1
|
Shoae-Hagh P, Razavi BM, Sadeghnia HR, Mehri S, Karimi G, Hosseinzadeh H. Molecular and Behavioral Neuroprotective Effects of Clavulanic Acid and Crocin in Haloperidol-Induced Tardive Dyskinesia in Rats. Mol Neurobiol 2025; 62:5156-5182. [PMID: 39520654 DOI: 10.1007/s12035-024-04566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Clavulanic acid (ClvA), a beta-lactamase inhibitor, is being explored for its significant neuroprotective potential. The effects of ClvA were assessed both individually and in combination with crocin (Cr), an antioxidant derived from saffron, in the context of tardive dyskinesia (TD). In rat haloperidol (Hp)-induced-TD (1 mg/kg, i.p. 21 days), the effects of ClvA (50, 100, 150 mg/kg) and Cr (10, 20, 40 mg/kg) were assessed via vacuous chewing movements (VCM) and tongue protrusion (TP). Striatal malondialdehyde (MDA) and glutathione (GSH) were measured spectrophotometrically. Based on the results, ClvA (100 mg/kg) and Cr (10 mg/kg) were determined with sub-effective doses. Glutamate transporter-subtype1 (GLT1), dopamine active transporter (DAT), vesicular monoamine transporter-type2 (VMAT2), Bax/Bcl2, cleaved Caspase3, phosphorylated AKT/AKT, IL1β, and TNFα levels were quantified using western blotting in sub-effective doses and their combination. The behavioral results of catalepsy and orofacial dyskinesia demonstrated model establishment. Hp decreased GLT1 (p < 0.05), DAT (p < 0.01), VMAT2 (p < 0.001), GSH and pAKT/AKT (p < 0.0001); increased TNFα (p < 0.05), IL1β, cleaved Caspase3 (p < 0.001); MDA and Bax/Bcl2 (p < 0.0001). ClvA 100 mg/kg reversed the decreased GLT1 and VMAT2 (p < 0.01), alongside the increased MDA (p < 0.0001) and VCM (p < 0.05). It also increased AKT phosphorylation (p < 0.05). No effects were noted on DAT, GSH, Bax/Bcl2, or inflammatory factors. However, the combination with Cr at 10 mg/kg influenced ClvA on DAT (p < 0.01) and resulted in a significant increase in GSH (p < 0.0001). Additionally, there was a marked decrease in TNFα (p < 0.0001) and IL1β (p < 0.001), enhancing its effects on reducing MDA and increasing pAKT/AKT (p < 0.0001). The combination adversely affected GLT1. ClvA protects against TD via GLT1 and VMAT2; combined with Cr, it enhances antioxidant effects, improves DAT, and requires dose optimization for GLT1 disruption.
Collapse
Affiliation(s)
- Parisa Shoae-Hagh
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hamid Reza Sadeghnia
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Peterson IL, Scholpa NE, Bachtle KJ, Frye JB, Loppi SH, Thompson AD, Doyle K, Largent-Milnes TM, Schnellmann RG. Formoterol alters chemokine expression and ameliorates pain behaviors after moderate spinal cord injury in female mice. J Pharmacol Exp Ther 2025; 392:100015. [PMID: 40023611 DOI: 10.1124/jpet.124.002171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Secondary spinal cord injury (SCI) is characterized by increased cytokines and chemokines at the site of injury that have been associated with the development of neuropathic pain. Nearly 80% of SCI patients report suffering from chronic pain, which is poorly managed with available analgesics. While treatment with the US Food and Drug Administration-approved β2-adrenergic receptor agonist formoterol improves various aspects of recovery post-SCI in vivo, its effects on cytokines, chemokines, and neuropathic pain remain unknown. Female mice were subjected to moderate (60 kilodynes [kdyn]) or severe (80 kdyn) SCI followed by daily treatment with vehicle or formoterol (0.3 mg/kg, i.p.) beginning 8 hours after injury. The expression of proinflammatory cytokines/chemokines, such as interferon gamma-induced protein 10, macrophage inflammatory protein 1a, monocyte chemoattractant protein 1, B-cell attracting chemokine 1, and nuclear factor kappa-light-chain-enhancer of activated B-cells, was increased in the injury site of vehicle-treated mice 24 hours post-SCI, which was ameliorated with formoterol treatment, regardless of injury severity. Thermal hyperalgesia and mechanical allodynia, as measured by Hargreaves infrared apparatus and von Frey filaments, respectively, were assessed prior to SCI and then weekly beginning 21 days post-injury (DPI). While all injured mice exhibited decreased withdrawal latency following thermal stimulation compared with baseline, formoterol treatment reduced this response ∼15% by 35 DPI. Vehicle-treated mice displayed significant mechanical allodynia, as evidenced by a 55% decrease in withdrawal threshold from baseline. In contrast, mice treated with formoterol maintained a consistent withdrawal time at all times tested. These data indicate that formoterol reduces inflammation post-SCI, likely contributing to mitigation of neuropathic pain and further supporting the therapeutic potential of this treatment strategy. SIGNIFICANCE STATEMENT: Chronic pain is a detrimental consequence of spinal cord injury (SCI). We show that treatment with the US Food and Drug Administration-approved drug formoterol after SCI decreases injury site proinflammatory chemo-/cytokines and alters markers of glial cell activation and infiltration. Additionally, formoterol treatment improves locomotor function and body composition, and decreases lesion volume. Finally, formoterol treatment decreased mechanical allodynia and thermal hyperalgesia post-SCI. These data are suggestive of the mechanism of formoterol-induced recovery, and further indicate its potential as a therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ingrid L Peterson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona; Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Natalie E Scholpa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona.
| | - Kiara J Bachtle
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jennifer B Frye
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Sanna H Loppi
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Austin D Thompson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Kristian Doyle
- Southern Arizona VA Health Care System, Tucson, Arizona; Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona; Department of Neuroscience, College of Medicine, University of Arizona, Tucson, Arizona; Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona; Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona.
| |
Collapse
|
3
|
Ramírez OA, Hellwig A, Zhang Z, Bading H. Pharmacological Targeting of the NMDAR/TRPM4 Death Signaling Complex with a TwinF Interface Inhibitor Prevents Excitotoxicity-Associated Dendritic Blebbing and Organelle Damage. Cells 2025; 14:195. [PMID: 39936986 PMCID: PMC11816953 DOI: 10.3390/cells14030195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Focal swellings of dendrites ("dendritic blebbing") together with structural damage of mitochondria and the endoplasmic reticulum (ER) are morphological hallmarks of glutamate neurotoxicity, also known as excitotoxicity. These pathological alterations are generally thought to be caused by the so-called "overactivation" of N-methyl-D-aspartate receptors (NMDARs). Here, we demonstrate that the activation of extrasynaptic NMDARs, specifically when forming a protein-protein complex with TRPM4, drives these pathological traits. In contrast, strong activation of synaptic NMDARs fails to induce cell damage despite evoking plateau-type calcium signals that are comparable to those generated by activation of the NMDAR/TRPM4 complex, indicating that high intracellular calcium levels per se are not toxic to neurons. Using confocal laser scanning microscopy and transmission electron microscopy, we show that disrupting the NMDAR/TRPM4 complex using the recently discovered small-molecule TwinF interface inhibitor FP802 inhibits the NMDA-induced neurotoxicity-associated dendritic blebbing and structural damage to mitochondria and the ER. It also prevents, at least in part, the disruption of ER-mitochondria contact sites. These findings establish the NMDAR/TRPM4 complex as the trigger for the structural damage of dendrites and intracellular organelles associated with excitotoxicity. They also suggest that activation of the NMDAR/TRPM4 complex, in addition to inducing high-amplitude, plateau-type calcium signals, generates a second signal required for glutamate neurotoxicity ("two-hit hypothesis"). As structural damage to organelles, particularly mitochondria, is a common feature of many human neurodegenerative diseases, including Alzheimer's disease and amyotrophic lateral sclerosis (ALS), TwinF interface inhibitors have the potential to provide neuroprotection across a broad spectrum of these diseases.
Collapse
Affiliation(s)
- Omar A. Ramírez
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (O.A.R.); (A.H.); (Z.Z.)
| | - Andrea Hellwig
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (O.A.R.); (A.H.); (Z.Z.)
| | - Zihong Zhang
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (O.A.R.); (A.H.); (Z.Z.)
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (O.A.R.); (A.H.); (Z.Z.)
- Network Aging Research, Heidelberg University, 69115 Heidelberg, Germany
| |
Collapse
|
4
|
Perrin F, Anderson LC, Mitchell SPC, Sinha P, Turchyna Y, Maesako M, Houser MCQ, Zhang C, Wagner SL, Tanzi RE, Berezovska O. PS1/gamma-secretase acts as rogue chaperone of glutamate transporter EAAT2/GLT-1 in Alzheimer's disease. Acta Neuropathol Commun 2024; 12:166. [PMID: 39434170 PMCID: PMC11492509 DOI: 10.1186/s40478-024-01876-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
The recently discovered interaction between presenilin 1 (PS1), a subunit of γ-secretase involved in amyloid-β (Aβ) peptide production, and GLT-1, the major brain glutamate transporter (EAAT2 in the human), may link two pathological aspects of Alzheimer's disease: abnormal Aβ occurrence and neuronal network hyperactivity. In the current study, we employed a FRET-based fluorescence lifetime imaging microscopy (FLIM) to characterize the PS1/GLT-1 interaction in brain tissue from sporadic AD (sAD) patients. sAD brains showed significantly less PS1/GLT-1 interaction than those with frontotemporal lobar degeneration or non-demented controls. Familial AD (fAD) PS1 mutations, inducing a "closed" PS1 conformation similar to that in sAD brain, and gamma-secretase modulators (GSMs), inducing a "relaxed" conformation, respectively reduced and increased the interaction. Furthermore, PS1 influences GLT-1 cell surface expression and homomultimer formation, acting as a chaperone but not affecting GLT-1 stability. The diminished PS1/GLT-1 interaction suggests that these functions may not work properly in AD.
Collapse
Affiliation(s)
- Florian Perrin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Lauren C Anderson
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shane P C Mitchell
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Priyanka Sinha
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Yuliia Turchyna
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Masato Maesako
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Mei C Q Houser
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Can Zhang
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
- VA San Diego Healthcare System, La Jolla, CA, 92161, USA
| | - Rudolph E Tanzi
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Oksana Berezovska
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
5
|
Riboldi JG, Correa J, Renfijes MM, Tintorelli R, Viola H. Arc and BDNF mediated effects of hippocampal astrocytic glutamate uptake blockade on spatial memory stages. Commun Biol 2024; 7:1032. [PMID: 39174690 PMCID: PMC11341830 DOI: 10.1038/s42003-024-06586-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
Glutamate is involved in fundamental functions, including neuronal plasticity and memory. Astrocytes are integral elements involved in synaptic function, and the GLT-1 transporter possesses a critical role in glutamate uptake. Here, we study the role of GLT-1, specifically located in astrocytes, in the consolidation, expression, reconsolidation and persistence of spatial object recognition memory in rats. Administration of dihydrokainic acid (DHK), a selective GLT-1 inhibitor, into the dorsal hippocampus around a weak training which only induces short-term memory, promotes long-term memory formation. This promotion is prevented by hippocampal administration of protein-synthesis translation inhibitor, blockade of Activity-regulated cytoskeleton-associated protein (Arc) translation or Brain-Derived Neurotrophic Factor (BDNF) action, which are plasticity related proteins necessary for memory consolidation. However, DHK around a strong training, which induces long-term memory, does not affect memory consolidation. Administration of DHK before the test session impairs the expression of long-term memory, and this effect is dependent of Arc translation. Furthermore, DHK impairs reconsolidation if applied before a reactivation session, and this effect is independent of Arc translation. These findings reveal specific consequences on spatial memory stages developed under hippocampal GLT-1 blockade, shedding light on the intricate molecular mechanisms, governed in part for the action of glia.
Collapse
Affiliation(s)
- Juan Gabriel Riboldi
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (FBMC), Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Correa
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (FBMC), Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina
| | - Matías M Renfijes
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ramiro Tintorelli
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (FBMC), Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina
| | - Haydee Viola
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias "Profesor Eduardo De Robertis" (IBCN), Buenos Aires, Argentina.
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (FBMC), Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.
- Instituto Tecnológico de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Franklin JP, Testen A, Mieczkowski PA, Hepperla A, Crynen G, Simon JM, Wood JD, Harder EV, Bellinger TJ, Witt EA, Powell NL, Reissner KJ. Investigating cocaine- and abstinence-induced effects on astrocyte gene expression in the nucleus accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606656. [PMID: 39149305 PMCID: PMC11326167 DOI: 10.1101/2024.08.05.606656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In recent years, astrocytes have been increasingly implicated in cellular mechanisms of substance use disorders (SUD). Astrocytes are structurally altered following exposure to drugs of abuse; specifically, astrocytes within the nucleus accumbens (NAc) exhibit significantly decreased surface area, volume, and synaptic colocalization after operant self-administration of cocaine and extinction or protracted abstinence (45 days). However, the mechanisms that elicit these morphological modifications are unknown. The current study aims to elucidate the molecular modifications that lead to observed astrocyte structural changes in rats across cocaine abstinence using astrocyte-specific RiboTag and RNA-seq, as an unbiased, comprehensive approach to identify genes whose transcription or translation change within NAc astrocytes following cocaine self-administration and extended abstinence. Using this method, our data reveal cellular processes including cholesterol biosynthesis that are altered specifically by cocaine self-administration and abstinence, suggesting that astrocyte involvement in these processes is changed in cocaine-abstinent rats. Overall, the results of this study provide insight into astrocyte functional adaptations that occur due to cocaine exposure or during cocaine withdrawal, which may pinpoint further mechanisms that contribute to cocaine-seeking behavior.
Collapse
Affiliation(s)
- Janay P Franklin
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Anze Testen
- Department of Neuroscience, Medical University of South Carolina
| | | | - Austin Hepperla
- Department of Genetics, University of North Carolina at Chapel Hill
| | - Gogce Crynen
- Bioinformatics and Statistics Core, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Institute Department of Biostatistics, Harvard T.H. Chan School of Public Health
| | - Jonathan D Wood
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Eden V Harder
- Neuroscience Center, University of North Carolina at Chapel Hill
| | - Tania J Bellinger
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Emily A Witt
- Department of Medical Neuroscience, Dalhousie University
| | - N LaShae Powell
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Kathryn J Reissner
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| |
Collapse
|
7
|
Poljak L, Miše B, Čičin-Šain L, Tvrdeić A. Ceftriaxone Inhibits Conditioned Fear and Compulsive-like Repetitive Marble Digging without Central Nervous System Side Effects Typical of Diazepam-A Study on DBA2/J Mice and a High-5HT Subline of Wistar-Zagreb 5HT Rats. Biomedicines 2024; 12:1711. [PMID: 39200176 PMCID: PMC11351474 DOI: 10.3390/biomedicines12081711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Ceftriaxone upregulates GLT1 glutamate transporter in the brain and may have anti-CFC and anti-OCD effects. Methods: Twenty WZ-5HT rats were used to investigate the effects of ceftriaxone on obsessive-compulsive (OCD)-like behaviour in the marble-burying (MB) test, freezing behaviour in contextual fear conditioning (CFC) and expression of GLT1 protein in the hippocampus or amygdala using immunoblots. Fifteen DBA/2J mice were used in the MB test. We also compared diazepam with ceftriaxone in open-field, beam-walking, and wire-hanging tests on 47 DBA/2J mice. Ceftriaxone (200 mg/kg) and saline were applied intraperitoneally, once daily for 7 (rats) or 5 (mice) consecutive days. A single dose of diazepam (1.5-3.0 mg/kg) or saline was injected 30 min before the behavioural tests. Results: Ceftriaxone significantly diminished OCD-like behaviour (↓ number of marbles buried) and freezing behaviour in CFC context session (↑ latencies, ↓ total duration, ↓ duration over four 2 min periods of the session) but increased GLT1 protein expression in the amygdala and hippocampus of rats. Diazepam induced sedation, ataxia and myorelaxation in mice. Ceftriaxone did not have these side effects. Conclusions: The results of this study confirm the anti-CFC and anti-OCD effects of ceftriaxone, which did not produce the unwanted effects typical of diazepam.
Collapse
Affiliation(s)
- Ljiljana Poljak
- Department of Physiology and Immunology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Branko Miše
- University Hospital for Infectious Diseases “Fran Mihaljević”, 10000 Zagreb, Croatia;
| | - Lipa Čičin-Šain
- Laboratory for Neurochemistry and Molecular Neurobiology, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Ante Tvrdeić
- Department of Pharmacology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
8
|
Torres-Carmona E, Nakajima S, Iwata Y, Ueno F, Stefan C, Song J, Abdolizadeh A, Koizumi MT, Kambari Y, Amaev A, Agarwal SM, Mar W, de Luca V, Remington G, Gerretsen P, Graff-Guerrero A. Clozapine treatment and astrocyte activity in treatment resistant schizophrenia: A proton magnetic resonance spectroscopy study. Schizophr Res 2024; 270:152-161. [PMID: 38909486 DOI: 10.1016/j.schres.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
Clozapine is the only antipsychotic approved for treating treatment-resistant schizophrenia (TRS), characterized by persistent positive symptoms despite adequate antipsychotic treatment. Unfortunately, clozapine demonstrates clinical efficacy in only ~30-60 % of patients with TRS (clozapine-responders; ClzR+), while the remaining ~40-70 % are left with no pharmacological recourse for improvement (clozapine-resistant; ClzR-). Mechanism(s) underlying clozapine's superior efficacy remain unclear. However, in vitro evidence suggests clozapine may mitigate glutamatergic dysregulations observed in TRS, by modulating astrocyte activity in ClzR+, but not ClzR-. A factor that if proven correct, may help the assessment of treatment response and development of more effective antipsychotics. To explore the presence of clozapine-astrocyte interaction and clinical improvement, we used 3 T proton-magnetic resonance spectroscopy to quantify levels of myo-Inositol, surrogate biomarker of astrocyte activity, in regions related to schizophrenia neurobiology: Dorsal-anterior-cingulate-cortex (dACC), left-dorsolateral-prefrontal-cortex (left-DLPFC), and left-striatum (left-striatum) of 157 participants (ClzR- = 30; ClzR+ = 37; responders = 38; controls = 52). Clozapine treatment was assessed using clozapine to norclozapine plasma levels, 11-12 h after last clozapine dose. Measures for symptom severity (i.e., Positive and Negative Symptoms Scale) and cognition (i.e., Mini-Mental State Examination) were also recorded. Higher levels of myo-Inositol were observed in TRS groups versus responders and controls (dACC (p < 0.001); left-striatum (p = 0.036); left-DLPFC (p = 0.023)). In ClzR+, but not ClzR-, clozapine to norclozapine ratios were positively associated with myo-Inositol levels (dACC (p = 0.004); left-DLPFC (p < 0.001)), and lower positive symptom severity (p < 0.001). Our results support growing in vitro evidence of clozapine-astrocyte interaction in clozapine-responders. Further research may determine the viability of clozapine-astrocyte interactions as an early marker of clozapine response.
Collapse
Affiliation(s)
- Edgardo Torres-Carmona
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Shinichiro Nakajima
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Neuropsychiatry, Keio University, Minato, Tokyo, Japan
| | - Yusuke Iwata
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Fumihiko Ueno
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Cristiana Stefan
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Jianmeng Song
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Ali Abdolizadeh
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | | | - Yasaman Kambari
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Aron Amaev
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sri Mahavir Agarwal
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Wanna Mar
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Vincenzo de Luca
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Gary Remington
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Philip Gerretsen
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Campbell Family Mental Health Research Institute, CAMH, Toronto, ON, Canada.
| |
Collapse
|
9
|
Sun M, Chen Z. Unveiling the Complex Role of Exosomes in Alzheimer's Disease. J Inflamm Res 2024; 17:3921-3948. [PMID: 38911990 PMCID: PMC11193473 DOI: 10.2147/jir.s466821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative illness, characterized by memory loss and cognitive decline, accounting for 60-80% of dementia cases. AD is characterized by senile plaques made up of amyloid β (Aβ) protein, intracellular neurofibrillary tangles caused by hyperphosphorylation of tau protein linked with microtubules, and neuronal loss. Currently, therapeutic treatments and nanotechnological developments are effective in treating the symptoms of AD, but a cure for the illness has not yet been found. Recently, the increased study of extracellular vesicles (EVs) has led to a growing awareness of their significant involvement in neurodegenerative disorders, including AD. Exosomes are small extracellular vesicles that transport various components including messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive compounds from one cell to another, facilitating information transmission and material movement. There is growing evidence indicating that exosomes have complex functions in AD. Exosomes may have a dual role in Alzheimer's disease by contributing to neuronal death and also helping to alleviate the pathological progression of the disease. Therefore, the primary aim of this review is to outline the updated understandings on exosomes biogenesis and many functions of exosomes in the generation, conveyance, distribution, and elimination of hazardous proteins related to Alzheimer's disease. This review is intended to provide novel insights for understanding the development, specific treatment, and early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Mingyue Sun
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| | - Zhuoyou Chen
- Department of Neurology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, 213000, People’s Republic of China
| |
Collapse
|
10
|
Virtuoso A, D’Amico G, Scalia F, De Luca C, Papa M, Maugeri G, D’Agata V, Caruso Bavisotto C, D’Amico AG. The Interplay between Glioblastoma Cells and Tumor Microenvironment: New Perspectives for Early Diagnosis and Targeted Cancer Therapy. Brain Sci 2024; 14:331. [PMID: 38671983 PMCID: PMC11048111 DOI: 10.3390/brainsci14040331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma multiforme (GBM) stands out as the most tremendous brain tumor, constituting 60% of primary brain cancers, accompanied by dismal survival rates. Despite advancements in research, therapeutic options remain limited to chemotherapy and surgery. GBM molecular heterogeneity, the intricate interaction with the tumor microenvironment (TME), and non-selective treatments contribute to the neoplastic relapse. Diagnostic challenges arise from GBM advanced-stage detection, necessitating the exploration of novel biomarkers for early diagnosis. Using data from the literature and a bioinformatic tool, the current manuscript delineates the molecular interplay between human GBM, astrocytes, and myeloid cells, underscoring selected protein pathways belonging to astroglia and myeloid lineage, which can be considered for targeted therapies. Moreover, the pivotal role of extracellular vesicles (EVs) in orchestrating a favorable microenvironment for cancer progression is highlighted, suggesting their utility in identifying biomarkers for GBM early diagnosis.
Collapse
Affiliation(s)
- Assunta Virtuoso
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Giuseppa D’Amico
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Federica Scalia
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
| | - Ciro De Luca
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.V.); (C.D.L.); (M.P.)
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (V.D.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), Human Anatomy Section, University of Palermo, 90127 Palermo, Italy; (G.D.); (F.S.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
| | - Agata Grazia D’Amico
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| |
Collapse
|
11
|
Perrin F, Anderson LC, Mitchell SPC, Sinha P, Turchyna Y, Maesako M, Houser MCQ, Zhang C, Wagner SL, Tanzi RE, Berezovska O. PS1/gamma-secretase acts as rogue chaperone of glutamate transporter EAAT2/GLT-1 in Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-3495211. [PMID: 37986905 PMCID: PMC10659539 DOI: 10.21203/rs.3.rs-3495211/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The recently discovered interaction between presenilin 1 (PS1), a catalytic subunit of γ-secretase responsible for the generation of amyloid-β(Aβ) peptides, and GLT-1, the major glutamate transporter in the brain (EAAT2 in the human) may provide a mechanistic link between two important pathological aspects of Alzheimer's disease (AD): abnormal Aβoccurrence and neuronal network hyperactivity. In the current study, we employed a FRET-based approach, fluorescence lifetime imaging microscopy (FLIM), to characterize the PS1/GLT-1 interaction in its native environment in the brain tissue of sporadic AD (sAD) patients. There was significantly less interaction between PS1 and GLT-1 in sAD brains, compared to tissue from patients with frontotemporal lobar degeneration (FTLD), or non-demented age-matched controls. Since PS1 has been shown to adopt pathogenic "closed" conformation in sAD but not in FTLD, we assessed the impact of changes in PS1 conformation on the interaction. Familial AD (fAD) PS1 mutations which induce a "closed" PS1 conformation similar to that in sAD brain and gamma-secretase modulators (GSMs) which induce a "relaxed" conformation, reduced and increased the interaction, respectively. This indicates that PS1 conformation seems to have a direct effect on the interaction with GLT-1. Furthermore, using biotinylation/streptavidin pull-down, western blotting, and cycloheximide chase assays, we determined that the presence of PS1 increased GLT-1 cell surface expression and GLT-1 homomultimer formation, but did not impact GLT-1 protein stability. Together, the current findings suggest that the newly described PS1/GLT-1 interaction endows PS1 with chaperone activity, modulating GLT-1 transport to the cell surface and stabilizing the dimeric-trimeric states of the protein. The diminished PS1/GLT-1 interaction suggests that these functions of the interaction may not work properly in AD.
Collapse
|
12
|
Davletshin AI, Matveeva AA, Poletaeva II, Evgen'ev MB, Garbuz DG. The role of molecular chaperones in the mechanisms of epileptogenesis. Cell Stress Chaperones 2023; 28:599-619. [PMID: 37755620 PMCID: PMC10746656 DOI: 10.1007/s12192-023-01378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Epilepsy is a group of neurological diseases which requires significant economic costs for the treatment and care of patients. The central point of epileptogenesis stems from the failure of synaptic signal transmission mechanisms, leading to excessive synchronous excitation of neurons and characteristic epileptic electroencephalogram activity, in typical cases being manifested as seizures and loss of consciousness. The causes of epilepsy are extremely diverse, which is one of the reasons for the complexity of selecting a treatment regimen for each individual case and the high frequency of pharmacoresistant cases. Therefore, the search for new drugs and methods of epilepsy treatment requires an advanced study of the molecular mechanisms of epileptogenesis. In this regard, the investigation of molecular chaperones as potential mediators of epileptogenesis seems promising because the chaperones are involved in the processing and regulation of the activity of many key proteins directly responsible for the generation of abnormal neuronal excitation in epilepsy. In this review, we try to systematize current data on the role of molecular chaperones in epileptogenesis and discuss the prospects for the use of chemical modulators of various chaperone groups' activity as promising antiepileptic drugs.
Collapse
Affiliation(s)
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
- Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Moscow Region, Russia
| | - Inga I Poletaeva
- Biology Department, Lomonosov Moscow State University, 119991, Moscow, Russia
| | | | - David G Garbuz
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
| |
Collapse
|
13
|
Villanueva CE, Hagenbuch B. Palmitoylation of solute carriers. Biochem Pharmacol 2023; 215:115695. [PMID: 37481134 PMCID: PMC10530500 DOI: 10.1016/j.bcp.2023.115695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Post-translational modifications are an important mechanism in the regulation of protein expression, function, and degradation. Well-known post-translational modifications are phosphorylation, glycosylation, and ubiquitination. However, lipid modifications, including myristoylation, prenylation, and palmitoylation, are poorly studied. Since the early 2000s, researchers have become more interested in lipid modifications, especially palmitoylation. The number of articles in PubMed increased from about 350 between 2000 and 2005 to more than 600 annually during the past ten years. S-palmitoylation, where the 16-carbon saturated (C16:0) palmitic acid is added to free cysteine residues of proteins, is a reversible protein modification that can affect the expression, membrane localization, and function of the modified proteins. Various diseases like Huntington's and Alzheimer's disease have been linked to changes in protein palmitoylation. In humans, the addition of palmitic acid is mediated by 23 palmitoyl acyltransferases, also called DHHC proteins. The modification can be reversed by a few thioesterases or hydrolases. Numerous soluble and membrane-attached proteins are known to be palmitoylated, but among the approximately 400 solute carriers that are classified in 66 families, only 15 found in 8 families have so far been documented to be palmitoylated. Among the best-characterized transporters are the glucose transporters GLUT1 (SLC2A1) and GLUT4 (SLC2A4), the three monoamine transporters norepinephrine transporter (NET; SLC6A2), dopamine transporter (DAT; SLC6A3), and serotonin transporter (SERT; SLC6A4), and the sodium-calcium exchanger NCX1 (SLC8A1). While there is evidence from recent proteomics experiments that numerous solute carriers are palmitoylated, no details beyond the 15 transporters covered in this review are available.
Collapse
Affiliation(s)
- Cecilia E Villanueva
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States.
| |
Collapse
|
14
|
Liu T, Bai M, Liu M, Li T, Liao Y, Zhao C, Yao M, Wang J, Wen A, Ding Y. Novel synergistic mechanism of 11-keto-β-boswellic acid and Z-Guggulsterone on ischemic stroke revealed by single-cell transcriptomics. Pharmacol Res 2023; 193:106803. [PMID: 37230158 DOI: 10.1016/j.phrs.2023.106803] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/11/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Although strides have been made, the challenge of preventing and treating ischemic stroke continues to persist globally. For thousands of years, the natural substances Frankincense and Myrrh have been employed in Chinese and Indian medicine to address cerebrovascular diseases, with the key components of 11-keto-β-boswellic acid (KBA) and Z-Guggulsterone (Z-GS) being the active agents. In this study, the synergistic effect and underlying mechanism of KBA and Z-GS on ischemic stroke were examined using single-cell transcriptomics. Fourteen cell types were identified in KBA-Z-GS-treated ischemic penumbra, and microglia and astrocytes account for the largest proportion. They were further re-clustered into six and seven subtypes, respectively. GSVA analysis reflected the distinct roles of each subtype. Pseudo-time trajectory indicated that Slc1a2 and Timp1 were core fate transition genes regulated by KBA-Z-GS. In addition, KBA-Z-GS synergistically regulated inflammatory reactions in microglia and cellular metabolism and ferroptosis in astrocytes. Most notably, we established an innovative drug-gene synergistic regulation pattern, and genes regulated by KBA-Z-GS were divided into four categories based on this pattern. Finally, Spp1 was demonstrated as the hub target of KBA-Z-GS. Taken together, this study reveals the synergistic mechanism of KBA and Z-GS on cerebral ischemia, and Spp1 may be the synergistic target for that. Precise drug development targeting Spp1 may offer a potential therapeutic approach for treating ischemic stroke.
Collapse
Affiliation(s)
- Tianlong Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Pharmacy, The 940th Hospital Joint Logistics Support Forces of PLA, Lanzhou 730050, China
| | - Min Bai
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Minna Liu
- Department of Nephrology, The 940th Hospital Joint Logistics Support Forces of PLA, Lanzhou 730050, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yucheng Liao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Minna Yao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
15
|
Pathak D, Sriram K. Neuron-astrocyte omnidirectional signaling in neurological health and disease. Front Mol Neurosci 2023; 16:1169320. [PMID: 37363320 PMCID: PMC10286832 DOI: 10.3389/fnmol.2023.1169320] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Astrocytes are an abundantly distributed population of glial cells in the central nervous system (CNS) that perform myriad functions in the normal and injured/diseased brain. Astrocytes exhibit heterogeneous phenotypes in response to various insults, a process known as astrocyte reactivity. The accuracy and precision of brain signaling are primarily based on interactions involving neurons, astrocytes, oligodendrocytes, microglia, pericytes, and dendritic cells within the CNS. Astrocytes have emerged as a critical entity within the brain because of their unique role in recycling neurotransmitters, actively modulating the ionic environment, regulating cholesterol and sphingolipid metabolism, and influencing cellular crosstalk in diverse neural injury conditions and neurodegenerative disorders. However, little is known about how an astrocyte functions in synapse formation, axon specification, neuroplasticity, neural homeostasis, neural network activity following dynamic surveillance, and CNS structure in neurological diseases. Interestingly, the tripartite synapse hypothesis came to light to fill some knowledge gaps that constitute an interaction of a subpopulation of astrocytes, neurons, and synapses. This review highlights astrocytes' role in health and neurological/neurodegenerative diseases arising from the omnidirectional signaling between astrocytes and neurons at the tripartite synapse. The review also recapitulates the disruption of the tripartite synapse with a focus on perturbations of the homeostatic astrocytic function as a key driver to modulate the molecular and physiological processes toward neurodegenerative diseases.
Collapse
|
16
|
Yeşilören E, Yalcin GD. The Regulation of GLT-1 Degradation Pathway by SIRT4. Neurochem Res 2023:10.1007/s11064-023-03947-3. [PMID: 37178383 DOI: 10.1007/s11064-023-03947-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/18/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Glial cells give rise to glioblastoma multiform as a primary brain tumor. In glioblastomas, neurons are destroyed via excitotoxicity which is the accumulation of excess glutamate in synaptic cavity. Glutamate Transporter 1 (GLT-1) is the main transporter that absorbs the excessive glutamate. Sirtuin 4 (SIRT4) was shown to have a potential protective role against excitotoxicity in previous studies. In this study, the regulation of dynamic GLT-1 expression by SIRT4 was analyzed in glia (immortalized human astrocytes) and glioblastoma (U87) cells. The expression of GLT-1 dimers and trimers were reduced and the ubiquitination of GLT-1 was increased in glioblastoma cells when SIRT4 was silenced; however GLT-1 monomer was not affected. In glia cells, SIRT4 reduction did not affect GLT-1 monomer, dimer, trimer expression or the ubiquitination of GLT-1. The phosphorylation of Nedd4-2 and the expression of PKC did not change in glioblastoma cells when SIRT4 was silenced but increased in glia cells. We also showed that SIRT4 deacetylates PKC in glia cells. In addition, GLT-1 was shown to be deacetylated by SIRT4 which might be a priority for ubiquitination. Therefore, we conclude that GLT-1 expression is regulated differently in glia and glioblastoma cells. SIRT4 activators or inhibitors of ubiquitination may be used to prevent excitotoxicity in glioblastomas.
Collapse
Affiliation(s)
- Emre Yeşilören
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Gizem Donmez Yalcin
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey.
| |
Collapse
|
17
|
Paton SE, Menard C. Glutamate shall not pass: a mechanistic role for astrocytic O-GlcNAc transferase in stress and depression. J Clin Invest 2023; 133:168662. [PMID: 37009895 PMCID: PMC10065068 DOI: 10.1172/jci168662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
Major depressive disorder, characterized by aberrant glutamatergic signaling in the prefrontal cortex (PFC), is a leading cause of disability worldwide. Depression is highly comorbid with metabolic disorders, but a mechanistic link is elusive. In this issue of the JCI, Fan and coauthors report that elevated posttranslational modification with the glucose metabolite N-acetylglucosamine (GlcNAc) by O-GlcNAc transferase (OGT) contributed to stress-induced establishment of depression-like behaviors in mice. This effect was specific to medial PFC (mPFC) astrocytes, with glutamate transporter-1 (GLT-1) identified as an OGT target. Specifically, O-GlcNAcylation of GLT-1 resulted in diminished glutamate clearance from excitatory synapses. Further, astrocytic OGT knockdown restored stress-induced deficits in glutamatergic signaling, promoting resilience. These findings provide a mechanistic link between metabolism and depression and have relevance for antidepressant targets.
Collapse
|
18
|
Petroianu GA, Aloum L, Adem A. Neuropathic pain: Mechanisms and therapeutic strategies. Front Cell Dev Biol 2023; 11:1072629. [PMID: 36727110 PMCID: PMC9884983 DOI: 10.3389/fcell.2023.1072629] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
The physiopathology and neurotransmission of pain are of an owe inspiring complexity. Our ability to satisfactorily suppress neuropathic or other forms of chronic pain is limited. The number of pharmacodynamically distinct and clinically available medications is low and the successes achieved modest. Pain Medicine practitioners are confronted with the ethical dichotomy imposed by Hippocrates: On one hand the mandate of primum non nocere, on the other hand, the promise of heavenly joys if successful divinum est opus sedare dolorem. We briefly summarize the concepts associated with nociceptive pain from nociceptive input (afferents from periphery), modulatory output [descending noradrenergic (NE) and serotoninergic (5-HT) fibers] to local control. The local control is comprised of the "inflammatory soup" at the site of pain origin and synaptic relay stations, with an ATP-rich environment promoting inflammation and nociception while an adenosine-rich environment having the opposite effect. Subsequently, we address the transition from nociceptor pain to neuropathic pain (independent of nociceptor activation) and the process of sensitization and pain chronification (transient pain progressing into persistent pain). Having sketched a model of pain perception and processing we attempt to identify the sites and modes of action of clinically available drugs used in chronic pain treatment, focusing on adjuvant (co-analgesic) medication.
Collapse
|
19
|
Bansal Y, Fee C, Misquitta KA, Codeluppi SA, Sibille E, Berman RM, Coric V, Sanacora G, Banasr M. Prophylactic Efficacy of Riluzole against Anxiety- and Depressive-Like Behaviors in Two Rodent Stress Models. Complex Psychiatry 2023; 9:57-69. [PMID: 37101541 PMCID: PMC10123365 DOI: 10.1159/000529534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Chronic stress-related illnesses such as major depressive disorder and post-traumatic stress disorder share symptomatology, including anxiety, anhedonia, and helplessness. Across disorders, neurotoxic dysregulated glutamate (Glu) signaling may underlie symptom emergence. Current first-line antidepressant drugs, which do not directly target Glu signaling, fail to provide adequate benefit for many patients and are associated with high relapse rates. Riluzole modulates glutamatergic neurotransmission by increasing metabolic cycling and modulating signal transduction. Clinical studies exploring riluzole's efficacy in stress-related disorders have provided varied results. However, the utility of riluzole for treating specific symptom dimensions or as a prophylactic treatment has not been comprehensively assessed. Methods We investigated whether chronic prophylactic riluzole (∼12-15 mg/kg/day p.o.) could prevent the emergence of behavioral deficits induced by unpredictable chronic mild stress (UCMS) in mice. We assessed (i) anxiety-like behavior using the elevated-plus maze, open-field test, and novelty-suppressed feeding, (ii) mixed anxiety/anhedonia-like behavior in the novelty-induced hypophagia test, and (iii) anhedonia-like behavior using the sucrose consumption test. Z-scoring summarized changes across tests measuring similar dimensions. In a separate learned helplessness (LH) cohort, we investigated whether chronic prophylactic riluzole treatment could block the development of helplessness-like behavior. Results UCMS induced an elevation in anhedonia-like behavior and overall behavioral emotionality that was blocked by prophylactic riluzole. In the LH cohort, prophylactic riluzole blocked the development of helplessness-like behavior. Discussion/Conclusion This study supports the utility of riluzole as a prophylactic medication for preventing anhedonia and helplessness symptoms associated with stress-related disorders.
Collapse
Affiliation(s)
- Yashika Bansal
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Corey Fee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Keith A. Misquitta
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Sierra A. Codeluppi
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | | | | | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
McClendon LK, Garcia RL, Lazaro T, Robledo A, Vasandani V, Luna ZAE, Rao AS, Srivatsan A, Lonard DM, Dacso CC, Kan P, O’Malley BW. A steroid receptor coactivator small molecule "stimulator" attenuates post-stroke ischemic brain injury. Front Mol Neurosci 2022; 15:1055295. [PMID: 36533127 PMCID: PMC9751323 DOI: 10.3389/fnmol.2022.1055295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2023] Open
Abstract
Introduction: Pathologic remodeling of the brain following ischemic stroke results in neuronal loss, increased inflammation, oxidative stress, astrogliosis, and a progressive decrease in brain function. We recently demonstrated that stimulation of steroid receptor coactivator 3 with the small-molecule stimulator MCB-613 improves cardiac function in a mouse model of myocardial ischemia. Since steroid receptor coactivators are ubiquitously expressed in the brain, we reasoned that an MCB-613 derivative (MCB-10-1), could protect the brain following ischemic injury. To test this, we administered MCB-10-1 to rats following middle cerebral artery occlusion and reperfusion. Methods: Neurologic impairment and tissue damage responses were evaluated on day 1 and day 4 following injury in rats treated with control or 10-1. Results: We show that 10-1 attenuates injury post-stroke. 10-1 decreases infarct size and mitigates neurologic impairment. When given within 30 min post middle cerebral artery occlusion and reperfusion, 10-1 induces lasting protection from tissue damage in the ischemic penumbra concomitant with: (1) promotion of reparative microglia; (2) an increase in astrocyte NRF2 and GLT-1 expression; (3) early microglia activation; and (4) attenuation of astrogliosis. Discussion: Steroid receptor coactivator stimulation with MCB-10-1 is a potential therapeutic strategy for reducing inflammation and oxidative damage that cause neurologic impairment following an acute ischemic stroke.
Collapse
Affiliation(s)
- Lisa K. McClendon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Roberto L. Garcia
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Tyler Lazaro
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - Ariadna Robledo
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Viren Vasandani
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Zean Aaron Evan Luna
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Abhijit S. Rao
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Aditya Srivatsan
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, United States
| | - David M. Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Clifford C. Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| | - Peter Kan
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- CoRegen, Inc., Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
21
|
Increased Extrasynaptic Glutamate Escape in Stochastically Shaped Probabilistic Synaptic Environment. Biomedicines 2022; 10:biomedicines10102406. [PMID: 36289667 PMCID: PMC9599130 DOI: 10.3390/biomedicines10102406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022] Open
Abstract
Excitatory synapses in the brain are often surrounded by nanoscopic astroglial processes that express high-affinity glutamate transporters at a high surface density. This ensures that the bulk of glutamate leaving the synaptic cleft is taken up for its subsequent metabolic conversion and replenishment in neurons. Furthermore, variations in the astroglial coverage of synapses can thus determine to what extent glutamate released into the synaptic cleft could activate its receptors outside the cleft. The biophysical determinants of extrasynaptic glutamate actions are complex because they involve a competition between transporters and target receptors of glutamate in the tortuous space of synaptic environment. To understand key spatiotemporal relationships between the extrasynaptic landscapes of bound and free glutamate, we explored a detailed Monte Carlo model for its release, diffusion, and uptake. We implemented a novel representation of brain neuropil in silico as a space filled with randomly scattered, overlapping spheres (spheroids) of distributed size. The parameters of perisynaptic space, astroglial presence, and glutamate transport were constrained by the empirical data obtained for the 'average' environment of common cortical synapses. Our simulations provide a glimpse of the perisynaptic concentration landscapes of free and transporter-bound glutamate relationship, suggesting a significant tail of space-average free glutamate within 3 ms post-release.
Collapse
|
22
|
Agnew-Svoboda W, Ubina T, Figueroa Z, Wong YC, Vizcarra EA, Roebini B, Wilson EH, Fiacco TA, Riccomagno MM. A genetic tool for the longitudinal study of a subset of post-inflammatory reactive astrocytes. CELL REPORTS METHODS 2022; 2:100276. [PMID: 36046623 PMCID: PMC9421582 DOI: 10.1016/j.crmeth.2022.100276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/01/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022]
Abstract
Astrocytes are vital support cells that ensure proper brain function. In brain disease, astrocytes reprogram into a reactive state that alters many of their cellular roles. A long-standing question in the field is whether downregulation of reactive astrocyte (RA) markers during resolution of inflammation is because these astrocytes revert back to a non-reactive state or die and are replaced. This has proven difficult to answer mainly because existing genetic tools cannot distinguish between healthy versus RAs. Here we describe the generation of an inducible genetic tool that can be used to specifically target and label a subset of RAs. Longitudinal analysis of an acute inflammation model using this tool revealed that the previously observed downregulation of RA markers after inflammation is likely due to changes in gene expression and not because of cell death. Our findings suggest that cellular changes associated with astrogliosis after acute inflammation are largely reversible.
Collapse
Affiliation(s)
- William Agnew-Svoboda
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Teresa Ubina
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Zoe Figueroa
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
| | - Yiu-Cheung Wong
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Edward A. Vizcarra
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Bryan Roebini
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Emma H. Wilson
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Todd A. Fiacco
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| | - Martin M. Riccomagno
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
- Biomedical Sciences Graduate Program, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
23
|
Gupta S, Bazargani N, Drew J, Howden JH, Modi S, Al Awabdh S, Marie H, Attwell D, Kittler JT. The non-adrenergic imidazoline-1 receptor protein nischarin is a key regulator of astrocyte glutamate uptake. iScience 2022; 25:104127. [PMID: 35434559 PMCID: PMC9010640 DOI: 10.1016/j.isci.2022.104127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 12/24/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Astrocytic GLT-1 is the main glutamate transporter involved in glutamate buffering in the brain, pivotal for glutamate removal at excitatory synapses to terminate neurotransmission and for preventing excitotoxicity. We show here that the surface expression and function of GLT-1 can be rapidly modulated through the interaction of its N-terminus with the nonadrenergic imidazoline-1 receptor protein, Nischarin. The phox domain of Nischarin is critical for interaction and internalization of surface GLT-1. Using live super-resolution imaging, we found that glutamate accelerated Nischarin-GLT-1 internalization into endosomal structures. The surface GLT-1 level increased in Nischarin knockout astrocytes, and this correlated with a significant increase in transporter uptake current. In addition, Nischarin knockout in astrocytes is neuroprotective against glutamate excitotoxicity. These data provide new molecular insights into regulation of GLT-1 surface level and function and suggest new drug targets for the treatment of neurological disorders. Nischarin phox domain interacts with the N-terminus of the glutamate transporter, GLT-1 Nischarin promotes internalization of GLT-1 to endosomes Glutamate modulates GLT-1 surface levels by regulating the Nischarin-GLT-1 interaction Nischarin loss enhances GLT-1 surface levels, transport currents, and neuroprotection
Collapse
Affiliation(s)
- Swati Gupta
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Narges Bazargani
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - James Drew
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Jack H. Howden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Souvik Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Sana Al Awabdh
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Hélène Marie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Josef T. Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
- Corresponding author
| |
Collapse
|
24
|
Avalos MP, Guzman AS, Rigoni D, Gorostiza EA, Sanchez MA, Mongi-Bragato B, Garcia-Keller C, Perassi EM, Virgolini MB, Peralta Ramos JM, Iribarren P, Calfa GD, Bollati FA, Cancela LM. Minocycline prevents chronic restraint stress-induced vulnerability to developing cocaine self-administration and associated glutamatergic mechanisms: a potential role of microglia. Brain Behav Immun 2022; 101:359-376. [PMID: 35065197 DOI: 10.1016/j.bbi.2022.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/24/2021] [Accepted: 01/16/2022] [Indexed: 12/14/2022] Open
Abstract
Stressful experience-induced cocaine-related behaviors are associated with a significant impairment of glutamatergic mechanisms in the Nucleus Accumbens core (NAcore). The hallmarks of disrupted glutamate homeostasis following restraint stress are the enduring imbalance of glutamate efflux after a cocaine stimulus and increased basal concentrations of extracellular glutamate attributed to GLT-1 downregulation in the NAcore. Glutamate transmission is tightly linked to microglia functioning. However, the role of microglia in the biological basis of stress-induced addictive behaviors is still unknown. By using minocycline, a potent inhibitor of microglia activation with anti-inflammatory properties, we determined whether microglia could aid chronic restraint stress (CRS)-induced glutamate homeostasis disruption in the NAcore, underpinning stress-induced cocaine self-administration. In this study, adult male rats were restrained for 2 h/day for seven days (day 1-7). From day 16 until completing the experimental protocol, animals received a vehicle or minocycline treatment (30 mg/Kg/12h i.p.). On day 21, animals were assigned to microscopic, biochemical, neurochemical or behavioral studies. We confirm that the CRS-induced facilitation of cocaine self-administration is associated with enduring GLT-1 downregulation, an increase of basal extracellular glutamate and postsynaptic structural plasticity in the NAcore. These alterations were strongly related to the CRS-induced reactive microglia and increased TNF-α mRNA and protein expression, since by administering minocycline, the impaired glutamate homeostasis and the facilitation of cocaine self-administration were prevented. Our findings are the first to demonstrate that minocycline suppresses the CRS-induced facilitation of cocaine self-administration and glutamate homeostasis disruption in the NAcore. A role of microglia is proposed for the development of glutamatergic mechanisms underpinning stress-induced vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Daiana Rigoni
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Ezequiel Axel Gorostiza
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Marianela Adela Sanchez
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Constanza Garcia-Keller
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC-CONICET), Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Miriam Beatriz Virgolini
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Javier María Peralta Ramos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Pablo Iribarren
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Gastón Diego Calfa
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
25
|
Rapid Regulation of Glutamate Transport: Where Do We Go from Here? Neurochem Res 2022; 47:61-84. [PMID: 33893911 PMCID: PMC8542062 DOI: 10.1007/s11064-021-03329-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). A family of five Na+-dependent transporters maintain low levels of extracellular glutamate and shape excitatory signaling. Shortly after the research group of the person being honored in this special issue (Dr. Baruch Kanner) cloned one of these transporters, his group and several others showed that their activity can be acutely (within minutes to hours) regulated. Since this time, several different signals and post-translational modifications have been implicated in the regulation of these transporters. In this review, we will provide a brief introduction to the distribution and function of this family of glutamate transporters. This will be followed by a discussion of the signals that rapidly control the activity and/or localization of these transporters, including protein kinase C, ubiquitination, glutamate transporter substrates, nitrosylation, and palmitoylation. We also include the results of our attempts to define the role of palmitoylation in the regulation of GLT-1 in crude synaptosomes. In some cases, the mechanisms have been fairly well-defined, but in others, the mechanisms are not understood. In several cases, contradictory phenomena have been observed by more than one group; we describe these studies with the goal of identifying the opportunities for advancing the field. Abnormal glutamatergic signaling has been implicated in a wide variety of psychiatric and neurologic disorders. Although recent studies have begun to link regulation of glutamate transporters to the pathogenesis of these disorders, it will be difficult to determine how regulation influences signaling or pathophysiology of glutamate without a better understanding of the mechanisms involved.
Collapse
|
26
|
NRM 2021 Abstract Booklet. J Cereb Blood Flow Metab 2021; 41:11-309. [PMID: 34905986 PMCID: PMC8851538 DOI: 10.1177/0271678x211061050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
27
|
Cieślik P, Siekierzycka A, Radulska A, Płoska A, Burnat G, Brański P, Kalinowski L, Wierońska JM. Nitric Oxide-Dependent Mechanisms Underlying MK-801- or Scopolamine-Induced Memory Dysfunction in Animals: Mechanistic Studies. Int J Mol Sci 2021; 22:12282. [PMID: 34830164 PMCID: PMC8624219 DOI: 10.3390/ijms222212282] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
MK-801, an NMDA receptor antagonist, and scopolamine, a cholinergic receptor blocker, are widely used as tool compounds to induce learning and memory deficits in animal models to study schizophrenia or Alzheimer-type dementia (AD), respectively. Memory impairments are observed after either acute or chronic administration of either compound. The present experiments were performed to study the nitric oxide (NO)-related mechanisms underlying memory dysfunction induced by acute or chronic (14 days) administration of MK-801 (0.3 mg/kg, i.p.) or scopolamine (1 mg/kg, i.p.). The levels of L-arginine and its derivatives, L-citrulline, L-glutamate, L-glutamine and L-ornithine, were measured. The expression of constitutive nitric oxide synthases (cNOS), dimethylaminohydrolase (DDAH1) and protein arginine N-methyltransferases (PMRTs) 1 and 5 was evaluated, and the impact of the studied tool compounds on cGMP production and NMDA receptors was measured. The studies were performed in both the cortex and hippocampus of mice. S-nitrosylation of selected proteins, such as GLT-1, APP and tau, was also investigated. Our results indicate that the availability of L-arginine decreased after chronic administration of MK-801 or scopolamine, as both the amino acid itself as well as its level in proportion to its derivatives (SDMA and NMMA) were decreased. Additionally, among all three methylamines, SDMA was the most abundant in the brain (~70%). Administration of either compound impaired eNOS-derived NO production, increasing the monomer levels, and had no significant impact on nNOS. Both compounds elevated DDAH1 expression, and slight decreases in PMRT1 and PMRT5 in the cortex after scopolamine (acute) and MK-801 (chronic) administration were observed in the PFC, respectively. Administration of MK-801 induced a decrease in the cGMP level in the hippocampus, accompanied by decreased NMDA expression, while increased cGMP production and decreased NMDA receptor expression were observed after scopolamine administration. Chronic MK-801 and scopolamine administration affected S-nitrosylation of GLT-1 transport protein. Our results indicate that the analyzed tool compounds used in pharmacological models of schizophrenia or AD induce changes in NO-related pathways in the brain structures involved in cognition. To some extent, the changes resemble those observed in human samples.
Collapse
Affiliation(s)
- Paulina Cieślik
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (P.C.); (A.S.); (G.B.); (P.B.)
| | - Anna Siekierzycka
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (P.C.); (A.S.); (G.B.); (P.B.)
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland; (A.R.); (A.P.)
| | - Adrianna Radulska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland; (A.R.); (A.P.)
- Biobanking and Biomoleclular Resources Research Infrastructure Consortium Poland (BBMRI.pl), 7 Dębinki Street, 80-211 Gdańsk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland; (A.R.); (A.P.)
- Biobanking and Biomoleclular Resources Research Infrastructure Consortium Poland (BBMRI.pl), 7 Dębinki Street, 80-211 Gdańsk, Poland
| | - Grzegorz Burnat
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (P.C.); (A.S.); (G.B.); (P.B.)
| | - Piotr Brański
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (P.C.); (A.S.); (G.B.); (P.B.)
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland; (A.R.); (A.P.)
- Biobanking and Biomoleclular Resources Research Infrastructure Consortium Poland (BBMRI.pl), 7 Dębinki Street, 80-211 Gdańsk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, University of Technology, 11/12 Narutowicza, 80-233 Gdańsk, Poland
| | - Joanna M. Wierońska
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Kraków, Poland; (P.C.); (A.S.); (G.B.); (P.B.)
| |
Collapse
|
28
|
Ren C, He KJ, Hu H, Zhang JB, Dong LG, Li D, Chen J, Mao CJ, Wang F, Liu CF. Induction of Parkinsonian-Like Changes via Targeted Downregulation of Astrocytic Glutamate Transporter GLT-1 in the Striatum. JOURNAL OF PARKINSONS DISEASE 2021; 12:295-314. [PMID: 34719508 DOI: 10.3233/jpd-212640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous investigations have suggested that decreased expression of glutamate transporter-1 (GLT-1) is involved in glutamate excitotoxicity and contribute to the development of Parkinson's disease (PD), GLT-1 is decreased in animal models of PD. GLT-1 is mainly expressed in astrocytes, and the striatum is a GLT-1-rich brain area. OBJECTIVE The aim was to explore the function and mechanism of astrocytic GLT-1 in PD-like changes. METHODS In the study, PD-like changes and their molecular mechanism in rodents were tested by a behavioral assessment, micro-positron emission tomography/computed tomography (PET/CT), western blotting, immunohistochemical and immunofluorescence staining, and high performance liquid chromatography pre-column derivatization with O-pthaldialdehida after downregulating astrocytic GLT-1 in vivo and in vitro. RESULTS In vivo, after 6 weeks of brain stereotactic injection of adeno-associated virus into the striatum, rats in the astrocytic GLT-1 knockdown group showed poorer motor performance, abnormal gait, and depression-like feature; but no olfactory disorders. The results of micro-PET/CT and western blotting indicated that the dopaminergic system was impaired in astrocytic GLT-1 knockdown rats. Similarly, tyrosine hydroxylase (TH) positive immune-staining in neurons of astrocytic GLT-1 knockdown rats showed deficit in cell count. In vitro, knockdown of astrocytic GLT-1 via RNA interference led to morphological injury of TH-positive neurons, which may be related to the abnormal calcium signal induced by glutamate accumulation after GLT-1 knockdown. Furthermore, the GLT-1 agonist ceftriaxone showed a protective effect on TH-positive neuron impairment. CONCLUSION The present findings may shed new light on the future prevention and treatment of PD based on blocking glutamate excitotoxicity.
Collapse
Affiliation(s)
- Chao Ren
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kai-Jie He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Hua Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin-Bao Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Li-Guo Dong
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dan Li
- Department of Neurology, Suqian First Hospital, Suqian, China
| | - Jing Chen
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Jie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.,Department of Neurology, Suqian First Hospital, Suqian, China.,Department of Neurology, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China.,Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
29
|
Soliman HM, Ghonaim GA, Gharib SM, Chopra H, Farag AK, Hassanin MH, Nagah A, Emad-Eldin M, Hashem NE, Yahya G, Emam SE, Hassan AEA, Attia MS. Exosomes in Alzheimer's Disease: From Being Pathological Players to Potential Diagnostics and Therapeutics. Int J Mol Sci 2021; 22:10794. [PMID: 34639135 PMCID: PMC8509246 DOI: 10.3390/ijms221910794] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Exosomes (EXOs) were given attention as an extracellular vesicle (EV) with a pivotal pathophysiological role in the development of certain neurodegenerative disorders (NDD), such as Parkinson's and Alzheimer's disease (AD). EXOs have shown the potential to carry pathological and therapeutic cargo; thus, researchers have harnessed EXOs in drug delivery applications. EXOs have shown low immunogenicity as natural drug delivery vehicles, thus ensuring efficient drug delivery without causing significant adverse reactions. Recently, EXOs provided potential drug delivery opportunities in AD and promising future clinical applications with the diagnosis of NDD and were studied for their usefulness in disease detection and prediction prior to the emergence of symptoms. In the future, the microfluidics technique will play an essential role in isolating and detecting EXOs to diagnose AD before the development of advanced symptoms. This review is not reiterative literature but will discuss why EXOs have strong potential in treating AD and how they can be used as a tool to predict and diagnose this disorder.
Collapse
Affiliation(s)
- Hagar M. Soliman
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Ghada A. Ghonaim
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Shaza M. Gharib
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aya K. Farag
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Mohamed H. Hassanin
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Abdalrazeq Nagah
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Mahmoud Emad-Eldin
- Department of Clinical, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Nevertary E. Hashem
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Galal Yahya
- Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Sherif E. Emam
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| | - Abdalla E. A. Hassan
- Applied Nucleic Acids Research Center & Chemistry, Faculty of Science, Zagazig 44519, Egypt;
| | - Mohamed S. Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (H.M.S.); (G.A.G.); (S.M.G.); (A.K.F.); (M.H.H.); (A.N.); (N.E.H.); (S.E.E.)
| |
Collapse
|
30
|
Codeluppi SA, Chatterjee D, Prevot TD, Bansal Y, Misquitta KA, Sibille E, Banasr M. Chronic Stress Alters Astrocyte Morphology in Mouse Prefrontal Cortex. Int J Neuropsychopharmacol 2021; 24:842-853. [PMID: 34346493 PMCID: PMC8538896 DOI: 10.1093/ijnp/pyab052] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/25/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Neuromorphological changes are consistently reported in the prefrontal cortex of patients with stress-related disorders and in rodent stress models, but the effects of stress on astrocyte morphology and the potential link to behavioral deficits are relatively unknown. METHODS To answer these questions, transgenic mice expressing green fluorescent protein (GFP) under the glial fibrillary acid protein (GFAP) promotor were subjected to 7, 21, or 35 days of chronic restraint stress (CRS). CRS-induced behavioral effects on anhedonia- and anxiety-like behaviors were measured using the sucrose intake and the PhenoTyper tests, respectively. Prefrontal cortex GFP+ or GFAP+ cell morphology was assessed using Sholl analysis, and associations with behavior were determined using correlation analysis. RESULTS CRS-exposed male and female mice displayed anxiety-like behavior at 7, 21, and 35 days and anhedonia-like behavior at 35 days. Analysis of GFAP+ cell morphology revealed significant atrophy of distal processes following 21 and 35 days of CRS. CRS induced similar decreases in intersections at distal radii for GFP+ cells accompanied by increased proximal processes. In males, the number of intersections at the most distal radius step significantly correlated with anhedonia-like behavior (r = 0.622, P < .05) for GFP+ cells and with behavioral emotionality calculated by z-scoring all behavioral measured deficits (r = -0.667, P < .05). Similar but not significant correlations were observed in females. No correlation between GFP+ cell atrophy with anxiety-like behavior was found. CONCLUSION Chronic stress exposure induces a progressive atrophy of cortical astroglial cells, potentially contributing to maladaptive neuroplastic and behavioral changes associated with stress-related disorders.
Collapse
Affiliation(s)
- Sierra A Codeluppi
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Dipashree Chatterjee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Thomas D Prevot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Yashika Bansal
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Keith A Misquitta
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, Canada,Correspondence: Mounira Banasr, PhD, CAMH, 250 College Street, Toronto, ON M5T 1R8, Canada ()
| |
Collapse
|
31
|
Peterson AR, Garcia TA, Cullion K, Tiwari-Woodruff SK, Pedapati EV, Binder DK. Targeted overexpression of glutamate transporter-1 reduces seizures and attenuates pathological changes in a mouse model of epilepsy. Neurobiol Dis 2021; 157:105443. [PMID: 34246771 DOI: 10.1016/j.nbd.2021.105443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/06/2023] Open
Abstract
Astrocytic glutamate transporters are crucial for glutamate homeostasis in the brain, and dysregulation of these transporters can contribute to the development of epilepsy. Glutamate transporter-1 (GLT-1) is responsible for the majority of glutamate uptake in the dorsal forebrain and has been shown to be reduced at epileptic foci in patients and preclinical models of temporal lobe epilepsy (TLE). Current antiepileptic drugs (AEDs) work primarily by targeting neurons directly through suppression of excitatory neurotransmission or enhancement of inhibitory neurotransmission, which can lead to both behavioral and psychiatric side effects. This study investigates the therapeutic capacity of astrocyte-specific AAV-mediated GLT-1 expression in the intrahippocampal kainic acid (IHKA) model of TLE. In this study, we used Western blot analysis, immunohistochemistry, and long-term-video EEG monitoring to demonstrate that cell-type-specific upregulation of GLT-1 in astrocytes is neuroprotective at early time points during epileptogenesis, reduces seizure frequency and total time spent in seizures, and eliminates large behavioral seizures in the IHKA model of epilepsy. Our findings suggest that targeting glutamate uptake is a promising therapeutic strategy for the treatment of epilepsy.
Collapse
Affiliation(s)
- Allison R Peterson
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, School of Medicine, University of California, Riverside, CA, USA
| | - Terese A Garcia
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, School of Medicine, University of California, Riverside, CA, USA
| | - Kyle Cullion
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, School of Medicine, University of California, Riverside, CA, USA
| | - Ernest V Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Devin K Binder
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, School of Medicine, University of California, Riverside, CA, USA.
| |
Collapse
|
32
|
Lyu S, Guo Y, Zhang L, Tang G, Li R, Yang J, Gao S, Li W, Liu J. Downregulation of astroglial glutamate transporter GLT-1 in the lateral habenula is associated with depressive-like behaviors in a rat model of Parkinson's disease. Neuropharmacology 2021; 196:108691. [PMID: 34197892 DOI: 10.1016/j.neuropharm.2021.108691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 01/11/2023]
Abstract
Recent studies show that neuron-glial communication plays an important role in neurological diseases. Particularly, dysfunction of astroglial glutamate transporter GLT-1 has been involved in various neuropsychiatric disorders, including Parkinson's disease (PD) and depression. Our previous studies indicated hyperactivity of neurons in the lateral habenula (LHb) of hemiparkinsonian rats with depressive-like behaviors. Thus, we hypothesized that impaired expression or function of GLT-1 in the LHb might be a potential contributor to LHb hyperactivity, which consequently induces PD-related depression. In the study, unilateral lesions of the substantia nigra pars compacta (SNc) by 6-hydroxydopamine in rats induced depressive-like behaviors and resulted in neuronal hyperactivity as well as increased glutamate levels in the LHb compared to sham-lesioned rats. Intra-LHb injection of GLT-1 inhibitor WAY-213613 induced the depressive-like behaviors in both groups, but the dose producing behavioral effects in the lesioned rats was lower than that of sham-lesioned rats. In the two groups of rats, WAY-213613 increased the firing rate of LHb neurons and extracellular levels of glutamate, and these excitatory effects in the lesioned rats lasted longer than those in sham-lesioned rats. The functional changes of the GLT-1 which primarily expresses in astrocytes in the LHb may attribute to its downregulation after degeneration of the nigrostriatal pathway. Bioinformatics analysis showed that GLT-1 is correlated with various biomarkers of PD and depression risks. Collectively, our study suggests that astroglial GLT-1 in the LHb regulates the firing activity of the neurons, whereupon its downregulation and dysfunction are closely associated with PD-related depression.
Collapse
Affiliation(s)
- Shuxuan Lyu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yuan Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Li Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Guoyi Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ruotong Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jie Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Shasha Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Wenjuan Li
- Department of Rehabilitation Medicine, The Second Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
33
|
Ohashi N, Terashima T, Katagi M, Nakae Y, Okano J, Suzuki Y, Kojima H. GLT1 gene delivery based on bone marrow-derived cells ameliorates motor function and survival in a mouse model of ALS. Sci Rep 2021; 11:12803. [PMID: 34140581 PMCID: PMC8211665 DOI: 10.1038/s41598-021-92285-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an intractable neurodegenerative disease. CD68-positive bone marrow (BM)-derived cells (BMDCs) accumulate in the pathological lesion in the SOD1(G93A) ALS mouse model after BM transplantation (BMT). Therefore, we investigated whether BMDCs can be applied as gene carriers for cell-based gene therapy by employing the accumulation of BMDCs. In ALS mice, YFP reporter signals were observed in 12-14% of white blood cells (WBCs) and in the spinal cord via transplantation of BM after lentiviral vector (LV) infection. After confirmation of gene transduction by LV with the CD68 promoter in 4-7% of WBCs and in the spinal cord of ALS mice, BM cells were infected with LVs expressing glutamate transporter (GLT) 1 that protects neurons from glutamate toxicity, driven by the CD68 promoter, which were transplanted into ALS mice. The treated mice showed improvement of motor behaviors and prolonged survival. Additionally, interleukin (IL)-1β was significantly suppressed, and IL-4, arginase 1, and FIZZ were significantly increased in the mice. These results suggested that GLT1 expression by BMDCs improved the spinal cord environment. Therefore, our gene therapy strategy may be applied to treat neurodegenerative diseases such as ALS in which BMDCs accumulate in the pathological lesion by BMT.
Collapse
Affiliation(s)
- Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan.
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Yuki Nakae
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| | - Junko Okano
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Yoshihisa Suzuki
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
34
|
Dagdelen DN, Akkulak A, Donmez Yalcin G. The investigation of glutamate transporter 1 (GLT-1) degradation pathway in glioblastoma cells. Mol Biol Rep 2021; 48:3495-3502. [PMID: 34003424 DOI: 10.1007/s11033-021-06407-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/08/2021] [Indexed: 11/25/2022]
Abstract
Glioblastoma multiform is a primary brain tumor derived from glial cells. The aim of this study is to investigate how glutamate metabolism is regulated by glutamate transporter 1 (GLT-1) degradation pathway in glioblastoma and glial cell lines. The protein expression levels of GLT-1, total ubiquitin, protein kinase C (PKC) proteins involved in the GLT-1 degradation pathway were measured by the western blot technique. Additionally, in glial and glioblastoma cells, the level of glutamate accumulated in the medium and the lysates was measured with the glutamate assay. GLT-1 protein expression was increased significantly in glioblastoma cells. The expression levels of the PKC protein and total ubiquitin were found to be decreased in glioblastoma cells although not significantly. The glutamate accumulated in the medium and lysates of glioblastoma cells is reduced compared to glial cells. Further research regarding excitotoxicity in glioblastoma focusing on GLT-1 degradation or activation pathway may create new opportunities of drug and treatment development.
Collapse
Affiliation(s)
- Duriye Nur Dagdelen
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Aysenur Akkulak
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey
| | - Gizem Donmez Yalcin
- Department of Medical Biology, Faculty of Medicine, Aydin Adnan Menderes University, Aydin, Turkey.
| |
Collapse
|
35
|
Michaluk P, Heller JP, Rusakov DA. Rapid recycling of glutamate transporters on the astroglial surface. eLife 2021; 10:e64714. [PMID: 33860761 PMCID: PMC8079145 DOI: 10.7554/elife.64714] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
Glutamate uptake by astroglial transporters confines excitatory transmission to the synaptic cleft. The efficiency of this mechanism depends on the transporter dynamics in the astrocyte membrane, which remains poorly understood. Here, we visualise the main glial glutamate transporter GLT1 by generating its pH-sensitive fluorescent analogue, GLT1-SEP. Fluorescence recovery after photobleaching-based imaging shows that 70-75% of GLT1-SEP dwell on the surface of rat brain astroglia, recycling with a lifetime of ~22 s. Genetic deletion of the C-terminus accelerates GLT1-SEP membrane turnover while disrupting its surface pattern, as revealed by single-molecule localisation microscopy. Excitatory activity boosts surface mobility of GLT1-SEP, involving its C-terminus, metabotropic glutamate receptors, intracellular Ca2+, and calcineurin-phosphatase activity, but not the broad-range kinase activity. The results suggest that membrane turnover, rather than lateral diffusion, is the main 'redeployment' route for the immobile fraction (20-30%) of surface-expressed GLT1. This finding reveals an important mechanism helping to control extrasynaptic escape of glutamate.
Collapse
Affiliation(s)
- Piotr Michaluk
- UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- BRAINCITY, Laboratory of Neurobiology, Nencki Institute of Experimental Biology PASWarsawPoland
| | - Janosch Peter Heller
- UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- School of Biotechnology and National Institute for Cellular Biotechnology (NICB), Dublin City UniversityGlasnevinIreland
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| |
Collapse
|
36
|
Martinez D, Kline DD. The role of astrocytes in the nucleus tractus solitarii in maintaining central control of autonomic function. Am J Physiol Regul Integr Comp Physiol 2021; 320:R418-R424. [PMID: 33439770 PMCID: PMC8238142 DOI: 10.1152/ajpregu.00254.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022]
Abstract
The nucleus tractus solitarii (nTS) is the first central site for the termination and integration of autonomic and respiratory sensory information. Sensory afferents terminating in the nTS as well as the embedded nTS neurocircuitry release and utilize glutamate that is critical for maintenance of baseline cardiorespiratory parameters and initiating cardiorespiratory reflexes, including those activated by bouts of hypoxia. nTS astrocytes contribute to synaptic and neuronal activity through a variety of mechanisms, including gliotransmission and regulation of glutamate in the extracellular space via membrane-bound transporters. Here, we aim to highlight recent evidence for the role of astrocytes within the nTS and their regulation of autonomic and cardiorespiratory processes under normal and hypoxic conditions.
Collapse
Affiliation(s)
- Diana Martinez
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
37
|
Brewster AL. Relationship Status Update on Astrocytic VEGFR-3 and mTOR Signaling: It's Complicated. Epilepsy Curr 2021; 21:117-119. [PMID: 34025288 PMCID: PMC8010874 DOI: 10.1177/1535759720988541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Vascular Endothelial Growth Factor Receptor-3 Regulates Astroglial Glutamate Transporter-1 Expression via mTOR Activation in Reactive Astrocytes Following Pilocarpine-Induced Status Epilepticus Jeong KH, Cho KO, Lee MY, Kim SY, Kim WJ. Glia. 2021;69(2):296-309. doi:10.1002/glia.23897. PMID: 32835451. Recent evidence has shown that the vascular endothelial growth factor (VEGF) system plays a crucial role in several neuropathological processes. We previously reported an upregulation of VEGF-C and its receptor, VEGFR-3, in reactive astrocytes after the onset of status epilepticus (SE). However, it remains unknown, which molecules act as downstream signals following VEGFR-3 upregulation and are involved in astrogliosis after SE. Therefore, we investigated whether VEGFR-3 upregulation within reactive astrocytes is associated with the activation of mammalian target of rapamycin (mTOR) signaling, which we confirmed by assaying for the phosphorylated form of S6 protein (pS6), and whether VEGFR-3-mediated mTOR activation induces astroglial glutamate transporter-1 (GLT-1) expression in the hippocampus after pilocarpine-induced SE. We found that spatiotemporal expression of pS6 was consistent with VEGFR-3 expression in the hippocampus after SE and that both pS6 and VEGFR-3 were highly expressed in SE-induced reactive astrocytes. Treatment with the mTOR inhibitor rapamycin decreased astroglial VEGFR-3 expression and GLT-1 expression after SE. Treatment with a selective inhibitor for VEGFR-3 attenuated astroglial pS6 expression as well as suppressed GLT-1 expression and astroglial reactivity in the hippocampus after SE. These findings demonstrate that VEGFR-3-mediated mTOR activation could contribute to the regulation of GLT-1 expression in reactive astrocytes during the subacute phase of epilepsy. In conclusion, the present study suggests that VEGFR-3 upregulation in reactive astrocytes may play a role in preventing hyperexcitability induced by continued seizure activity.
Collapse
|
38
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
39
|
Perez-Jimenez E, Viana R, Muñoz-Ballester C, Vendrell-Tornero C, Moll-Diaz R, Garcia-Gimeno MA, Sanz P. Endocytosis of the glutamate transporter 1 is regulated by laforin and malin: Implications in Lafora disease. Glia 2020; 69:1170-1183. [PMID: 33368637 DOI: 10.1002/glia.23956] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 01/13/2023]
Abstract
Lafora disease (LD) is a fatal rare type of progressive myoclonus epilepsy that appears during early adolescence. The disease is caused by mutations in EPM2A or EPM2B genes, which encode laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase, respectively. Although the exact roles of laforin and malin are still not well understood, it is known that they work as a complex in which laforin recruits targets that will be ubiquitinated by malin. Recently, we suggested that the type of epilepsy that accompanies LD could be due to deficiencies in the function of the astrocytic glutamate transporter GLT-1. We described that astrocytes from LD mouse models presented decreased levels of GLT-1 at the plasma membrane, leading to increased levels of glutamate in the brain parenchyma. In this work, we present evidence indicating that in the absence of a functional laforin/malin complex (as in LD cellular models) there is an alteration in the ubiquitination of GLT-1, which could be the cause of the reduction in the levels of GLT-1 at the plasma membrane. On the contrary, overexpression of the laforin/malin complex promotes the retention of GLT-1 at the plasma membrane. This retention may be due to the direct ubiquitination of GLT-1 and/or to an opposite effect of this complex on the dynamics of the Nedd4.2-mediated endocytosis of the transporter. This work, therefore, presents new pieces of evidence on the regulation of GLT-1 by the laforin/malin complex, highlighting its value as a therapeutic target for the amelioration of the type of epilepsy that accompanies LD.
Collapse
Affiliation(s)
- Eva Perez-Jimenez
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Rosa Viana
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Carmen Muñoz-Ballester
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Carlos Vendrell-Tornero
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Raquel Moll-Diaz
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | | | - Pascual Sanz
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| |
Collapse
|
40
|
Cheng H, Zhang L, Xia F, Jin L, Liu S, Ren H, Zhu C, Ji Q, Tang J. Astrocytic NDRG2 is critical in the maintenance of neuropathic pain. Brain Behav Immun 2020; 89:300-313. [PMID: 32688030 DOI: 10.1016/j.bbi.2020.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Activation of astrocytes and abnormal synaptic glutamate metabolism are closely associated with the induction and maintenance of neuropathic pain (NP), but the exact mechanism underlying this association remains unclear. N-myc downstream-regulated gene 2 (NDRG2), a novel tumor-suppressor protein and stress-response gene, is involved in the pathogenesis of several neurodegenerative diseases. However, its role in nociceptive transduction has rarely been investigated. Here, we found that NDRG2, which was mainly expressed in the astrocytes in the central nervous system (CNS), was increased in the spinal cord of a spinal nerve ligation (SNL) rat model for NP. Suppression of NDRG2 by intrathecal injection of an NDRG2-RNAi-adenovirus significantly alleviated SNL-induced mechanical and thermal hypersensitivity, as well as elevated astrocytic glutamate transporter 1 (GLT-1) expression and downregulated pro-inflammatory cytokine levels, in the spinal dorsal horn of rats on Day 10 after SNL. Furthermore, in lipopolysaccharide (LPS)-stimulated primary astrocytic cultures derived from neonatal rats, inhibition of NDRG2 significantly reversed both the LPS-induced activation of astrocytes and decreased expression of GLT-1. By contrast, overexpression of NDRG2 by an adenoviral vector carrying NDRG2 resulted in astrocytic activation, aberrant glutamatergic neurotransmission, and spontaneous nociceptive responses in rats. Intrathecal injection of AG490, which is an inhibitor of the Janus tyrosine kinase and signal transducer and activator of the transcription 3 (JAK/STAT3) signaling pathway, significantly attenuated both mechanical and thermal hyperalgesia, as well as inhibited reactive astrocytes and restored normal expression levels of astrocytic GLT-1, in the spinal dorsal horn of NDRG2-overexpression rats. In conclusion, spinal astrocytic NDRG2 is critical in the maintenance of NP. Moreover, NDRG2 modulates astrocytic activation and inflammatory responses via regulating GLT-1 expression through the JAK/STAT3 signaling pathway. Our findings suggested that NDRG2 could be a novel therapeutic target for the treatment of NP.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fei Xia
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Jin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Suting Liu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hongwei Ren
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chao Zhu
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Ji
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Jun Tang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
41
|
Gunes ZI, Kan VWY, Ye X, Liebscher S. Exciting Complexity: The Role of Motor Circuit Elements in ALS Pathophysiology. Front Neurosci 2020; 14:573. [PMID: 32625051 PMCID: PMC7311855 DOI: 10.3389/fnins.2020.00573] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease, characterized by the degeneration of both upper and lower motor neurons. Despite decades of research, we still to date lack a cure or disease modifying treatment, emphasizing the need for a much-improved insight into disease mechanisms and cell type vulnerability. Altered neuronal excitability is a common phenomenon reported in ALS patients, as well as in animal models of the disease, but the cellular and circuit processes involved, as well as the causal relevance of those observations to molecular alterations and final cell death, remain poorly understood. Here, we review evidence from clinical studies, cell type-specific electrophysiology, genetic manipulations and molecular characterizations in animal models and culture experiments, which argue for a causal involvement of complex alterations of structure, function and connectivity of different neuronal subtypes within the cortical and spinal cord motor circuitries. We also summarize the current knowledge regarding the detrimental role of astrocytes and reassess the frequently proposed hypothesis of glutamate-mediated excitotoxicity with respect to changes in neuronal excitability. Together, these findings suggest multifaceted cell type-, brain area- and disease stage- specific disturbances of the excitation/inhibition balance as a cardinal aspect of ALS pathophysiology.
Collapse
Affiliation(s)
- Zeynep I Gunes
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig Maximilians University Munich, Munich, Germany.,Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Vanessa W Y Kan
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig Maximilians University Munich, Munich, Germany.,Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | - XiaoQian Ye
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany.,Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Munich, Germany.,Biomedical Center, Ludwig Maximilians University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
42
|
Martinez D, Rogers RC, Hasser EM, Hermann GE, Kline DD. Loss of excitatory amino acid transporter restraint following chronic intermittent hypoxia contributes to synaptic alterations in nucleus tractus solitarii. J Neurophysiol 2020; 123:2122-2135. [PMID: 32347148 PMCID: PMC7311725 DOI: 10.1152/jn.00766.2019] [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: 01/16/2023] Open
Abstract
Peripheral viscerosensory afferent signals are transmitted to the nucleus tractus solitarii (nTS) via release of glutamate. Following release, glutamate is removed from the extrasynaptic and synaptic cleft via excitatory amino acid transporters (EAATs), thus limiting glutamate receptor activation or over activation, and maintaining its working range. We have shown that EAAT block with the antagonist threo-β-benzyloxyaspartic acid (TBOA) depolarized nTS neurons and increased spontaneous excitatory postsynaptic current (sEPSC) frequency yet reduced the amplitude of afferent (TS)-evoked EPSCs (TS-EPSCs). Interestingly, chronic intermittent hypoxia (CIH), a model of obstructive sleep apnea (OSA), produces similar synaptic responses as EAAT block. We hypothesized EAAT expression or function are downregulated after CIH, and this reduction in glutamate removal contributes to the observed neurophysiological responses. To test this hypothesis, we used brain slice electrophysiology and imaging of glutamate release and TS-afferent Ca2+ to compare nTS properties of rats exposed to 10 days of normoxia (Norm; 21%O2) or CIH. Results show that EAAT blockade with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]-amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) in Norm caused neuronal depolarization, generation of an inward current, and increased spontaneous synaptic activity. The latter augmentation was eliminated by inclusion of tetrodotoxin in the perfusate. TS stimulation during TFB-TBOA also elevated extracellular glutamate and decreased presynaptic Ca2+ and TS-EPSC amplitude. In CIH, the effects of EAAT block are eliminated or attenuated. CIH reduced EAAT expression in nTS, which may contribute to the attenuated function seen in this condition. Therefore, CIH reduces EAAT influence on synaptic and neuronal activity, which may lead to the physiological consequences seen in OSA and CIH.NEW & NOTEWORTHY Removal of excitatory amino acid transporter (EAAT) restraint increases spontaneous synaptic activity yet decreases afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. In the chronic intermittent hypoxia model of obstructive sleep apnea, this restraint is lost due to reduction in EAAT expression and function. Thus EAATs are important in controlling elevated glutamatergic signaling, and loss of such control results in maladaptive synaptic signaling.
Collapse
Affiliation(s)
- Diana Martinez
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | - Eileen M. Hasser
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | | | - David D. Kline
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
43
|
Hammad AM, Sari Y. Effects of Cocaine Exposure on Astrocytic Glutamate Transporters and Relapse-Like Ethanol-Drinking Behavior in Male Alcohol-Preferring Rats. Alcohol Alcohol 2020; 55:254-263. [PMID: 32099993 PMCID: PMC7171926 DOI: 10.1093/alcalc/agaa010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/14/2022] Open
Abstract
AIM Glutamate has been considered as neurotransmitter that is critical in triggering relapse to drugs of abuse, including ethanol and cocaine. Extracellular glutamate concentrations are tightly regulated by several mechanisms, including reuptake through glutamate transporters. Glutamate transporter type 1 (GLT-1) is responsible for clearing the majority of extracellular glutamate. The astrocytic cystine/glutamate antiporter (xCT) regulates also glutamate homeostasis. In this study, we investigated the effects of cocaine exposure and ampicillin/sulbactam (AMP/SUL), a β-lactam antibiotic known to upregulate GLT-1 and xCT, on relapse-like ethanol intake and the expression of astrocytic glutamate transporters in mesocorticolimbic brain regions. METHODS Male alcohol-preferring (P) rats had free access to ethanol for 5 weeks. On Week 6, rats were exposed to either cocaine (20 mg/kg, i.p.) or saline for 12 consecutive days. Ethanol bottles were then removed for 7 days; during the last 5 days, either AMP/SUL (100 or 200 mg/kg, i.p.) or saline was administered to the P rats. Ethanol bottles were reintroduced, and ethanol intake was measured for 4 days. RESULTS Cocaine exposure induced an alcohol deprivation effect (ADE), which was associated in part by a decrease in the expression of GLT-1 and xCT in the nucleus accumbens (NAc) core. AMP/SUL (100 mg/kg, i.p.) attenuated the ADE, while AMP/SUL (200 mg/kg, i.p.) reduced ethanol intake during 4 days of ethanol re-exposure and upregulated GLT-1 and xCT expression in the NAc core, NAc shell and dorsomedial prefrontal cortex (dmPFC). CONCLUSION This study suggests that these astrocytic glutamate transporters might be considered as potential targets for the treatment of polysubstance abuse.
Collapse
Affiliation(s)
- Alaa M Hammad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave, Toledo, OH, USA
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130 Amman, 11733, Jordan
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, 3000 Arlington Ave, Toledo, OH, USA
| |
Collapse
|
44
|
Matott MP, Hasser EM, Kline DD. Sustained Hypoxia Alters nTS Glutamatergic Signaling and Expression and Function of Excitatory Amino Acid Transporters. Neuroscience 2020; 430:131-140. [PMID: 32032667 PMCID: PMC7560968 DOI: 10.1016/j.neuroscience.2020.01.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/11/2020] [Accepted: 01/23/2020] [Indexed: 01/16/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the nucleus tractus solitarii (nTS) and mediates chemoreflex function during periods of low oxygen (i.e. hypoxia). We have previously shown that nTS excitatory amino acid transporters (EAATs), specifically EAAT-2, located on glia modulate neuronal activity, cardiorespiratory and chemoreflex function under normal conditions via its tonic uptake of extracellular glutamate. Chronic sustained hypoxia (SH) elevates nTS synaptic transmission and chemoreflex function. The goal of this study was to determine the extent to which glial EAAT-2 contributes to SH-induced nTS synaptic alterations. To do so, male Sprague-Dawley rats (4-7 weeks) were exposed to either 1, 3, or 7 days of SH (10% O2, 24 h/day) and compared to normoxic controls (21% O2, 24 h/day, i.e., 0 days SH). After which, the nTS was harvested for patch clamp electrophysiology, quantitative real-time PCR, immunohistochemistry and immunoblots. SH induced time- and parameter-dependent increases in excitatory postsynaptic currents (EPSCs). TS-evoked EPSC amplitude increased after 1D SH which returned at 3D and 7D SH. Spontaneous EPSC frequency increased only after 3D SH, which returned to normoxic levels at 7D SH. EPSC enhancement occurred primarily by presynaptic mechanisms. Inhibition of EAAT-2 with dihydrokainate (DHK, 300 µM) did not alter EPSCs following 1D SH but induced depolarizing inward currents (Ihold). After 3D SH, DHK decreased TS-EPSC amplitude yet its resulting Ihold was eliminated. EAAT-2 mRNA and protein increased after 3D and 7D SH, respectively. These data suggest that SH alters the expression and function of EAAT-2 which may have a neuroprotective effect.
Collapse
Affiliation(s)
- Michael P Matott
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA
| | - Eileen M Hasser
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA
| | - David D Kline
- Dept. of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
45
|
Martinez D, Rogers RC, Hermann GE, Hasser EM, Kline DD. Astrocytic glutamate transporters reduce the neuronal and physiological influence of metabotropic glutamate receptors in nucleus tractus solitarii. Am J Physiol Regul Integr Comp Physiol 2020; 318:R545-R564. [PMID: 31967862 PMCID: PMC7099463 DOI: 10.1152/ajpregu.00319.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Astrocytic excitatory amino acid transporters (EAATs) are critical to restraining synaptic and neuronal activity in the nucleus tractus solitarii (nTS). Relief of nTS EAAT restraint generates two opposing effects, an increase in neuronal excitability that reduces blood pressure and breathing and an attenuation in afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. Although the former is due, in part, to activation of ionotropic glutamate receptors, there remains a substantial contribution from another unidentified glutamate receptor. In addition, the mechanism(s) by which EAAT inhibition reduced TS-EPSC amplitude is unknown. Metabotropic glutamate receptors (mGluRs) differentially modulate nTS excitability. Activation of group I mGluRs on nTS neuron somas leads to depolarization, whereas group II/III mGluRs on sensory afferents decrease TS-EPSC amplitude. Thus we hypothesize that EAATs control postsynaptic excitability and TS-EPSC amplitude via restraint of mGluR activation. To test this hypothesis, we used in vivo recording, brain slice electrophysiology, and imaging of glutamate release and TS-afferent Ca2+. Results show that EAAT blockade in the nTS with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) induced group I mGluR-mediated depressor, bradycardic, and apneic responses that were accompanied by neuronal depolarization, elevated discharge, and increased spontaneous synaptic activity. Conversely, upon TS stimulation TFB-TBOA elevated extracellular glutamate to decrease presynaptic Ca2+ and TS-EPSC amplitude via activation of group II/III mGluRs. Together, these data suggest an important role of EAATs in restraining mGluR activation and overall cardiorespiratory function.
Collapse
Affiliation(s)
- Diana Martinez
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | | | - Eileen M. Hasser
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri,2Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - David D. Kline
- 1Department of Biomedical Sciences and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|