1
|
McIntyre RS, Jain R. Glutamatergic Modulators for Major Depression from Theory to Clinical Use. CNS Drugs 2024; 38:869-890. [PMID: 39150594 PMCID: PMC11486832 DOI: 10.1007/s40263-024-01114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Major depressive disorder (MDD) is a chronic, burdensome, highly prevalent disease that is characterized by depressed mood and anhedonia. MDD is especially burdensome as approved monoamine antidepressant treatments have weeks-long delays before clinical benefit and low remission rates. In the past 2 decades, a promising target emerged to improve patient outcomes in depression treatment: glutamatergic signaling. This narrative review provides a high-level overview of glutamate signaling in synaptogenesis and neural plasticity and the implications of glutamate dysregulation in depression. Based on this preclinical evidence implicating glutamate in depression and the rapid improvement of depression with ketamine treatment in a proof-of-concept trial, a range of N-methyl-D-aspartate (NMDA)-targeted therapies have been investigated. While an array of treatments has been investigated in registered phase 2 or 3 clinical trials, the development of most of these agents has been discontinued. Multiple glutamate-targeted antidepressants are actively in development, and two are approved. Nasal administration of esketamine (Spravato®) was approved by the US Food and Drug Administration (FDA) in 2019 to treat adults with treatment-resistant depression and in 2020 for adults with MDD with acute suicidal ideation or behavior. Oral combination dextromethorphan-bupropion (AXS-05, Auvelity® extended-release tablet) was FDA approved in 2022 for the treatment of MDD in adults. These approvals bolster the importance of glutamate in depression and represent an exciting breakthrough in contemporary psychiatry, providing new avenues of treatment for patients as first-line therapy or with either poor response or unacceptable side effects to monoaminergic antidepressants.
Collapse
Affiliation(s)
- Roger S McIntyre
- University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Rakesh Jain
- Department of Psychiatry, Texas Tech University School of Medicine-Permian Basin, Midland, TX, USA.
- Texas Tech University School of Medicine-Permian Basin, 2500 W William Cannon Drive, Suite 505, Austin, Texas, 78745, USA.
| |
Collapse
|
2
|
Kukułowicz J, Pietrzak-Lichwa K, Klimończyk K, Idlin N, Bajda M. The SLC6A15-SLC6A20 Neutral Amino Acid Transporter Subfamily: Functions, Diseases, and Their Therapeutic Relevance. Pharmacol Rev 2023; 76:142-193. [PMID: 37940347 DOI: 10.1124/pharmrev.123.000886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/07/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
The neutral amino acid transporter subfamily that consists of six members, consecutively SLC6A15-SLC620, also called orphan transporters, represents membrane, sodium-dependent symporter proteins that belong to the family of solute carrier 6 (SLC6). Primarily, they mediate the transport of neutral amino acids from the extracellular milieu toward cell or storage vesicles utilizing an electric membrane potential as the driving force. Orphan transporters are widely distributed throughout the body, covering many systems; for instance, the central nervous, renal, or intestinal system, supplying cells into molecules used in biochemical, signaling, and building pathways afterward. They are responsible for intestinal absorption and renal reabsorption of amino acids. In the central nervous system, orphan transporters constitute a significant medium for the provision of neurotransmitter precursors. Diseases related with aforementioned transporters highlight their significance; SLC6A19 mutations are associated with metabolic Hartnup disorder, whereas altered expression of SLC6A15 has been associated with a depression/stress-related disorders. Mutations of SLC6A18-SLCA20 cause iminoglycinuria and/or hyperglycinuria. SLC6A18-SLC6A20 to reach the cellular membrane require an ancillary unit ACE2 that is a molecular target for the spike protein of the SARS-CoV-2 virus. SLC6A19 has been proposed as a molecular target for the treatment of metabolic disorders resembling gastric surgery bypass. Inhibition of SLC6A15 appears to have a promising outcome in the treatment of psychiatric disorders. SLC6A19 and SLC6A20 have been suggested as potential targets in the treatment of COVID-19. In this review, we gathered recent advances on orphan transporters, their structure, functions, related disorders, and diseases, and in particular their relevance as therapeutic targets. SIGNIFICANCE STATEMENT: The following review systematizes current knowledge about the SLC6A15-SLCA20 neutral amino acid transporter subfamily and their therapeutic relevance in the treatment of different diseases.
Collapse
Affiliation(s)
- Jędrzej Kukułowicz
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Krzysztof Pietrzak-Lichwa
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Klaudia Klimończyk
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Nathalie Idlin
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Marek Bajda
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
3
|
Tsugiyama LE, Macedo Moraes RC, Cavalcante Moraes YA, Francis-Oliveira J. Promising new pharmacological targets for depression: The search for efficacy. Drug Discov Today 2023; 28:103804. [PMID: 37865307 DOI: 10.1016/j.drudis.2023.103804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Pharmacological treatment of major depressive disorder (MDD) still relies on the use of serotonergic drugs, despite their limited efficacy. A few mechanistically new drugs have been developed in recent years, but many fail in clinical trials. Several hypotheses have been proposed to explain MDD pathophysiology, indicating that physiological processes such as neuroplasticity, circadian rhythms, and metabolism are potential targets. Here, we review the current state of pharmacological treatments for MDD, as well as the preclinical and clinical evidence for an antidepressant effect of molecules that target non-serotonergic systems. We offer some insights into the challenges facing the development of new antidepressant drugs, and the prospect of finding more effectiveness for each target discussed.
Collapse
Affiliation(s)
- Lucila Emiko Tsugiyama
- Kansai Medical University, Graduate School of Medicine, iPS Cell Applied Medicine, Hirakata, Osaka, Japan
| | - Ruan Carlos Macedo Moraes
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil
| | | | - Jose Francis-Oliveira
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil.
| |
Collapse
|
4
|
Abstract
Rett syndrome is a neurodevelopmental disorder caused by loss-of-function mutations in the methyl-CpG binding protein-2 (MeCP2) gene that is characterized by epilepsy, intellectual disability, autistic features, speech deficits, and sleep and breathing abnormalities. Neurologically, patients with all three disorders display microcephaly, aberrant dendritic morphology, reduced spine density, and an imbalance of excitatory/inhibitory signaling. Loss-of-function mutations in the cyclin-dependent kinase-like 5 (CDKL5) and FOXG1 genes also cause similar behavioral and neurobiological defects and were referred to as congenital or variant Rett syndrome. The relatively recent realization that CDKL5 deficiency disorder (CDD), FOXG1 syndrome, and Rett syndrome are distinct neurodevelopmental disorders with some distinctive features have resulted in separate focus being placed on each disorder with the assumption that distinct molecular mechanisms underlie their pathogenesis. However, given that many of the core symptoms and neurological features are shared, it is likely that the disorders share some critical molecular underpinnings. This review discusses the possibility that deregulation of common molecules in neurons and astrocytes plays a central role in key behavioral and neurological abnormalities in all three disorders. These include KCC2, a chloride transporter, vGlut1, a vesicular glutamate transporter, GluD1, an orphan-glutamate receptor subunit, and PSD-95, a postsynaptic scaffolding protein. We propose that reduced expression or activity of KCC2, vGlut1, PSD-95, and AKT, along with increased expression of GluD1, is involved in the excitatory/inhibitory that represents a key aspect in all three disorders. In addition, astrocyte-derived brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), and inflammatory cytokines likely affect the expression and functioning of these molecules resulting in disease-associated abnormalities.
Collapse
Affiliation(s)
- Santosh R D’Mello
- Department of Biological Sciences, Louisiana State University Shreveport, Shreveport, LA 71104, USA
| |
Collapse
|
5
|
Moulard J, Rouach N. [Astrocytes, suppliers of neuronal glutamine for physiological synaptic activity and memory]. Med Sci (Paris) 2023; 39:830-832. [PMID: 38018925 DOI: 10.1051/medsci/2023157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Affiliation(s)
- Julien Moulard
- Interactions neurogliales dans la physiologie et les pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université Paris sciences & lettres, Paris, France
| | - Nathalie Rouach
- Interactions neurogliales dans la physiologie et les pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université Paris sciences & lettres, Paris, France
| |
Collapse
|
6
|
Belloch FDB, Cortés-Erice M, Herzog E, Zhang XM, Díaz-Perdigon T, Puerta E, Tordera RM. Fast antidepressant action of ketamine in mouse models requires normal VGLUT1 levels from prefrontal cortex neurons. Prog Neuropsychopharmacol Biol Psychiatry 2023; 121:110640. [PMID: 36209771 DOI: 10.1016/j.pnpbp.2022.110640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 09/03/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022]
Abstract
The NMDA antagonist ketamine demonstrated a fast antidepressant activity in treatment-resistant depression. Pre-clinical studies suggest that de novo synthesis of the brain-derived neurotrophic factor (BDNF) in the PFC might be involved in the rapid antidepressant action of ketamine. Applying a genetic model of impaired glutamate release, this study aims to further identify the molecular mechanisms that could modulate antidepressant action and resistance to treatment. To that end, mice knocked-down for the vesicular glutamate transporter 1 (VGLUT1+/-) were used. We analyzed anhedonia and helpless behavior as well as the expression of the proteins linked to glutamate transmission in the PFC of mice treated with ketamine or the reference antidepressant reboxetine. Moreover, we analyzed the acute effects of ketamine in VGLUT1+/- mice pretreated with chronic reboxetine or those that received a PFC rescue expression of VGLUT1. Chronic reboxetine rescued the depressive-like phenotype of the VGLUT1+/- mice. In addition, it enhanced the expression of the proteins linked to the AMPA signaling pathway as well as the immature form of BDNF (pro-BDNF). Unlike WT mice, ketamine had no effect on anhedonia or pro-BDNF expression in VGLUT1+/- mice; it also failed to decrease phosphorylated eukaryote elongation factor 2 (p-eEF2). Nevertheless, we found that reboxetine administered as pretreatment or PFC overexpression of VGLUT1 did rescue the antidepressant-like activity of acute ketamine in the mice. Our results strongly suggest that not only do PFC VGLUT1 levels modulate the rapid-antidepressant action of ketamine, but also highlight a possible mechanism for antidepressant resistance in some patients.
Collapse
Affiliation(s)
| | - María Cortés-Erice
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Etienne Herzog
- Université de Bordeaux, Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Xiao Min Zhang
- Université de Bordeaux, Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000 Bordeaux, France
| | - Teresa Díaz-Perdigon
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Elena Puerta
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain
| | - Rosa M Tordera
- Department of Pharmacology and Toxicology, University of Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
7
|
Zhang CC, Zhu LX, Shi HJ, Zhu LJ. The Role of Vesicle Release and Synaptic Transmission in Depression. Neuroscience 2022; 505:171-185. [DOI: 10.1016/j.neuroscience.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/19/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
|
8
|
Bauminger H, Gaisler-Salomon I. Beyond NMDA Receptors: Homeostasis at the Glutamate Tripartite Synapse and Its Contributions to Cognitive Dysfunction in Schizophrenia. Int J Mol Sci 2022; 23:8617. [PMID: 35955750 PMCID: PMC9368772 DOI: 10.3390/ijms23158617] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Cognitive deficits are core symptoms of schizophrenia but remain poorly addressed by dopamine-based antipsychotic medications. Glutamate abnormalities are implicated in schizophrenia-related cognitive deficits. While the role of the NMDA receptor has been extensively studied, less attention was given to other components that control glutamate homeostasis. Glutamate dynamics at the tripartite synapse include presynaptic and postsynaptic components and are tightly regulated by neuron-astrocyte crosstalk. Here, we delineate the role of glutamate homeostasis at the tripartite synapse in schizophrenia-related cognitive dysfunction. We focus on cognitive domains that can be readily measured in humans and rodents, i.e., working memory, recognition memory, cognitive flexibility, and response inhibition. We describe tasks used to measure cognitive function in these domains in humans and rodents, and the relevance of glutamate alterations in these domains. Next, we delve into glutamate tripartite synaptic components and summarize findings that implicate the relevance of these components to specific cognitive domains. These collective findings indicate that neuron-astrocyte crosstalk at the tripartite synapse is essential for cognition, and that pre- and postsynaptic components play a critical role in maintaining glutamate homeostasis and cognitive well-being. The contribution of these components to cognitive function should be considered in order to better understand the role played by glutamate signaling in cognition and develop efficient pharmacological treatment avenues for schizophrenia treatment-resistant symptoms.
Collapse
Affiliation(s)
- Hagar Bauminger
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Inna Gaisler-Salomon
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
9
|
Mizera J, Pomierny B, Sadakierska-Chudy A, Bystrowska B, Pomierny-Chamiolo L. Disruption of Glutamate Homeostasis in the Brain of Rat Offspring Induced by Prenatal and Early Postnatal Exposure to Maternal High-Sugar Diet. Nutrients 2022; 14:nu14112184. [PMID: 35683984 PMCID: PMC9182612 DOI: 10.3390/nu14112184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
A high-calorie diet has contributed greatly to the prevalence of overweight and obesity worldwide for decades. These conditions also affect pregnant women and have a negative impact on the health of both the woman and the fetus. Numerous studies indicate that an unbalanced maternal diet, rich in sugars and fats, can influence the in utero environment and, therefore, the future health of the child. It has also been shown that prenatal exposure to an unbalanced diet might permanently alter neurotransmission in offspring. In this study, using a rat model, we evaluated the effects of a maternal high-sugar diet on the level of extracellular glutamate and the expression of key transporters crucial for maintaining glutamate homeostasis in offspring. Glutamate concentration was assessed in extracellular fluid samples collected from the medial prefrontal cortex and hippocampus of male and female offspring. Analysis showed significantly increased glutamate levels in both brain structures analyzed, regardless of the sex of the offspring. These changes were accompanied by altered expression of the EAAT1, VGLUT1, and xc− proteins in these brain structures. This animal study further confirms our previous findings that a maternal high-sugar diet has a detrimental effect on the glutamatergic system.
Collapse
Affiliation(s)
- Jozef Mizera
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland;
| | - Bartosz Pomierny
- Department of Biochemical Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (B.P.); (B.B.)
| | - Anna Sadakierska-Chudy
- Department of Genetics, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705 Krakow, Poland;
| | - Beata Bystrowska
- Department of Biochemical Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland; (B.P.); (B.B.)
| | - Lucyna Pomierny-Chamiolo
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland;
- Correspondence: ; Tel.: +48-(12)-620-56-30
| |
Collapse
|
10
|
Physiological synaptic activity and recognition memory require astroglial glutamine. Nat Commun 2022; 13:753. [PMID: 35136061 PMCID: PMC8826940 DOI: 10.1038/s41467-022-28331-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
Presynaptic glutamate replenishment is fundamental to brain function. In high activity regimes, such as epileptic episodes, this process is thought to rely on the glutamate-glutamine cycle between neurons and astrocytes. However the presence of an astroglial glutamine supply, as well as its functional relevance in vivo in the healthy brain remain controversial, partly due to a lack of tools that can directly examine glutamine transfer. Here, we generated a fluorescent probe that tracks glutamine in live cells, which provides direct visual evidence of an activity-dependent glutamine supply from astroglial networks to presynaptic structures under physiological conditions. This mobilization is mediated by connexin43, an astroglial protein with both gap-junction and hemichannel functions, and is essential for synaptic transmission and object recognition memory. Our findings uncover an indispensable recruitment of astroglial glutamine in physiological synaptic activity and memory via an unconventional pathway, thus providing an astrocyte basis for cognitive processes. The authors present a fluorescent probe that tracks glutamine in live cells. They demonstrate the capabilities of the probe by providing direct visual evidence of an activity-dependent glutamine supply from astroglial networks to presynaptic structures under physiological conditions.
Collapse
|
11
|
Lindström SH, Sundberg SC, Larsson M, Andersson FK, Broman J, Granseth B. VGluT1 Deficiency Impairs Visual Attention and Reduces the Dynamic Range of Short-Term Plasticity at Corticothalamic Synapses. Cereb Cortex 2021; 30:1813-1829. [PMID: 31711131 PMCID: PMC7132919 DOI: 10.1093/cercor/bhz204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
The most common excitatory neurotransmitter in the central nervous system, glutamate, is loaded into synaptic vesicles by vesicular glutamate transporters (VGluTs). The primary isoforms, VGluT1 and 2, are expressed in complementary patterns throughout the brain and correlate with short-term synaptic plasticity. VGluT1 deficiency is observed in certain neurological disorders, and hemizygous (VGluT1+/−) mice display increased anxiety and depression, altered sensorimotor gating, and impairments in learning and memory. The synaptic mechanisms underlying these behavioral deficits are unknown. Here, we show that VGluT1+/− mice had decreased visual processing speeds during a sustained visual-spatial attention task. Furthermore, in vitro recordings of corticothalamic (CT) synapses revealed dramatic reductions in short-term facilitation, increased initial release probability, and earlier synaptic depression in VGluT1+/− mice. Our electron microscopy results show that VGluT1 concentration is reduced at CT synapses of hemizygous mice, but other features (such as vesicle number and active zone size) are unchanged. We conclude that VGluT1-haploinsuficiency decreases the dynamic range of gain modulation provided by CT feedback to the thalamus, and this deficiency contributes to the observed attentional processing deficit. We further hypothesize that VGluT1 concentration regulates release probability by applying a “brake” to an unidentified presynaptic protein that typically acts as a positive regulator of release.
Collapse
Affiliation(s)
- Sarah H Lindström
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Sofie C Sundberg
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Max Larsson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Fredrik K Andersson
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Jonas Broman
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| | - Björn Granseth
- Department of Clinical and Experimental Medicine, Division of Neurobiology, Linköping University, Linköping, 58185, Sweden
| |
Collapse
|
12
|
Khoramjouy M, Naderi N, Kobarfard F, Heidarli E, Faizi M. An Intensified Acrolein Exposure Can Affect Memory and Cognition in Rat. Neurotox Res 2021; 39:277-291. [PMID: 32876917 DOI: 10.1007/s12640-020-00278-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022]
Abstract
Acrolein is a clear, colorless liquid and a highly reactive α, β-unsaturated aldehyde. Acrolein, a byproduct and initiator of oxidative stress, has a major role in the pathogenesis of disorders including pulmonary, cardiovascular, atherosclerosis, and neurodegenerative diseases. Environmental or dietary exposure and endogenous production are common sources of acrolein. Widespread exposure to acrolein is a major risk for human health; therefore, we decided to investigate the neurological effects of acrolein. In this study, we used male Sprague-Dawley rats and exposed them orally to acrolein (0.5, 1, 3, and 5 mg/kg/day) for 90 days and investigated the neurobehavioral and electrophysiological disturbances. We also assessed the correlation between neurotoxicity and CSF concentration of acrolein in the rats. The results showed that chronic oral administration of acrolein at 5 mg/kg/day impaired learning and memory in the neurobehavioral tests. In addition, acrolein decreased the release of excitatory neurotransmitters such as glutamate in electrophysiological studies. Our data demonstrated that chronic oral exposure of acrolein at a dose of 5 mg/kg leads to a direct correlation between neurotoxicity and its CSF concentration. In conclusion, exposure to acrolein as a major pollutant in the environment may cause cognitive problems and may have serious neurocognitive effects on humans.
Collapse
Affiliation(s)
- Mona Khoramjouy
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elmira Heidarli
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran.
| |
Collapse
|
13
|
Primary role for melatonin MT 2 receptors in the regulation of anhedonia and circadian temperature rhythm. Eur Neuropsychopharmacol 2021; 44:51-65. [PMID: 33451856 DOI: 10.1016/j.euroneuro.2020.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/09/2020] [Accepted: 12/31/2020] [Indexed: 01/10/2023]
Abstract
Circadian rhythms disturbance is widely observable in patients with major depression (MD) and is also associated with depression vulnerability. Of them, disturbed melatonin secretion rhythm is particularly relevant to MD and is strongly phase-locked to core body temperature (CBT) rhythm. Here we aim to study the specific role of each melatonin receptor (MT1 and MT2) subtype in melatonin regulation of circadian CBT and its possible relationship with depressive-like behaviors. MT1-/- , MT2-/- and WT (C57BL/6) mice were used. Anhedonia, using the sucrose intake test, circadian CBT, environmental place preference (EPP) conditioning and vulnerability to chronic social defeat stress (CSDS) procedure were studied. Moreover, the antidepressant effects of reboxetine (15 mg/kg/day, i.p.) for three weeks or ketamine (15 mg/kg i.p. every four days, 4 doses in total) were studied. Further, exposure to ultra-mild stress induced by individual housing for several weeks was also studied in these mice. MT2-/- mice showed anhedonia and lower CBT compared to WT and MT1-/-. In addition, while reward exposure raised nocturnal CBT in WT this increase did not take place in MT2-/- mice. Further, MT2-/- mice showed an enhanced vulnerability to stress-induced anhedonia and social avoidance as well as an impaired acquisition of novelty seeking behavior. Both reboxetine and ketamine reverted anhedonia and induced a clear anti-helpless behavior in the tail suspension test (TST). Reboxetine raised CBT in mice and reverted ultra-mild stress-induced anhedonia. Our findings show a primary role for MT2 receptors in the regulation of circadian CBT as well as anhedonia and suggest that these receptors could be involved in depressive disorders associated to disturbed melatonin function.
Collapse
|
14
|
Dysregulation of miR-185, miR-193a, and miR-450a in the skin are linked to the depressive phenotype. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110052. [PMID: 32738353 DOI: 10.1016/j.pnpbp.2020.110052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/23/2020] [Accepted: 07/26/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Dysregulated microRNAs (miRNAs) in dermal fibroblasts of depressive subjects, indicate biomarker potential and can possibly aid clinical diagnostics. To overcome methodological challenges related to human experiments and fibroblast cultures, we here validate 38 miRNAs previously observed to be dysregulated in human fibroblasts from depressed subjects, in the skin of four distinct rat models of depression. METHODS In the presented study male rats from the adrenocorticotropic hormone (ACTH) model (n = 10/group), the chronic mild stress model (n = 10/group), Wistar Kyoto/Wistar Hannover rats (n = 10/group), and Flinders Resistant/Flinders Sensitive Line rats (n = 8/group) were included. Real-time qPCR was utilized to investigate miRNA alterations in flash-frozen skin-biopsies from the ear and fibroblast cultures. RESULTS In the ACTH rat model of depression, we identified nine dysregulated miRNAs in the skin and three in the fibroblasts. As the skin presented three times the amount of dysregulated miRNAs compared to the fibroblasts, skin instead of fibroblasts were continuously used for studies with the other rat models. In the skin from the four rat models of depression, 15 out of 38 miRNAs re-exhibited significant dysregulation in at least one of the rat models of depression and 67% were regulated in the same direction as in the human study. miR-450a and miR-193a presented dysregulation across rat models and miR-193a and miR-185 exhibited very strong dysregulation (30-fold and 50-fold, respectively). Lastly, an Ingenuity Pathway Analysis indicated functional overlap between dysregulated miRNAs, and common regulated pathways. CONCLUSION Flash-frozen skin is a valid alternative to fibroblast cultures as the skin appear to retain more of the miRNA dysregulation present in vivo. A sub-population of 15 miRNAs appear to be specific for the depressive phenotype, as they are dysregulated in both human depressed patients and distinct rat models of depression. We propose miR-450a, miR-185, and miR-193a as biomarker candidates of particular interest.
Collapse
|
15
|
De Giacomo V, Ruehle S, Lutz B, Häring M, Remmers F. Cell type-specific genetic reconstitution of CB1 receptor subsets to assess their role in exploratory behaviour, sociability, and memory. Eur J Neurosci 2020; 55:939-951. [PMID: 33253450 DOI: 10.1111/ejn.15069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/11/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
Several studies support the notion that exploratory behaviour depends on the functionality of the cannabinoid type 1 (CB1) receptor in a cell type-specific manner. Mice lacking the CB1 receptor in forebrain GABAergic or dorsal telencephalic glutamatergic neurons have served as essential tools revealing the necessary CB1 receptor functions in these two neuronal populations. However, whether these specific CB1 receptor populations are also sufficient within the endocannabinoid system for wild-type-like exploratory behaviour has remained unknown. To evaluate cell-type-specific sufficiency of CB1 receptor signalling exclusively in dorsal telencephalic glutamatergic neurons (Glu-CB1-RS) or in forebrain GABAergic neurons (GABA-CB1-RS), we utilised a mouse model in which CB1 receptor expression can be reactivated conditionally at endogenous levels from a complete CB1-KO background. The two types of conditional CB1-rescue mice were compared with CB1 receptor-deficient [no reactivation (Stop-CB1)] and wild-type [ubiquitous reactivation of endogenous CB1 receptor (CB1-RS)] controls to investigate the behavioural consequences. We evaluated social and object exploratory behaviour in four different paradigms. Remarkably, the reduced exploration observed in Stop-CB1 animals was rescued in Glu-CB1-RS mice and sometimes even surpassed CB1-RS (wild-type) exploration. In contrast, GABA-CB1-RS animals showed the lowest exploratory drive in all paradigms, with an even stronger phenotype than Stop-CB1 mice. Interestingly, these effects weakened with increasing familiarity with the environment, suggesting a causal role for altered neophobia in the observed phenotypes. Taken together, using our genetic approach, we were able to substantiate the opposing role of the CB1 receptor in dorsal telencephalic glutamatergic versus forebrain GABAergic neurons regarding exploratory behaviour.
Collapse
Affiliation(s)
- Vanessa De Giacomo
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sabine Ruehle
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Martin Häring
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.,Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Floortje Remmers
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
16
|
Frizzo ME, Ohno Y. Perisynaptic astrocytes as a potential target for novel antidepressant drugs. J Pharmacol Sci 2020; 145:60-68. [PMID: 33357781 DOI: 10.1016/j.jphs.2020.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that dysfunctions in glutamatergic signaling are associated with the pathophysiology of depression. Several molecules that act on glutamate binding sites, so-called glutamatergic modulators, are rapid-acting antidepressants that stimulate synaptogenesis. Their antidepressant response involves the elevation of both extracellular glutamate and brain-derived neurotrophic factor (BDNF) levels, as well as the postsynaptic activation of the mammalian target of rapamycin complex 1. The mechanisms involved in the antidepressant outcomes of glutamatergic modulators, including ketamine, suggest that astrocytes must be considered a cellular target for developing rapid-acting antidepressants. It is well known that extracellular glutamate levels and glutamate intrasynaptic time-coursing are maintained by perisynaptic astrocytes, where inwardly rectifying potassium channels 4.1 (Kir4.1 channels) regulate both potassium and glutamate uptake. In addition, ketamine reduces membrane expression of Kir4.1 channels, which raises extracellular potassium and glutamate levels, increasing postsynaptic neural activities. Furthermore, inhibition of Kir4.1 channels stimulates BDNF expression in astrocytes, which may enhance synaptic connectivity. In this review, we discuss glutamatergic modulators' actions in regulating extracellular glutamate and BDNF levels, and reinforce the importance of perisynaptic astrocytes for the development of novel antidepressant drugs.
Collapse
Affiliation(s)
- Marcos E Frizzo
- Department of Morphological Sciences, Federal University of Rio Grande Do Sul, Sarmento Leite Street, 500, CEP 90050-170, Porto Alegre, Brazil.
| | - Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, Japan
| |
Collapse
|
17
|
Shen M, Lv D, Li S, Zhang Y, Wang Z, Zhao C, Chen X, Wang C. Positive Allosteric Modulation of AMPAR by PF-4778574 Produced Rapid Onset Antidepressant Actions in Mice. Cereb Cortex 2020; 29:4438-4451. [PMID: 30566581 DOI: 10.1093/cercor/bhy324] [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: 08/27/2018] [Revised: 11/20/2018] [Accepted: 11/26/2018] [Indexed: 01/30/2023] Open
Abstract
It has been reported that fast-acting antidepressants enhance glutamatergic neurotransmission in the prefrontal cortex (PFC) regions via alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) activation. However, the precise mechanisms underlying the fast-acting antidepressants lead to an activation of AMPAR pathways remain largely unclear. To address this issue, a novel AMPAR positive allosteric agonist, PF-4778574, was used to test the rapid effects and the role of VGF (nonacronymic)/brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB)/AKT signaling in these actions in mice. We found that PF-4778574 rapidly alleviated chronic unpredictable stress-induced depression-like behaviors in a concentration-dependent manner. In addition, knock down of vesicular glutamate transporter 1 (VGLUT1) in the PFC of mice induced depression-like behaviors, whereas treatment with PF-4778574 was sufficient to alleviate it, indicating a presynaptic VGLUT1 independent effect. Furthermore, we demonstrate that pharmacological inhibitors of AMPAR or of L-type voltage-dependent Ca2+ channel (L-VDCC) blocked the antidepressants' effect on behaviors and the upregulation on the AMPAR-mediated VGF/BDNF/TrkB/AKT signaling of PF-4778574. Together, our findings indicate that postsynaptic AMPAR activation followed by activation of L-VDCC and subsequent VGF/BDNF/TrkB/AKT signaling are required for the rapid antidepressant effects of PF-4778574. Our data support a promising therapeutic profile for PF-4778574 as a new fast-acting antidepressant.
Collapse
Affiliation(s)
- Mengxin Shen
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Dan Lv
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Shuting Li
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Yanhua Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Zhen Wang
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chiyu Zhao
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Xuejie Chen
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China.,Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang, China
| |
Collapse
|
18
|
Pietrancosta N, Djibo M, Daumas S, El Mestikawy S, Erickson JD. Molecular, Structural, Functional, and Pharmacological Sites for Vesicular Glutamate Transporter Regulation. Mol Neurobiol 2020; 57:3118-3142. [PMID: 32474835 PMCID: PMC7261050 DOI: 10.1007/s12035-020-01912-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) control quantal size of glutamatergic transmission and have been the center of numerous studies over the past two decades. VGLUTs contain two independent transport modes that facilitate glutamate packaging into synaptic vesicles and phosphate (Pi) ion transport into the synaptic terminal. While a transmembrane proton electrical gradient established by a vacuolar-type ATPase powers vesicular glutamate transport, recent studies indicate that binding sites and flux properties for chloride, potassium, and protons within VGLUTs themselves regulate VGLUT activity as well. These intrinsic ionic binding and flux properties of VGLUTs can therefore be modulated by neurophysiological conditions to affect levels of glutamate available for release from synapses. Despite their extraordinary importance, specific and high-affinity pharmacological compounds that interact with these sites and regulate VGLUT function, distinguish between the various modes of transport, and the different isoforms themselves, are lacking. In this review, we provide an overview of the physiologic sites for VGLUT regulation that could modulate glutamate release in an over-active synapse or in a disease state.
Collapse
Affiliation(s)
- Nicolas Pietrancosta
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Laboratoire des Biomolécules, Sorbonne Université, CNRS, ENS, LBM, 75005, Paris, France.
| | - Mahamadou Djibo
- Sorbonne Paris Cité, Université Paris Descartes, LCBPT, UMR 8601, 75006, Paris, France
| | - Stephanie Daumas
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Salah El Mestikawy
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 boulevard Lasalle, Verdun, Montreal, QC, Canada.
| | - Jeffrey D Erickson
- Neuroscience Center, Louisiana State University, New Orleans, LA, 70112, USA. .,Department of Pharmacology, Louisiana State University, New Orleans, LA, 70112, USA.
| |
Collapse
|
19
|
Turovskaya MV, Gaidin SG, Vedunova MV, Babaev AA, Turovsky EA. BDNF Overexpression Enhances the Preconditioning Effect of Brief Episodes of Hypoxia, Promoting Survival of GABAergic Neurons. Neurosci Bull 2020; 36:733-760. [PMID: 32219700 PMCID: PMC7340710 DOI: 10.1007/s12264-020-00480-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxia causes depression of synaptic plasticity, hyperexcitation of neuronal networks, and the death of specific populations of neurons. However, brief episodes of hypoxia can promote the adaptation of cells. Hypoxic preconditioning is well manifested in glutamatergic neurons, while this adaptive mechanism is virtually suppressed in GABAergic neurons. Here, we show that brain-derived neurotrophic factor (BDNF) overexpression in neurons enhances the preconditioning effect of brief episodes of hypoxia. The amplitudes of the NMDAR- and AMPAR-mediated Ca2+ responses of glutamatergic and GABAergic neurons gradually decreased after repetitive brief hypoxia/reoxygenation cycles in cell cultures transduced with the (AAV)-Syn-BDNF-EGFP virus construct. In contrast, the amplitudes of the responses of GABAergic neurons increased in non-transduced cultures after preconditioning. The decrease of the amplitudes in GABAergic neurons indicated the activation of mechanisms of hypoxic preconditioning. Preconditioning suppressed apoptotic or necrotic cell death. This effect was most pronounced in cultures with BDNF overexpression. Knockdown of BDNF abolished the effect of preconditioning and promoted the death of GABAergic neurons. Moreover, the expression of the anti-apoptotic genes Stat3, Socs3, and Bcl-xl substantially increased 24 h after hypoxic episodes in the transduced cultures compared to controls. The expression of genes encoding the pro-inflammatory cytokines IL-10 and IL-6 also increased. In turn, the expression of pro-apoptotic (Bax, Casp-3, and Fas) and pro-inflammatory (IL-1β and TNFα) genes decreased after hypoxic episodes in cultures with BDNF overexpression. Inhibition of vesicular BDNF release abolished its protective action targeting inhibition of the oxygen-glucose deprivation (OGD)-induced [Ca2+]i increase in GABAergic and glutamatergic neurons, thus promoting their death. Bafilomycin A1, Brefeldin A, and tetanus toxin suppressed vesicular release (including BDNF) and shifted the gene expression profile towards excitotoxicity, inflammation, and apoptosis. These inhibitors of vesicular release abolished the protective effects of hypoxic preconditioning in glutamatergic neurons 24 h after hypoxia/reoxygenation cycles. This finding indicates a significant contribution of vesicular BDNF release to the development of the mechanisms of hypoxic preconditioning. Thus, our results demonstrate that BDNF plays a pivotal role in the activation and enhancement of the preconditioning effect of brief episodes of hypoxia and promotes tolerance of the most vulnerable populations of GABAergic neurons to hypoxia/ischemia.
Collapse
Affiliation(s)
- M V Turovskaya
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - S G Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - M V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - A A Babaev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - E A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| |
Collapse
|
20
|
Du X, Li J, Li M, Yang X, Qi Z, Xu B, Liu W, Xu Z, Deng Y. Research progress on the role of type I vesicular glutamate transporter (VGLUT1) in nervous system diseases. Cell Biosci 2020; 10:26. [PMID: 32158532 PMCID: PMC7057577 DOI: 10.1186/s13578-020-00393-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Glutamate (Glu) is the predominant excitatory neurotransmitter in the central nervous system (CNS). Glutamatergic transmission is critical for controlling neuronal activity. In presynaptic neurons, Glu is stored in synaptic vesicles and released by stimulation. The homeostasis of glutamatergic system is maintained by a set of transporters in the membrane of synaptic vesicles. The family of vesicular Glu transporters in mammals is comprised of three highly homologous proteins: VGLUT1-3. Among them, VGLUT1 accounts for the largest proportion. However, most of the Glu is transported into the synaptic vesicles via the type 1 vesicle Glu transporter (VGLUT1). So, the expression of particular VGLUT1 is largely complementary with limited overlap and so far it is most specific markers for neurons that use Glu as neurotransmitter. Controlling the activity of VGLUT1 could potentially modulate the efficiency of excitatory neuro-transmission and change the filling level of synaptic vesicles. This review summarizes the recent knowledge concerning molecular and functional characteristic of VGLUT1, their development, contribution to a series of central nervous system and peripheral nervous system diseases such as learning and memory disorders, Alzheimer's disease, Parkinson's disease and sensitized nociception or pain pathology et al.
Collapse
Affiliation(s)
- Xianchao Du
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Jiashuo Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Minghui Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Xinxin Yang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Zhipeng Qi
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Bin Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Wei Liu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Zhaofa Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| | - Yu Deng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122 Liaoning People’s Republic of China
| |
Collapse
|
21
|
The solute carrier transporters and the brain: Physiological and pharmacological implications. Asian J Pharm Sci 2019; 15:131-144. [PMID: 32373195 PMCID: PMC7193445 DOI: 10.1016/j.ajps.2019.09.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/17/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023] Open
Abstract
Solute carriers (SLCs) are the largest family of transmembrane transporters that determine the exchange of various substances, including nutrients, ions, metabolites, and drugs across biological membranes. To date, the presence of about 287 SLC genes have been identified in the brain, among which mutations or the resultant dysfunctions of 71 SLC genes have been reported to be correlated with human brain disorders. Although increasing interest in SLCs have focused on drug development, SLCs are currently still under-explored as drug targets, especially in the brain. We summarize the main substrates and functions of SLCs that are expressed in the brain, with an emphasis on selected SLCs that are important physiologically, pathologically, and pharmacologically in the blood-brain barrier, astrocytes, and neurons. Evidence suggests that a fraction of SLCs are regulated along with the occurrences of brain disorders, among which epilepsy, neurodegenerative diseases, and autism are representative. Given the review of SLCs involved in the onset and procession of brain disorders, we hope these SLCs will be screened as promising drug targets to improve drug delivery to the brain.
Collapse
|
22
|
Consequences of VGluT3 deficiency on learning and memory in mice. Physiol Behav 2019; 212:112688. [PMID: 31622610 DOI: 10.1016/j.physbeh.2019.112688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/22/2019] [Accepted: 09/22/2019] [Indexed: 01/06/2023]
Abstract
The aim of the present study was to test the hypothesis that vesicular glutamate transporter 3 (VGluT3) deficiency is associated with cognitive impairments. Male VGluT3 knockout (KO) and wild type (WT) mice were exposed to a behavioral test battery covering paradigms based on spontaneous exploratory behavior and reinforcement-based learning tests. Reversal learning was examined to test the cognitive flexibility. The VGluT3 KO mice clearly exhibited the ability to learn. The social recognition memory of KO mice was intact. The y-maze test revealed weaker working memory of VGluT3 KO mice. No significant learning impairments were noticed in operant conditioning or holeboard discrimination paradigm. In avoidance-based learning tests (Morris water maze and active avoidance), KO mice exhibited slightly slower learning process compared to WT mice, but not a complete learning impairment. In tests based on simple associations (operant conditioning, avoidance learning) an attenuation of cognitive flexibility was observed in KO mice. In conclusion, knocking out VGluT3 results in mild disturbances in working memory and learning flexibility. Apparently, this glutamate transporter is not a major player in learning and memory formation in general. Based on previous characteristics of VGluT3 KO mice we would have expected a stronger deficit. The observed hypolocomotion did not contribute to the mild cognitive disturbances herein reported, either.
Collapse
|
23
|
Huang S, Dong W, Jiao Z, Liu J, Li K, Wang H, Xu D. Prenatal Dexamethasone Exposure Induced Alterations in Neurobehavior and Hippocampal Glutamatergic System Balance in Female Rat Offspring. Toxicol Sci 2019; 171:369-384. [PMID: 31518422 DOI: 10.1093/toxsci/kfz163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epidemiological investigations have suggested that periodic use of dexamethasone during pregnancy is a risk factor for abnormal behavior in offspring, but the potential mechanism remains unclear. In this study, we investigated the changes in the glutamatergic system and neurobehavior in female offspring with prenatal dexamethasone exposure (PDE) to explore intrauterine programing mechanisms. Compared with the control group, rat offspring with PDE exhibited spatial memory deficits and anxiety-like behavior. The expression of hippocampal glucocorticoid receptors (GR) and histone deacetylase 2 (HDAC2) increased, whereas histone H3 lysine 14 acetylation (H3K14ac) of brain-derived neurotrophic factor (BDNF) exon IV (BDNF IV) and expression of BDNF decreased. The glutamatergic system also changed. We further observed that changes in the fetal hippocampus were consistent with those in adult offspring. In vitro, the administration of 0.5 μM dexamethasone to the H19-7 fetal hippocampal neuron cells directly led to a cascade of changes in the GR/HDAC2/BDNF pathway, whereas the GR antagonist RU486 and the HDAC2 inhibitor romidepsin (Rom) reversed changes caused by dexamethasone to the H3K14ac level of BDNF IV and to the expression of BDNF. The increase in HDAC2 can be reversed by RU486, and the changes in the glutamatergic system can be partially reversed after supplementation with BDNF. It is suggested that PDE increases the expression of HDAC2 by activating GR, reducing the H3K14ac level of BDNF IV, inducing alterations in neurobehavior and hippocampal glutamatergic system balance. The findings suggest that BDNF supplementation and glutamatergic system improvement are potential therapeutic targets for the fetal origins of abnormal neurobehavior.
Collapse
Affiliation(s)
- Songqiang Huang
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Wanting Dong
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Zhexiao Jiao
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Jie Liu
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Ke Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, Hubei Province, China
| | - Hui Wang
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| | - Dan Xu
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| |
Collapse
|
24
|
Liu X, Zhou KX, Yin NN, Zhang CK, Shi MH, Zhang HY, Wang DM, Xu ZJ, Zhang JD, Li JL, Wang MQ. Malocclusion Generates Anxiety-Like Behavior Through a Putative Lateral Habenula-Mesencephalic Trigeminal Nucleus Pathway. Front Mol Neurosci 2019; 12:174. [PMID: 31427925 PMCID: PMC6689965 DOI: 10.3389/fnmol.2019.00174] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/01/2019] [Indexed: 01/06/2023] Open
Abstract
Malocclusion is an important risk factor for temporomandibular disorder (TMD), a series of disorders characterized by dysfunction in the orofacial region involving the temporomandibular joint (TMJ) and jaw muscles. We recently showed that experimental unilateral anterior crossbite (UAC) produced masseter hyperactivity through a circuit involving the periodontal proprioception, trigeminal mesencephalic nucleus (Vme), and trigeminal motor nucleus (Vmo). Anxiety is a common complication in patients with TMD. The lateral habenula (LHb) is involved in emotional modulation and has direct projections to the Vme. Therefore, the present research examined whether UAC facilitates excitatory input from the LHb to the Vme and, subsequently, anxiety-like behaviors in rats. The LHb activation was evaluated by the electrophysiological recording, assessment of vesicular glutamate transporter-2 (VGLUT2) mRNA expression, and measurement of anxiety-like behaviors. The effects of LHb activity on Vme were evaluated by electrophysiological recording from Vme neurons and local changes in VGLUT2 protein density. UAC produced anxiety in modeled rats and increased neuronal activity in the LHb. VGLUT2 mRNA expression was also increased in the LHb. Further, VGLUT2-positive boutons were observed in close apposite upon parvalbumin (PV)-labeled Vme neurons. VGLUT2 protein expression was also increased in the Vme. Significantly, injection of VGLUT2-targeted shRNA into the LHb reduced the expression of VGLUT2 protein in the Vme, attenuated UAC-associated anxiety-like behaviors, and attenuated electrophysiological changes in the Vme neurons. In conclusion, we show that UAC activates the LHb neurons as well as the periodontal proprioceptive pathway to provide excitatory input to the Vme and produce anxiety in rats. These findings provide a rationale for suppressing activity of the LHb to attenuate both the physical and psychological effects of TMD.
Collapse
Affiliation(s)
- Xin Liu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an, China.,Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Kai-Xiang Zhou
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Center, The Fourth Military Medical University, Xi'an, China
| | - Nan-Nan Yin
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, China
| | - Chun-Kui Zhang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Center, The Fourth Military Medical University, Xi'an, China
| | - Ming-Hong Shi
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Hong-Yun Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Dong-Mei Wang
- Department of Oral Medicine, School of Stomatology, Xinxiang Medical University, Xinxiang, China
| | - Zi-Jun Xu
- School of Clinical Medicine, University of South China, Hengyang, China
| | - Jing-Dong Zhang
- Department of Anesthesiology, University of Cincinnati Medical College of Medicine, Cincinnati, OH, United States
| | - Jin-Lian Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Center, The Fourth Military Medical University, Xi'an, China
| | - Mei-Qing Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology, National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
25
|
Glutamate Within the Marmoset Anterior Hippocampus Interacts with Area 25 to Regulate the Behavioral and Cardiovascular Correlates of High-Trait Anxiety. J Neurosci 2019; 39:3094-3107. [PMID: 30718320 PMCID: PMC6468106 DOI: 10.1523/jneurosci.2451-18.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 11/21/2022] Open
Abstract
High-trait anxiety is a risk factor for the development of affective disorders and has been associated with decreased cardiovascular and behavioral responsivity to acute stressors in humans that may increase the risk of developing cardiovascular disease. Although human neuroimaging studies of high-trait anxiety reveals dysregulation in primate cingulate areas 25 and 32 and the anterior hippocampus (aHipp) and rodent studies reveal the importance of aHipp glutamatergic hypofunction, the causal involvement of aHipp glutamate and its interaction with these areas in the primate brain is unknown. Accordingly, we correlated marmoset trait anxiety scores to their postmortem aHipp glutamate levels and showed that low glutamate in the right aHipp is associated with high-trait anxiety in marmosets. Moreover, pharmacologically increasing aHipp glutamate reduced anxiety levels in highly anxious marmosets in two uncertainty-based tests of anxiety: exposure to a human intruder with uncertain intent and unpredictable loud noise. In the human intruder test, increasing aHipp glutamate decreased anxiety by increasing approach to the intruder. In the unpredictable threat test, animals showed blunted behavioral and cardiovascular responsivity after control infusions, which was normalized by increasing aHipp glutamate. However, this aHipp-mediated anxiolytic effect was blocked by simultaneous pharmacological inactivation of area 25, but not area 32, areas which when inactivated independently reduced and had no effect on anxiety, respectively. These findings provide causal evidence in male and female primates that aHipp glutamatergic hypofunction and its regulation by area 25 contribute to the behavioral and cardiovascular symptoms of endogenous high-trait anxiety.SIGNIFICANCE STATEMENT High-trait anxiety predisposes sufferers to the development of anxiety and depression. Although neuroimaging of these disorders and rodent modeling implicate dysregulation in hippocampal glutamate and the subgenual/perigenual cingulate cortices (areas 25/32), the causal involvement of these structures in endogenous high-trait anxiety and their interaction are unknown. Here, we demonstrate that increased trait anxiety in marmoset monkeys correlates with reduced hippocampal glutamate and that increasing hippocampal glutamate release in high-trait-anxious monkeys normalizes the aberrant behavioral and cardiovascular responsivity to potential threats. This normalization was blocked by simultaneous inactivation of area 25, but not area 32. These findings provide casual evidence in primates that hippocampal glutamatergic hypofunction regulates endogenous high-trait anxiety and the hippocampal-area 25 circuit is a potential therapeutic target.
Collapse
|
26
|
Dutar P, Tolle V, Kervern M, Carcenac C, Carola V, Gross C, Savasta M, Darmon M, Masson J. GLS1 Mutant Mice Display Moderate Alterations of Hippocampal Glutamatergic Neurotransmission Associated with Specific Adaptive Behavioral Changes. Neuroscience 2019; 396:175-186. [PMID: 30472430 DOI: 10.1016/j.neuroscience.2018.11.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 11/29/2022]
Abstract
Significant alterations in glutamatergic neurotransmission have been reported in major depressive disorder (MDD) that could underlie psychiatric traits. Studies were mainly interested in synaptic dysfunction in the prefrontal cortex, a key structure involved in depressive-like behavior, however hippocampus has been shown to be important in MDD. As cognitive deficits such as hippocampus-memory process were observed in MDD, we investigated in a mild hypoglutamatergic model behaviors related to depression and memory, synaptic transmission parameters and glutamatergic state specifically in the hippocampus. We thus characterized these phenotypes in adult male mice partially depleted in glutaminase type 1 or GLS1 (GLS1 HET), the enzyme responsible for glutamate synthesis in neurons, that we previously characterized as displaying moderate lower levels of glutamate in brain. We showed that GLS1 mutant mice display AMPA-R-mediated response deficits after prolonged repetitive stimulation with electrophysiological recording and inability to sustain glutamate release by microdialysis experiments with no consequences on behavioral spatial learning performances. However, their ability to escape from unpleasant but repeated escapable condition was attenuated whereas they were more immobile in the unescapable situation in the FST during re-test. These results show that GLS1 mutant mice display moderate impairments of hippocampal glutamatergic neurotransmission and moderate changes in adaptive behaviors that have been shown to participate to the development of depressive-like state.
Collapse
Affiliation(s)
- Patrick Dutar
- Centre de Psychiatrie et Neuroscience, INSERM UMR894, Paris F-75014 France Université Paris Descartes, Sorbonne Paris Cité, Paris 5, France
| | - Virginie Tolle
- Centre de Psychiatrie et Neuroscience, INSERM UMR894, Paris F-75014 France Université Paris Descartes, Sorbonne Paris Cité, Paris 5, France
| | - Myriam Kervern
- Centre de Psychiatrie et Neuroscience, INSERM UMR894, Paris F-75014 France Université Paris Descartes, Sorbonne Paris Cité, Paris 5, France
| | - Carole Carcenac
- Université Grenoble Alpes, Grenoble Institut des neuroscience (GIN), 38000 Grenoble, France
| | - Valeria Carola
- Department of Experimental Neuroscience, Fondazione Santa Lucia, Rome, Italy; Epigenetics & Neurobiology Unit, European Molecular Biology Laboratory, EMBL-Rome, Italy
| | - Cornelius Gross
- Epigenetics & Neurobiology Unit, European Molecular Biology Laboratory, EMBL-Rome, Italy
| | - Marc Savasta
- Université Grenoble Alpes, Grenoble Institut des neuroscience (GIN), 38000 Grenoble, France
| | - Michèle Darmon
- Centre de Psychiatrie et Neuroscience, INSERM UMR894, Paris F-75014 France Université Paris Descartes, Sorbonne Paris Cité, Paris 5, France
| | - Justine Masson
- Centre de Psychiatrie et Neuroscience, INSERM UMR894, Paris F-75014 France Université Paris Descartes, Sorbonne Paris Cité, Paris 5, France.
| |
Collapse
|
27
|
Moriguchi S, Takamiya A, Noda Y, Horita N, Wada M, Tsugawa S, Plitman E, Sano Y, Tarumi R, ElSalhy M, Katayama N, Ogyu K, Miyazaki T, Kishimoto T, Graff-Guerrero A, Meyer JH, Blumberger DM, Daskalakis ZJ, Mimura M, Nakajima S. Glutamatergic neurometabolite levels in major depressive disorder: a systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Mol Psychiatry 2019; 24:952-964. [PMID: 30315224 PMCID: PMC6755980 DOI: 10.1038/s41380-018-0252-9] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/13/2018] [Accepted: 08/10/2018] [Indexed: 12/22/2022]
Abstract
Alterations in glutamatergic neurotransmission are implicated in the pathophysiology of depression, and the glutamatergic system represents a treatment target for depression. To summarize the nature of glutamatergic alterations in patients with depression, we conducted a meta-analysis of proton magnetic resonance (1H-MRS) spectroscopy studies examining levels of glutamate. We used the search terms: depress* AND (MRS OR "magnetic resonance spectroscopy"). The search was performed with MEDLINE, Embase, and PsycINFO. The inclusion criteria were 1H-MRS studies comparing levels of glutamate + glutamine (Glx), glutamate, or glutamine between patients with depression and healthy controls. Standardized mean differences (SMD) were calculated to assess group differences in the levels of glutamatergic neurometabolites. Forty-nine studies met the eligibility criteria, which included 1180 patients and 1066 healthy controls. There were significant decreases in Glx within the medial frontal cortex (SMD = -0.38; 95% CI, -0.69 to -0.07) in patients with depression compared with controls. Subanalyses revealed that there was a significant decrease in Glx in the medial frontal cortex in medicated patients with depression (SMD = -0.50; 95% CI, -0.80 to -0.20), but not in unmedicated patients (SMD = -0.27; 95% CI, -0.76 to 0.21) compared with controls. Overall, decreased levels of glutamatergic metabolites in the medial frontal cortex are linked with the pathophysiology of depression. These findings are in line with the hypothesis that depression may be associated with abnormal glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Sho Moriguchi
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan ,0000 0001 2157 2938grid.17063.33Research Imaging Centre, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Akihiro Takamiya
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| | - Nobuyuki Horita
- 0000 0001 1033 6139grid.268441.dDepartment of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masataka Wada
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sakiko Tsugawa
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Eric Plitman
- 0000 0001 2157 2938grid.17063.33Research Imaging Centre, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Yasunori Sano
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Ryosuke Tarumi
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Muhammad ElSalhy
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Nariko Katayama
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kamiyu Ogyu
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Miyazaki
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Taishiro Kishimoto
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Ariel Graff-Guerrero
- 0000 0001 2157 2938grid.17063.33Research Imaging Centre, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Jeffrey H. Meyer
- 0000 0001 2157 2938grid.17063.33Research Imaging Centre, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| | - Daniel M. Blumberger
- 0000 0001 2157 2938grid.17063.33Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Zafiris J. Daskalakis
- 0000 0001 2157 2938grid.17063.33Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Masaru Mimura
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- 0000 0004 1936 9959grid.26091.3cDepartment of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan ,0000 0001 2157 2938grid.17063.33Research Imaging Centre, Centre for Addiction and Mental Health, University of Toronto, Toronto, Canada
| |
Collapse
|
28
|
Nucleocytoplasmic export of HDAC5 and SIRT2 downregulation: two epigenetic mechanisms by which antidepressants enhance synaptic plasticity markers. Psychopharmacology (Berl) 2018; 235:2831-2846. [PMID: 30091005 DOI: 10.1007/s00213-018-4975-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 07/17/2018] [Indexed: 10/28/2022]
Abstract
RATIONALE Antidepressant action has been linked to increased synaptic plasticity in which epigenetic mechanisms such as histone posttranslational acetylation could be involved. Interestingly, the histone deacetylases HDAC5 and SIRT2 are oppositely regulated by stress and antidepressants in mice prefrontal cortex (PFC). Besides, the neuroblastoma SH-SY5Y line is an in vitro neuronal model reliable to study drug effects with clear advantages over animals. OBJECTIVES We aimed to characterize in vitro the role of HDAC5 and SIRT2 in antidepressant regulation of neuroplasticity. METHODS SH-SY5Y cultures were incubated with imipramine, fluoxetine, and reboxetine (10 μM, 2 and 24 h) as well as the selective HDAC5 (MC3822, 5 μM, 24 h) or SIRT2 (33i, 5 μM, 24 h) inhibitors. The regulation of the brain-derived neurotrophic factor (BDNF), the vesicular glutamate transporter 1 (VGLUT1), the acetylated histones 3 (AcH3) and 4 (AcH4), HDAC5, and SIRT2 was studied. Comparatively, the long-term effects of these antidepressants (21 days, i.p.) in the mice (C57BL6, 8 weeks) PFC were studied. RESULTS Antidepressants increased both in vitro and in vivo expression of BDNF, VGLUT1, AcH3, and AcH4. Moreover, imipramine and reboxetine increased the phosphorylated form of HDAC5 (P-HDAC5), mediating its cytoplasmic export. Further, SIRT2 was downregulated by all antidepressants. Finally, specific inhibition of HDAC5 and SIRT2 increased neuroplasticity markers. CONCLUSIONS This study supports the validity of the SH-SY5Y model for studying epigenetic changes linked to synaptic plasticity induced by antidepressants as well as the effect of selective HDAC inhibitors. Particularly, nucleocytoplasmic export of HDAC5 and SIRT2 downregulation mediated by antidepressants could enhance synaptic plasticity markers leading to antidepressant action.
Collapse
|
29
|
Zhang FX, Ge SN, Dong YL, Shi J, Feng YP, Li Y, Li YQ, Li JL. Vesicular glutamate transporter isoforms: The essential players in the somatosensory systems. Prog Neurobiol 2018; 171:72-89. [PMID: 30273635 DOI: 10.1016/j.pneurobio.2018.09.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/28/2018] [Accepted: 09/23/2018] [Indexed: 02/08/2023]
Abstract
In nervous system, glutamate transmission is crucial for centripetal conveyance and cortical perception of sensory signals of different modalities, which necessitates vesicular glutamate transporters 1-3 (VGLUT 1-3), the three homologous membrane-bound protein isoforms, to load glutamate into the presysnaptic vesicles. These VGLUTs, especially VGLUT1 and VGLUT2, selectively label and define functionally distinct neuronal subpopulations at each relay level of the neural hierarchies comprising spinal and trigeminal sensory systems. In this review, by scrutinizing each structure of the organism's fundamental hierarchies including dorsal root/trigeminal ganglia, spinal dorsal horn/trigeminal sensory nuclear complex, somatosensory thalamic nuclei and primary somatosensory cortex, we summarize and characterize in detail within each relay the neuronal clusters expressing distinct VGLUT protein/transcript isoforms, with respect to their regional distribution features (complementary distribution in some structures), axonal terminations/peripheral innervations and physiological functions. Equally important, the distribution pattern and characteristics of VGLUT1/VGLUT2 axon terminals within these structures are also epitomized. Finally, the correlation of a particular VGLUT isoform and its physiological role, disclosed thus far largely via studying the peripheral receptors, is generalized by referring to reports on global and conditioned VGLUT-knockout mice. Also, researches on VGLUTs relating to future direction are tentatively proposed, such as unveiling the elusive differences between distinct VGLUTs in mechanism and/or pharmacokinetics at ionic/molecular level, and developing VGLUT-based pain killers.
Collapse
Affiliation(s)
- Fu-Xing Zhang
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Shun-Nan Ge
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yu-Lin Dong
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Juan Shi
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yu-Peng Feng
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yang Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yun-Qing Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, PR China.
| | - Jin-Lian Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
30
|
Carter BM, Sullivan BJ, Landers JR, Kadam SD. Dose-dependent reversal of KCC2 hypofunction and phenobarbital-resistant neonatal seizures by ANA12. Sci Rep 2018; 8:11987. [PMID: 30097625 PMCID: PMC6086916 DOI: 10.1038/s41598-018-30486-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/31/2018] [Indexed: 01/22/2023] Open
Abstract
Neonatal seizures have an incidence of 3.5 per 1000 newborns; while hypoxic-ischemic encephalopathy (HIE) accounts for 50-60% of cases, half are resistant to 1st-line anti-seizure drugs such as phenobarbital (PB). Tyrosine receptor kinase B (TrkB) activation following ischemic injury is known to increase neuronal excitability by downregulation of K-Cl co-transporter 2 (KCC2); a neuronal chloride (Cl-) co-transporter. In this study, three graded doses of ANA12, a small-molecule selective TrkB antagonist, were tested in CD1 mice at P7 and P10 following induction of neonatal ischemia by a unilateral carotid ligation. The PB loading dose remained the same in all treatment groups at both ages. Evaluation criteria for the anti-seizure efficacy of ANA12 were: (1) quantitative electroencephalographic (EEG) seizure burden and power, (2) rescue of post-ischemic KCC2 and pKCC2-S940 downregulation and (3) reversal of TrkB pathway activation following ischemia. ANA12 significantly rescued PB resistant seizures in a dose-dependent manner at P7 and improved PB efficacy at P10. Additionally, female pups responded better to lower doses of ANA12 compared to males. ANA12 significantly reversed post-ischemic KCC2 downregulation and TrkB pathway activation at P7 when PB alone was inefficacious. Rescuing KCC2 hypofunction may be critical for preventing emergence of refractory seizures.
Collapse
Affiliation(s)
- B M Carter
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - B J Sullivan
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - J R Landers
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - S D Kadam
- Neuroscience Laboratory, Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
31
|
Matrov D, Kaart T, Lanfumey L, Maldonado R, Sharp T, Tordera RM, Kelly PA, Deakin B, Harro J. Cerebral oxidative metabolism mapping in four genetic mouse models of anxiety and mood disorders. Behav Brain Res 2018; 356:435-443. [PMID: 29885846 DOI: 10.1016/j.bbr.2018.05.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
Abstract
The psychopathology of depression is highly complex and the outcome of studies on animal models is divergent. In order to find brain regions that could be metabolically distinctively active across a variety of mouse depression models and to compare the interconnectivity of brain regions of wild-type and such genetically modified mice, histochemical mapping of oxidative metabolism was performed by the measurement of cytochrome oxidase activity. We included mice with the heterozygous knockout of the vesicular glutamate transporter (VGLUT1-/+), full knockout of the cannabinoid 1 receptor (CB1-/-), an anti-sense knockdown of the glucocorticoid receptor (GRi) and overexpression of the human 5-hydroxytryptamine transporter (h5-HTT). Altogether 76 mouse brains were studied to measure oxidative metabolism in one hundred brain regions, and the obtained dataset was submitted to a variety of machine learning algorithms and multidimensional scaling. Overall, the top brain regions having the largest contribution to classification into depression model were the lateroanterior hypothalamic nucleus, the anterior part of the basomedial amygdaloid nucleus, claustrum, the suprachiasmatic nucleus, the ventromedial hypothalamic nucleus, and the anterior hypothalamic area. In terms of the patterns of inter-regional relationship between wild-type and genetically modified mice there was little overall difference, while the most deviating brain regions were cortical amygdala and ventrolateral and ventral posteromedial thalamic nuclei. The GRi mice that most clearly differed from their controls exhibited deviation of connectivity for a number of brain regions, such as ventrolateral thalamic nucleus, the intermediate part of the lateral septal nucleus, the anteriodorsal part of the medial amygdaloid nucleus, the medial division of the central amygdaloid nucleus, ventral pallidum, nucleus of the vertical limb of the diagonal band, anteroventral parts of the thalamic nucleus and parts of the bed nucleus of the stria terminalis. Conclusively, the GRi mouse model was characterized by changes in the functional connectivity of the extended amygdala and stress response circuits.
Collapse
Affiliation(s)
- Denis Matrov
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia; Department of Neuroscience, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tanel Kaart
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Kreutzwaldi 1, 51014 Tartu, Estonia
| | - Laurence Lanfumey
- Centre de Psychiatrie et Neuroscience, INSERM U 894, 2 ter rue d'Alésia, 75014 Paris, France
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| | - Rosa M Tordera
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona, Spain
| | - Paul A Kelly
- Centre for Cognitive and Neural Systems, University of Edinburgh, Scotland, United Kingdom
| | - Bill Deakin
- Neuroscience and Psychiatry Unit, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, United Kingdom
| | - Jaanus Harro
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia.
| |
Collapse
|
32
|
Jung HY, Yoo DY, Park JH, Kim JW, Chung JY, Kim DW, Won MH, Yoon YS, Hwang IK. Age-dependent changes in vesicular glutamate transporter 1 and 2 expression in the gerbil hippocampus. Mol Med Rep 2018. [PMID: 29532891 PMCID: PMC5928628 DOI: 10.3892/mmr.2018.8705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Glutamate is a major excitatory neurotransmitter that is stored in vesicles located in the presynaptic terminal. Glutamate is transported into vesicles via the vesicular glutamate transporter (VGLUT). In the present study, the age‑associated changes of the major VGLUTs, VGLUT1 and VGLUT2, in the hippocampus were investigated, based on immunohistochemistry and western blot analysis at postnatal month 1 (PM1; adolescent), PM6, PM12 (adult group), PM18 and PM24 (the aged groups). VGLUT1 immunoreactivity was primarily detected in the mossy fibers, Schaffer collaterals and stratum lacunosum‑moleculare. By contrast, VGLUT2 immunoreactivity was observed in the granule cell layer and the outer molecular layer of the dentate gyrus, stratum pyramidale, Schaffer collaterals and stratum lacunosum‑moleculare in the hippocampal CA1‑3 regions. VGLUT1 immunoreactivity and protein levels remained constant across all age groups. However, VGLUT2 immunoreactivity and protein levels decreased in the PM3 group when compared with the PM1 group. VGLUT2 immunoreactivity and protein levels were not altered in the PM12 group; however, they increased in the PM18 group. In addition, in the PM18 group, highly immunoreactive VGLUT2 cells were also identified in the stratum radiatum and oriens of the hippocampal CA1 region. In the PM24 group, VGLUT2 immunoreactivity and protein levels were significantly decreased and were the lowest levels observed amongst the different groups. These results suggested that VGLUT1 may be less susceptible to the aging process; however, the increase of VGLUT2 in the non‑pyramidal cells in the PM18 group, and the consequent decrease in VGLUT2, may be closely linked to age‑associated memory impairment in the hippocampus.
Collapse
Affiliation(s)
- Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin Young Chung
- Department of Veterinary Internal Medicine and Geriatrics, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
33
|
Balázsfi D, Fodor A, Török B, Ferenczi S, Kovács KJ, Haller J, Zelena D. Enhanced innate fear and altered stress axis regulation in VGluT3 knockout mice. Stress 2018; 21:151-161. [PMID: 29310485 DOI: 10.1080/10253890.2017.1423053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Glutamatergic neurons, characterized by vesicular glutamate transporters (VGluT1-3) provide the main excitation in the brain. Their disturbances have been linked to various brain disorders, which could be also modeled by the contextual fear test in rodents. We aimed to characterize the participation of VGluT3 in the development of contextual fear through its contribution to hypothalamic-pituitary-adrenocortical axis (HPA) regulation using knockout (KO) mice. Contextual fear conditioning was induced by foot shock and mice were examined 1 and 7 d later in the same environment comparing wild type with KO. Foot shock increased the immobility time without context specificity. Additionally, foot shock reduced open arm time in the elevated plus maze (EPM) test, and distance traveled in the open field (OF) test, representing the generalization of fear. Moreover, KO mice spent more time with freezing during the contextual fear test, less time in the open arm of the EPM, and traveled a smaller distance in the OF, with less entries into the central area. However, there was no foot shock and genotype interaction suggesting that VGluT3 does not influence the fear conditioning, rather determines anxiety-like characteristic of the mice. The resting hypothalamic CRH mRNA was higher in KO mice with reduced stressor-induced corticosterone elevations. Immunohistochemistry revealed the presence of VGluT3 positive fibers in the paraventricular nucleus of hypothalamus, but not on the hypophysis. As a summary, we confirmed the involvement of VGluT3 in innate fear, but not in the development of fear memory and generalization, with a significant contribution to HPA alterations. Highlights VGluT3 KO mice show innate fear without significant influence on fear memory and generalization. A putative background is the higher resting CRH mRNA level in their PVN and reduced stress-reactivity.
Collapse
Affiliation(s)
- Diána Balázsfi
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
- b János Szentágothai School of Neurosciences , Semmelweis University , Budapest , Hungary
| | - Anna Fodor
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
- b János Szentágothai School of Neurosciences , Semmelweis University , Budapest , Hungary
| | - Bibiána Török
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
- b János Szentágothai School of Neurosciences , Semmelweis University , Budapest , Hungary
| | - Szilamér Ferenczi
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
| | - Krisztina J Kovács
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
| | - József Haller
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
| | - Dóra Zelena
- a Hungarian Academy of Sciences , Institute of Experimental Medicine , Budapest , Hungary
| |
Collapse
|
34
|
Mori Y, Takamori S. Molecular Signatures Underlying Synaptic Vesicle Cargo Retrieval. Front Cell Neurosci 2018; 11:422. [PMID: 29379416 PMCID: PMC5770824 DOI: 10.3389/fncel.2017.00422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/15/2017] [Indexed: 12/31/2022] Open
Abstract
Efficient retrieval of the synaptic vesicle (SV) membrane from the presynaptic plasma membrane, a process called endocytosis, is crucial for the fidelity of neurotransmission, particularly during sustained neural activity. Although multiple modes of endocytosis have been identified, it is clear that the efficient retrieval of the major SV cargos into newly formed SVs during any of these modes is fundamental for synaptic transmission. It is currently believed that SVs are eventually reformed via a clathrin-dependent pathway. Various adaptor proteins recognize SV cargos and link them to clathrin, ensuring the efficient retrieval of the cargos into newly formed SVs. Here, we summarize our current knowledge of the molecular signatures within individual SV cargos that underlie efficient retrieval into SV membranes, as well as discuss possible contributions of the mechanisms under physiological conditions.
Collapse
Affiliation(s)
- Yasunori Mori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
35
|
Horváth HR, Fazekas CL, Balázsfi D, Jain SK, Haller J, Zelena D. Contribution of Vesicular Glutamate Transporters to Stress Response and Related Psychopathologies: Studies in VGluT3 Knockout Mice. Cell Mol Neurobiol 2018; 38:37-52. [PMID: 28776199 DOI: 10.1007/s10571-017-0528-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
Abstract
Maintenance of the homeostasis in a constantly changing environment is a fundamental process of life. Disturbances of the homeostatic balance is defined as stress response and is induced by wide variety of challenges called stressors. Being the main excitatory neurotransmitter of the central nervous system glutamate is important in the adaptation process of stress regulating both the catecholaminergic system and the hypothalamic-pituitary-adrenocortical axis. Data are accumulating about the role of different glutamatergic receptors at all levels of these axes, but little is known about the contribution of different vesicular glutamate transporters (VGluT1-3) characterizing the glutamatergic neurons. Here we summarize basic knowledge about VGluTs, their role in physiological regulation of stress adaptation, as well as their contribution to stress-related psychopathology. Most of our knowledge comes from the VGluT3 knockout mice, as VGluT1 and 2 knockouts are not viable. VGluT3 was discovered later than, and is not as widespread as the VGluT1 and 2. It may co-localize with other transmitters, and participate in retrograde signaling; as such its role might be unique. Previous reports using VGluT3 knockout mice showed enhanced anxiety and innate fear compared to wild type. Moreover, these knockout animals had enhanced resting corticotropin-releasing hormone mRNA levels in the hypothalamus and disturbed glucocorticoid stress responses. In conclusion, VGluT3 participates in stress adaptation regulation. The neuroendocrine changes observed in VGluT3 knockout mice may contribute to their anxious, fearful phenotype.
Collapse
Affiliation(s)
- Hanga Réka Horváth
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Diána Balázsfi
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 26, Üllői út, 1085, Budapest, Hungary
| | | | - József Haller
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary
| | - Dóra Zelena
- Institute of Experimental Medicine, Hungarian Academy of Sciences, 43, Szigony utca, Szigony 43, 1083, Budapest, Hungary.
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary.
| |
Collapse
|
36
|
Li MX, Zheng HL, Luo Y, He JG, Wang W, Han J, Zhang L, Wang X, Ni L, Zhou HY, Hu ZL, Wu PF, Jin Y, Long LH, Zhang H, Hu G, Chen JG, Wang F. Gene deficiency and pharmacological inhibition of caspase-1 confers resilience to chronic social defeat stress via regulating the stability of surface AMPARs. Mol Psychiatry 2018; 23:556-568. [PMID: 28416811 PMCID: PMC5822452 DOI: 10.1038/mp.2017.76] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022]
Abstract
Both inflammatory processes and glutamatergic systems have been implicated in the pathophysiology of mood-related disorders. However, the role of caspase-1, a classic inflammatory caspase, in behavioral responses to chronic stress remains largely unknown. To address this issue, we examined the effects and underlying mechanisms of caspase-1 on preclinical murine models of depression. We found that loss of caspase-1 expression in Caspase-1-/- knockout mice alleviated chronic stress-induced depression-like behaviors, whereas overexpression of caspase-1 in the hippocampus of wild-type (WT) mice was sufficient to induce depression- and anxiety-like behaviors. Furthermore, chronic stress reduced glutamatergic neurotransmission and decreased surface expression of glutamate receptors in hippocampal pyramidal neurons of WT mice, but not Caspase-1-/- mice. Importantly, pharmacological inhibition of caspase-1-interleukin-1β (IL-1β) signaling pathway prevented the depression-like behaviors and the decrease in surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in stressed WT mice. Finally, the effects of chronic stress on both depression- and anxiety-like behaviors can be mimicked by exogenous intracerebroventricular (i.c.v.) administration of IL-1β in both WT and Caspase-1-/- mice. Taken together, our findings demonstrate that an increase in the caspase-1/IL-1β axis facilitates AMPAR internalization in the hippocampus, which dysregulates glutamatergic synaptic transmission, eventually resulting in depression-like behaviors. These results may represent an endophenotype for chronic stress-induced depression.
Collapse
Affiliation(s)
- M-X Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H-L Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Luo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J-G He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - W Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Han
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - X Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L Ni
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H-Y Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Z-L Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - P-F Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - Y Jin
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - L-H Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China
| | - H Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - G Hu
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - J-G Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China,The Collaborative-Innovation Center for Brain Science (HUST), Wuhan, China,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China. E-mail: or
| | - F Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China,The Collaborative-Innovation Center for Brain Science (HUST), Wuhan, China,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China. E-mail: or
| |
Collapse
|
37
|
Yu H, Li M, Zhou D, Lv D, Liao Q, Lou Z, Shen M, Wang Z, Li M, Xiao X, Zhang Y, Wang C. Vesicular glutamate transporter 1 (VGLUT1)-mediated glutamate release and membrane GluA1 activation is involved in the rapid antidepressant-like effects of scopolamine in mice. Neuropharmacology 2017; 131:209-222. [PMID: 29274366 DOI: 10.1016/j.neuropharm.2017.12.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/12/2017] [Accepted: 12/17/2017] [Indexed: 12/19/2022]
Abstract
Emerging data have identified certain drugs such as scopolamine as rapidly acting antidepressants for major depressive disorder (MDD) that increase glutamate release and induce neurotrophic factors through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) activation in rodent models. However, little research has addressed the direct mechanisms of scopolamine on AMPAR activation or vesicular glutamate transporter 1 (VGLUT1)-mediated glutamate release in the prefrontal cortex (PFC) of mice. Herein, using a chronic unpredictable stress (CUS) paradigm, acute treatment with scopolamine rapidly reversed stress-induced depression-like behaviors in mice. Our results showed that CUS-induced depression-like behaviors, accompanied by a decrease in membrane AMPAR subunit 1 (GluA1), phosphorylated GluA1 Ser845 (pGluA1 Ser845), brain-derived neurotrophic factor (BDNF) and VGF (non-acronymic) and an increase in bicaudal C homolog 1 gene (BICC1) in the PFC of mice, and these biochemical and behavioral abnormalities were ameliorated by acute scopolamine treatments. However, pharmacological block of AMPAR by NBQX infusion into the PFC significantly abolished these effects of scopolamine. In addition, knock down of VGLUT1 by lentiviral-mediated RNA interference in the PFC of mice was sufficient to induce depression-like phenotype, to decrease extracellular glutamate accumulation and to cause similar molecular changes with CUS in mice. Remarkably, VGLUT1 knockdown alleviated the rapid antidepressant-like actions of scopolamine and the effects of scopolamine on membrane GluA1-mediated BDNF, VGF and BICC1 changes. Altogether, our findings suggest that VGLUT1-mediated glutamate release and membrane GluA1 activation may play a critical role in the rapid-acting antidepressant-like effects of scopolamine in mice.
Collapse
Affiliation(s)
- Hanjie Yu
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Mengmeng Li
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Dongsheng Zhou
- Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China
| | - Dan Lv
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Qi Liao
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Zhongze Lou
- Department of Psychosomatic Medicine, Ningbo First Hospital, 59 Liuting Str., Ningbo, Zhejiang 315010, China
| | - Mengxin Shen
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Zhen Wang
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, 32 East Jiao-Chang Rd, Kunming, Yunnan 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, 32 East Jiao-Chang Rd, Kunming, Yunnan 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanhua Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
38
|
Groves NJ, Zhou M, Jhaveri DJ, McGrath JJ, Burne THJ. Adult vitamin D deficiency exacerbates impairments caused by social stress in BALB/c and C57BL/6 mice. Psychoneuroendocrinology 2017; 86:53-63. [PMID: 28915381 DOI: 10.1016/j.psyneuen.2017.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/18/2017] [Accepted: 09/03/2017] [Indexed: 02/06/2023]
Abstract
Vitamin D deficiency is prevalent in adults throughout the world. Epidemiological studies have shown significant associations between vitamin D deficiency and an increased risk of various neuropsychiatric and neurodegenerative disorders, such as schizophrenia, depression, Alzheimer's disease and cognitive impairment. However, studies based on observational epidemiology cannot address questions of causality; they cannot determine if vitamin D deficiency is a causal factor leading to the adverse health outcome. The main aim of this study was to determine if AVD deficiency would exacerbate the effects of a secondary exposure, in this case social stress, in BALB/c mice and in the more resilient C57BL/6 mice. Ten-week old male BALB/c and C57BL/6 mice were fed a control or vitamin D deficient diet for 10 weeks, and the mice were further separated into one of two groups for social treatment, either Separated (SEP) or Social Defeat (DEF). SEP mice were placed two per cage with a perforated Plexiglas divider, whereas the DEF mice underwent 10days of social defeat prior to behavioural testing. We found that AVD-deficient mice were more vulnerable to the effects of social stress using a social avoidance test, and this was dependent on strain. These results support the hypothesis that vitamin D deficiency may exacerbate behavioural outcomes in mice vulnerable to stress, a finding that can help guide future studies. Importantly, these discoveries support the epidemiological link between vitamin D deficiency and neuropsychiatric and neurodegenerative disorders; and has provided clues that can guide future studies related to unravelling the mechanisms of action linking adult vitamin D deficiency and adverse brain related outcomes.
Collapse
Affiliation(s)
- Natalie J Groves
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Mei Zhou
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia; Mater Research Institute, The University of Queensland, South Brisbane, QLD, Australia
| | - John J McGrath
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Richlands, QLD, Australia; National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Richlands, QLD, Australia.
| |
Collapse
|
39
|
Riku Y, Watanabe H, Yoshida M, Mimuro M, Iwasaki Y, Masuda M, Ishigaki S, Katsuno M, Sobue G. Pathologic Involvement of Glutamatergic Striatal Inputs From the Cortices in TAR DNA-Binding Protein 43 kDa-Related Frontotemporal Lobar Degeneration and Amyotrophic Lateral Sclerosis. J Neuropathol Exp Neurol 2017; 76:759-768. [PMID: 28859339 DOI: 10.1093/jnen/nlx055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS), recent studies have presumed relationships between cognitive declines and striatal dysfunctions. The striatum contributes to socio-cognitive functions by receiving glutamatergic inputs from the cerebral cortices. However, the vulnerability of these cortico-striatal inputs is unclear in these diseases. This study aimed to evaluate the glutamatergic inputs to the striatum from the cerebral cortices in patients with sporadic TDP-43-related FTLD (FTLD-TDP) and ALS (ALS-TDP). We examined 46 consecutively autopsied patients (31 FTLD-TDP and 15 ALS patients) and 10 normal controls. The axon terminals of the glutamatergic cortico-striatal projection neurons were quantified at the striatum using antivesicular glutamate transporter-1 (VGLUT-1) immunohistochemistry. In results, all FTLD-TDP patients displayed marked depletion of VGLUT-1-positive axon terminals in the caudate head and putamen. Particularly, the patients with type C pathology showed a severe loss. The nondemented ALS patients displayed loss of VGLUT-1-positive axon terminals in the putamen, but those were relatively spared in the caudate head. Confocal microscopy revealed TDP-43 aggregations within VGLUT-1-positive axon terminals in a subset of the patients. Our results indicate marked involvement of glutamatergic striatal inputs from the cerebral cortices in association with socio-cognitive declines in a disease spectrum of TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Yuichi Riku
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Mari Yoshida
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Maya Mimuro
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yasushi Iwasaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Michihito Masuda
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan; and Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
40
|
Hari A, Cruz SA, Qin Z, Couture P, Vilmundarson RO, Huang H, Stewart AFR, Chen HH. IRF2BP2-deficient microglia block the anxiolytic effect of enhanced postnatal care. Sci Rep 2017; 7:9836. [PMID: 28852125 PMCID: PMC5575313 DOI: 10.1038/s41598-017-10349-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/03/2017] [Indexed: 12/21/2022] Open
Abstract
Enhanced postnatal care (EPC) increases resilience to adversity in adulthood. Since microglia participate in shaping neural circuits, we asked how ablation of an inflammation-suppressing factor IRF2BP2 (Interferon Regulatory Factor 2 Binding Protein 2) in microglia would affect the responses to EPC. Mice lacking IRF2BP2 in microglia (KO) and littermate controls (WT) were subjected to EPC during the first 3 weeks after birth. EPC reduced anxiety in WT but not KO mice. This was associated with reduced inflammatory cytokine expression in the hypothalamus. Whole genome RNAseq profiling of the hypothalamus identified 101 genes whose expression was altered by EPC: 95 in WT, 11 in KO, with 5 in common that changed in opposite directions. Proteoglycan 4 (Prg4), prostaglandin D2 synthase (Ptgds) and extracellular matrix protease inhibitor Itih2 were suppressed by EPC in WT but elevated in KO mice. On the other hand, the glutamate transporter VGLUT1 (Slc17a7) was increased by EPC in WT but not KO mice. Prostaglandin D2 (PGD2) is known to enhance microglial inflammation and promote Gfap expression. ELISA confirmed reduced PGD2 in the hypothalamus of WT mice after EPC, associated with reduced Gfap expression. Our study suggests that the anxiety-reducing effect of EPC operates by suppressing microglial inflammation, likely by reducing neuronal prostaglandin D2 production.
Collapse
Affiliation(s)
- Aswin Hari
- Ottawa Hospital Research Institute, Ottawa, Canada.,University of Ottawa Heart Institute, Ottawa, Canada
| | | | - Zhaohong Qin
- Ottawa Hospital Research Institute, Ottawa, Canada
| | | | | | - Hua Huang
- Ottawa Hospital Research Institute, Ottawa, Canada.,University of Ottawa Heart Institute, Ottawa, Canada
| | - Alexandre F R Stewart
- University of Ottawa Heart Institute, Ottawa, Canada.,Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.,Medicine, University of Ottawa, Ottawa, Canada.,University of Ottawa, Center for Infection, Immunity and Inflammation (CI3), Ottawa, Canada
| | - Hsiao-Huei Chen
- Ottawa Hospital Research Institute, Ottawa, Canada. .,University of Ottawa, Brain and Mind Institute, Ottawa, Canada. .,Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada. .,Medicine, University of Ottawa, Ottawa, Canada. .,Canadian Partnership for Stroke Recovery, Ottawa, Canada. .,University of Ottawa, Center for Infection, Immunity and Inflammation (CI3), Ottawa, Canada.
| |
Collapse
|
41
|
SIRT2 inhibition reverses anhedonia in the VGLUT1+/- depression model. Behav Brain Res 2017; 335:128-131. [PMID: 28778545 DOI: 10.1016/j.bbr.2017.07.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 07/21/2017] [Accepted: 07/29/2017] [Indexed: 11/21/2022]
Abstract
Some histone deacetylase (HDACs) enzymes have been proposed as epigenetic targets involved in the pathophysiology of depression and antidepressant-like action. Among them, we have recently identified SIRT2, a class III NAD+-dependent HDAC, as being oppositely regulated by stress and antidepressants. Moreover, SIRT2 inhibition has shown antianhedonic-like action in the chronic mild stress model of depression. Here we have extended the study using an alternative model of depression based in a genetic manipulation of glutamate function. Specifically, mice heterozygous for the vesicular glutamate transporter 1 (VGLUT1+/-) were used. Firstly, mRNA expression of the different members of the HDAC superfamily in the prefrontal cortex (PFC) of VGLUT1+/- mice and WT littermates were studied by RT-PCR. Secondly, the effect of repeated treatment with the selective SIRT2 inhibitor 33i and the antidepressant imipramine on anhedonic behaviour of VGLUT1+/- mice was studied by weekly monitoring of sucrose intake. Further, the interaction of 33i towards specific monoaminergic targets such as serotonin or noradrenaline transporters as well as the monoaminooxidase enzyme was studied. The mRNA occurance of the different members of HDAC superfamily was not altered in the PFC of VGLUT1+/- mice. While repeated imipramine showed an anti-anhedonic action in both VGLUT1+/- and WT, the selective SIRT2 inhibitor 33i fully reversed anhedonia of VGLUT1+/-. Further, 33i showed no interaction with the above mentioned monoaminergic molecular targets. These results confirm that SIRT2 inhibition is able to reverse anhedonia in different animal models and highlight the need to further investigate the role of SIRT2 inhibitors as new antidepressant agents.
Collapse
|
42
|
Characterization of a Human Point Mutation of VGLUT3 (p.A211V) in the Rodent Brain Suggests a Nonuniform Distribution of the Transporter in Synaptic Vesicles. J Neurosci 2017; 37:4181-4199. [PMID: 28314816 DOI: 10.1523/jneurosci.0282-16.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 11/21/2022] Open
Abstract
The atypical vesicular glutamate transporter type 3 (VGLUT3) is expressed by subpopulations of neurons using acetylcholine, GABA, or serotonin as neurotransmitters. In addition, VGLUT3 is expressed in the inner hair cells of the auditory system. A mutation (p.A211V) in the gene that encodes VGLUT3 is responsible for progressive deafness in two unrelated families. In this study, we investigated the consequences of the p.A211V mutation in cell cultures and in the CNS of a mutant mouse. The mutation substantially decreased VGLUT3 expression (-70%). We measured VGLUT3-p.A211V activity by vesicular uptake in BON cells, electrophysiological recording of isolated neurons, and its ability to stimulate serotonergic accumulation in cortical synaptic vesicles. Despite a marked loss of expression, the activity of the mutated isoform was only minimally altered. Furthermore, mutant mice displayed none of the behavioral alterations that have previously been reported in VGLUT3 knock-out mice. Finally, we used stimulated emission depletion microscopy to analyze how the mutation altered VGLUT3 distribution within the terminals of mice expressing the mutated isoform. The mutation appeared to reduce the expression of the VGLUT3 transporter by simultaneously decreasing the number of VGLUT3-positive synaptic vesicles and the amount of VGLUT3 per synapses. These observations suggested that VGLUT3 global activity is not linearly correlated with VGLUT3 expression. Furthermore, our data unraveled a nonuniform distribution of VGLUT3 in synaptic vesicles. Identifying the mechanisms responsible for this complex vesicular sorting will be critical to understand VGLUT's involvement in normal and pathological conditions.SIGNIFICANCE STATEMENT VGLUT3 is an atypical member of the vesicular glutamate transporter family. A point mutation of VGLUT3 (VGLUT3-p.A211V) responsible for a progressive loss of hearing has been identified in humans. We observed that this mutation dramatically reduces VGLUT3 expression in terminals (∼70%) without altering its function. Furthermore, using stimulated emission depletion microscopy, we found that reducing the expression levels of VGLUT3 diminished the number of VGLUT3-positive vesicles at synapses. These unexpected findings challenge the vision of a uniform distribution of synaptic vesicles at synapses. Therefore, the overall activity of VGLUT3 is not proportional to the level of VGLUT3 expression. These data will be key in interpreting the role of VGLUTs in human pathologies.
Collapse
|
43
|
Effective Mechanism for Synthesis of Neurotransmitter Glutamate and its Loading into Synaptic Vesicles. Neurochem Res 2016; 42:64-76. [DOI: 10.1007/s11064-016-2037-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 08/14/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023]
|
44
|
Hollins SL, Zavitsanou K, Walker FR, Cairns MJ. Alteration of transcriptional networks in the entorhinal cortex after maternal immune activation and adolescent cannabinoid exposure. Brain Behav Immun 2016; 56:187-96. [PMID: 26923065 DOI: 10.1016/j.bbi.2016.02.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 02/16/2016] [Accepted: 02/24/2016] [Indexed: 02/04/2023] Open
Abstract
Maternal immune activation (MIA) and adolescent cannabinoid exposure (ACE) have both been identified as major environmental risk factors for schizophrenia. We examined the effects of these two risk factors alone, and in combination, on gene expression during late adolescence. Pregnant rats were exposed to the viral infection mimic polyriboinosinic-polyribocytidylic acid (poly I:C) on gestational day (GD) 15. Adolescent offspring received daily injections of the cannabinoid HU210 for 14days starting on postnatal day (PND) 35. Gene expression was examined in the left entorhinal cortex (EC) using mRNA microarrays. We found prenatal treatment with poly I:C alone, or HU210 alone, produced relatively minor changes in gene expression. However, following combined treatments, offspring displayed significant changes in transcription. This dramatic and persistent alteration of transcriptional networks enriched with genes involved in neurotransmission, cellular signalling and schizophrenia, was associated with a corresponding perturbation in the expression of small non-coding microRNA (miRNA). These results suggest that a combination of environmental exposures during development leads to significant genomic remodeling that disrupts maturation of the EC and its associated circuitry with important implications as the potential antecedents of memory and learning deficits in schizophrenia and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sharon L Hollins
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia
| | - Katerina Zavitsanou
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Frederick Rohan Walker
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia; Centre for Brain and Mental Health Research, Hunter Medical Research Institute, Newcastle, NSW 2305, Australia.
| |
Collapse
|
45
|
Inbar R, Weiss R, Tomljenovic L, Arango MT, Deri Y, Shaw CA, Chapman J, Blank M, Shoenfeld Y. Behavioral abnormalities in female mice following administration of aluminum adjuvants and the human papillomavirus (HPV) vaccine Gardasil. Immunol Res 2016; 65:136-149. [DOI: 10.1007/s12026-016-8826-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
46
|
Weiss R, Bitton A, Ben Shimon M, Elhaik Goldman S, Nahary L, Cooper I, Benhar I, Pick CG, Chapman J. Annexin A2, autoimmunity, anxiety and depression. J Autoimmun 2016; 73:92-9. [PMID: 27372915 DOI: 10.1016/j.jaut.2016.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/21/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Antiphospholipid syndrome (APS) is associated with neurological manifestations and one of the novel autoantigens associated with this disease is Annexin A2 (ANXA2). In this work we have examined the effect of high levels of autoantibodies to ANXA2 on the brain in a mouse model. METHODS Recombinant ANXA2 emulsified in adjuvant was used to immunize mice while mice immunized with adjuvant only served as controls. At peak antibody levels the animal underwent behavioral and cognitive tests and their brains were examined for ANXA2 immunoglobulin G (IgG) and expression of ANXA2 and the closely linked protein p11. RESULTS Very high levels of anti-ANXA2 antibodies (Abs) were associated with reduced anxiety in the open field 13.14% ± 0.89% of the time in the center compared to 8.64% ± 0.91% observed in the control mice (p < 0.001 by t-test). A forced swim test found significantly less depression manifested by immobility in the ANXA2 group. The changes in behavior were accompanied by a significant reduction in serum corticosteroid levels of ANXA2 group compared to controls. Moreover, higher levels of total IgG and p11 expression were found in ANXA2 group brains. Lower levels of circulating anti-ANXA2 Abs were not associated with behavioral changes. CONCLUSIONS We have established an animal model with high levels of anti-ANXA2 Abs which induced IgG accumulation in the brain and specific anxiolytic and anti-depressive effects. This model promises to further our understanding of autoimmune disease such as APS and to provide better understanding of the role of the ANXA2-p11 complex in the brain.
Collapse
Affiliation(s)
- R Weiss
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - A Bitton
- Department of Molecular Microbiology and Biotechnology, Tel-Aviv University, Tel-Aviv, Israel
| | - M Ben Shimon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - S Elhaik Goldman
- BBB-Group, The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, 52621, Israel
| | - L Nahary
- Department of Molecular Microbiology and Biotechnology, Tel-Aviv University, Tel-Aviv, Israel
| | - I Cooper
- BBB-Group, The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, 52621, Israel; The Interdisciplinary Center, Herzliya, Israel
| | - I Benhar
- Department of Molecular Microbiology and Biotechnology, Tel-Aviv University, Tel-Aviv, Israel
| | - C G Pick
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Anatomy, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Chapman
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Neurology, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Hashomer, Israel; Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
47
|
Rodriguez-Perdigon M, Tordera RM, Gil-Bea FJ, Gerenu G, Ramirez MJ, Solas M. Down-regulation of glutamatergic terminals (VGLUT1) driven by Aβ in Alzheimer's disease. Hippocampus 2016; 26:1303-12. [PMID: 27258819 DOI: 10.1002/hipo.22607] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2016] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is characterized phenotypically by memory impairment, histologically by accumulation of pTau and β-amyloid peptide and morphologically by a loss of nerve terminals in cortical and hippocampal regions. As glutamate is the principle excitatory neurotransmitter of the central nervous system (CNS), the glutamatergic system may play an important role in AD. To date, not many studies have addressed the deleterious effects of Aβ on glutamatergic terminals; therefore the aim of this study was to investigate how Aβ affects glutamatergic terminals and to assess the extent to which alterations in the glutamatergic neurotransmission could impact susceptibility to the illness. The present study shows that Aβ caused a loss of glutamatergic terminals, measured by VGLUT1 protein levels, in Tg2576 primary cell cultures, Tg2576 mice and AD patient brains, and also when Aβ was added exogenously to hippocampal cell cultures. Interestingly, no correlation was found between cognition and decreased VGLUT1 levels. Moreover, when Aβ1-42 was intracerebroventricularlly administered into VGLUT1+/- mice, altered synaptic plasticity and increased neuroinflammation was observed in the hippocampus of those animals. In conclusion, the present study not only revealed susceptibility of glutamatergic nerve terminals to Aβ induced toxicity but also underlined the importance of VGLUT1 in the progression of AD, as the decrease of this protein levels could increase the susceptibility to subsequent deleterious inputs by exacerbating Aβ induced neuroinflammation and synaptic plasticity disruption. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Rosa María Tordera
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona 31008, Spain.,IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Francisco Javier Gil-Bea
- Center for Applied Medical Research (CIMA), Neuroscience, University of Navarra, Pamplona, Spain
| | - Gorka Gerenu
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona 31008, Spain
| | - Maria Javier Ramirez
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona 31008, Spain.,IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Maite Solas
- Department of Pharmacology and Toxicology, University of Navarra, Pamplona 31008, Spain. .,IdiSNA Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
48
|
Cutforth T, DeMille MM, Agalliu I, Agalliu D. CNS autoimmune disease after Streptococcus pyogenes infections: animal models, cellular mechanisms and genetic factors. FUTURE NEUROLOGY 2016; 11:63-76. [PMID: 27110222 DOI: 10.2217/fnl.16.4] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Streptococcus pyogenes infections have been associated with two autoimmune diseases of the CNS: Sydenham's chorea (SC) and Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus infections (PANDAS). Despite the high frequency of pharyngeal streptococcus infections among children, only a small fraction develops SC or PANDAS. This suggests that several factors in combination are necessary to trigger autoimmune complications: specific S. pyogenes strains that induce a strong immune response toward the host nervous system; genetic susceptibility that predispose children toward an autoimmune response involving movement or tic symptoms; and multiple infections of the throat or tonsils that lead to a robust Th17 cellular and humoral immune response when untreated. In this review, we summarize the evidence for each factor and propose that all must be met for the requisite neurovascular pathology and behavioral deficits found in SC/PANDAS.
Collapse
Affiliation(s)
- Tyler Cutforth
- Department of Neurology, Columbia University Medical Center, 650 West 168 Street, Room 310E, New York, NY 10032, USA
| | - Mellissa Mc DeMille
- Department of Pediatrics, Yale Child Health Research Center, Yale University School of Medicine, 464 Congress Avenue, Suite S208, New Haven, CT 06519, USA
| | - Ilir Agalliu
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Ave., Room 1315-B, Bronx, NY 10461, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, 650 West 168 Street, Room 310E, New York, NY 10032, USA; Departments of Pathology & Cell Biology & Pharmacology, Columbia University Medical Center, 650 West 168 Street, Room 310E, New York, NY 10032, USA
| |
Collapse
|
49
|
Inbar R, Weiss R, Tomljenovic L, Arango MT, Deri Y, Shaw CA, Chapman J, Blank M, Shoenfeld Y. WITHDRAWN: Behavioral abnormalities in young female mice following administration of aluminum adjuvants and the human papillomavirus (HPV) vaccine Gardasil. Vaccine 2016:S0264-410X(16)00016-5. [PMID: 26778424 DOI: 10.1016/j.vaccine.2015.12.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/15/2015] [Accepted: 12/31/2015] [Indexed: 11/22/2022]
Abstract
This article has been withdrawn at the request of the Editor-in-Chief due to serious concerns regarding the scientific soundness of the article. Review by the Editor-in-Chief and evaluation by outside experts, confirmed that the methodology is seriously flawed, and the claims that the article makes are unjustified. As an international peer-reviewed journal we believe it is our duty to withdraw the article from further circulation, and to notify the community of this issue. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Rotem Inbar
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel; Department of Obstetrics and Gynecology, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel
| | - Ronen Weiss
- Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv, 69978 Tel-Aviv, Israel; Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978 Tel-Aviv, Israel
| | - Lucija Tomljenovic
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel; Neural Dynamics Research Group, Department of Ophthalmology and Visual Sciences, University of British Columbia, 828 W. 10th Ave, Vancouver, BC, Canada V5Z 1L8
| | - Maria-Teresa Arango
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel; Doctoral Program in Biomedical Sciences, Universidad del Rosario, Bogota 111221, Colombia
| | - Yael Deri
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel
| | - Christopher A Shaw
- Neural Dynamics Research Group, Department of Ophthalmology and Visual Sciences, University of British Columbia, 828 W. 10th Ave, Vancouver, BC, Canada V5Z 1L8
| | - Joab Chapman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel; Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv, 69978 Tel-Aviv, Israel; Department of Neurology, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, 52621 Ramat Gan, Israel; Incumbent of the Laura Schwarz-Kip Chair for Research of Autoimmune Diseases, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, 69978 Tel-Aviv, Israel.
| |
Collapse
|
50
|
|