients, including 4 synonymous and 4 missense variants located at protein coding regions. One of these rare variants (rs117295512) showed significant association with the risk of glaucoma (OR = 10.44, P = 0.005). Furthermore, the allele frequency for loss-of-function EAAT1 variants, pAla169Gly and pAla329Thr, was 5.5 folds higher in the glaucoma (1.1%) compared with the control cohort (0.2%). These findings suggest that these rare variants may contribute to the pathogenesis of glaucoma and that loss-of-function variants in EAAT1 are present in a small number of patients with glaucoma.
- Published
- 2020
- Full Text
- View/download PDF
24. Circadian vulnerability of cisplatin‐induced ototoxicity in the cochlea
- Author
-
Evangelia Tserga, Erik Björn, Christian Göritz, Rocio Moreno‐Paublete, Heela Sarlus, Christopher R. Cederroth, Eduardo L Guimarães, and Barbara Canlon
- Subjects
Male ,0301 basic medicine ,afferent synapse ,Oto-rino-laryngologi ,cisplatin ototoxicity ,Pharmacology ,Biochemistry ,Synapse ,Mice ,0302 clinical medicine ,Glutamate aspartate transporter ,biology ,PER2 ,Circadian Rhythm ,Cochlea ,3. Good health ,Excitatory Amino Acid Transporter 1 ,Female ,medicine.symptom ,Biotechnology ,medicine.drug ,Hearing loss ,Antineoplastic Agents ,GLAST ,Article ,03 medical and health sciences ,Ototoxicity ,Evoked Potentials, Auditory, Brain Stem ,Genetics ,medicine ,Animals ,Circadian rhythm ,Molecular Biology ,hearing loss ,Cisplatin ,business.industry ,Auditory Threshold ,medicine.disease ,Mice, Inbred C57BL ,circadian ,030104 developmental biology ,Otorhinolaryngology ,biology.protein ,business ,synaptic pairing ,030217 neurology & neurosurgery - Abstract
The chemotherapeutic agent cisplatin is renowned for its ototoxic effects. While hair cells in the cochlea are established targets of cisplatin, less is known regarding the afferent synapse, which is an essential component in the faithful temporal transmission of sound. The glutamate aspartate transporter (GLAST) shields the auditory synapse from excessive glutamate release, and its loss of function increases the vulnerability to noise, salicylate, and aminoglycosides. Until now, the involvement of GLAST in cisplatin-mediated ototoxicity remains unknown. Here, we test in mice lacking GLAST the effects of a low-dose cisplatin known not to cause any detectable change in hearing thresholds. When administered at nighttime, a mild hearing loss in GLAST KO mice was found but not at daytime, revealing a potential circadian regulation of the vulnerability to cisplatin-mediated ototoxicity. We show that the auditory synapse of GLAST KO mice is more vulnerable to cisplatin administration during the active phase (nighttime) when compared to WT mice and treatment during the inactive phase (daytime). This effect was not related to the abundance of platinum compounds in the cochlea, rather cisplatin had a dose-dependent impact on cochlear clock rhythms only after treatment at nighttime suggesting that cisplatin can modulate the molecular clock. Our findings suggest that the current protocols of cisplatin administration in humans during daytime may cause a yet undetectable damage to the auditory synapse, more so in already damaged ears, and severely impact auditory sensitivity in cancer survivors.
- Published
- 2020
- Full Text
- View/download PDF
25. Fluoxetine and Riluzole Mitigates Manganese-Induced Disruption of Glutamate Transporters and Excitotoxicity via Ephrin-A3/GLAST-GLT-1/Glu Signaling Pathway in Striatum of Mice
- Author
-
Yanan Liu, Miao He, Wei Liu, Bin Xu, Yanqi Sang, Jinghai Zhu, Zhaofa Xu, Zhipeng Qi, Yu Deng, Jiashuo Li, and Xinxin Yang
- Subjects
0301 basic medicine ,Excitotoxicity ,Glutamic Acid ,Striatum ,Pharmacology ,Toxicology ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Fluoxetine ,medicine ,Animals ,Manganese ,Messenger RNA ,Riluzole ,Chemistry ,General Neuroscience ,Glutamate receptor ,Corpus Striatum ,Ephrin-A3 ,Excitatory Amino Acid Transporter 1 ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,Signal transduction ,Ephrin A3 ,Excitatory Amino Acid Antagonists ,Selective Serotonin Reuptake Inhibitors ,030217 neurology & neurosurgery ,Signal Transduction ,medicine.drug - Abstract
Manganese (Mn) is an essential element required for many biological processes and systems in the human body. Mn intoxication increases brain glutamate (Glu) levels causing neuronal damage. Recent studies have reported that ephrin-A3 regulates this glutamate transporter. However, none has explored the role of this crucial molecule in Mn-induced excitotoxicity. The present study investigated whether ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity using astrocytes and Kunming mice. The mechanisms were explored using fluoxetine (ephrin-A3 inhibitor) and riluzole (a Glu release inhibitor). Firstly, we demonstrated that Mn exposure (500 μM or 50 mg/kg MnCl2) significantly increased Mn, ephrin-A3, and Glu levels, and inhibited Na+-K+ ATPase activity, as well as mRNA and protein levels of GLAST and GLT-1. Secondly, we found that astrocytes and mice pretreated with fluoxetine (100 μM or 15 mg/kg) and riluzole (100 μM or 32 μmol/kg) prior to Mn exposure had lower ephrin-A3 and Glu levels, but higher Na+-K+ ATPase activity, expression levels of GLAST and GLT-1 than those exposed to 500 μM or 50 mg/kg MnCl2. Moreover, the morphology of cells and the histomorphology of mice striatum were injured. Results showed that pretreatment with fluoxetine and riluzole attenuated the Mn-induced motor dysfunctions. Together, these results suggest that the ephrin-A3/GLAST-GLT-1/Glu signaling pathway participates in Mn-induced excitotoxicity, and fluoxetine and riluzole can mitigate the Mn-induced excitotoxicity in mice brain.
- Published
- 2020
- Full Text
- View/download PDF
26. Effects of Cocaine Exposure on Astrocytic Glutamate Transporters and Relapse-Like Ethanol-Drinking Behavior in Male Alcohol-Preferring Rats
- Author
-
Alaa M. Hammad and Youssef Sari
- Subjects
Male ,0301 basic medicine ,Alcohol Drinking ,Amino Acid Transport System X-AG ,Blotting, Western ,Cystine ,Nucleus accumbens ,Pharmacology ,Article ,Reuptake ,Acetylcysteine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cocaine ,Glutamate homeostasis ,Recurrence ,medicine ,Animals ,Neurotransmitter ,Ethanol ,Chemistry ,Glutamate receptor ,Brain ,General Medicine ,Rats ,Excitatory Amino Acid Transporter 1 ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,Astrocytes ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Aim Glutamate has been considered as neurotransmitter that is critical in triggering relapse to drugs of abuse, including ethanol and cocaine. Extracellular glutamate concentrations are tightly regulated by several mechanisms, including reuptake through glutamate transporters. Glutamate transporter type 1 (GLT-1) is responsible for clearing the majority of extracellular glutamate. The astrocytic cystine/glutamate antiporter (xCT) regulates also glutamate homeostasis. In this study, we investigated the effects of cocaine exposure and ampicillin/sulbactam (AMP/SUL), a β-lactam antibiotic known to upregulate GLT-1 and xCT, on relapse-like ethanol intake and the expression of astrocytic glutamate transporters in mesocorticolimbic brain regions. Methods Male alcohol-preferring (P) rats had free access to ethanol for 5 weeks. On Week 6, rats were exposed to either cocaine (20 mg/kg, i.p.) or saline for 12 consecutive days. Ethanol bottles were then removed for 7 days; during the last 5 days, either AMP/SUL (100 or 200 mg/kg, i.p.) or saline was administered to the P rats. Ethanol bottles were reintroduced, and ethanol intake was measured for 4 days. Results Cocaine exposure induced an alcohol deprivation effect (ADE), which was associated in part by a decrease in the expression of GLT-1 and xCT in the nucleus accumbens (NAc) core. AMP/SUL (100 mg/kg, i.p.) attenuated the ADE, while AMP/SUL (200 mg/kg, i.p.) reduced ethanol intake during 4 days of ethanol re-exposure and upregulated GLT-1 and xCT expression in the NAc core, NAc shell and dorsomedial prefrontal cortex (dmPFC). Conclusion This study suggests that these astrocytic glutamate transporters might be considered as potential targets for the treatment of polysubstance abuse.
- Published
- 2020
- Full Text
- View/download PDF
27. Protocol for purification and culture of astrocytes: useful not only in 2 days postnatal but also in adult rat brain
- Author
-
Sahar Kiani, Sara Mirsadeghi, Masoumeh Zarei-Kheirabadi, Alexander R. Vaccaro, and Vafa Rahimi-Movaghar
- Subjects
0301 basic medicine ,Patch-Clamp Techniques ,Primary Cell Culture ,Population ,Cell Separation ,S100 Calcium Binding Protein beta Subunit ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Glial Fibrillary Acidic Protein ,Genetics ,medicine ,Animals ,education ,Molecular Biology ,Cells, Cultured ,education.field_of_study ,Meninges ,Brain ,General Medicine ,Rat brain ,Oligodendrocyte ,Pathophysiology ,Rats ,Cell biology ,Excitatory Amino Acid Transporter 1 ,030104 developmental biology ,medicine.anatomical_structure ,Animals, Newborn ,Cell culture ,Cerebral cortex ,Astrocytes ,030220 oncology & carcinogenesis ,Astrocyte - Abstract
Astrocytes play the key roles in the physiology and pathology of the CNS. Thereupon, in this manuscript, we aim to demonstrate that the protocol for purification and culture of astrocytes is useful not only in 2 days postnatal but also in adult rat brain. Also, the mentioned protocol is a simple and efficient primary cell culture technique. The whole-brain was isolated from the skull and the meninges were removed carefully. Afterward, the cerebral hemispheres were mechanically and enzymatically digested. Then, the cell suspension was seeded in T25 culture flask and was incubated at 37 °C in the CO2 incubator. The first shaking was performed after 7–8 days and on day 14, second shaking was done. After 2–3 passage, the culture was analyzed. By passaging, the majority of extracted cells were astrocytes presenting with a polygonal to fusiform and flat morphology that expressed GFAP, GLAST, and S100β. The expression of neural, neuronal and oligodendrocyte markers was not detected in extracted cells. The patch-clamp recording comfirmed the purity of isolated astrocytes as well. The isolated cells from adult rat brain were astrocytes that expressed specific astrocyte markers after 3 and 10 passages. This method is suggested to obtain a population of astrocytes that may provide a beneficial tool for different neurophysiological and pathophysiological studies.
- Published
- 2020
- Full Text
- View/download PDF
28. The ion‐coupling mechanism of human excitatory amino acid transporters
- Author
-
Julia Chamot-Rooke, Jonathan Dhenin, Pierre Legrand, Martial Rey, Nicolas Reyes, Anand Kumar, Reda Assal, Juan Carlos Canul-Tec, Mécanismes des Protéines Membranaires - Membrane Protein Mechanisms, Institut Pasteur [Paris] (IP), Institut Européen de Chimie et de Biologie, Microbiologie Fondamentale et Pathogénicité (MFP), Université Bordeaux Segalen - Bordeaux 2-Centre National de la Recherche Scientifique (CNRS), Spectrométrie de Masse pour la Biologie – Mass Spectrometry for Biology (UTechS MSBio), Institut Pasteur [Paris] (IP)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Synchrotron SOLEIL (SSOLEIL), Centre National de la Recherche Scientifique (CNRS), The work was funded by the European Research Council (ERC grant 309657 to NR). JCC-T was partly supported by a Pasteur-Roux Postdoctoral Fellowship. Further support from Institut Pasteur, INCA 2017-44 Grant, CACSICE grant (ANR-11-EQPX-008), and CNRS UMR3528 to NR and JC-R is acknowledged., The authors thank Ahmed Haouz and the staff at the crystallogenesis core facility of the Institut Pasteur for assistance with crystallization screens and staff at Synchrotron Soleil and the European Synchrotron Radiation Facility for assistance with data collection. The IECB cryoEM imaging facility is acknowledged for support in cryo-EM sample screening and initial data acquisition, the EMBL-Heildelberg Cryo-Electron Microscopy Service Platform for support in cryoEM data collection., ANR-11-EQPX-0008,CACSICE,Centre d'analyse de systèmes complexes dans les environnements complexes(2011), and European Project: 309657,EC:FP7:ERC,ERC-2012-StG_20111109,HEAATS(2012)
- Subjects
Models, Molecular ,Cations, Divalent ,Protein Conformation ,solute carrier ,neurotransmitter transport ,Neurotransmission ,Biology ,permeation and transport ,General Biochemistry, Genetics and Molecular Biology ,Neurotransmitter binding ,03 medical and health sciences ,0302 clinical medicine ,Ion binding ,Humans ,Molecular Biology ,030304 developmental biology ,X-ray crystallography ,chemistry.chemical_classification ,0303 health sciences ,Binding Sites ,Ion Transport ,General Immunology and Microbiology ,[SDV.BBM.BS]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] ,General Neuroscience ,Cryoelectron Microscopy ,Sodium ,Glutamate receptor ,Articles ,Amino acid ,Excitatory Amino Acid Transporter 1 ,chemistry ,Biophysics ,Excitatory postsynaptic potential ,cryo-EM ,Calcium ,Protons ,Neurotransmitter transport ,Energy source ,030217 neurology & neurosurgery - Abstract
International audience; Excitatory amino acid transporters (EAATs) maintain glutamate gradients in the brain essential for neurotransmission and to prevent neuronal death. They use ionic gradients as energy source and co-transport transmitter into the cytoplasm with Na+ and H+, while counter-transporting K+ to re-initiate the transport cycle. However, the molecular mechanisms underlying ion-coupled transport remain incompletely understood. Here, we present 3D X-ray crystallographic and cryo-EM structures, as well as thermodynamic analysis of human EAAT1 in different ion bound conformations, including elusive counter-transport ion bound states. Binding energies of Na+ and H+, and unexpectedly Ca2+, are coupled to neurotransmitter binding. Ca2+ competes for a conserved Na+ site, suggesting a regulatory role for Ca2+ in glutamate transport at the synapse, while H+ binds to a conserved glutamate residue stabilizing substrate occlusion. The counter-transported ion binding site overlaps with that of glutamate, revealing the K+-based mechanism to exclude the transmitter during the transport cycle and to prevent its neurotoxic release on the extracellular side.
- Published
- 2022
- Full Text
- View/download PDF
29. Molecular Basis of Coupled Transport and Anion Conduction in Excitatory Amino Acid Transporters
- Author
-
Daniel Kortzak, Christoph Fahlke, Ingo Weyand, Jan-Philipp Machtens, and Claudia Alleva
- Subjects
Anions ,Synaptic cleft ,Amino Acid Transport System X-AG ,Glutamic Acid ,Biochemistry ,Glutamate Plasma Membrane Transport Proteins ,Cellular and Molecular Neuroscience ,Animals ,ddc:610 ,EAAT anion channel ,Fluorescence spectroscopy ,Mammals ,Induced fit ,chemistry.chemical_classification ,Original Paper ,MD simulations ,Tryptophan ,Glutamate receptor ,Biological Transport ,Transporter ,General Medicine ,Hairpin 2 ,Dual function protein ,Transport stoichiometry ,Amino acid ,Excitatory Amino Acid Transporter 1 ,Coupled transport ,chemistry ,Conformational selection ,Symporter ,GltPh ,Excitatory postsynaptic potential ,Biophysics ,Heterologous expression ,Glutamate transporter - Abstract
Neurochemical research 47(1), 9-22 (2022). doi:10.1007/s11064-021-03252-x special issue: "Special Issue in Honor of Baruch Kanner", Published by Springer Science + Business Media B.V, Dordrecht [u.a.]
- Published
- 2022
- Full Text
- View/download PDF
30. Microscopic Characterization of the Chloride Permeation Pathway in the Human Excitatory Amino Acid Transporter 1 (EAAT1)
- Author
-
Shashank Pant, Qianyi Wu, Renae Ryan, and Emad Tajkhorshid
- Subjects
chemistry.chemical_classification ,Aqueous solution ,Physiology ,Cognitive Neuroscience ,Sodium ,Glutamic Acid ,Substrate (chemistry) ,Conductance ,Transporter ,Cell Biology ,General Medicine ,Biochemistry ,Article ,Solute carrier family ,Amino acid ,Excitatory Amino Acid Transporter 1 ,Molecular dynamics ,Excitatory Amino Acid Transporter 3 ,Membrane ,Chlorides ,Excitatory Amino Acid Transporter 2 ,chemistry ,Biophysics ,Humans - Abstract
Excitatory amino acid transporters (EAATs) are glutamate transporters that belong to the solute carrier 1A (SLC1A) family. They couple glutamate transport to the co-transport of three sodium (Na+) ions and one proton (H+) and the counter-transport of one potassium (K+) ion. In addition to this coupled transport, binding of substrate and Na+ ions to EAATs activates a thermodynamically uncoupled chloride (Cl−) conductance. Structures of SLC1A family members have revealed that these transporters use a twisting elevator mechanism of transport, where a mobile transport domain carries substrate and coupled ions across the membrane, while a static scaffold domain anchors the transporter in the membrane. We have recently demonstrated that the uncoupled Cl− conductance is activated by the formation of an aqueous pore at the domain interface during the transport cycle in archaeal GltPh. However, a pathway for the uncoupled Cl− conductance has not been reported for the EAATs and it is unclear if such a pathway is conserved. Here, we employ all-atom molecular dynamics (MD) simulations combined with enhanced sampling, free-energy calculations, and experimental mutagenesis to approximate large-scale conformational changes during the transport process and identified a Cl− conducting conformation in human EAAT1. We were able to extensively sample the large-scale structural transitions, allowing us to capture an intermediate conformation formed during the transport cycle with a continuous aqueous pore at the domain interface. The free-energy calculations performed for the conduction of Cl− and Na+ ions through the captured conformation, highlight the presence of two hydrophobic gates which control the selective movement of Cl− through the aqueous pathway. Overall, our findings provide insights into the mechanism by which a human glutamate transporter can support the dual functions of active transport and passive Cl− permeation and confirming the commonality of this mechanism in different members of the SLC1A family.
- Published
- 2021
- Full Text
- View/download PDF
31. Reduction of Glutamate Neurotoxicity: A Novel Therapeutic Approach for Niemann-Pick disease, type C1
- Author
-
Christopher A. Wassif, Antony Cougnoux, Niamh X. Cawley, Fatemeh Navid, Jenny Serra-Vinardell, Forbes D. Porter, Julia C Yerger, and Mason Fellmeth
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Endocrinology, Diabetes and Metabolism ,Glutamic Acid ,Pharmacology ,Biochemistry ,Article ,chemistry.chemical_compound ,Mice ,Endocrinology ,Niemann-Pick C1 Protein ,hemic and lymphatic diseases ,Genetics ,medicine ,Animals ,Humans ,Glutamate receptor antagonist ,Amyotrophic lateral sclerosis ,Molecular Biology ,Cells, Cultured ,Mice, Inbred BALB C ,Riluzole ,biology ,Cerebellar ataxia ,business.industry ,Ceftriaxone ,SLC1A2 ,Glutamate receptor ,Neurotoxicity ,nutritional and metabolic diseases ,Niemann-Pick Disease, Type C ,medicine.disease ,Excitatory Amino Acid Transporter 1 ,Disease Models, Animal ,chemistry ,Astrocytes ,biology.protein ,Female ,NPC1 ,medicine.symptom ,business ,medicine.drug - Abstract
Niemann-Pick disease, type C1 is a progressive, lethal, neurodegenerative disorder due to endolysosomal storage of unesterified cholesterol. Cerebellar ataxia, as a result of progressive loss of cerebellar Purkinje neurons, is a major symptom of Nieman-Pick disease, type C1. Comparing single cell RNAseq data from control (Npc1(+/+)) and mutant (Npc1(−/−)) mice, we observed significantly decreased expression of Slc1a3 in Npc1(−/−) astrocytes. Slc1a3 encodes a glutamate transporter (GLAST, EAAT1) which functions to decrease glutamate concentrations in the post synaptic space after neuronal firing. Glutamate is an excitatory neurotransmitter and elevated extracellular levels of glutamate can be neurotoxic. Impaired EAAT1 function underlies type-6 episodic ataxia, a rare disorder with progressive cerebellar dysfunction, thus suggesting that impaired glutamate uptake in Niemann-Pick disease, type C1 could contribute to disease progression. We now show that decreased expression of Slc1a3 in Npc1(−/−) mice has functional consequences that include decreased surface protein expression and decreased glutamate uptake by Npc1(−/−) astrocytes. To test whether glutamate neurotoxicity plays a role in Niemann-Pick disease, type C1 progression, we treated NPC1 deficient mice with ceftriaxone and riluzole. Ceftriaxone is a β-lactam antibiotic that is known to upregulate the expression of Slc1a2, an alternative glial glutamate transporter. Although ceftriaxone increased Slc1a2 expression, we did not observe a treatment effect in NPC1 mutant mice. Riluzole is a glutamate receptor antagonist that inhibits postsynaptic glutamate receptor signaling and reduces the release of glutamate. We found that treatment with riluzole increased median survival in Npc1(−/−) by 12%. Given that riluzole is an approved drug for the treatment of amyotrophic lateral sclerosis, repurposing of this drug may provide a novel therapeutic approach to decrease disease progression in Niemann-Pick disease type, C1 patients.
- Published
- 2021
32. The 4b-4c loop of excitatory amino acid transporter 1 containing four critical residues essential for substrate transport
- Author
-
Suifen He, Xiuping Zhang, Mei Hong, Shaogang Qu, Wenlong Zhang, and Pingyi Xu
- Subjects
Amino Acid Transport System X-AG ,030303 biophysics ,Glutamic Acid ,03 medical and health sciences ,Pyrococcus horikoshii ,Structural Biology ,Animals ,Humans ,Molecular Biology ,Gene ,chemistry.chemical_classification ,0303 health sciences ,biology ,Chemistry ,Glutamate receptor ,Substrate (chemistry) ,Biological Transport ,Transporter ,General Medicine ,biology.organism_classification ,Amino acid ,Excitatory Amino Acid Transporter 1 ,Loop (topology) ,Biochemistry ,Mutagenesis ,Excitatory postsynaptic potential - Abstract
In the mammalians, the 4b-4c loop of excitatory amino acid transporters (EAATs) spans more than 50 amino-acid residues that are absent in glutamate transporter homologue of Pyrococcus horikoshii (GltPh). This part of insertion is unique for metazoans and indispensable to the localization of EAATs. The excitatory amino acid transporter (EAAT) 1 is one of the two glial glutamate transporters, which are responsible for efficiently clearing glutamate from the synaptic cleft to prevent neurotoxicity and cell death. Although the crystal structure of EAAT1cryst (a human thermostable EAAT1) was resolved in 2017, the structure-function relationship of the 4b-4c loop has not been elucidated in EAAT1cryst. To investigate the role of the 4b-4c loop, we performed alanine-scanning mutagenesis in the mutants and observed dramatically decreased transport activities in T192A, Y194A, N242A, and G245A mutants. The surface expression of T192A and Y194A mutants even decreased by more than 80%, and most of them were detained in the cytoplasm. However, when T192 and Y194 were substituted with conservative residues, the transport activities and the surface expressions of T192S and Y194F were largely recovered, and their kinetic parameters (Km values) were comparable to the wild-type EAAT1 as well. In contrast, N242 and G245 substituted with conservative residues could not rescue the uptake function, suggesting that N242 and G245 may play irreplaceable roles in the glutamate uptake process. These results indicate that the 4b-4c loop of EAAT1 may not only affect the glutamate uptake activity, but also influence the surface localization of EAAT1 by T192 and Y194. Communicated by Ramaswamy H. Sarma
- Published
- 2019
- Full Text
- View/download PDF
33. Regulation of Synaptosomal GLT-1 and GLAST during Epileptogenesis
- Author
-
Allison R. Peterson and Devin K. Binder
- Subjects
0301 basic medicine ,Kainic acid ,medicine.medical_specialty ,Hippocampus ,Status epilepticus ,Epileptogenesis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Epilepsy ,0302 clinical medicine ,Seizures ,Internal medicine ,medicine ,Glutamate aspartate transporter ,Animals ,Kainic Acid ,biology ,Chemistry ,General Neuroscience ,Glutamate receptor ,medicine.disease ,Excitatory Amino Acid Transporter 1 ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Epilepsy, Temporal Lobe ,Excitatory Amino Acid Transporter 2 ,Astrocytes ,biology.protein ,medicine.symptom ,030217 neurology & neurosurgery ,Synaptosomes ,Astrocyte - Abstract
Astrocytes regulate extracellular glutamate homeostasis in the central nervous system through the Na+-dependent glutamate transporters glutamate transporter-1 (GLT-1) and glutamate aspartate transporter (GLAST). Impaired astrocyte glutamate uptake could contribute to the development of epilepsy but the regulation of glutamate transporters in epilepsy is not well understood. In this study, we investigate the expression of GLT-1 and GLAST in the mouse intrahippocampal kainic acid (IHKA) model of temporal lobe epilepsy (TLE). We used immunohistochemistry, synaptosomal fractionation and Western blot analysis at 1, 3, 7 and 30 days post-IHKA induced status epilepticus (SE) to examine changes in GLT-1 and GLAST immunoreactivity and synaptosomal expression during the development of epilepsy. We found a significant upregulation in GLT-1 immunoreactivity at 1 and 3 days post-IHKA in the ipsilateral dorsal hippocampus. However, GLT-1 immunoreactivity and synaptosomal protein levels were significantly downregulated at 7 days post-IHKA in the ipsilateral hippocampus, a time point corresponding to the onset of spontaneous seizures in this model. GLAST immunoreactivity was increased in specific layers at 1 and 3 days post-IHKA in the ipsilateral hippocampus. GLAST synaptosomal protein levels were significantly elevated at 30 days compared to 7 days post-IHKA in the ipsilateral hippocampus. Our findings suggest that astrocytic glutamate transporter dysregulation could contribute to the development of epilepsy.
- Published
- 2019
- Full Text
- View/download PDF
34. Neuronal excitatory amino acid transporter EAAT3: Emerging functions in health and disease
- Author
-
Susanne E. Ahmari, Susan G. Amara, Susan L. Ingram, Jeremy Veenstra-VanderWeele, and Suzanne M. Underhill
- Subjects
0301 basic medicine ,Aging ,Synaptic cleft ,Amino Acid Transport System X-AG ,Glutamic Acid ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Glutamatergic ,0302 clinical medicine ,medicine ,Animals ,Humans ,Premovement neuronal activity ,Receptor ,Neurons ,Chemistry ,Neurodegeneration ,Age Factors ,Glutamate receptor ,Transporter ,Cell Biology ,medicine.disease ,Cell biology ,Excitatory Amino Acid Transporter 1 ,Excitatory Amino Acid Transporter 3 ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,Knockout mouse ,030217 neurology & neurosurgery - Abstract
Plasma membrane neurotransmitter transporters maintain extracellular concentrations of neurotransmitters by facilitating transport into the cytosol. This regulation of extracellular neurotransmitters limits binding to receptors and activation of downstream signaling pathways. In addition to this critical function, transporters modulate neuronal activity via direct gating of transporter-associated ion channels and indirectly through trafficking of transporters to and from the plasma membrane. These functions are dependent on diverse expression patterns and levels of the transporters throughout the brain. This diversity is highlighted within the excitatory amino acid transporter family, which consists of five excitatory amino acid transporters found in the mammalian central nervous system (CNS). EAAT1 (GLAST) and EAAT2 (GLT-1) are primarily expressed in astrocytes while EAAT3 expression is mainly observed in many neurons throughout the brain (Holmseth et al., 2012). In contrast, EAAT4 is most prominently expressed in cerebellar Purkinje neurons and EAAT5 is exclusively found in the retina. In general, the astroglial transporters are highly expressed in the brain; EAAT2 is the most abundant, followed by EAAT1 with approximately a 4-fold lower expression (Holmseth et al., 2012). High expression levels of these glial transporters are consistent with their role in glutamate clearance (Lehre and Danbolt, 1998; Rothstein et al., 1996; Tanaka et al., 1997). The role of the neuronal transporter EAAT3 in brain has been more difficult to elucidate. Levels of EAAT3 are approximately 100-fold lower than EAAT2 (Holmseth et al., 2012) but EAAT3 expression is observed throughout the CNS, with enriched expression in the cerebral cortex, hippocampus, cerebellum and basal ganglia (Rothstein et al., 1994; Shashidharan et al., 1997). Given the lack of selective EAAT3 inhibitors, studies have relied on EAAT3 transporters expressed in various cells and endogenous transporters expressed in cultured hippocampal neurons (Diamond and Jahr, 1997; Grewer et al., 2000; Wadiche et al., 1995b), as well as EAAT3 knockout mice (Scimemi et al., 2009) to dertmine the physiological functions of EAAT3. These studies determined that the time course of glutamate in the synaptic cleft is a function of the binding of glutamate to EAATs and that transport of glutamate does not significantly contribute to the amplitude or kinetics of synaptic responses due to the relatively slow transport cycle (Diamond and Jahr, 1997; Tong and Jahr, 1994; Wadiche and von Gersdorff, 2006). Interestingly, EAAT3 knockout mice exhibit few behavioral deficits (Peghini et al., 1997), and antisense oligonucleotide knockdown in the striatum results in minimal elevation of extracellular glutamate levels or neurodegeneration, in contrast to knockdown of EAATs 1 and 2 (Rothstein et al., 1996). The lack of neurodegeneration is particularly surprising given that EAAT3 also serves as a cysteine transporter (Aoyama et al., 2006; Watts et al., 2014; Zerangue and Kavanaugh, 1996). Cysteine is the rate-limiting substrate for the synthesis of the antioxidant glutathione and its extracellular depletion is hypothesized to contribute to neurodegeneration. EAAT3 is also the dominant glutamate transporter in the intestines and provides nutrient absorption from the diet (Hu et al., 2018), but knockout animals appear to grow at a comparable rate to their litter mates (Peghini et al., 1997). The most remarkable initial observation from EAAT3 knockout mice was aminoaciduria due to the absence of EAAT3 in the kidneys (Peghini et al., 1997). The reported lack of overt behavioral abnormalities in the EAAT3 knockout mice suggest that either EAAT3 is not integral to regulation of glutamatergic signaling in the brain or that substantial developmental compensatory changes are induced in these mice. Human genetic studies and constitutive deletion mouse models have now provided evidence that EAAT3 has important roles in regulating neuronal signaling.
- Published
- 2019
- Full Text
- View/download PDF
35. Variability with Astroglial Glutamate Transport Genetics Is Associated with Increased Risk for Post-Traumatic Seizures
- Author
-
Yvette P. Conley, Anne C. Ritter, Kristen B Breslin, Amy K. Wagner, and Raj G. Kumar
- Subjects
Adult ,Male ,030506 rehabilitation ,Genotype ,Traumatic brain injury ,Excitotoxicity ,macromolecular substances ,Bioinformatics ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Epileptogenesis ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Seizures ,Brain Injuries, Traumatic ,Humans ,Medicine ,Genetic Predisposition to Disease ,Post-traumatic epilepsy ,business.industry ,Glutamate receptor ,Original Articles ,Epilepsy, Post-Traumatic ,medicine.disease ,Excitatory Amino Acid Transporter 1 ,Increased risk ,Excitatory Amino Acid Transporter 2 ,Astrocytes ,Female ,Neurology (clinical) ,0305 other medical science ,business ,Clinical risk factor ,030217 neurology & neurosurgery - Abstract
Excitotoxicity contributes to epileptogenesis after severe traumatic brain injury (sTBI). Demographic and clinical risk factors for post-traumatic seizures (PTS) have been identified, but genetic risk remains largely unknown. Thus, we investigated whether genetic variation in astroglial glutamate transporter genes is associated with accelerated epileptogenesis and PTS risk after sTBI. Adults (n = 267) 18–75 years old were assessed over a three-year period post-TBI. Single nucleotide polymorphisms (SNPs) throughout the SLC1A2 and SLC1A3 genes were assayed. Kaplan-Meier estimates and log-rank statistics were used to compare seizure frequencies by genotype. Multivariate Cox proportional hazards regression was used to estimate hazard ratios (HRs) for genotypes significant in Kaplan-Meier analyses. Thirty-nine tagging SNPs were examined (SLC1A2: n = 21, SLC1A3: n = 18). PTS developed in 57 (21.4%) individuals. Of those with PTS, n = 20 (35.7%) had an immediate/early seizure within the first seven days, and n = 36 (64.3%) had a late seizure occurring between eight days and three years post-TBI. When adjusting for multiple comparisons, rs4869682 genotypes (SLC1A3, GG vs. T-carriers) were associated with time to first seizure (p = 0.003). Median time until first seizure was 20.4 days for individuals with a GG genotype and 44.8 days for T-carriers. After adjusting for covariates, rs4869682 GG-homozygotes had a 2.05 times increased PTS risk versus T-carriers (aHR = 2.08, 95% confidence interval: 1.20, 3.62, p = 0.009). Variation within SLC1A3 is associated with accelerated epileptogenesis and clinical PTS development after sTBI. Future studies should validate these findings and examine how genetic variation at rs4869682 may be a target for PTS prevention and treatment.
- Published
- 2019
- Full Text
- View/download PDF
36. Anxiolytic activity of paraoxon is associated with alterations in rat brain glutamatergic system
- Author
-
Zohreh Zare, Noorollah Rezaei, Moslem Mohammadi, Mohsen Tehrani, and Babak Dana Ghalebarzand
- Subjects
Male ,medicine.medical_specialty ,Elevated plus maze ,medicine.drug_class ,Prefrontal Cortex ,Anxiety ,Motor Activity ,010501 environmental sciences ,Toxicology ,Hippocampus ,01 natural sciences ,Anxiolytic ,Paraoxon ,Open field ,Glutamate Plasma Membrane Transport Proteins ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Developmental Neuroscience ,Internal medicine ,medicine ,Animals ,Cholinesterases ,Hippocampus (mythology) ,Maze Learning ,Prefrontal cortex ,0105 earth and related environmental sciences ,Cholinesterase ,Behavior, Animal ,Dose-Response Relationship, Drug ,biology ,Chemistry ,Organophosphate ,Brain ,Rats ,Excitatory Amino Acid Transporter 1 ,Endocrinology ,Excitatory Amino Acid Transporter 2 ,biology.protein ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Exposure to organophosphate (OP) compounds leads to behavioral alterations. To determine whether paraoxon has effects on anxiety, anxiety-like behaviors were assessed in paraoxon-exposed rats. Protein expression of glutamate transporters has also been measured in hippocampus and prefrontal cortex. Three doses of paraoxon (0.3, 0.7, or 1 mg/kg) or corn oil (vehicle) were intraperitoneally injected to adult male rats. At 14 or 28 days after exposure, behavioral tests were done using elevated plus-maze (EPM) or open field tests. Thereafter, animals were sacrificed and both hippocampi and prefrontal cortices were extracted for cholinesterase assay and western blotting. Animals treated with convulsive doses of paraoxon (0.7 and 1 mg/kg) showed an increase in percentage of time spent in open arms and percentage of open arm entries in the EPM. In the open field test, an increase in the time spent in central area was observed in rats treated with the same doses of paraoxon. These effects of paraoxon were independent of any changes in locomotor activity. There was an increase in both astrocytic glutamate transporter proteins (GLAST and GLT-1) in the hippocampus of animals treated with 0.7 and 1 mg/kg of paraoxon. In the prefrontal cortex, protein levels of the GLAST and GLT-1 increased in 0.7 and decreased in 1 mg/kg groups. Only a significant decrease in EAAC1 protein was observed in the prefrontal cortex at 14 days following exposure to 1 mg/kg of paraoxon. Collectively, this study showed that exposure to convulsive doses of paraoxon induced anxiolytic-like behaviors in both behavioral tests. This effect may be attributed to alterations of glutamate transporter proteins in the rat hippocampus and prefrontal cortex.
- Published
- 2019
- Full Text
- View/download PDF
37. Punicalagin increases glutamate absorption in differentiated Caco-2 cells by a mechanism involving gene expression regulation of an EAAT3 transporter
- Author
-
Giuseppe Bee, Corinne Jud, Claudine Biolley, Paolo Silacci, and Marco Tretola
- Subjects
0301 basic medicine ,Enterocyte ,Glutamic Acid ,03 medical and health sciences ,chemistry.chemical_compound ,Gene expression ,medicine ,Humans ,Punicalagin ,Regulation of gene expression ,030109 nutrition & dietetics ,Ussing chamber ,Chemistry ,Glutamate receptor ,Biological Transport ,Cell Differentiation ,General Medicine ,Transfection ,Hydrolyzable Tannins ,Cell biology ,Excitatory Amino Acid Transporter 1 ,Enterocytes ,Excitatory Amino Acid Transporter 3 ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Caco-2 ,Caco-2 Cells ,Food Science - Abstract
This study investigates, for the first time, the ability of punicalagin to modulate intestinal glutamate uptake by upregulation of the expression of one of its transporters present on the enterocyte membrane. The use of an Ussing chamber revealed an increase in glutamate transport in differentiated Caco-2 cells after punicalagin treatment for 24 h. This cell line constitutively expresses two glutamate transporters: EAAT1 and EAAT3. In response to punicalagin, the expression of EAAT3 was increased, at both mRNA and protein levels, but not that of EAAT1. Transfection with EAAT3-targeting siRNA specifically altered basal and induced EAAT3 gene expression, decreasing the positive effect of punicalagin on glutamate uptake. These data confirmed the involvement of EAAT3 in increasing glutamate uptake by enterocytes after punicalagin treatment.
- Published
- 2019
- Full Text
- View/download PDF
38. BMP-dependent, injury-induced stem cell niche as a mechanism of heterotopic ossification
- Author
-
Haimei Lu, Jiazhao Yang, John A. Kessler, Zhenya Tan, Lixin Kan, Keqin Zhang, Diana M. Palila Berger, Na Ding, Tammy L. McGuire, and Chen Kan
- Subjects
0301 basic medicine ,Medicine (miscellaneous) ,Mice ,0302 clinical medicine ,Loss of Function Mutation ,Osteogenesis ,Diphtheria Toxin ,Niche supportive cells ,lcsh:QD415-436 ,Stem Cell Niche ,lcsh:R5-920 ,Cell Differentiation ,Receptor, TIE-2 ,Cell biology ,Excitatory Amino Acid Transporter 1 ,030220 oncology & carcinogenesis ,Molecular Medicine ,Stem cell ,Signal transduction ,lcsh:Medicine (General) ,Signal Transduction ,Neurite ,Mesenchymal stem cells (MSCs) ,Biology ,Bone morphogenetic protein (BMP) ,Zinc Finger Protein GLI1 ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Heterotopic ossification (HO) ,lcsh:Biochemistry ,03 medical and health sciences ,Niche supportive molecules ,In vivo ,Niche ,medicine ,Animals ,Humans ,Hedgehog ,Cell Proliferation ,Progenitor ,Research ,Hedgehog (Hh) ,Ossification, Heterotopic ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,medicine.disease ,Peptide Fragments ,Disease Models, Animal ,030104 developmental biology ,Myositis Ossificans ,Fibrodysplasia ossificans progressiva - Abstract
Background Heterotopic ossification (HO), either acquired (aHO) or hereditary, such as fibrodysplasia ossificans progressiva (FOP), is a serious condition without effective treatment. Understanding of the core process of injury-induced HO is still severely limited. Methods Double-pulse thymidine analog labeling was used to explore the distinctive domains evolved in injury-induced lesions in an animal model of HO (Nse-BMP4). Histological studies were performed to see whether a similar zonal pattern is also consistently found in biopsies from patients with aHO and FOP. In vivo clonal analysis with Rainbow mice, genetic loss-of-function studies with diphtheria toxin A (DTA)-mediated depletion and lineage tracing with Zsgreen reporter mice were used to obtain further evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells. Immunohistochemistry was used to test whether vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Similar methods also were employed to further understand the signaling pathways that regulate the niche and the resultant HO. Results We found that distinctive domains evolved in injury-induced lesions, including, from outside-in, a mesenchymal stem cell (MSC) niche, a transient domain and an inner differentiated core in an animal model of HO (Nse-BMP4). A similar zonal structure was found in patients with aHO and FOP. In vivo clonal analysis with Rainbow mice and genetic loss-of-function studies with DTA provided evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells; consistently, vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Further mechanistic study found that BMP and hedgehog (Hh) signaling co-regulate the niche and the resultant HO. Conclusions Available data provide evidence of a potential core mechanism in which multiple disease-specific cellular and extracellular molecular elements form a unique local microenvironment, i.e., an injury-induced stem cell niche, which regulates the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs). The implication for HO is that therapeutic approaches must consider several different disease specific factors as parts of a functional unit, instead of treating one factor at a time. Electronic supplementary material The online version of this article (10.1186/s13287-018-1107-7) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
39. Early postnatal inhibition of GLAST causes abnormalities of psychobehaviors and neuronal morphology in adult mice
- Author
-
Akihiro Mouri, Yukihiro Noda, Sho Hasegawa, Kohichi Tanaka, Tomomi Aida, Kiyofumi Yamada, Toshitaka Nabeshima, Masayuki Taniguchi, Norio Ozaki, Hirotake Hida, Mizuki Uchida, and Akira Yoshimi
- Subjects
Male ,Neurons ,Aspartic Acid ,Period (gene) ,Mental Disorders ,Glutamate receptor ,Hippocampus ,Cognition ,Cell Biology ,Neurotransmission ,Biology ,Cortex (botany) ,Excitatory Amino Acid Transporter 1 ,Mice, Inbred C57BL ,Cellular and Molecular Neuroscience ,Glutamatergic ,Mice ,Animals, Newborn ,Pregnancy ,Facilitation ,Animals ,Female ,Maze Learning ,Neuroscience ,Injections, Intraventricular - Abstract
The importance of glutamate transporters in learning, memory, and emotion remains poorly understood; hence, in the present study, we investigated whether deficiency of pharmacological GLAST in neurodevelopmental processes affects cognitive and/or emotional behaviors in mice. The mice were injected with a glutamate transporter inhibitor, dl -threo-β-benzyloxyaspartate ( dl -TBOA), during the early postnatal period. At 8 weeks of age, they showed impairments in cognitive or emotional behaviors; dysfunction of glutamatergic neurotransmission (increased expressions of GLAST, GLT-1, or GFAP protein, and decreased ability of glutamate release) in the cortex or hippocampus; morphological changes (decreased cell size in the cortex and thickness of the pyramidal neuronal layer of the CA1 area in the hippocampus). Such behavioral and morphological changes were not observed in adult mice injected with dl- TBOA. These results suggest that GLAST plays an important role in the regulation of cognitive and emotional behaviors. Early postnatal glutamatergic facilitation by GLAST dysfunction leads to cognitive and emotional abnormalities due to neurodevelopmental abnormalities such as morphological changes.
- Published
- 2021
40. Antibody Characterization Report for Excitatory amino acid transporter 1 SLC1A3 (EAAT1)
- Author
-
Ayoubi, Riham, McPherson, Peter S., and Laflamme, Carl
- Subjects
EAAT1 ,Excitatory amino acid transporter 1 ,P43003 ,antibody characterization ,Sodium-dependent glutamate/aspartate transporter 1 ,SLC1A3 ,Solute carrier family 1 member 3 ,antibody validation ,GLAST-1 - Abstract
Head to head comparison of available commercial antibodies against Excitatory amino acid transporter 1 SLC1A3 (EAAT1) by immunoblot (Western blot). Work performed in collaboration with the RESOLUTE consortium (re-solute.eu). RESOLUTE received support from the EU/EFPIA Innovative Medicines Initiative 2 Joint Undertaking (RESOLUTE grant No 777372).
- Published
- 2021
- Full Text
- View/download PDF
41. Modulation of Glutamate Transporter EAAT1 and Inward-Rectifier Potassium Channel K
- Author
-
Leo, Markus, Schmitt, Linda-Isabell, Steffen, Rebecca, Kutritz, Andrea, Kleinschnitz, Christoph, and Hagenacker, Tim
- Subjects
EAAT1 ,Male ,cisplat ,Kir4.1 ,QH301-705.5 ,Medizinische Fakultät » Universitätsklinikum Essen » Klinik für Neurologie ,cisplatin ,Minocycline ,Cell Separation ,Article ,Glial Fibrillary Acidic Protein ,Animals ,ddc:610 ,Biology (General) ,Potassium Channels, Inwardly Rectifying ,Rats, Wistar ,QD1-999 ,Cells, Cultured ,Platinum ,GFAP ,chemotherapy-induced neuropath ,spinal cor ,oxaliplatin ,astrocytes ,chemotherapy-induced neuropathy ,Anti-Bacterial Agents ,Excitatory Amino Acid Transporter 1 ,Chemistry ,Spinal Cord ,Astrocytes ,Female ,potas-sium channel ,potassium channel - Abstract
Platinum-based chemotherapeutics still play an essential role in cancer treatment. Despite their high effectiveness, severe side effects such as chemotherapy-induced neuropathy (CIPN) occur frequently. The pathophysiology of CIPN by platinum-based chemotherapeutics is not fully understood yet, but primarily the disturbance of dorsal root ganglion cells is discussed. However, there is increasing evidence of central nervous system involvement with activation of spinal cord astrocytes after treatment with chemotherapeutics. We investigated the influence of cis- or oxaliplatin on the functionality of cultured rat spinal cord astrocytes by using immunocytochemistry and patch-clamp electrophysiology. Cis- or oxaliplatin activated spinal astrocytes and led to downregulation of the excitatory amino acid transporter 1 (EAAT1) expression. Furthermore, the expression and function of potassium channel Kir4.1 were modulated. Pre-exposure to a specific Kir4.1 blocker in control astrocytes led to a reduced immune reactivity (IR) of EAAT1 and a nearly complete block of the current density. When spinal astrocytes were pre-exposed to antibiotic minocycline, all effects of cis- or oxaliplatin were abolished. Taken together, the modulation of Kir4.1 and EAAT1 proteins in astrocytes could be linked to the direct impact of cis- or oxaliplatin, identifying spinal astrocytes as a potential target in the prevention and treatment of chemotherapy-induced neuropathy.
- Published
- 2021
42. Rapid Regulation of Glutamate Transport: Where do we go from here?
- Author
-
Michael B. Robinson, Alain M Guillem, and Elizabeth N. Krizman
- Subjects
0301 basic medicine ,Central Nervous System ,Amino Acid Transport System X-AG ,Glutamic Acid ,Biology ,Biochemistry ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Glutamatergic ,0302 clinical medicine ,Palmitoylation ,Ubiquitin ,Animals ,Humans ,Protein kinase C ,Mammals ,Nitrosylation ,Sodium ,Glutamate receptor ,Transporter ,General Medicine ,Excitatory Amino Acid Transporter 1 ,030104 developmental biology ,Excitatory Amino Acid Transporter 2 ,biology.protein ,Excitatory postsynaptic potential ,Neuroscience ,030217 neurology & neurosurgery ,Synaptosomes - Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). A family of five Na(+)-dependent transporters maintain low levels of extracellular glutamate and shape excitatory signaling. Shortly after the research group of the person being honored in this special issue (Dr. Baruch Kanner) cloned one of these transporters, his group and several others showed that their activity can be acutely (within minutes to hours) regulated. Since this time, several different signals and post-translational modifications have been implicated in the regulation of these transporters. In this review, we will provide a brief introduction to the distribution and function of this family of glutamate transporters. This will be followed by a discussion of the signals that rapidly control the activity and/or localization of these transporters, including protein kinase C, ubiquitination, glutamate transporter substrates, nitrosylation, and palmitoylation. We also include the results of our attempts to define the role of palmitoylation in the regulation of GLT-1 in crude synaptosomes. In some cases, the mechanisms have been fairly well-defined, but in others, the mechanisms are not understood. In several cases, contradictory phenomena have been observed by more than one group; we describe these studies with the goal of identifying the opportunities for advancing the field. Abnormal glutamatergic signaling has been implicated in a wide variety of psychiatric and neurologic disorders. Although recent studies have begun to link regulation of glutamate transporters to the pathogenesis of these disorders, it will be difficult to determine how regulation influences signaling or pathophysiology of glutamate without a better understanding of the mechanisms involved.
- Published
- 2021
43. Hippocampal astrocytic neogenin regulating glutamate uptake, a critical pathway for preventing epileptic response
- Author
-
Lin Mei, Xiang-Dong Sun, Daehoon Lee, Leena Milibari, Jin-Xiu Pan, Zhihui Huang, Zhi-Peng Liu, Wen Cheng Xiong, Xiao Ren, Chen Shen, Zhibing Tan, Ling-Ling Yao, Dong Sun, and Wenbing Chen
- Subjects
Male ,Glutamic Acid ,Biology ,Hippocampal formation ,Inhibitory postsynaptic potential ,Hippocampus ,Mice ,Epilepsy ,Seizures ,medicine ,Animals ,Mice, Knockout ,Neurons ,Multidisciplinary ,Dentate gyrus ,Membrane Proteins ,Biological Transport ,Synaptic Potentials ,Biological Sciences ,medicine.disease ,Excitatory Amino Acid Transporter 1 ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Excitatory Amino Acid Transporter 2 ,nervous system ,Astrocytes ,Excitatory postsynaptic potential ,biology.protein ,NMDA receptor ,Female ,Neuroscience ,Parvalbumin ,Signal Transduction ,Astrocyte - Abstract
Epilepsy, a common neurological disorder, is featured with recurrent seizures. Its underlying pathological mechanisms remain elusive. Here, we provide evidence for loss of neogenin (NEO1), a coreceptor for multiple ligands, including netrins and bone morphological proteins, in the development of epilepsy. NEO1 is reduced in hippocampi from patients with epilepsy based on transcriptome and proteomic analyses. Neo1 knocking out (KO) in mouse brains displays elevated epileptiform spikes and seizure susceptibility. These phenotypes were undetectable in mice, with selectively depleted NEO1 in excitatory (NeuroD6-Cre(+)) or inhibitory (parvalbumin(+)) neurons, but present in mice with specific hippocampal astrocytic Neo1 KO. Additionally, neurons in hippocampal dentate gyrus, a vulnerable region in epilepsy, in mice with astrocyte-specific Neo1 KO show reductions in inhibitory synaptic vesicles and the frequency of miniature inhibitory postsynaptic current(mIPSC), but increase of the duration of miniature excitatory postsynaptic current and tonic NMDA receptor currents, suggesting impairments in both GABAergic transmission and extracellular glutamate clearance. Further proteomic and cell biological analyses of cell-surface proteins identified GLAST, a glutamate–aspartate transporter that is marked reduced in Neo1 KO astrocytes and the hippocampus. NEO1 interacts with GLAST and promotes GLAST surface distribution in astrocytes. Expressing NEO1 or GLAST in Neo1 KO astrocytes in the hippocampus abolishes the epileptic phenotype. Taken together, these results uncover an unrecognized pathway of NEO1-GLAST in hippocampal GFAP(+) astrocytes, which is critical for GLAST surface distribution and function, and GABAergic transmission, unveiling NEO1 as a valuable therapeutic target to protect the brain from epilepsy.
- Published
- 2021
- Full Text
- View/download PDF
44. Bisphenol A exposure disrupts aspartate transport in HepG2 cells.
- Author
-
UCL - SSS/IREC/PEDI - Pôle de Pédiatrie, UCL - (SLuc) Service de cardiologie pédiatrique, Jiménez-Torres, Catya, Hernández-Kelly, Luisa C, Najimi, Mustapha, Ortega, Arturo, UCL - SSS/IREC/PEDI - Pôle de Pédiatrie, UCL - (SLuc) Service de cardiologie pédiatrique, Jiménez-Torres, Catya, Hernández-Kelly, Luisa C, Najimi, Mustapha, and Ortega, Arturo
- Abstract
The liver is the organ responsible for bisphenol A (BPA) metabolism, an environmental chemical agent. Exposure to this toxin is associated with liver abnormalities and dysfunction. An important role played by excitatory amino acid transporters (EAATs) of the slc1 gene family has been reported in liver injuries. To gain insight into a plausible effect of BPA exposure in the liver glutamate/aspartate transport, using the human hepatoblastoma cell line HepG2, we report a BPA-dependent dynamic regulation of SLC1A3 and SLC1A2. Through the use of radioactive [ H]- d-aspartate uptake experiments and immunochemical approaches, we characterized time and dose-dependent regulation of the protein levels and function of these transporters after acute exposure to BPA. An increase in nuclear Yin Yang 1 was found. These results suggest an important involvement of the EAATs in liver physiology and its disruption after acute BPA exposure.
- Published
- 2020
45. Glutamate transporters: Critical components of glutamatergic transmission
- Author
-
Arturo Ortega and Ada G. Rodríguez-Campuzano
- Subjects
0301 basic medicine ,Nervous system ,Synaptic cleft ,Amino Acid Transport System X-AG ,Excitotoxicity ,Glutamic Acid ,Biology ,medicine.disease_cause ,Synaptic Transmission ,Protein Structure, Secondary ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Glutamatergic ,0302 clinical medicine ,Glutamate homeostasis ,medicine ,Animals ,Humans ,Receptor ,Pharmacology ,Glutamate receptor ,Cell biology ,Protein Structure, Tertiary ,Excitatory Amino Acid Transporter 1 ,030104 developmental biology ,medicine.anatomical_structure ,Excitatory Amino Acid Transporter 2 ,Signal transduction ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
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.
- Published
- 2021
46. Genetic interactions regulate hypoxia tolerance conferred by activating Notch in excitatory amino acid transporter 1-positive glial cells inDrosophila melanogaster
- Author
-
Dan Zhou, DeeAnn W. Visk, Gabriel G. Haddad, Jin Xue, Tsering Stobdan, and Ramaswami, M
- Subjects
AcademicSubjects/SCI01140 ,genetic interactions ,Candidate gene ,Notch ,AcademicSubjects/SCI00010 ,1.1 Normal biological development and functioning ,Notch signaling pathway ,Biology ,AcademicSubjects/SCI01180 ,Fibroblast growth factor ,03 medical and health sciences ,0302 clinical medicine ,Underpinning research ,Receptors ,Genetics ,medicine ,Animals ,Drosophila Proteins ,2.1 Biological and endogenous factors ,Aetiology ,Hypoxia ,Molecular Biology ,Transcription factor ,Gene ,Notch signaling ,Genetics (clinical) ,030304 developmental biology ,Investigation ,0303 health sciences ,Receptors, Notch ,Human Genome ,Hypoxia (medical) ,biology.organism_classification ,Phenotype ,Cell biology ,Excitatory Amino Acid Transporter 1 ,eaat1-posive glia ,Stroke ,Drosophila melanogaster ,AcademicSubjects/SCI00960 ,medicine.symptom ,Neuroglia ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Hypoxia is a critical pathological element in many human diseases, including ischemic stroke, myocardial infarction, and solid tumors. Of particular significance and interest of ours are the cellular and molecular mechanisms that underlie susceptibility or tolerance to low O2. Previous studies have demonstrated that Notch signaling pathway regulates hypoxia tolerance in both Drosophila melanogaster and humans. However, the mechanisms mediating Notch-conferred hypoxia tolerance are largely unknown. In this study, we delineate the evolutionarily conserved mechanisms underlying this hypoxia tolerant phenotype. We determined the role of a group of conserved genes that were obtained from a comparative genomic analysis of hypoxia-tolerant D.melanogaster populations and human highlanders living at the high-altitude regions of the world (Tibetans, Ethiopians, and Andeans). We developed a novel dual-UAS/Gal4 system that allows us to activate Notch signaling in the Eaat1-positive glial cells, which remarkably enhances hypoxia tolerance in D.melanogaster, and, simultaneously, knock down a candidate gene in the same set of glial cells. Using this system, we discovered that the interactions between Notch signaling and bnl (fibroblast growth factor), croc (forkhead transcription factor C), or Mkk4 (mitogen-activated protein kinase kinase 4) are important for hypoxia tolerance, at least in part, through regulating neuronal development and survival under hypoxic conditions. Becausethese genetic mechanisms are evolutionarily conserved, this group of genes may serve as novel targets for developing therapeutic strategies and have a strong potential to be translated to humans to treat/prevent hypoxia-related diseases.
- Published
- 2021
- Full Text
- View/download PDF
47. Slc1a3-2A-CreERT2 mice reveal unique features of Bergmann glia and augment a growing collection of Cre drivers and effectors in the 129S4 genetic background
- Author
-
Nicole Reichenbach, Maria Jonson, Lech Kaczmarczyk, Lars Dittrich, Nelli Blank, Walker S. Jackson, and Gabor C. Petzold
- Subjects
Cell type ,Science ,Transgene ,Mice, Transgenic ,metabolism [Neurodegenerative Diseases] ,Biology ,Article ,Mice ,Inbred strain ,Genetic variation ,Genetics ,Animals ,genetics [Excitatory Amino Acid Transporter 1] ,Genetik ,metabolism [Neuroglia] ,Gene ,Multidisciplinary ,Integrases ,Effector ,Strain (biology) ,metabolism [Excitatory Amino Acid Transporter 1] ,Glial biology ,Neurodegenerative Diseases ,Phenotype ,Excitatory Amino Acid Transporter 1 ,genetics [Neurodegenerative Diseases] ,Medicine ,Astrocyte ,ddc:600 ,Neuroglia ,Neuroscience - Abstract
Genetic variation is a primary determinant of phenotypic diversity. In laboratory mice, genetic variation can be a serious experimental confounder, and thus minimized through inbreeding. However, generalizations of results obtained with inbred strains must be made with caution, especially when working with complex phenotypes and disease models. Here we compared behavioral characteristics of C57Bl/6-the strain most widely used in biomedical research-with those of 129S4. In contrast to 129S4, C57Bl/6 demonstrated high within-strain and intra-litter behavioral hyperactivity. Although high consistency would be advantageous, the majority of disease models and transgenic tools are in C57Bl/6. We recently established six Cre driver lines and two Cre effector lines in 129S4. To augment this collection, we genetically engineered a Cre line to study astrocytes in 129S4. It was validated with two Cre effector lines: calcium indicator gCaMP5g-tdTomato and RiboTag-a tool widely used to study cell type-specific translatomes. These reporters are in different genomic loci, and in both the Cre was functional and astrocyte-specific. We found that calcium signals lasted longer and had a higher amplitude in cortical compared to hippocampal astrocytes, genes linked to a single neurodegenerative disease have highly divergent expression patterns, and that ribosome proteins are non-uniformly expressed across brain regions and cell types. Funding Agencies|Wallenberg Center for Molecular Medicine; German Science Foundation DFGGerman Research Foundation (DFG) [FOR 2795/PE1193/6-1]
- Published
- 2021
- Full Text
- View/download PDF
48. Excitatory Amino Acid Transporter EAAT5 Improves Temporal Resolution in the Retina
- Author
-
Jana Gehlen, Frank Müller, Christoph Fahlke, Anja Mataruga, and Christoph Aretzweiler
- Subjects
retina ,Synaptic cleft ,genetic structures ,EAAT5 ,Amino Acid Transport System X-AG ,Glutamic Acid ,Mice ,chemistry.chemical_compound ,Glutamatergic ,Postsynaptic potential ,medicine ,Animals ,ddc:610 ,Ion channel ,Retina ,Chemistry ,General Neuroscience ,Glutamate receptor ,Retinal ,General Medicine ,eye diseases ,Excitatory Amino Acid Transporter 1 ,medicine.anatomical_structure ,Excitatory Amino Acid Transporter 2 ,Excitatory postsynaptic potential ,Sensory and Motor Systems ,sense organs ,glutamate transporter ,Excitatory Amino Acid Transporter 5 ,Neuroscience ,Research Article: New Research - Abstract
Excitatory amino acid transporters (EAATs) remove glutamate from the synaptic cleft. In the retina, EAAT1 and EAAT2 are considered the major glutamate transporters. However, it has not yet been possible to determine how EAAT5 shapes the retinal light responses because of the lack of a selective EAAT5 blocker or EAAT5 knock-out animal model. In this study, EAAT5 was found to be expressed in a punctate manner close to release sites of glutamatergic synapses in the mouse retina. Light responses from retinae of wild-type and of a newly generated model with a targeted deletion of EAAT5 (EAAT5-/-) were recorded in vitro using multi-electrode arrays. Flicker resolution was considerably lower in EAAT5-/- retinae than in wild-type retinae. The close proximity to the glutamate release site makes EAAT5 an ideal tool to improve temporal information processing in the retina by controlling information transfer at glutamatergic synapses.Significance statementNeurons communicate with other neurons at synaptic connections by release of neurotransmitters acting at postsynaptic receptors. Neurotransmitters are removed from the synaptic cleft by transporters. Using the mouse retina as a model for the central nervous system, the role of EAAT5 that functions as glutamate transporter and as glutamate-gated ion channel was investigated in retinal information processing. EAAT5 was found highly localized to the glutamate release site at retinal synapses, suggesting a role in shaping of synaptic responses. In a mouse model devoid of EAAT5, temporal resolution of the retina was severely compromised. The results demonstrate that glutamate transporters like EAAT5 can exert a tremendous effect on information processing in neuronal networks.
- Published
- 2021
- Full Text
- View/download PDF
49. Identification of 3'-UTR single nucleotide variants and prediction of select protein imbalance in mesial temporal lobe epilepsy patients
- Author
-
Janaki Chintalapati, M.V. Hosur, and Tanusree Chaudhuri
- Subjects
Untranslated region ,Molecular biology ,RNA Stability ,RNA-binding proteins ,RNA-binding protein ,CACNB4 ,Biochemistry ,Physical Chemistry ,Medicine and Health Sciences ,Missense mutation ,RNA structure ,3' Untranslated Regions ,Genetics ,Multidisciplinary ,Symporters ,Messenger RNA ,Software Engineering ,Genomics ,Aldehyde Oxidoreductases ,Nucleic acids ,Excitatory Amino Acid Transporter 1 ,Chemistry ,Neurology ,Ribonucleoproteins ,Physical Sciences ,Engineering and Technology ,Medicine ,Research Article ,Computer and Information Sciences ,Science ,MiRNA binding ,Biology ,Polymorphism, Single Nucleotide ,Receptors, N-Methyl-D-Aspartate ,Human Genomics ,Computer Software ,microRNA ,Humans ,Non-coding RNA ,Gene ,Natural antisense transcripts ,Epilepsy ,SKP Cullin F-Box Protein Ligases ,Biology and life sciences ,Chemical Bonding ,Proteins ,Hydrogen Bonding ,Gene regulation ,Fibroblast Growth Factors ,MicroRNAs ,Macromolecular structure analysis ,Epilepsy, Temporal Lobe ,biology.protein ,RNA ,GRIN2A ,Gene expression ,Calcium Channels - Abstract
The genetic influence in epilepsy, characterized by unprovoked and recurrent seizures, is through variants in genes critical to brain development and function. We have carried out variant calling in Mesial Temporal Lobe Epilepsy (MTLE) patients by mapping the RNA-Seq data available at SRA, NCBI, USA onto human genome assembly hg-19. We have identified 1,75,641 SNVs in patient samples. These SNVs are distributed over 14700 genes of which 655 are already known to be associated with epilepsy. Large number of variants occur in the 3’-UTR, which is one of the regions involved in the regulation of protein translation through binding of miRNAs and RNA-binding proteins (RBP). We have focused on studying the structure-function relationship of the 3’-UTR SNVs that are common to at-least 10 of the 35 patient samples. For the first time we find SNVs exclusively in the 3’-UTR of FGF12, FAR1, NAPB, SLC1A3, SLC12A6, GRIN2A, CACNB4 and FBXO28 genes. Structural modelling reveals that the variant 3’-UTR segments possess altered secondary and tertiary structures which could affect mRNA stability and binding of RBPs to form proper ribonucleoprotein (RNP) complexes. Secondly, these SNVs have either created or destroyed miRNA-binding sites, and molecular modeling reveals that, where binding sites are created, the additional miRNAs bind strongly to 3’-UTR of only variant mRNAs. These two factors affect protein production thereby creating an imbalance in the amounts of select proteins in the cell. We suggest that in the absence of missense and nonsense variants, protein-activity imbalances associated with MTLE patients can be caused through 3’-UTR variants in relevant genes by the mechanisms mentioned above. 3’-UTR SNV has already been identified as causative variant in the neurological disorder, Tourette syndrome. Inhibition of these miRNA-mRNA bindings could be a novel way of treating drug-resistant MTLE patients. We also suggest that joint occurrence of these SNVs could serve as markers for MTLE. We find, in the present study, SNV-mediated destruction of miRNA binding site in the 3’-UTR of the gene encoding glutamate receptor subunit, and, interestingly, overexpression of one of this receptor subunit is also associated with Febrile Seizures.
- Published
- 2021
50. Observing spontaneous, accelerated substrate binding in molecular dynamics simulations of glutamate transporters
- Author
-
Peifan Li, Xiaozhen Yu, Christof Grewer, and Jiali Wang
- Subjects
Amino Acid Transport System X-AG ,Plasma protein binding ,Molecular Dynamics ,Biochemistry ,Physical Chemistry ,Substrate Specificity ,Computational Chemistry ,0302 clinical medicine ,Biochemical Simulations ,0303 health sciences ,Crystallography ,Multidisciplinary ,Chemistry ,Physics ,Simulation and Modeling ,Glutamate receptor ,Software Engineering ,Neurochemistry ,Glutamate binding ,Neurotransmitters ,Condensed Matter Physics ,Excitatory Amino Acid Transporter 1 ,Aspartate binding ,Physical Sciences ,Crystal Structure ,Engineering and Technology ,Medicine ,Glutamate ,Research Article ,Protein Binding ,Computer and Information Sciences ,Synaptic cleft ,Science ,Geometry ,Glutamic Acid ,Molecular Dynamics Simulation ,Research and Analysis Methods ,Computer Software ,03 medical and health sciences ,Solid State Physics ,Humans ,Binding site ,030304 developmental biology ,Intermolecular Forces ,Aspartic Acid ,Binding Sites ,Biology and Life Sciences ,Computational Biology ,Substrate (chemistry) ,Transporter ,Chemical Properties ,Dihedral Angles ,Biophysics ,Mathematics ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Glutamate transporters are essential for removing the neurotransmitter glutamate from the synaptic cleft. Glutamate transport across the membrane is associated with elevator-like structural changes of the transport domain. These structural changes require initial binding of the organic substrate to the transporter. Studying the binding pathway of ligands to their protein binding sites using molecular dynamics (MD) simulations requires micro-second level simulation times. Here, we used three methods to accelerate aspartate binding to the glutamate transporter homologue Gltph and to investigate the binding pathway. 1) Two methods using user-defined forces to prevent the substrate from diffusing too far from the binding site. 2) Conventional MD simulations using very high substrate concentrations in the 0.1 M range. The final, substrate bound states from these methods are comparable to the binding pose observed in crystallographic studies, although they show more flexibility in the side chain carboxylate function. We also captured an intermediate on the binding pathway, where conserved residues D390 and D394 stabilize the aspartate molecule. Finally, we investigated glutamate binding to the mammalian glutamate transporter, excitatory amino acid transporter 1 (EAAT1), for which a crystal structure is known, but not in the glutamate-bound state. Overall, the results obtained in this study reveal new insights into the pathway of substrate binding to glutamate transporters, highlighting intermediates on the binding pathway and flexible conformational states of the side chain, which most likely become locked in once the hairpin loop 2 closes to occlude the substrate.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.