1
|
陈 洁, 刘 晨, 王 春, 李 丽, 陶 伟, 徐 婧, 唐 红, 黄 丽. [Exogenous leptin improves cerebral ischemia-reperfusion-induced glutamate excitotoxic injury in mice by up-regulating GLT-1 and GLAST expression in astrocytes]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1079-1087. [PMID: 38977337 PMCID: PMC11237293 DOI: 10.12122/j.issn.1673-4254.2024.06.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To investigate the protective effect of exogenous leptin against focal cerebral ischemia-reperfusion (I/R) injury in mice and explore the underlying mechanism. METHODS A total of 100 C57BL/6 mice were randomly divided into 5 groups, including a sham-operated group, cerebral I/R model group, and 3 leptin treatment groups with intraperitoneal injections of 0.5, 1.0 or 2.0 leptin immediately after occlusion of the internal carotid artery. At 24 h after reperfusion, neurological function scores of the mice were assessed, and TTC staining was used to determine the area of cerebral infarction. The pathological changes in the cortical brain tissue of the mice were observed using HE staining, and degenerative damage of the cortical neurons were assessed with Fluoro-Jade C staining. The expression of glial fibrillary acidic protein in cortical brain tissues was detected using immunohistochemistry and Western blotting. In another 45 C57BL/6 mice with sham operation, I/R modeling, or leptin (1 mg/kg) treatment, glutamic acid in the cortical brain tissue was detected using glutamate assay, and cortical glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) protein expressions were detected using immunohistochemistry. RESULTS Compared with the I/R model mice, the leptin-treated mice had significantly lower neurological deficit scores, smaller cerebral infarct area, milder pathologies in the cortical brain tissue, and lessened cortical neuronal damage with normal morphology and less excessive proliferation of the astrocytes. Leptin treatment significantly up-regulated the expressions of GLT-1 and GLAST and lowered the content of glutamic acid in the brain tissue of the I/R mice. CONCLUSION Exogenous leptin has obvious neuroprotective effect against cerebral I/R injury in mice, mediated probably by controlling excessive astrocyte proliferation and up-regulating cortical GLT-1 and GLAST expressions to reduce glutamate-mediated excitotoxic injury of the astrocytes.
Collapse
|
2
|
Mueller SM, McFarland White K, Fass SB, Chen S, Shi Z, Ge X, Engelbach JA, Gaines SH, Bice AR, Vasek MJ, Garbow JR, Culver JP, Martinez-Lozada Z, Cohen-Salmon M, Dougherty JD, Sapkota D. Evaluation of gliovascular functions of AQP4 readthrough isoforms. Front Cell Neurosci 2023; 17:1272391. [PMID: 38077948 PMCID: PMC10701521 DOI: 10.3389/fncel.2023.1272391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/06/2023] [Indexed: 12/20/2023] Open
Abstract
Aquaporin-4 (AQP4) is a water channel protein that links the astrocytic endfeet to the blood-brain barrier (BBB) and regulates water and potassium homeostasis in the brain, as well as the glymphatic clearance of waste products that would otherwise potentiate neurological diseases. Recently, translational readthrough was shown to generate a C-terminally extended variant of AQP4, known as AQP4x, which preferentially localizes around the BBB through interaction with the scaffolding protein α-syntrophin, and loss of AQP4x disrupts waste clearance from the brain. To investigate the function of AQP4x, we generated a novel AQP4 mouse line (AllX) to increase relative levels of the readthrough variant above the ~15% of AQP4 in the brain of wild-type (WT) mice. We validated the line and assessed characteristics that are affected by the presence of AQP4x, including AQP4 and α-syntrophin localization, integrity of the BBB, and neurovascular coupling. We compared AllXHom and AllXHet mice to WT and to previously characterized AQP4 NoXHet and NoXHom mice, which cannot produce AQP4x. An increased dose of AQP4x enhanced perivascular localization of α-syntrophin and AQP4, while total protein expression of the two was unchanged. However, at 100% readthrough, AQP4x localization and the formation of higher order complexes were disrupted. Electron microscopy showed that overall blood vessel morphology was unchanged except for an increased proportion of endothelial cells with budding vesicles in NoXHom mice, which may correspond to a leakier BBB or altered efflux that was identified in NoX mice using MRI. These data demonstrate that AQP4x plays a small but measurable role in maintaining BBB integrity as well as recruiting structural and functional support proteins to the blood vessel. This also establishes a new set of genetic tools for quantitatively modulating AQP4x levels.
Collapse
Affiliation(s)
- Shayna M. Mueller
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| | - Kelli McFarland White
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| | - Stuart B. Fass
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| | - Siyu Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Zhan Shi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - John A. Engelbach
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Seana H. Gaines
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Annie R. Bice
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Michael J. Vasek
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
| | - Joel R. Garbow
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Joseph P. Culver
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Physics, Washington University in St. Louis, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, United States
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, United States
- Imaging Science PhD Program, Washington University in St. Louis, Saint Louis, MO, United States
| | - Zila Martinez-Lozada
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, The National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université PSL, Paris, France
| | - Joseph D. Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, United States
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Darshan Sapkota
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, United States
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
3
|
Wang X, Su M, Wang L, Zhou Y, Li N, Yang B. NEDD4 Like E3 Ubiquitin Protein Ligase Represses Astrocyte Activation and Aggravates Neuroinflammation in Mice with Depression via Paired Box 6 Ubiquitination. Neuroscience 2023; 530:144-157. [PMID: 37661017 DOI: 10.1016/j.neuroscience.2023.08.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Astrocytes are implicated in stress-induced neuroinflammatory responses in depression. This paper was to explore the molecular mechanism of the E3 ubiquitin ligase NEDD4L (NEDD4 like E3 ubiquitin protein ligase) in depressed mice by regulating astrocyte activation, and to find a new target for depression. A mouse model of depression was established by CUMS (chronic mild unpredictable stress) in 48 6-week male C57BL/6 mice and injected with sh-NEDD4L vector for testing behavioral and cognitive abilities, histopathological changes, and the number of GFAP-positive cells. The mRNA and protein levels of NEDD4L, PAX6 (paired box 6) and P2X7R (purinergic ligand-gated ion channel 7 receptor) were measured. Inflammation model was established by lipopolysaccharide treatment of mouse astrocyte line C8-D1A and infected with sh-NEDD4L. After CUMS induction, mice showed depression-like symptoms, increased inflammatory infiltration, decreased glial fibrillary acidic protein (GFAP)-positive cells in brain tissue, and increased NEDD4L protein levels. NEDD4L inhibition increased GFAP-positive cells, increased PAX6 protein levels and decreased P2X7R mRNA and protein levels, and decreased inflammatory factor secretion in brain tissue and in vitro cells. PAX6 knockdown or P2X7R overexpression partially reversed the effects of NEDD4L inhibition on astrocyte activation and neuroinflammation. To conclude, highly-expressed NEDD4L in depression-like mouse brain inhibits astrocyte activation and exacerbates neuroinflammation by ubiquitinating PAX6 and promoting P2X7R level.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an710000, China
| | - Mingming Su
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an710000, China
| | - Lesheng Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yixuan Zhou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an710000, China
| | - Bangkun Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Martinez-Lozada Z, Todd FW, Schober AL, Krizman E, Robinson MB, Murai KK. Cooperative and competitive regulation of the astrocytic transcriptome by neurons and endothelial cells: Impact on astrocyte maturation. J Neurochem 2023; 167:52-75. [PMID: 37525469 PMCID: PMC10543513 DOI: 10.1111/jnc.15908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023]
Abstract
Astrocytes have essential roles in central nervous system (CNS) health and disease. During development, immature astrocytes show complex interactions with neurons, endothelial cells, and other glial cell types. Our work and that of others have shown that these interactions are important for astrocytic maturation. However, whether and how these cells work together to control this process remains poorly understood. Here, we test the hypothesis that cooperative interactions of astrocytes with neurons and endothelial cells promote astrocytic maturation. Astrocytes were cultured alone, with neurons, endothelial cells, or a combination of both. This was followed by astrocyte sorting, RNA sequencing, and bioinformatic analysis to detect transcriptional changes. Across culture configurations, 7302 genes were differentially expressed by 4 or more fold and organized into 8 groups that demonstrate cooperative and antagonist effects of neurons and endothelia on astrocytes. We also discovered that neurons and endothelial cells caused splicing of 200 and 781 mRNAs, respectively. Changes in gene expression were validated using quantitative PCR, western blot (WB), and immunofluorescence analysis. We found that the transcriptomic data from the three-culture configurations correlated with protein expression of three representative targets (FAM107A, GAT3, and GLT1) in vivo. Alternative splicing results also correlated with cortical tissue isoform representation of a target (Fibronectin 1) at different developmental stages. By comparing our results to published transcriptomes of immature and mature astrocytes, we found that neurons or endothelia shift the astrocytic transcriptome toward a mature state and that the presence of both cell types has a greater effect on maturation than either cell alone. These results increase our understanding of cellular interactions/pathways that contribute to astrocytic maturation. They also provide insight into how alterations to neurons and/or endothelial cells may alter astrocytes with implications for astrocytic changes in CNS disorders and diseases.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Farmer W. Todd
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| | - Alexandra L. Schober
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| | - Elizabeth Krizman
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Michael B. Robinson
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Keith K. Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada H3G 1A4
| |
Collapse
|
5
|
Mueller SM, White KM, Fass SB, Chen S, Shi Z, Ge X, Engelbach JA, Gaines SH, Bice AR, Vasek MJ, Garbow JR, Culver JP, Zila Martinez-Lozada, Cohen-Salmon M, Dougherty JD, Sapkota D. Evaluation of gliovascular functions of Aqp4 readthrough isoforms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.549379. [PMID: 37546949 PMCID: PMC10401933 DOI: 10.1101/2023.07.21.549379] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Aquaporin-4 (AQP4) is a water channel protein that links astrocytic endfeet to the blood-brain barrier (BBB) and regulates water and potassium homeostasis in the brain, as well as the glymphatic clearance of waste products that would otherwise potentiate neurological diseases. Recently, translational readthrough was shown to generate a C-terminally extended variant of AQP4, known as AQP4x, that preferentially localizes around the BBB through interaction with the scaffolding protein α-syntrophin, and loss of AQP4x disrupts waste clearance from the brain. To investigate the function of AQP4x, we generated a novel mouse AQP4 line (AllX) to increase relative levels of the readthrough variant above the ~15% of AQP4 in the brain of wildtype (WT) mice. We validated the line and assessed characteristics that are affected by the presence of AQP4x, including AQP4 and α-syntrophin localization, integrity of the BBB, and neurovascular coupling. We compared AllXHom and AllXHet mice to wildtype, and to previously characterized AQP4 NoXHet and NoXHom mice, which cannot produce AQP4x. Increased dose of AQP4x enhanced perivascular localization of α-syntrophin and AQP4, while total protein expression of the two were unchanged. However, at 100% readthrough, AQP4x localization and formation of higher-order complexes was disrupted. Electron microscopy showed that overall blood vessel morphology was unchanged except for increased endothelial cell vesicles in NoXHom mice, which may correspond to a leakier BBB or altered efflux that was identified in NoX mice using MRI. These data demonstrate that AQP4x plays a small but measurable role in maintaining BBB integrity as well as recruiting structural and functional support proteins to the blood vessel. This also establishes a new set of genetic tools for quantitatively modulating AQP4x levels.
Collapse
Affiliation(s)
- Shayna M. Mueller
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kelli McFarland White
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Stuart B. Fass
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Siyu Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Zhan Shi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Intellectual and Development Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - John A. Engelbach
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Intellectual and Development Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Seana H Gaines
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Annie R Bice
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael J. Vasek
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Joel R. Garbow
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Intellectual and Development Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Joseph P. Culver
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Physics, Washington University in St. Louis, Saint Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Imaging Science PhD Program, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Zila Martinez-Lozada
- Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Joseph D. Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO 63110, USA
- Intellectual and Development Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Darshan Sapkota
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
6
|
Boot J, Rosser G, Kancheva D, Vinel C, Lim YM, Pomella N, Zhang X, Guglielmi L, Sheer D, Barnes M, Brandner S, Nelander S, Movahedi K, Marino S. Global hypo-methylation in a proportion of glioblastoma enriched for an astrocytic signature is associated with increased invasion and altered immune landscape. eLife 2022; 11:e77335. [PMID: 36412091 PMCID: PMC9681209 DOI: 10.7554/elife.77335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
We describe a subset of glioblastoma, the most prevalent malignant adult brain tumour, harbouring a bias towards hypomethylation at defined differentially methylated regions. This epigenetic signature correlates with an enrichment for an astrocytic gene signature, which together with the identification of enriched predicted binding sites of transcription factors known to cause demethylation and to be involved in astrocytic/glial lineage specification, point to a shared ontogeny between these glioblastomas and astroglial progenitors. At functional level, increased invasiveness, at least in part mediated by SRPX2, and macrophage infiltration characterise this subset of glioblastoma.
Collapse
Affiliation(s)
- James Boot
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Gabriel Rosser
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Dailya Kancheva
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Claire Vinel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Yau Mun Lim
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Nicola Pomella
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Loredana Guglielmi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Denise Sheer
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| | - Michael Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Sebastian Brandner
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, and Department of Neurodegenerative Disease, Queen Square, Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Sven Nelander
- Department of Immunology Genetics and Pathology, Uppsala UniversityUppsalaSweden
| | - Kiavash Movahedi
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit BrusselBrusselsBelgium
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary UniversityLondonUnited Kingdom
| |
Collapse
|
7
|
Reis L, Raciti M, Rodriguez PG, Joseph B, Al Rayyes I, Uhlén P, Falk A, da Cunha Lima ST, Ceccatelli S. Glyphosate-based herbicide induces long-lasting impairment in neuronal and glial differentiation. ENVIRONMENTAL TOXICOLOGY 2022; 37:2044-2057. [PMID: 35485992 PMCID: PMC9541419 DOI: 10.1002/tox.23549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 05/09/2023]
Abstract
Glyphosate-based herbicides (GBH) are among the most sold pesticides in the world. There are several formulations based on the active ingredient glyphosate (GLY) used along with other chemicals to improve the absorption and penetration in plants. The final composition of commercial GBH may modify GLY toxicological profile, potentially enhancing its neurotoxic properties. The developing nervous system is particularly susceptible to insults occurring during the early phases of development, and exposure to chemicals in this period may lead to persistent impairments on neurogenesis and differentiation. The aim of this study was to evaluate the long-lasting effects of a sub-cytotoxic concentration, 2.5 parts per million of GBH and GLY, on the differentiation of human neuroepithelial stem cells (NES) derived from induced pluripotent stem cells (iPSC). We treated NES cells with each compound and evaluated the effects on key cellular processes, such as proliferation and differentiation in daughter cells never directly exposed to the toxicants. We found that GBH induced a more immature neuronal profile associated to increased PAX6, NESTIN and DCX expression, and a shift in the differentiation process toward glial cell fate at the expense of mature neurons, as shown by an increase in the glial markers GFAP, GLT1, GLAST and a decrease in MAP2. Such alterations were associated to dysregulation of key genes critically involved in neurogenesis, including PAX6, HES1, HES5, and DDK1. Altogether, the data indicate that subtoxic concentrations of GBH, but not of GLY, induce long-lasting impairments on the differentiation potential of NES cells.
Collapse
Affiliation(s)
- Luã Reis
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Marilena Raciti
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | | | - Bertrand Joseph
- Institute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Per Uhlén
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Anna Falk
- Department of NeuroscienceKarolinska InstitutetStockholmSweden
| | - Suzana Telles da Cunha Lima
- Laboratório de Bioprospecção e Biotecnologia, Instituto de BiologiaUniversidade Federal da Bahia (UFBA)SalvadorBrazil
| | | |
Collapse
|
8
|
Nuclear Transporter IPO13 Is Central to Efficient Neuronal Differentiation. Cells 2022; 11:cells11121904. [PMID: 35741036 PMCID: PMC9221400 DOI: 10.3390/cells11121904] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Molecular transport between the nucleus and cytoplasm of the cell is mediated by the importin superfamily of transport receptors, of which the bidirectional transporter Importin 13 (IPO13) is a unique member, with a critical role in early embryonic development through nuclear transport of key regulators, such as transcription factors Pax6, Pax3, and ARX. Here, we examined the role of IPO13 in neuronal differentiation for the first time, using a mouse embryonic stem cell (ESC) model and a monolayer-based differentiation protocol to compare IPO13−/− to wild type ESCs. Although IPO13−/− ESCs differentiated into neural progenitor cells, as indicated by the expression of dorsal forebrain progenitor markers, reduced expression of progenitor markers Pax6 and Nestin compared to IPO13−/− was evident, concomitant with reduced nuclear localisation/transcriptional function of IPO13 import cargo Pax6. Differentiation of IPO13−/− cells into neurons appeared to be strongly impaired, as evidenced by altered morphology, reduced expression of key neuronal markers, and altered response to the neurotransmitter glutamate. Our findings establish that IPO13 has a key role in ESC neuronal differentiation, in part through the nuclear transport of Pax6.
Collapse
|
9
|
Ochi S, Manabe S, Kikkawa T, Osumi N. Thirty Years' History since the Discovery of Pax6: From Central Nervous System Development to Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:6115. [PMID: 35682795 PMCID: PMC9181425 DOI: 10.3390/ijms23116115] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a sequence-specific DNA binding transcription factor that positively and negatively regulates transcription and is expressed in multiple cell types in the developing and adult central nervous system (CNS). As indicated by the morphological and functional abnormalities in spontaneous Pax6 mutant rodents, Pax6 plays pivotal roles in various biological processes in the CNS. At the initial stage of CNS development, Pax6 is responsible for brain patterning along the anteroposterior and dorsoventral axes of the telencephalon. Regarding the anteroposterior axis, Pax6 is expressed inversely to Emx2 and Coup-TF1, and Pax6 mutant mice exhibit a rostral shift, resulting in an alteration of the size of certain cortical areas. Pax6 and its downstream genes play important roles in balancing the proliferation and differentiation of neural stem cells. The Pax6 gene was originally identified in mice and humans 30 years ago via genetic analyses of the eye phenotypes. The human PAX6 gene was discovered in patients who suffer from WAGR syndrome (i.e., Wilms tumor, aniridia, genital ridge defects, mental retardation). Mutations of the human PAX6 gene have also been reported to be associated with autism spectrum disorder (ASD) and intellectual disability. Rodents that lack the Pax6 gene exhibit diverse neural phenotypes, which might lead to a better understanding of human pathology and neurodevelopmental disorders. This review describes the expression and function of Pax6 during brain development, and their implications for neuropathology.
Collapse
Affiliation(s)
| | | | | | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (S.O.); (S.M.); (T.K.)
| |
Collapse
|
10
|
Hirschberg S, Dvorzhak A, Rasooli-Nejad SMA, Angelov S, Kirchner M, Mertins P, Lättig-Tünnemann G, Harms C, Schmitz D, Grantyn R. Uncoupling the Excitatory Amino Acid Transporter 2 From Its C-Terminal Interactome Restores Synaptic Glutamate Clearance at Corticostriatal Synapses and Alleviates Mutant Huntingtin-Induced Hypokinesia. Front Cell Neurosci 2022; 15:792652. [PMID: 35173582 PMCID: PMC8841566 DOI: 10.3389/fncel.2021.792652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/21/2021] [Indexed: 02/05/2023] Open
Abstract
Rapid removal of glutamate from the sites of glutamate release is an essential step in excitatory synaptic transmission. However, despite many years of research, the molecular mechanisms underlying the intracellular regulation of glutamate transport at tripartite synapses have not been fully uncovered. This limits the options for pharmacological treatment of glutamate-related motor disorders, including Huntington’s disease (HD). We therefore investigated the possible binding partners of transgenic EAAT2 and their alterations under the influence of mutant huntingtin (mHTT). Mass spectrometry analysis after pull-down of striatal YFP-EAAT2 from wild-type (WT) mice and heterozygote (HET) Q175 mHTT-knock-in mice identified a total of 148 significant (FDR < 0.05) binders to full-length EAAT2. Of them 58 proteins exhibited mHTT-related differences. Most important, in 26 of the 58 mHTT-sensitive cases, protein abundance changed back toward WT levels when the mice expressed a C-terminal-truncated instead of full-length variant of EAAT2. These findings motivated new attempts to clarify the role of astrocytic EAAT2 regulation in cortico-basal movement control. Striatal astrocytes of Q175 HET mice were targeted by a PHP.B vector encoding EAAT2 with different degree of C-terminal modification, i.e., EAAT2-S506X (truncation at S506), EAAT2-4KR (4 lysine to arginine substitutions) or EAAT2 (full-length). The results were compared to HET and WT injected with a tag-only vector (CTRL). It was found that the presence of a C-terminal-modified EAAT2 transgene (i) increased the level of native EAAT2 protein in striatal lysates and perisynaptic astrocyte processes, (ii) enhanced the glutamate uptake of transduced astrocytes, (iii) stimulated glutamate clearance at individual corticostriatal synapses, (iv) increased the glutamate uptake of striatal astrocytes and (iv) alleviated the mHTT-related hypokinesia (open field indicators of movement initiation). In contrast, over-expression of full-length EAAT2 neither facilitated glutamate uptake nor locomotion. Together, our results support the new hypothesis that preventing abnormal protein-protein interactions at the C-terminal of EAAT2 could eliminate the mHTT-related deficits in corticostriatal synaptic glutamate clearance and movement initiation.
Collapse
Affiliation(s)
- Stefan Hirschberg
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anton Dvorzhak
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Seyed M. A. Rasooli-Nejad
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Svilen Angelov
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Marieluise Kirchner
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Gilla Lättig-Tünnemann
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Rosemarie Grantyn
- Synaptic Dysfunction Lab, Neuroscience Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Department of Experimental Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Cluster of Excellence NeuroCure, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- *Correspondence: Rosemarie Grantyn,
| |
Collapse
|
11
|
Rodríguez-Campuzano AG, Ortega A. Glutamate transporters: Critical components of glutamatergic transmission. Neuropharmacology 2021; 192:108602. [PMID: 33991564 DOI: 10.1016/j.neuropharm.2021.108602] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Ada G Rodríguez-Campuzano
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, Ciudad de México, 07000, Mexico
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, Ciudad de México, 07000, Mexico.
| |
Collapse
|
12
|
Todd AC, Hardingham GE. The Regulation of Astrocytic Glutamate Transporters in Health and Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E9607. [PMID: 33348528 PMCID: PMC7766851 DOI: 10.3390/ijms21249607] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
The astrocytic glutamate transporters excitatory amino acid transporters 1 and 2 (EAAT1 and EAAT2) play a key role in nervous system function to maintain extracellular glutamate levels at low levels. In physiology, this is essential for the rapid uptake of synaptically released glutamate, maintaining the temporal fidelity of synaptic transmission. However, EAAT1/2 hypo-expression or hypo-function are implicated in several disorders, including epilepsy and neurodegenerative diseases, as well as being observed naturally with aging. This not only disrupts synaptic information transmission, but in extremis leads to extracellular glutamate accumulation and excitotoxicity. A key facet of EAAT1/2 expression in astrocytes is a requirement for signals from other brain cell types in order to maintain their expression. Recent evidence has shown a prominent role for contact-dependent neuron-to-astrocyte and/or endothelial cell-to-astrocyte Notch signalling for inducing and maintaining the expression of these astrocytic glutamate transporters. The relevance of this non-cell-autonomous dependence to age- and neurodegenerative disease-associated decline in astrocytic EAAT expression is discussed, plus the implications for disease progression and putative therapeutic strategies.
Collapse
Affiliation(s)
- Alison C. Todd
- UK Dementia Research Institute at the University of Edinburgh, Chancellor’s Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK;
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Giles E. Hardingham
- UK Dementia Research Institute at the University of Edinburgh, Chancellor’s Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK;
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
13
|
Smaga I, Gawlińska K, Frankowska M, Wydra K, Sadakierska-Chudy A, Suder A, Piechota M, Filip M. Extinction Training after Cocaine Self-Administration Influences the Epigenetic and Genetic Machinery Responsible for Glutamatergic Transporter Gene Expression in Male Rat Brain. Neuroscience 2020; 451:99-110. [PMID: 33065231 DOI: 10.1016/j.neuroscience.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022]
Abstract
Glutamate is a key excitatory neurotransmitter in the central nervous system. The balance of glutamatergic transporter proteins allows long-term maintenance of glutamate homeostasis in the brain, which is impaired during cocaine use disorder. The aim of this study was to investigate changes in the gene expression of SLC1A2 (encoding GLT-1), and SLC7A11 (encoding xCT), in rat brain structures after short-term (3 days) and long-term (10 days) extinction training using microarray analysis and quantitative real-time PCR. Furthermore, we analyzed the expression of genes encoding transcription factors, i.e., NFKB1 and NFKB2 (encoding NF-κB), PAX6, (encoding Pax6), and NFE2L2 (encoding Nrf2), to verify the correlation between changes in glutamatergic transporters and changes in their transcriptional factors and microRNAs (miRNAs; miR-124a, miR-543-3p and miR-342-3p) and confirm the epigenetic mechanism. We found reduced GLT-1 transcript and mRNA level in the prefrontal cortex (PFCTX) and dorsal striatum (DSTR) in rats that had previously self-administered cocaine after 3 days of extinction training, which was associated with downregulation of PAX6 (transcript and mRNA) and NFKB2 (mRNA) level in the PFCTX and with upregulation of miR-543-3p and miR-342-3p in the DSTR. The xCT mRNA level was reduced in the PFCTX and DSTR, and NFE2L2 transcript level in the PFCTX was decreased on the 3rd day of extinction training. In conclusion, 3-day drug-free period modulates GLT-1 and xCT gene expression through genetic and epigenetic mechanisms, and such changes in expression seem to be potential molecular targets for developing a treatment for cocaine-seeking behavior.
Collapse
Affiliation(s)
- Irena Smaga
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland.
| | - Kinga Gawlińska
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Małgorzata Frankowska
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Karolina Wydra
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Anna Sadakierska-Chudy
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Agata Suder
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| | - Marcin Piechota
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Molecular Neuropharmacology, Smętna 12, 31-343 Kraków, Poland
| | - Małgorzata Filip
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Smętna 12, PL 31-343 Kraków, Poland
| |
Collapse
|
14
|
Martinez-Lozada Z, Robinson MB. Reciprocal communication between astrocytes and endothelial cells is required for astrocytic glutamate transporter 1 (GLT-1) expression. Neurochem Int 2020; 139:104787. [PMID: 32650029 DOI: 10.1016/j.neuint.2020.104787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/15/2020] [Accepted: 06/14/2020] [Indexed: 12/12/2022]
Abstract
Astrocytes have diverse functions that are supported by their anatomic localization between neurons and blood vessels. One of these functions is the clearance of extracellular glutamate. Astrocytes clear glutamate using two Na+-dependent glutamate transporters, GLT-1 (also called EAAT2) and GLAST (also called EAAT1). GLT-1 expression increases during synaptogenesis and is a marker of astrocyte maturation. Over 20 years ago, several groups demonstrated that astrocytes in culture express little or no GLT-1 and that neurons induce expression. We recently demonstrated that co-culturing endothelia with mouse astrocytes also induced expression of GLT-1 and GLAST. These increases were blocked by an inhibitor of γ-secretase. This and other observations are consistent with the hypothesis that Notch signaling is required, but the ligands involved were not identified. In the present study, we used rat astrocyte cultures to further define the mechanisms by which endothelia induce expression of GLT-1 and GLAST. We found that co-cultures of astrocytes and endothelia express higher levels of GLT-1 and GLAST protein and mRNA. That endothelia activate Hes5, a transcription factor target of Notch, in astrocytes. Using recombinant Notch ligands, anti-Notch ligand neutralizing antibodies, and shRNAs, we provide evidence that both Dll1 and Dll4 contribute to endothelia-dependent regulation of GLT-1. We also provide evidence that astrocytes secrete a factor(s) that induces expression of Dll4 in endothelia and that this effect is required for Notch-dependent induction of GLT-1. Together these studies indicate that reciprocal communication between astrocytes and endothelia is required for appropriate astrocyte maturation and that endothelia likely deploy additional non-Notch signals to induce GLT-1.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA, 19104-4318; Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA.
| |
Collapse
|
15
|
Peng M, Ling X, Song R, Gao X, Liang Z, Fang F, Cang J. Upregulation of GLT-1 via PI3K/Akt Pathway Contributes to Neuroprotection Induced by Dexmedetomidine. Front Neurol 2019; 10:1041. [PMID: 31611842 PMCID: PMC6776610 DOI: 10.3389/fneur.2019.01041] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022] Open
Abstract
Perioperative ischemic stroke usually leads to neurological dysfunction caused by neuron death. During the ischemic condition, excitotoxity due to extracellular glutamate accumulation is a main mechanism of neuron damage. The clearance of glutamate mainly depends on glutamate transporter-1 (GLT-1) which is expressed in astrocytes. Dexmedetomidine, an α2 adrenergic receptor agonist, is proved to induce neuroprotection. This study was set out to investigate the glutamate-related mechanism involved in the neuroprotective effect of dexmedetomidine. Middle cerebral artery occlusion (MCAO) was used as a model of ischemic stroke in our study. We determined Neurological deficit scores (NDS) and Magnetic resonance imaging (MRI) at three points (2, 6, and 24 h) after middle cerebral artery occlusion (MCAO) to evaluate the neuroprotective effect of dexmedetomidine. Besides, we performed western blot (6 and 24 h after MACO) and immunofluorescent staining (24 h after MCAO) to observe the expression of GLT-1. The effect and mechanism of dexmedetomidine on GLT-1 in primary cultured astrocytes were investigated using western blot and RT-PCR. Our results showed that pretreatment with dexmedetomidine improved NDS and reduced infarct volume as well as upregulating GLT-1 expression. Furthermore, using Atipamezole and LY294002, we found that dexmedetomidine significantly increased GLT-1 levels in astrocytes via activating α2 adrenergic receptor and PI3K/AKT pathway both in vitro and in vivo study. Overall, our present study indicated that dexmedetomidine had neuroprotective effects on ischemia stroke and upregulation of GLT-1 levels by PI3K/AKT dependent pathway might be the potential mechanism.
Collapse
Affiliation(s)
- Mengyuan Peng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruixue Song
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuan Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Liang
- Comparative Nerve Imaging Study Group, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fang Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
The Effect of Glutamatergic Modulators on Extracellular Glutamate: How Does this Information Contribute to the Discovery of Novel Antidepressants? Curr Ther Res Clin Exp 2019; 91:25-32. [PMID: 31871505 PMCID: PMC6911922 DOI: 10.1016/j.curtheres.2019.100566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/29/2019] [Indexed: 01/19/2023] Open
Abstract
The complexity of glutamatergic signaling challenges glutamate modulator usage. Functional biomarkers are needed to understand the MOA of glutamate modulators. Evaluating drug effect on EAATs' kinetics may add to antidepressant discovery.
Background In the search for new antidepressants, clinical researchers have been using drugs that simultaneously modulate multiple targets. During preclinical and clinical trials, the glutamatergic modulators riluzole and ketamine have received particular attention. Glutamatergic agents have a modulatory effect on synaptic transmission, so they can act on both neurons and astrocytes. In addition to influencing the quantity of glutamate released, these modulators can also affect the expression, localization, and functionality of glutamate-binding sites. Objective This review discusses the complexity of the glutamatergic system, the ambiguity of data regarding glutamate levels in patients with depression, as well as the mechanisms of action for riluzole and ketamine, which includes their relation to the physiology of glutamatergic transmission. The principal aim is to contribute to the development of novel glutamatergic antidepressant medications whilst emphasizing the need for innovative approaches that evaluate their effects on extracellular glutamate. Methods Literature was obtained via PubMed by searching the term depression in combination with each of the following terms: riluzole, ketamine, and glutamate. The search was restricted to full-text articles published in English between 1985 and 2018 relating to both the modulatory mechanisms of glutamatergic-binding proteins and the antidepressant actions of these medicines. Articles about mechanisms associated with synaptic plasticity and antidepressant effects were excluded. Results Although experimental data relates glutamatergic signaling to the pathophysiology of major depression and bipolar disorder, the role of glutamate—as well as its extracellular concentration in patients with said disorders—is still unclear. Riluzole's antidepressant action is ascribed to its capacity to reduce glutamate levels in the synaptic cleft, and ketamine's effect has been associated with increased extracellular glutamate levels. Conclusions The strategy of using glutamatergic modulators as therapeutic agents requires a better understanding of the role of glutamate in the pathophysiology of depression. Gaining such understanding is a challenge because it entails evaluating different targets as well as the effects of these modulators on the kinetics of glutamate uptake. Essentially, glutamate transport is a dynamic process and, currently, it is still necessary to develop new approaches to assay glutamate in the synaptic cleft. ORCID: 0000-0002-3358-6939.
Collapse
|
17
|
Niedzielska-Andres E, Mizera J, Sadakierska-Chudy A, Pomierny-Chamioło L, Filip M. Changes in the glutamate biomarker expression in rats vulnerable or resistant to the rewarding effects of cocaine and their reversal by ceftriaxone. Behav Brain Res 2019; 370:111945. [DOI: 10.1016/j.bbr.2019.111945] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
|
18
|
Olivares-Bañuelos TN, Chí-Castañeda D, Ortega A. Glutamate transporters: Gene expression regulation and signaling properties. Neuropharmacology 2019; 161:107550. [PMID: 30822498 DOI: 10.1016/j.neuropharm.2019.02.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 12/24/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the vertebrate central nervous system. During synaptic activity, glutamate is released and binds to specific membrane receptors and transporters activating, in the one hand, a wide variety of signal transduction cascades, while in the other hand, its removal from the synaptic cleft. Extracellular glutamate concentrations are maintained within physiological levels mainly by glia glutamate transporters. Inefficient clearance of this amino acid is neurotoxic due to a prolonged hyperactivation of its postsynaptic receptors, exacerbating a wide array of intracellular events linked to an ionic imbalance, that results in neuronal cell death. This process is known as excitotoxicity and is the underlying mechanisms of an important number of neurodegenerative diseases. Therefore, it is important to understand the regulation of glutamate transporters function. The transporter activity can be regulated at different levels: gene expression, transporter protein targeting and trafficking, and post-translational modifications of the transporter protein. The identification of these mechanisms has paved the way to our current understanding the role of glutamate transporters in brain physiology and will certainly provide the needed biochemical information for the development of therapeutic strategies towards the establishment of novel therapeutic approaches for the treatment and/or prevention of pathologies associated with excitotoxicity insults. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Tatiana N Olivares-Bañuelos
- Instituto de Investigaciones Oceanológicas, Universidad Autónoma de Baja California, Carretera Tijuana-Ensenada No. 3917, Fraccionamiento Playitas, 22860, Ensenada, Baja California, Mexico
| | - Donají Chí-Castañeda
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, Ciudad de México, 07000, Mexico
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado Postal 14-740, Ciudad de México, 07000, Mexico.
| |
Collapse
|
19
|
Mahmoud S, Gharagozloo M, Simard C, Gris D. Astrocytes Maintain Glutamate Homeostasis in the CNS by Controlling the Balance between Glutamate Uptake and Release. Cells 2019; 8:E184. [PMID: 30791579 PMCID: PMC6406900 DOI: 10.3390/cells8020184] [Citation(s) in RCA: 355] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 01/26/2023] Open
Abstract
Glutamate is one of the most prevalent neurotransmitters released by excitatory neurons in the central nervous system (CNS); however, residual glutamate in the extracellular space is, potentially, neurotoxic. It is now well-established that one of the fundamental functions of astrocytes is to uptake most of the synaptically-released glutamate, which optimizes neuronal functions and prevents glutamate excitotoxicity. In the CNS, glutamate clearance is mediated by glutamate uptake transporters expressed, principally, by astrocytes. Interestingly, recent studies demonstrate that extracellular glutamate stimulates Ca2+ release from the astrocytes' intracellular stores, which triggers glutamate release from astrocytes to the adjacent neurons, mostly by an exocytotic mechanism. This released glutamate is believed to coordinate neuronal firing and mediate their excitatory or inhibitory activity. Therefore, astrocytes contribute to glutamate homeostasis in the CNS, by maintaining the balance between their opposing functions of glutamate uptake and release. This dual function of astrocytes represents a potential therapeutic target for CNS diseases associated with glutamate excitotoxicity. In this regard, we summarize the molecular mechanisms of glutamate uptake and release, their regulation, and the significance of both processes in the CNS. Also, we review the main features of glutamate metabolism and glutamate excitotoxicity and its implication in CNS diseases.
Collapse
Affiliation(s)
- Shaimaa Mahmoud
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Marjan Gharagozloo
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Camille Simard
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Denis Gris
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
20
|
Vasilyeva TA, Voskresenskaya AA, Pozdeyeva NA, Marakhonov AV, Zinchenko RA. PAX6 Gene Characteristic and Causative Role of PAX6 Mutations in Inherited Eye Pathologies. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418090156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017; 143:489-506. [PMID: 28771710 DOI: 10.1111/jnc.14135] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Lee ML, Martinez-Lozada Z, Krizman EN, Robinson MB. Brain endothelial cells induce astrocytic expression of the glutamate transporter GLT-1 by a Notch-dependent mechanism. J Neurochem 2017. [PMID: 28771710 DOI: 10.1111/jnc.13825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Neuron-secreted factors induce astrocytic expression of the glutamate transporter, GLT-1 (excitatory amino acid transporter 2). In addition to their elaborate anatomic relationships with neurons, astrocytes also have processes that extend to and envelop the vasculature. Although previous studies have demonstrated that brain endothelia contribute to astrocyte differentiation and maturation, the effects of brain endothelia on astrocytic expression of GLT-1 have not been examined. In this study, we tested the hypothesis that endothelia induce expression of GLT-1 by co-culturing astrocytes from mice that utilize non-coding elements of the GLT-1 gene to control expression of reporter proteins with the mouse endothelial cell line, bEND.3. We found that endothelia increased steady state levels of reporter and GLT-1 mRNA/protein. Co-culturing with primary rat brain endothelia also increases reporter protein, GLT-1 protein, and GLT-1-mediated glutamate uptake. The Janus kinase/signal transducer and activator of transcription 3, bone morphogenic protein/transforming growth factor β, and nitric oxide pathways have been implicated in endothelia-to-astrocyte signaling; we provide multiple lines of evidence that none of these pathways mediate the effects of endothelia on astrocytic GLT-1 expression. Using transwells with a semi-permeable membrane, we demonstrate that the effects of the bEND.3 cell line are dependent upon contact. Notch has also been implicated in endothelia-astrocyte signaling in vitro and in vivo. The first step of Notch signaling requires cleavage of Notch intracellular domain by γ-secretase. We demonstrate that the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester blocks endothelia-induced increases in GLT-1. We show that the levels of Notch intracellular domain are higher in nuclei of astrocytes co-cultured with endothelia, an effect also blocked by N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. Finally, infection of co-cultures with shRNA directed against recombination signal binding protein for immunoglobulin kappa J, a Notch effector, also reduces endothelia-dependent increases in enhanced green fluorescent protein and GLT-1. Together, these studies support a novel role for Notch in endothelia-dependent induction of GLT-1 expression. Cover Image for this issue: doi. 10.1111/jnc.13825.
Collapse
Affiliation(s)
- Meredith L Lee
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zila Martinez-Lozada
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth N Krizman
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael B Robinson
- Departments of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Kardos J, Héja L, Jemnitz K, Kovács R, Palkovits M. The nature of early astroglial protection-Fast activation and signaling. Prog Neurobiol 2017; 153:86-99. [PMID: 28342942 DOI: 10.1016/j.pneurobio.2017.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/22/2016] [Accepted: 03/05/2017] [Indexed: 12/14/2022]
Abstract
Our present review is focusing on the uniqueness of balanced astroglial signaling. The balance of excitatory and inhibitory signaling within the CNS is mainly determined by sharp synaptic transients of excitatory glutamate (Glu) and inhibitory γ-aminobutyrate (GABA) acting on the sub-second timescale. Astroglia is involved in excitatory chemical transmission by taking up i) Glu through neurotransmitter-sodium transporters, ii) K+ released due to presynaptic action potential generation, and iii) water keeping osmotic pressure. Glu uptake-coupled Na+ influx may either ignite long-range astroglial Ca2+ transients or locally counteract over-excitation via astroglial GABA release and increased tonic inhibition. Imbalance of excitatory and inhibitory drives is associated with a number of disease conditions, including prevalent traumatic and ischaemic injuries or the emergence of epilepsy. Therefore, when addressing the potential of early therapeutic intervention, astroglial signaling functions combating progress of Glu excitotoxicity is of critical importance. We suggest, that excitotoxicity is linked primarily to over-excitation induced by the impairment of astroglial Glu uptake and/or GABA release. Within this framework, we discuss the acute alterations of Glu-cycling and metabolism and conjecture the therapeutic promise of regulation. We also confer the role played by key carrier proteins and enzymes as well as their interplay at the molecular, cellular, and organ levels. Moreover, based on our former studies, we offer potential prospect on the emerging theme of astroglial succinate sensing in course of Glu excitotoxicity.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary.
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Hungary
| | - Richárd Kovács
- Institute of Neurophysiology, Charité - Universitätsmedizin, Berlin, Germany
| | - Miklós Palkovits
- Human Brain Tissue Bank and Laboratory, Semmelweis University, Budapest, Hungary
| |
Collapse
|
24
|
Murphy-Royal C, Dupuis J, Groc L, Oliet SHR. Astroglial glutamate transporters in the brain: Regulating neurotransmitter homeostasis and synaptic transmission. J Neurosci Res 2017; 95:2140-2151. [PMID: 28150867 DOI: 10.1002/jnr.24029] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/20/2016] [Accepted: 01/02/2017] [Indexed: 12/29/2022]
Abstract
Astrocytes, the major glial cell type in the central nervous system (CNS), are critical for brain function and have been implicated in various disorders of the central nervous system. These cells are involved in a wide range of cerebral processes including brain metabolism, control of central blood flow, ionic homeostasis, fine-tuning synaptic transmission, and neurotransmitter clearance. Such varied roles can be efficiently carried out due to the intimate interactions astrocytes maintain with neurons, the vasculature, as well as with other glial cells. Arguably, one of the most important functions of astrocytes in the brain is their control of neurotransmitter clearance. This is particularly true for glutamate whose timecourse in the synaptic cleft needs to be controlled tightly under physiological conditions to maintain point-to-point excitatory transmission, thereby limiting spillover and activation of more receptors. Most importantly, accumulation of glutamate in the extracellular space can trigger excessive activation of glutamatergic receptors and lead to excitotoxicity, a trademark of many neurodegenerative diseases. It is thus of utmost importance for both physiological and pathophysiological reasons to understand the processes that control glutamate time course within the synaptic cleft and regulate its concentrations in the extracellular space. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ciaran Murphy-Royal
- Neurocentre Magendie, Inserm U1215, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Julien Dupuis
- Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Stéphane H R Oliet
- Neurocentre Magendie, Inserm U1215, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| |
Collapse
|
25
|
Regulation of Glutamate Transporter Expression in Glial Cells. ADVANCES IN NEUROBIOLOGY 2017; 16:199-224. [DOI: 10.1007/978-3-319-55769-4_10] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
EAAT2 and the Molecular Signature of Amyotrophic Lateral Sclerosis. ADVANCES IN NEUROBIOLOGY 2017; 16:117-136. [PMID: 28828608 DOI: 10.1007/978-3-319-55769-4_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapid and fatal neurodegenerative disease, primarily affecting upper and lower motor neurons. It is an extremely heterogeneous disease in both cause and symptom development, and its mechanisms of pathogenesis remain largely unknown. Excitotoxicity, a process caused by excessive glutamate signaling, is believed to play a substantial role, however. Excessive glutamate release, changes in postsynaptic glutamate receptors, and reduction of functional astrocytic glutamate transporters contribute to excitotoxicity in ALS. Here, we explore the roles of each, with a particular emphasis on glutamate transporters and attempts to increase them as therapy for ALS. Screening strategies have been employed to find compounds that increase the functional excitatory amino acid transporter EAAT2 (GLT1), which is responsible for the vast majority of glutamate clearance. One such compound, ceftriaxone, was recently tested in clinical trials but unfortunately did not modify disease course, though its effect on EAAT2 expression in patients was not measured.
Collapse
|
27
|
Martinez-Lozada Z, Guillem AM, Robinson MB. Transcriptional Regulation of Glutamate Transporters: From Extracellular Signals to Transcription Factors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:103-45. [PMID: 27288076 DOI: 10.1016/bs.apha.2016.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian CNS. It mediates essentially all rapid excitatory signaling. Dysfunction of glutamatergic signaling contributes to developmental, neurologic, and psychiatric diseases. Extracellular glutamate is cleared by a family of five Na(+)-dependent glutamate transporters. Two of these transporters (GLAST and GLT-1) are relatively selectively expressed in astrocytes. Other of these transporters (EAAC1) is expressed by neurons throughout the nervous system. Expression of the last two members of this family (EAAT4 and EAAT5) is almost exclusively restricted to specific populations of neurons in cerebellum and retina, respectively. In this review, we will discuss our current understanding of the mechanisms that control transcriptional regulation of the different members of this family. Over the last two decades, our understanding of the mechanisms that regulate expression of GLT-1 and GLAST has advanced considerably; several specific transcription factors, cis-elements, and epigenetic mechanisms have been identified. For the other members of the family, little or nothing is known about the mechanisms that control their transcription. It is assumed that by defining the mechanisms involved, we will advance our understanding of the events that result in cell-specific expression of these transporters and perhaps begin to define the mechanisms by which neurologic diseases are changing the biology of the cells that express these transporters. This approach might provide a pathway for developing new therapies for a wide range of essentially untreatable and devastating diseases that kill neurons by an excitotoxic mechanism.
Collapse
Affiliation(s)
- Z Martinez-Lozada
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - A M Guillem
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - M B Robinson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|