1
|
Lord B, Simavorian S, Fraser I, Welty N, Wyatt R, Pritchard R, Fletcher L, Van Der Linde H, Bounkhoun D, Libiger O, Maher M, Drevets W, Bischoff F, Bonaventure P, Neff RA. Pharmacological characterisation of JNJ-78911118, a novel, centrally-penetrant, selective GluN2A antagonist. Br J Pharmacol 2025. [PMID: 40361296 DOI: 10.1111/bph.70069] [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: 08/12/2024] [Revised: 03/01/2025] [Accepted: 03/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Non-selective NMDA receptor antagonism produces rapid symptom improvement in treatment-resistant depression; however, associated side effects necessitate medical oversight during administration. We hypothesised that selective GluN2A antagonism could provide similar efficacy with an improved side effect profile. Here, we report the pharmacology of JNJ-78911118, a brain-penetrant, GluN2A selective antagonist. EXPERIMENTAL APPROACH JNJ-78911118 pharmacology and mechanism of action was characterised in vitro using fluorescence, voltage clamp and radioligand binding assays. Target engagement was measured using ex vivo receptor autoradiography, and effects on rat prefrontal cortex monoamine levels were measured using microdialysis. Synaptogenesis assays and patch clamp studies were used to demonstrate effects on synaptic plasticity. Cardiovascular safety and neurotoxicity were assessed in rats. KEY RESULTS JNJ-78911118 blocked GluN1/2A receptors with an IC50 of 44 nM and showed selectivity against GluN1/2B, 2C and 2D receptors. Systemic administration produced concentration-dependent receptor occupancy, increased prefrontal cortex monoamine levels in wild type, but not in GluN2A knockout mice, and blocked theta burst induced LTP in the hippocampus. In addition, it produced increases in dendritic complexity and synapse number in vitro, and increased mEPSC frequency in rat cortical neurons in vivo. In rat toxicological studies, no Olney's lesions were observed, but acute increases in heart rate and blood pressure were detected. CONCLUSIONS AND IMPLICATIONS JNJ-78911118 is a potent and selective GluN2A antagonist that reproduces the effect of known rapidly acting antidepressants (RAADs) on neurotransmitter levels and synaptic plasticity. This molecule is a powerful in vivo tool that will enhance understanding of GluN2A biology.
Collapse
Affiliation(s)
- Brian Lord
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Sirak Simavorian
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ian Fraser
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Natalie Welty
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ryan Wyatt
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Rory Pritchard
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Lauren Fletcher
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Henk Van Der Linde
- CoE for Cardiovascular Safety Research, Janssen Research and Development, LLC, Beerse, Belgium
| | - Dominic Bounkhoun
- Preclinical Sciences and Translational Safety, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ondrej Libiger
- Statistics and Decision Sciences, Janssen Research and Development, LLC, San Diego, California, USA
| | - Michael Maher
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Wayne Drevets
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - François Bischoff
- Discovery, Product Development and Supply, Janssen Research and Development, LLC, Beerse, Belgium
| | - Pascal Bonaventure
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Robert A Neff
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| |
Collapse
|
2
|
Xie L, Sheehy RN, Muneer A, Xiong Y, Wrobel JA, Zhang F, Park KS, Velez J, Liu J, Luo YJ, Asrican B, Dong P, Li YD, Damian C, Quintanilla L, Li Y, Xu C, Deshmukh M, Coleman LG, Ming GL, Song H, Wen Z, Jin J, Song J, Chen X. Development of a brain-penetrant G9a methylase inhibitor to target Alzheimer's disease-associated proteopathology. Nat Commun 2025; 16:4222. [PMID: 40328756 PMCID: PMC12056044 DOI: 10.1038/s41467-025-59128-z] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Current Aβ-targeting therapeutics for Alzheimer's disease (AD) only slow cognitive decline due to poor understanding of AD pathogenesis. Here we describe a mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of hippocampal proteins associated with AD pathology. Correspondingly, we developed a brain-penetrant inhibitor of G9a, MS1262, which restored both age-related learning & memory and noncognitive functions in multiple AD mouse models. Further, comparison of AD pathology-correlated mouse proteomes with those of AD patients found G9a regulates pathological pathways that promote Aβ and neurofibrillary tangles. This mouse-to-human overlap of G9a regulated AD-associated pathologic proteins supports at the molecular level the efficacy of targeting G9a translational mechanism for treating AD patients. Additionally, MS1262 treatment reversed the AD-characteristic expression or phosphorylation of multiple clinically validated biomarkers of AD that have the potential to be used for early-stage AD diagnosis and companion diagnosis of individualized drug effects.
Collapse
Affiliation(s)
- Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adil Muneer
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Velez
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brent Asrican
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ping Dong
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corina Damian
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luis Quintanilla
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongyi Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chongchong Xu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Kipiani EE, Burjanadze MA, Dashniani MG, Chkhikvishvili NC, Naneishvili TL, Chighladze MR, Nozadze BG, Beselia GV. Medial septum deep brain stimulation enhances memory and hippocampal neurogenesis in the D-galactose induced rat model of aging: behavioral and immunohistochemical study. Exp Brain Res 2025; 243:95. [PMID: 40100345 DOI: 10.1007/s00221-025-07051-6] [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: 10/17/2024] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
One of the cardinal features of aging is brain aging, which manifests itself in impaired cognitive functions. Experimental data suggest that deep brain stimulation (DBS) can improve memory functions when stimulating specific brain regions. In present study we tested the hypothesis that medial septum (MS) DBS enhances memory function by modulating the hippocampal neurogenesis in the D-galactose (D-gal) induced rat model of aging. Rats were randomly assigned to four experimental groups: (1) control, (2) administration of D-gal, (3) administration of D-gal and electrode implantation and (4) administration of D-gal, electrode implantation and stimulation. Our results showed that MS DBS significantly enhanced the memory functions in an animal model of aging induced by D-gal administration, which impaired long-term spatial memory in the Morris water maze and impaired spatial and object novelty recognition memory in the open field. The immunohistochemical studies showed that in the Dentate Gyrus (DG) of rats with D-gal administration or D-gal combined with electrode implantation, the number of NeuN (neuronal nuclear antigen) or Doublecortin-immunopositive cells decreased (Doublecortin - a biomarker for the post-mitotic phase of cells); MS stimulation increases the number of these cells in the DG to levels comparable to the control group. Thus, MS-DBS restores the level of hippocampal neurogenesis. The present data demonstrate for the first time that chronic DBS of the MS restores memory functions in a D-gal-induced animal model of aging, and that one of the important underlying mechanisms is mediated by enhanced neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Ekaterine E Kipiani
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
- Teaching University Geomedi LLC, King Solomon II str,4, Tbilisi, 0114, Georgia
| | - Maia A Burjanadze
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Manana G Dashniani
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Nino C Chkhikvishvili
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Temur L Naneishvili
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Mariam R Chighladze
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Barbare G Nozadze
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia
| | - Gela V Beselia
- Department of Behavior and Cognitive Function, Ivane Beritashvili Center of Experimental Biomedicine, Gotua14, Tbilisi, 0160, Georgia.
- Department of Physiology and Pharmacology, Petre Shotadze Tbilisi Medical Academy, Ketevan Dedofali Ave51/2, Tbilisi, 0144, Georgia.
| |
Collapse
|
4
|
Salarvandian S, Digaleh H, Khodagholi F, Javadpour P, Asadi S, Zaman AAO, Dargahi L. Harmonic activity of glutamate dehydrogenase and neuroplasticity: The impact on aging, cognitive dysfunction, and neurodegeneration. Behav Brain Res 2025; 480:115399. [PMID: 39675635 DOI: 10.1016/j.bbr.2024.115399] [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: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
In recent years, glutamate has attracted significant attention for its roles in various brain processes. However, one of its key regulators, glutamate dehydrogenase (GDH), remains understudied despite its pivotal role in several biochemical pathways. Dysfunction or dysregulation of GDH has been implicated in aging and various neurological disorders, such as Alzheimer's disease and Parkinson's disease. In this review, the impact of GDH on aging, cognitive impairment, and neurodegenerative conditions, as exemplars of the phenomena that may affected by neuroplasticity, has been reviewed. Despite extensive research on synaptic plasticity, the precise influence of GDH on brain structure and function remains undiscovered. This review of existing literature on GDH and neuroplasticity reveals diverse and occasionally conflicting effects. Future research endeavors should aim to describe the precise mechanisms by which GDH influences neuroplasticity (eg. synaptic plasticity and neurogenesis), particularly in the context of human aging and disease progression. Studies on GDH activity have been limited by factors such as insufficient sample sizes and varying experimental conditions. Researchers should focus on investigating the molecular mechanisms by which GDH modulates neuroplasticity, utilizing various animal strains and species, ages, sexes, GDH isoforms, brain regions, and cell types. Understanding GDH's role in neuroplasticity may offer innovative therapeutic strategies for neurodegenerative and psychiatric diseases, potentially slowing the aging process and promoting brain regeneration.
Collapse
Affiliation(s)
- Shakiba Salarvandian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Digaleh
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ali Orang Zaman
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Stokes EG, Vasquez JJ, Azouz G, Nguyen M, Tierno A, Zhuang Y, Galinato VM, Hui M, Toledano M, Tyler I, Shi X, Hunt RF, Aoto J, Beier KT. Cationic peptides cause memory loss through endophilin-mediated endocytosis. Nature 2025; 638:479-489. [PMID: 39814881 DOI: 10.1038/s41586-024-08413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 11/14/2024] [Indexed: 01/18/2025]
Abstract
The zeta inhibitory peptide (ZIP) interferes with memory maintenance and long-term potentiation (LTP)1 when administered to mice. However, mice lacking its putative target, protein kinase PKMζ, exhibit normal learning and memory as well as LTP2,3, making the mechanism of ZIP unclear. Here we show that ZIP disrupts LTP by removing surface AMPA receptors through its cationic charge alone. This effect requires endophilin-A2-mediated endocytosis and is fully blocked by drugs suppressing macropinocytosis. ZIP and other cationic peptides remove newly inserted AMPA receptor nanoclusters at potentiated synapses, providing a mechanism by which these peptides erase memories without altering basal synaptic function. When delivered in vivo, cationic peptides can modulate memories on local and brain-wide scales, and these mechanisms can be leveraged to prevent memory loss in a model of traumatic brain injury. Our findings uncover a previously unknown synaptic mechanism by which memories are maintained or lost.
Collapse
Affiliation(s)
- Eric G Stokes
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO, USA
| | - Jose J Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Megan Nguyen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Alexa Tierno
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Yinyin Zhuang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Vivienne Mae Galinato
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Michael Toledano
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Xiaoyu Shi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Robert F Hunt
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
- Epilepsy Research Center, University of California, Irvine, Irvine, CA, USA
| | - Jason Aoto
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz, Aurora, CO, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
6
|
Guzman L, Parcerisas A, Cano A, Sánchez-López E, Verdaguer E, Auladell C, Cajal Y, Barenys M, Camins A, Rabanal F, Ettcheto M. Colistin treatment causes neuronal loss and cognitive impairment via ros accumulation and neuronal plasticity alterations. Biomed Pharmacother 2025; 183:117839. [PMID: 39823721 DOI: 10.1016/j.biopha.2025.117839] [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: 11/12/2024] [Revised: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
The rise of antimicrobial resistance has made necessary the increase of the antibacterial arsenal against multidrug-resistant bacteria. In this context, colistin has re-emerged as a first-line antibiotic in critical situations despite its nephro- and neuro- toxicity at peripheral level. However, the mechanism underlying its toxicity remains unknown, particularly in relation to the central nervous system (CNS). Therefore, this study aimed to characterize the molecular mechanisms underlying colistin-induced neurotoxicity in the CNS through a combination of in vitro and in vivo molecular studies along with several in vivo behavioral tests. Following colistin treatment, mice exhibited a significant reduction in body weight together with renal impairment, and locomotor dysfunction. Moreover, our results demonstrated that colistin disrupted the blood-brain barrier, inducing astrogliosis, and triggering apoptosis-related processes probably through the accumulation of reactive oxygen species (ROS) and mitochondrial dysfunction. Further analysis on mice and primary neuronal cultures revealed that colistin administration altered neuronal plasticity by reducing the number of immature neurons in adult neurogenesis and altering the synaptic function through a reduction of the post-synaptic protein PSD95. All these alterations together finally lead to cognitive impairment and depression-like symptoms in mice. These findings provide novel insights into the mechanisms of colistin-induced neurotoxicity in the CNS, highlighting the need for careful monitoring of cognitive function in patients undergoing colistin treatment.
Collapse
Affiliation(s)
- Laura Guzman
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d'Hebron, 171, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain
| | - Antoni Parcerisas
- Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC) Ctra. de Roda, 70, Vic 08500, Spain; Universitat de Vic - Universitat Central de Catalunya (UVic-UCC), Sagrada Família, 7, Vic 08500, Spain; Facultat de Ciències, Tecnologia i Enginyeria, Sagrada Família, 7, Vic 08500, Spain; Departament de Biociències, Sagrada Família, 7, Vic 08500, Spain
| | - Amanda Cano
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Ace Alzheimer Center Barcelona, C/Marquès de Sentmenat, 57, Barcelona 08029, Spain
| | - Elena Sánchez-López
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Departament de Farmàcia, Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), Av. Diagonal, 64, Barcelona 08028, Spain
| | - Ester Verdaguer
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d'Hebron, 171, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain
| | - Carme Auladell
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d'Hebron, 171, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain
| | - Yolanda Cajal
- Departament de Farmàcia, Tecnologia Farmacèutica i Fisicoquímica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), Av. Diagonal, 64, Barcelona 08028, Spain
| | - Marta Barenys
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Recerca en Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Av. Prat de la Riba, 171, Barcelona 08921, Spain; German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment (BfR), Germany
| | - Antoni Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d'Hebron, 171, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, Reus 43204, Spain
| | - Francesc Rabanal
- Secció de Química Orgànica, Departament de Química Inorgànica i Orgànica, Facultat de Química, Universitat de Barcelona (UB), C/Martí i Franquès, 1-11, Barcelona 08028, Spain
| | - Miren Ettcheto
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, Barcelona 08028, Spain; Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d'Hebron, 171, Barcelona 08035, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, Madrid 28029, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, Reus 43204, Spain.
| |
Collapse
|
7
|
Fischer QS, Kalikulov D, Viana Di Prisco G, Williams CA, Baldwin PR, Friedlander MJ. Synaptic Plasticity in the Injured Brain Depends on the Temporal Pattern of Stimulation. J Neurotrauma 2024; 41:2455-2477. [PMID: 38818799 DOI: 10.1089/neu.2024.0129] [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] [Indexed: 06/01/2024] Open
Abstract
Neurostimulation protocols are increasingly used as therapeutic interventions, including for brain injury. In addition to the direct activation of neurons, these stimulation protocols are also likely to have downstream effects on those neurons' synaptic outputs. It is well known that alterations in the strength of synaptic connections (long-term potentiation, LTP; long-term depression, LTD) are sensitive to the frequency of stimulation used for induction; however, little is known about the contribution of the temporal pattern of stimulation to the downstream synaptic plasticity that may be induced by neurostimulation in the injured brain. We explored interactions of the temporal pattern and frequency of neurostimulation in the normal cerebral cortex and after mild traumatic brain injury (mTBI), to inform therapies to strengthen or weaken neural circuits in injured brains, as well as to better understand the role of these factors in normal brain plasticity. Whole-cell (WC) patch-clamp recordings of evoked postsynaptic potentials in individual neurons, as well as field potential (FP) recordings, were made from layer 2/3 of visual cortex in response to stimulation of layer 4, in acute slices from control (naive), sham operated, and mTBI rats. We compared synaptic plasticity induced by different stimulation protocols, each consisting of a specific frequency (1 Hz, 10 Hz, or 100 Hz), continuity (continuous or discontinuous), and temporal pattern (perfectly regular, slightly irregular, or highly irregular). At the individual neuron level, dramatic differences in plasticity outcome occurred when the highly irregular stimulation protocol was used at 1 Hz or 10 Hz, producing an overall LTD in controls and shams, but a robust overall LTP after mTBI. Consistent with the individual neuron results, the plasticity outcomes for simultaneous FP recordings were similar, indicative of our results generalizing to a larger scale synaptic network than can be sampled by individual WC recordings alone. In addition to the differences in plasticity outcome between control (naive or sham) and injured brains, the dynamics of the changes in synaptic responses that developed during stimulation were predictive of the final plasticity outcome. Our results demonstrate that the temporal pattern of stimulation plays a role in the polarity and magnitude of synaptic plasticity induced in the cerebral cortex while highlighting differences between normal and injured brain responses. Moreover, these results may be useful for optimization of neurostimulation therapies to treat mTBI and other brain disorders, in addition to providing new insights into downstream plasticity signaling mechanisms in the normal brain.
Collapse
Affiliation(s)
- Quentin S Fischer
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Djanenkhodja Kalikulov
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | | | - Carrie A Williams
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
| | - Philip R Baldwin
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Michael J Friedlander
- Fralin Biomedical Research Institute at VTC, Roanoke, Virginia, USA
- FBRI Center for Neurobiology Research, Roanoke, Virginia, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
- Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- Faculty of Health Sciences, Virginia Tech, Roanoke, Virginia, USA
| |
Collapse
|
8
|
Allende LG, Natalí L, Cragnolini AB, Bollo M, Musri MM, de Mendoza D, Martín MG. Lysosomal cholesterol accumulation in aged astrocytes impairs cholesterol delivery to neurons and can be rescued by cannabinoids. Glia 2024; 72:1746-1765. [PMID: 38856177 DOI: 10.1002/glia.24580] [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: 11/29/2023] [Revised: 05/25/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Cholesterol is crucial for the proper functioning of eukaryotic cells, especially neurons, which rely on cholesterol to maintain their complex structure and facilitate synaptic transmission. However, brain cells are isolated from peripheral cholesterol by the blood-brain barrier and mature neurons primarily uptake the cholesterol synthesized by astrocytes for proper function. This study aimed to investigate the effect of aging on cholesterol trafficking in astrocytes and its delivery to neurons. We found that aged astrocytes accumulated high levels of cholesterol in the lysosomal compartment, and this cholesterol buildup can be attributed to the simultaneous occurrence of two events: decreased levels of the ABCA1 transporter, which impairs ApoE-cholesterol export from astrocytes, and reduced expression of NPC1, which hinders cholesterol release from lysosomes. We show that these two events are accompanied by increased microR-33 in aged astrocytes, which targets ABCA1 and NPC1. In addition, we demonstrate that the microR-33 increase is triggered by oxidative stress, one of the hallmarks of aging. By coculture experiments, we show that cholesterol accumulation in astrocytes impairs the cholesterol delivery from astrocytes to neurons. Remarkably, we found that this altered transport of cholesterol could be alleviated through treatment with endocannabinoids as well as cannabidiol or CBD. Finally, according to data demonstrating that aged astrocytes develop an A1 phenotype, we found that cholesterol buildup is also observed in reactive C3+ astrocytes. Given that reduced neuronal cholesterol affects synaptic plasticity, the ability of cannabinoids to restore cholesterol transport from aged astrocytes to neurons holds significant implications in aging and inflammation.
Collapse
Affiliation(s)
- Leandro G Allende
- Departamento de Neurobiología Molecular y celular, Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lautaro Natalí
- Departamento de Bioquímica y Biofísica, Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea B Cragnolini
- Instituto de Investigaciones Biológicas y Tecnológicas, CONICET-UNC, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Bollo
- Departamento de Bioquímica y Biofísica, Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Melina M Musri
- Departamento de Bioquímica y Biofísica, Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Diego de Mendoza
- Laboratorio de Fisiología Microbiana, Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Mauricio G Martín
- Departamento de Neurobiología Molecular y celular, Instituto Ferreyra, INIMEC-CONICET-UNC, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
9
|
Zhvania MG, Japaridze N, Tizabi Y, Pochkhidze N, Lobzhanidze G. Effects of high-intensity chronic noise on spatial memory in male versus female rats. Eur J Neurosci 2024; 60:5581-5590. [PMID: 39180282 DOI: 10.1111/ejn.16514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/25/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024]
Abstract
The detrimental effects of high-intensity noise on the auditory system and emotional status, including the induction of anxiety, are well documented. Preclinical as well as epidemiological and clinical studies have solidly established differential responses between males and females to various stressful stimuli, including high-intensity white noise (HIWN). However, whether chronic exposure to noise affects cognitive functions and whether this effect is sex dependent has not been adequately addressed. In this study, we used two cognitive test paradigms, such as the Morris water maze (MWM) and the multi-branch maze (MBM), to test the effect of chronic HIWN on indices of spatial learning and memory in both male and female Wistar rats. Our findings indicate that daily (1 h) exposure to 100 dB of noise for 30 consecutive days induces different task-dependent responses in male versus female rats. For example, in the acquisition phase of MWM, female rats exposed to noise outperformed their male counterparts at twice the speed. Similarly, in the MBM test, noise-exposed female rats outperformed the male rats in reaching the nest box. It is clear from these studies that noise impairs cognitive functions twice as negatively in male rats as in female rats. Thus, sex-related differences in spatial learning and memory in response to HIWN must be taken into consideration when investigating the neurobiological components and/or treatment modalities.
Collapse
Affiliation(s)
- Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
- Carl Zeiss Scientific and Education Center, New Vision University, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Nino Pochkhidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Giorgi Lobzhanidze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
10
|
Bridi MCD, Hong S, Severin D, Moreno C, Contreras A, Kirkwood A. Blockade of GluN2B-Containing NMDA Receptors Prevents Potentiation and Depression of Responses during Ocular Dominance Plasticity. J Neurosci 2024; 44:e0021232024. [PMID: 39117456 PMCID: PMC11376332 DOI: 10.1523/jneurosci.0021-23.2024] [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: 12/14/2022] [Revised: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Monocular deprivation (MD) causes an initial decrease in synaptic responses to the deprived eye in juvenile mouse primary visual cortex (V1) through Hebbian long-term depression (LTD). This is followed by a homeostatic increase, which has been attributed either to synaptic scaling or to a slide threshold for Hebbian long-term potentiation (LTP) rather than scaling. We therefore asked in mice of all sexes whether the homeostatic increase during MD requires GluN2B-containing NMDA receptor activity, which is required to slide the plasticity threshold but not for synaptic scaling. Selective GluN2B blockade from 2-6 d after monocular lid suture prevented the homeostatic increase in miniature excitatory postsynaptic current (mEPSC) amplitude in monocular V1 of acute slices and prevented the increase in visually evoked responses in binocular V1 in vivo. The decrease in mEPSC amplitude and visually evoked responses during the first 2 d of MD also required GluN2B activity. Together, these results support the idea that GluN2B-containing NMDA receptors first play a role in LTD immediately following eye closure and then promote homeostasis during prolonged MD by sliding the plasticity threshold in favor of LTP.
Collapse
Affiliation(s)
- Michelle C D Bridi
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Su Hong
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Daniel Severin
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Cristian Moreno
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Altagracia Contreras
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Alfredo Kirkwood
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| |
Collapse
|
11
|
Harris KM, Kuwajima M, Flores JC, Zito K. Synapse-specific structural plasticity that protects and refines local circuits during LTP and LTD. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230224. [PMID: 38853547 PMCID: PMC11529630 DOI: 10.1098/rstb.2023.0224] [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: 10/17/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 06/11/2024] Open
Abstract
Synapses form trillions of connections in the brain. Long-term potentiation (LTP) and long-term depression (LTD) are cellular mechanisms vital for learning that modify the strength and structure of synapses. Three-dimensional reconstruction from serial section electron microscopy reveals three distinct pre- to post-synaptic arrangements: strong active zones (AZs) with tightly docked vesicles, weak AZs with loose or non-docked vesicles, and nascent zones (NZs) with a postsynaptic density but no presynaptic vesicles. Importantly, LTP can be temporarily saturated preventing further increases in synaptic strength. At the onset of LTP, vesicles are recruited to NZs, converting them to AZs. During recovery of LTP from saturation (1-4 h), new NZs form, especially on spines where AZs are most enlarged by LTP. Sentinel spines contain smooth endoplasmic reticulum (SER), have the largest synapses and form clusters with smaller spines lacking SER after LTP recovers. We propose a model whereby NZ plasticity provides synapse-specific AZ expansion during LTP and loss of weak AZs that drive synapse shrinkage during LTD. Spine clusters become functionally engaged during LTP or disassembled during LTD. Saturation of LTP or LTD probably acts to protect recently formed memories from ongoing plasticity and may account for the advantage of spaced over massed learning. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Kristen M. Harris
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Masaaki Kuwajima
- Department of Neuroscience and Center for Learning and Memory, The University of Texas at Austin, Austin, TX78712, USA
| | - Juan C. Flores
- Center for Neuroscience, University of California, Davis, CA95618, USA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, CA95618, USA
| |
Collapse
|
12
|
Gendron WH, Fertan E, Roddick KM, Wong AA, Maliougina M, Hiani YE, Anini Y, Brown RE. Intranasal insulin treatment ameliorates spatial memory, muscular strength, and frailty deficits in 5xFAD mice. Physiol Behav 2024; 281:114583. [PMID: 38750806 DOI: 10.1016/j.physbeh.2024.114583] [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: 11/29/2023] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
The 5xFAD mouse model shows age-related weight loss as well as cognitive and motor deficits. Metabolic dysregulation, especially impaired insulin signaling, is also present in AD. This study examined whether intranasal delivery of insulin (INI) at low (0.875 U) or high (1.750 U) doses would ameliorate these deficits compared to saline in 10-month-old female 5xFAD and B6SJL wildtype (WT) mice. INI increased forelimb grip strength in the wire hang test in 5xFAD mice in a dose-dependent manner but did not improve the performance of 5xFAD mice on the balance beam. High INI doses reduced frailty scores in 5xFAD mice and improved spatial memory in both acquisition and reversal probe trials in the Morris water maze. INI increased swim speed in 5xFAD mice but had no effect on object recognition memory or working memory in the spontaneous alternation task, nor did it improve memory in the contextual or cued fear memory tasks. High doses of insulin increased the liver, spleen, and kidney weights and reduced brown adipose tissue weights. P-Akt signaling in the hippocampus was increased by insulin in a dose-dependent manner. Altogether, INI increased strength, reduced frailty scores, and improved visual spatial memory. Hypoglycemia was not present after INI, however alterations in tissue and organ weights were present. These results are novel and important as they indicate that intra-nasal insulin can reverse cognitive, motor and frailty deficits found in this mouse model of AD.
Collapse
Affiliation(s)
- William H Gendron
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Emre Fertan
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Kyle M Roddick
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Aimée A Wong
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Maria Maliougina
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Yassine El Hiani
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Younes Anini
- Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Departments of Obstetrics and Gynecology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Richard E Brown
- Departments of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Departments of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
13
|
Hoang TH, Manahan-Vaughan D. Differentiated somatic gene expression is triggered in the dorsal hippocampus and the anterior retrosplenial cortex by hippocampal synaptic plasticity prompted by spatial content learning. Brain Struct Funct 2024; 229:639-655. [PMID: 37690045 PMCID: PMC10978647 DOI: 10.1007/s00429-023-02694-z] [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: 04/05/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023]
Abstract
Hippocampal afferent inputs, terminating on proximal and distal subfields of the cornus ammonis (CA), enable the functional discrimination of 'what' (item identity) and 'where' (spatial location) elements of a spatial representation. This kind of information is supported by structures such as the retrosplenial cortex (RSC). Spatial content learning promotes the expression of hippocampal synaptic plasticity, particularly long-term depression (LTD). In the CA1 region, this is specifically facilitated by the learning of item-place features of a spatial environment. Gene-tagging, by means of time-locked fluorescence in situ hybridization (FISH) to detect nuclear expression of immediate early genes, can reveal neuronal populations that engage in experience-dependent information encoding. In the current study, using FISH, we examined if learning-facilitated LTD results in subfield-specific information encoding in the hippocampus and RSC. Rats engaged in novel exploration of small items during stimulation of Schaffer collateral-CA1 synapses. This resulted in LTD (> 24 h). FISH, to detect nuclear expression of Homer1a, revealed that the distal-CA1 and proximal-CA3 subcompartments were particularly activated by this event. By contrast, all elements of the proximodistal cornus ammonis-axis showed equal nuclear Homer1a expression following LTD induction solely by means of afferent stimulation. The RSC exhibited stronger nuclear Homer1a expression in response to learning-facilitated LTD, and to novel item-place experience, compared to LTD induced by sole afferent stimulation in CA1. These results show that both the cornus ammonis and RSC engage in differentiated information encoding of item-place learning that is salient enough, in its own right, to drive the expression of hippocampal LTD. These results also reveal a novel role of the RSC in item-place learning.
Collapse
Affiliation(s)
- Thu-Huong Hoang
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Universitätsstr. 150, MA 4/150, 44780, Bochum, Germany
| | - Denise Manahan-Vaughan
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Universitätsstr. 150, MA 4/150, 44780, Bochum, Germany.
| |
Collapse
|
14
|
Aidil-Carvalho F, Caulino-Rocha A, Ribeiro JA, Cunha-Reis D. Mismatch novelty exploration training shifts VPAC 1 receptor-mediated modulation of hippocampal synaptic plasticity by endogenous VIP in male rats. J Neurosci Res 2024; 102:e25333. [PMID: 38656542 DOI: 10.1002/jnr.25333] [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: 12/28/2023] [Revised: 03/04/2024] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
Novelty influences hippocampal-dependent memory through metaplasticity. Mismatch novelty detection activates the human hippocampal CA1 area and enhances rat hippocampal-dependent learning and exploration. Remarkably, mismatch novelty training (NT) also enhances rodent hippocampal synaptic plasticity while inhibition of VIP interneurons promotes rodent exploration. Since VIP, acting on VPAC1 receptors (Rs), restrains hippocampal LTP and depotentiation by modulating disinhibition, we now investigated the impact of NT on VPAC1 modulation of hippocampal synaptic plasticity in male Wistar rats. NT enhanced both CA1 hippocampal LTP and depotentiation unlike exploring an empty holeboard (HT) or a fixed configuration of objects (FT). Blocking VIP VPAC1Rs with PG 97269 (100 nM) enhanced both LTP and depotentiation in naïve animals, but this effect was less effective in NT rats. Altered endogenous VIP modulation of LTP was absent in animals exposed to the empty environment (HT). HT and FT animals showed mildly enhanced synaptic VPAC1R levels, but neither VIP nor VPAC1R levels were altered in NT animals. Conversely, NT enhanced the GluA1/GluA2 AMPAR ratio and gephyrin synaptic content but not PSD-95 excitatory synaptic marker. In conclusion, NT influences hippocampal synaptic plasticity by reshaping brain circuits modulating disinhibition and its control by VIP-expressing hippocampal interneurons while upregulation of VIP VPAC1Rs is associated with the maintenance of VIP control of LTP in FT and HT animals. This suggests VIP receptor ligands may be relevant to co-adjuvate cognitive recovery therapies in aging or epilepsy, where LTP/LTD imbalance occurs.
Collapse
Affiliation(s)
- Fatima Aidil-Carvalho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Caulino-Rocha
- BioISI-Instituto de Biossistemas e Ciências Integrativas, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim Alexandre Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Diana Cunha-Reis
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- BioISI-Instituto de Biossistemas e Ciências Integrativas, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
15
|
Nascimento C, Guerreiro-Pinto V, Pawlak S, Caulino-Rocha A, Amat-Garcia L, Cunha-Reis D. Impaired Response to Mismatch Novelty in the Li 2+-Pilocarpine Rat Model of TLE: Correlation with Hippocampal Monoaminergic Inputs. Biomedicines 2024; 12:631. [PMID: 38540244 PMCID: PMC10968540 DOI: 10.3390/biomedicines12030631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 11/11/2024] Open
Abstract
Novelty detection, crucial to episodic memory formation, is impaired in epileptic patients with mesial temporal lobe resection. Mismatch novelty detection, that activates the hippocampal CA1 area in humans and is vital for memory reformulation and reconsolidation, is also impaired in patients with hippocampal lesions. In this work, we investigated the response to mismatch novelty, as occurs with the new location of known objects in a familiar environment, in the Li2+-pilocarpine rat model of TLE and its correlation with hippocampal monoaminergic markers. Animals showing spontaneous recurrent seizures (SRSs) for at least 4 weeks at the time of behavioural testing showed impaired spatial learning in the radial arm maze, as described. Concurrently, SRS rats displayed impaired exploratory responses to mismatch novelty, yet novel object recognition was not significantly affected in SRS rats. While the levels of serotonin and dopamine transporters were mildly decreased in hippocampal membranes from SRS rats, the levels on the norepinephrine transporter, tyrosine hydroxylase and dopamine-β-hydroxylase were enhanced, hinting for an augmentation, rather than an impairment in noradrenergic function in SRS animals. Altogether, this reveals that mismatch novelty detection is particularly affected by hippocampal damage associated to the Li2+-pilocarpine model of epilepsy 4-8 weeks after the onset of SRSs and suggests that deficits in mismatch novelty detection may substantially contribute to cognitive impairment in MTLE. As such, behavioural tasks based on these aspects of mismatch novelty may prove useful in the development of cognitive therapy strategies aiming to rescue cognitive deficits observed in epilepsy.
Collapse
Affiliation(s)
- Carlos Nascimento
- Unidade de Fisiologia Clínica e Translacional, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Vasco Guerreiro-Pinto
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Seweryn Pawlak
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Ana Caulino-Rocha
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Laia Amat-Garcia
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Diana Cunha-Reis
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
- Departamento de Biologia Vegetal, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
16
|
Atarashi N, Morishita M, Matsuda S. Activation of innate immune receptor TLR9 by mitochondrial DNA plays essential roles in the chemical long-term depression of hippocampal neurons. J Biol Chem 2024; 300:105744. [PMID: 38354781 PMCID: PMC10943477 DOI: 10.1016/j.jbc.2024.105744] [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: 09/18/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/16/2024] Open
Abstract
Synaptic plasticity is believed to be the cellular basis for experience-dependent learning and memory. Although long-term depression (LTD), a form of synaptic plasticity, is caused by the activity-dependent reduction of cell surface α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptors (AMPA receptors) at postsynaptic sites, its regulation by neuronal activity is not completely understood. In this study, we showed that the inhibition of toll-like receptor-9 (TLR9), an innate immune receptor, suppresses N-methyl-d-aspartate (NMDA)-induced reduction of cell surface AMPA receptors in cultured hippocampal neurons. We found that inhibition of TLR9 also blocked NMDA-induced activation of caspase-3, which plays an essential role in the induction of LTD. siRNA-based knockdown of TLR9 also suppressed the NMDA-induced reduction of cell surface AMPA receptors, although the scrambled RNA had no effect on the NMDA-induced trafficking of AMPA receptors. Overexpression of the siRNA-resistant form of TLR9 rescued the AMPA receptor trafficking abolished by siRNA. Furthermore, NMDA stimulation induced rapid mitochondrial morphological changes, mitophagy, and the binding of mitochondrial DNA (mtDNA) to TLR9. Treatment with dideoxycytidine and mitochondrial division inhibitor-1, which block mtDNA replication and mitophagy, respectively, inhibited NMDA-dependent AMPA receptor internalization. These results suggest that mitophagy induced by NMDA receptor activation releases mtDNA and activates TLR9, which plays an essential role in the trafficking of AMPA receptors during the induction of LTD.
Collapse
Affiliation(s)
- Naoya Atarashi
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Misaki Morishita
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Shinji Matsuda
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan; Center for Neuroscience and Biomedical Engineering (CNBE), The University of Electro-Communications, Tokyo, Japan.
| |
Collapse
|
17
|
Chandrasekaran J, Caldwell KK, Brigman JL. Dynamic regulation of corticostriatal glutamatergic synaptic expression during reversal learning in male mice. Neurobiol Learn Mem 2024; 208:107892. [PMID: 38242226 PMCID: PMC10936219 DOI: 10.1016/j.nlm.2024.107892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
Behavioral flexibility, one of the core executive functions of the brain, has been shown to be an essential skill for survival across species. Corticostriatal circuits play a critical role in mediating behavioral flexibility. The molecular mechanisms underlying these processes are still unclear. Here, we measured how synaptic glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and N-methyl-D-aspartic acid receptor (NMDAR) expression dynamically changed during specific stages of learning and reversal. Following training to well-established stages of discrimination and reversal learning on a touchscreen visual task, lateral orbitofrontal cortex (OFC), dorsal striatum (dS) as well as medial prefrontal cortex (mPFC), basolateral amygdala (BLA) and piriform cortex (Pir) were micro dissected from male mouse brain and the expression of glutamatergic receptor subunits in the synaptic fraction were measured via immunoblotting. We found that the GluN2B subunit of NMDAR in the OFC remained stable during initial discrimination learning but significantly increased in the synaptic fraction during mid-reversal stages, the period during which the OFC has been shown to play a critical role in updating outcome expectancies. In contrast, both GluA1 and GluA2 subunits of the AMPAR significantly increased in the dS synaptic fraction as new associations were learned late in reversal. Expression of NMDAR and AMPAR subunits did not significantly differ across learning stages in any other brain region. Together, these findings further support the involvement of OFC-dS circuits in moderating well-learned associations and flexible behavior and suggest that dynamic synaptic expression of NMDAR and AMPAR in these circuits may play a role in mediating efficient learning during discrimination and the ability to update previously learned associations as environmental contingencies change.
Collapse
Affiliation(s)
- Jayapriya Chandrasekaran
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kevin K Caldwell
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque NM 87131, USA
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque NM 87131, USA.
| |
Collapse
|
18
|
Wei M, Yang L, Su F, Liu Y, Zhao X, Luo L, Sun X, Liu S, Dong Z, Zhang Y, Shi YS, Liang J, Zhang C. ABHD6 drives endocytosis of AMPA receptors to regulate synaptic plasticity and learning flexibility. Prog Neurobiol 2024; 233:102559. [PMID: 38159878 DOI: 10.1016/j.pneurobio.2023.102559] [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: 06/23/2023] [Revised: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Trafficking of α-Amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors (AMPARs), mediated by AMPAR interacting proteins, enabled neurons to maintain tuning capabilities at rest or active state. α/β-Hydrolase domain-containing 6 (ABHD6), an endocannabinoid hydrolase, was an AMPAR auxiliary subunit found to negatively regulate the surface delivery of AMPARs. While ABHD6 was found to prevent AMPAR tetramerization in endoplasmic reticulum, ABHD6 was also reported to localize at postsynaptic site. Yet, the role of ABHD6 interacting with AMPAR at postsynaptic site, and the physiological significance of ABHD6 regulating AMPAR trafficking remains elusive. Here, we generated the ABHD6 knockout (ABHD6KO) mice and found that deletion of ABHD6 selectively enhanced AMPAR-mediated basal synaptic responses and the surface expression of postsynaptic AMPARs. Furthermore, we found that loss of ABHD6 impaired hippocampal long-term depression (LTD) and synaptic downscaling in hippocampal synapses. AMPAR internalization assays revealed that ABHD6 was essential for neuronal activity-dependent endocytosis of surface AMPARs, which is independent of ABHD6's hydrolase activity. The defects of AMPAR endocytosis and LTD are expressed as deficits in learning flexibility in ABHD6KO mice. Collectively, we demonstrated that ABHD6 is an endocytic accessory protein promoting AMPAR endocytosis, thereby contributes to the formation of LTD, synaptic downscaling and reversal learning.
Collapse
Affiliation(s)
- Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China.
| | - Lei Yang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Feng Su
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ying Liu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xinyi Zhao
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Lin Luo
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Xinyue Sun
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Zhaoqi Dong
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Yong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission of the PR China, IDG/McGovern Institute for Brain Research at PKU, Peking University, Beijing 100083, China
| | - Yun Stone Shi
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing 210032, China
| | - Jing Liang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China.
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210000, Jiangsu, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
19
|
Stokes E, Zhuang Y, Toledano M, Vasquez J, Azouz G, Hui M, Tyler I, Shi X, Aoto J, Beier KT. Cationic peptides erase memories by removing synaptic AMPA receptors through endophilin-mediated endocytosis. RESEARCH SQUARE 2023:rs.3.rs-3559525. [PMID: 38045269 PMCID: PMC10690331 DOI: 10.21203/rs.3.rs-3559525/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Administration of the Zeta Inhibitory Peptide (ZIP) interferes with memory maintenance and long-term potentiation (LTP). However, mice lacking its putative target, the protein kinase PKMζ, exhibit normal learning and memory as well as LTP, making ZIP's mechanism unclear. Here, we show that ZIP disrupts LTP by removing surface AMPA receptors through its cationic charge alone. This effect was fully blocked by drugs that block macropinocytosis and is dependent on endophilin A2 (endoA2)-mediated endocytosis. ZIP and other cationic peptides selectively removed newly inserted AMPAR nanoclusters, providing a mechanism by which these peptides erase memories without effects on basal synaptic function. Lastly, cationic peptides can be administered locally and/or systemically and can be combined with local microinjection of macropinocytosis inhibitors to modulate memories on local and brain-wide scales. Our findings have critical implications for an entire field of memory mechanisms and highlight a previously unappreciated mechanism by which memories can be lost.
Collapse
Affiliation(s)
- Eric Stokes
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Yinyin Zhuang
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | - Michael Toledano
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - Jose Vasquez
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - Ghalia Azouz
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - Isabella Tyler
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - Xiaoyu Shi
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Department of Chemistry, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA 92697-4560
| | - Jason Aoto
- Pharmacology Graduate Program, University of Colorado Anschutz, Aurora, CO 80045, USA
- University of Colorado Anschutz, Department of Pharmacology, Aurora, CO 80045, USA
| | - Kevin T. Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA 92697-4560
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA 92697-4560
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA 92697-4560
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA 92697-4560
| |
Collapse
|
20
|
Jo D, Arjunan A, Choi S, Jung YS, Park J, Jo J, Kim OY, Song J. Oligonol ameliorates liver function and brain function in the 5 × FAD mouse model: transcriptional and cellular analysis. Food Funct 2023; 14:9650-9670. [PMID: 37843873 DOI: 10.1039/d3fo03451h] [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: 10/17/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease worldwide and is accompanied by memory deficits, personality changes, anxiety, depression, and social difficulties. For treatment of AD, many researchers have attempted to find medicinal resources with high effectiveness and without side effects. Oligonol is a low molecular weight polypeptide derived from lychee fruit extract. We investigated the effects of oligonol in 5 × FAD transgenic AD mice, which developed severe amyloid pathology, through behavioral tests (Barnes maze, marble burying, and nestle shredding) and molecular experiments. Oligonol treatment attenuated blood glucose levels and increased the antioxidant response in the livers of 5 × FAD mice. Moreover, the behavioral score data showed improvements in anxiety, depressive behavior, and cognitive impairment following a 2-month course of orally administered oligonol. Oligonol treatment not only altered the circulating levels of cytokines and adipokines in 5 × FAD mice, but also significantly enhanced the mRNA and protein levels of antioxidant enzymes and synaptic plasticity in the brain cortex and hippocampus. Therefore, we highlight the therapeutic potential of oligonol to attenuate neuropsychiatric problems and improve memory deficits in the early stage of AD.
Collapse
Affiliation(s)
- Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Seoyoon Choi
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Jihyun Park
- Department of Food Science and Nutrition, Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
- Department of Health Sciences, Graduate School of Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea
| | - Jihoon Jo
- Department of Biomedical Science, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
- Department of Health Sciences, Graduate School of Dong-A University, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
- Biomedical Science Graduate Program (BMSGP), Chonnam National University, Seoyangro 264, Hwasun 58128, Republic of Korea
| |
Collapse
|
21
|
Arriagada-Diaz J, Flores-Muñoz C, Gómez-Soto B, Labraña-Allende M, Mattar-Araos M, Prado-Vega L, Hinostroza F, Gajardo I, Guerra-Fernández MJ, Bevilacqua JA, Cárdenas AM, Bitoun M, Ardiles AO, Gonzalez-Jamett AM. A centronuclear myopathy-causing mutation in dynamin-2 disrupts neuronal morphology and excitatory synaptic transmission in a murine model of the disease. Neuropathol Appl Neurobiol 2023; 49:e12918. [PMID: 37317811 DOI: 10.1111/nan.12918] [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: 09/12/2022] [Revised: 04/30/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
AIMS Dynamin-2 is a large GTPase, a member of the dynamin superfamily that regulates membrane remodelling and cytoskeleton dynamics. Mutations in the dynamin-2 gene (DNM2) cause autosomal dominant centronuclear myopathy (CNM), a congenital neuromuscular disorder characterised by progressive weakness and atrophy of the skeletal muscles. Cognitive defects have been reported in some DNM2-linked CNM patients suggesting that these mutations can also affect the central nervous system (CNS). Here we studied how a dynamin-2 CNM-causing mutation influences the CNS function. METHODS Heterozygous mice harbouring the p.R465W mutation in the dynamin-2 gene (HTZ), the most common causing autosomal dominant CNM, were used as disease model. We evaluated dendritic arborisation and spine density in hippocampal cultured neurons, analysed excitatory synaptic transmission by electrophysiological field recordings in hippocampal slices, and evaluated cognitive function by performing behavioural tests. RESULTS HTZ hippocampal neurons exhibited reduced dendritic arborisation and lower spine density than WT neurons, which was reversed by transfecting an interference RNA against the dynamin-2 mutant allele. Additionally, HTZ mice showed defective hippocampal excitatory synaptic transmission and reduced recognition memory compared to the WT condition. CONCLUSION Our findings suggest that the dynamin-2 p.R465W mutation perturbs the synaptic and cognitive function in a CNM mouse model and support the idea that this GTPase plays a key role in regulating neuronal morphology and excitatory synaptic transmission in the hippocampus.
Collapse
Affiliation(s)
- Jorge Arriagada-Diaz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Bárbara Gómez-Soto
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias Médicas, Mención Biología Celular y Molecular, Universidad de Valparaíso, Valparaíso, Chile
| | - Marjorie Labraña-Allende
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias Médicas, Mención Biología Celular y Molecular, Universidad de Valparaíso, Valparaíso, Chile
| | - Michelle Mattar-Araos
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Prado-Vega
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Magister en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Fernando Hinostroza
- Centro de Investigación de Estudios Avanzados del Maule, CIEAM, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
- Centro de Investigación en Neuropsicología y Neurociencias Cognitivas, Facultad de Ciencias de la Salud, Universidad Católica del Maule, Talca, Chile
- Escuela de Química y Farmacia, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Ivana Gajardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Jorge A Bevilacqua
- Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Marc Bitoun
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, F-75013, France
| | - Alvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
22
|
Poorgholam P, Yaghmaei P, Noureddini M, Hajebrahimi Z. Artemisin and human endometrial-derived stem cells improve cognitive function and synaptic plasticity in a rat model of Alzheimer disease and diabetes. Metab Brain Dis 2023; 38:1925-1936. [PMID: 37043150 DOI: 10.1007/s11011-023-01200-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 03/10/2023] [Indexed: 04/13/2023]
Abstract
Alzheimer disease (AD) is a common form of dementia associated with loss of memory and disruption of synaptic plasticity. There is a strong correlation between the pathophysiological features of AD and diabetes, including induction of oxidative stress, inflammation, and abnormality in blood vessels. Considering the brain's limited capacity to repair damage and the potential of stem cell-derived neural cells in the repair of neurodegenerative disease, we investigated the effects of artemisinin and TSP‑1‑human endometrial-derived-derived stem cells (TSP‑1‑hEDSCs) on the cognitive function and synaptic plasticity in AD-diabetes rats. The authors previously showed that artemisinin and TSP‑1‑hEDSCs suppressed oxidative stress and inflammation in AD-diabetes rats. Thrombospondins-1 (TSPs-1) is a glycoprotein that inhibits angiogenesis. AD and diabetes were induced using streptozotocin. Synaptic plasticity and learning and memory function were studied using the Morris water maze and electrophysiological test, respectively. Streptozotocin increased traveled swimming distance and escape latency in the morris water maze test, decreased the percent time spent in the target quadrant, inhibited the long-term potentiation (LTP), and increased the blood glucose levels. Simultaneous or separate administration of artemisinin and TSP‑1‑hEDSCs decreased the blood levels of glucose and improved cognitive tasks and synaptic plasticity by considerably reducing traveled swimming distance and escape latency, increasing the percent time spent in the target quadrant, and retrieval of the LTP; therefore, they could be utilized as an adjunct treatment for AD treatment. These results may be due to a decrease in oxidative stress and inflammation.
Collapse
Affiliation(s)
- Parvin Poorgholam
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parichehreh Yaghmaei
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mehdi Noureddini
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Hajebrahimi
- A&S Research Institute, Ministry of Science Research and Technology, Tehran, Iran
| |
Collapse
|
23
|
Sridhar S, Khamaj A, Asthana MK. Cognitive neuroscience perspective on memory: overview and summary. Front Hum Neurosci 2023; 17:1217093. [PMID: 37565054 PMCID: PMC10410470 DOI: 10.3389/fnhum.2023.1217093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
This paper explores memory from a cognitive neuroscience perspective and examines associated neural mechanisms. It examines the different types of memory: working, declarative, and non-declarative, and the brain regions involved in each type. The paper highlights the role of different brain regions, such as the prefrontal cortex in working memory and the hippocampus in declarative memory. The paper also examines the mechanisms that underlie the formation and consolidation of memory, including the importance of sleep in the consolidation of memory and the role of the hippocampus in linking new memories to existing cognitive schemata. The paper highlights two types of memory consolidation processes: cellular consolidation and system consolidation. Cellular consolidation is the process of stabilizing information by strengthening synaptic connections. System consolidation models suggest that memories are initially stored in the hippocampus and are gradually consolidated into the neocortex over time. The consolidation process involves a hippocampal-neocortical binding process incorporating newly acquired information into existing cognitive schemata. The paper highlights the role of the medial temporal lobe and its involvement in autobiographical memory. Further, the paper discusses the relationship between episodic and semantic memory and the role of the hippocampus. Finally, the paper underscores the need for further research into the neurobiological mechanisms underlying non-declarative memory, particularly conditioning. Overall, the paper provides a comprehensive overview from a cognitive neuroscience perspective of the different processes involved in memory consolidation of different types of memory.
Collapse
Affiliation(s)
- Sruthi Sridhar
- Department of Psychology, Mount Allison University, Sackville, NB, Canada
| | - Abdulrahman Khamaj
- Department of Industrial Engineering, College of Engineering, Jazan University, Jazan, Saudi Arabia
| | - Manish Kumar Asthana
- Department of Humanities and Social Sciences, Indian Institute of Technology Roorkee, Roorkee, India
- Department of Design, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
24
|
Ge Y, Wang YT. GluN2B-containing NMDARs in the mammalian brain: pharmacology, physiology, and pathology. Front Mol Neurosci 2023; 16:1190324. [PMID: 37324591 PMCID: PMC10264587 DOI: 10.3389/fnmol.2023.1190324] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) is critical for promoting physiological synaptic plasticity and neuronal viability. As a major subpopulation of the NMDAR, the GluN2B subunit-containing NMDARs have distinct pharmacological properties, physiological functions, and pathological relevance to neurological diseases compared with other NMDAR subtypes. In mature neurons, GluN2B-containing NMDARs are likely expressed as both diheteromeric and triheteromeric receptors, though the functional importance of each subpopulation has yet to be disentangled. Moreover, the C-terminal region of the GluN2B subunit forms structural complexes with multiple intracellular signaling proteins. These protein complexes play critical roles in both activity-dependent synaptic plasticity and neuronal survival and death signaling, thus serving as the molecular substrates underlying multiple physiological functions. Accordingly, dysregulation of GluN2B-containing NMDARs and/or their downstream signaling pathways has been implicated in neurological diseases, and various strategies to reverse these deficits have been investigated. In this article, we provide an overview of GluN2B-containing NMDAR pharmacology and its key physiological functions, highlighting the importance of this receptor subtype during both health and disease states.
Collapse
Affiliation(s)
- Yang Ge
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Nino P, Mzia Z, Nadezhda J, Yousef T, Giorgi L, Tamar L. Short- and long-term effects of chronic toluene exposure on spatial memory in adolescent and adult male Wistar rats. Neurosci Lett 2023; 805:137238. [PMID: 37037302 DOI: 10.1016/j.neulet.2023.137238] [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: 02/03/2023] [Revised: 03/12/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023]
Abstract
Addiction to toluene-containing volatile inhalants is of significant medical and social concern, particularly among youth. These concerns are underscored by the fact that the majority of adult abusers of toluene started as teenagers. Surprisingly, however, the lasting effects of chronic toluene exposure, especially in various age groups, have not been well investigated. Recently, we reported that adolescent and adult male Wistar rats show differential responses to chronic toluene exposure in recognition memory tasks. Since different cognitive functions may be differentially affected by drugs of abuse, we used the same model to evaluate the short- and long-term effects of chronic toluene on spatial learning and memory using Morris water maze. Daily exposure to toluene (2000 ppm) for 40 days (5 min/day) resulted in age-dependent behavioral changes. For example, only adolescent animals showed a decrease in time and distance travelled to find the hidden platform 24 h after the last toluene exposure. In contrast, only adult rats exhibited a decrease in acquisition time and distance travelled at 90 days' post toluene exposure. Our data provide further support for the contention that age-dependent responses should be taken into consideration in interventional attempts to overcome specific detrimental consequences of chronic toluene exposure.
Collapse
Affiliation(s)
- Pochkhidze Nino
- School of Natural Sciences and Medicine, Ilia State University. Tbilisi, Georgia; Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Zhvania Mzia
- School of Natural Sciences and Medicine, Ilia State University. Tbilisi, Georgia; Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia.
| | - Japaridze Nadezhda
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia; Medical School of New Vision University, Tbilisi, Georgia
| | - Tizabi Yousef
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Lobzhanidze Giorgi
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Lordkipanidze Tamar
- School of Natural Sciences and Medicine, Ilia State University. Tbilisi, Georgia
| |
Collapse
|
26
|
Dainauskas JJ, Marie H, Migliore M, Saudargiene A. GluN2B-NMDAR subunit contribution on synaptic plasticity: A phenomenological model for CA3-CA1 synapses. Front Synaptic Neurosci 2023; 15:1113957. [PMID: 37008680 PMCID: PMC10050887 DOI: 10.3389/fnsyn.2023.1113957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/17/2023] Open
Abstract
Synaptic plasticity is believed to be a key mechanism underlying learning and memory. We developed a phenomenological N-methyl-D-aspartate (NMDA) receptor-based voltage-dependent synaptic plasticity model for synaptic modifications at hippocampal CA3-CA1 synapses on a hippocampal CA1 pyramidal neuron. The model incorporates the GluN2A-NMDA and GluN2B-NMDA receptor subunit-based functions and accounts for the synaptic strength dependence on the postsynaptic NMDA receptor composition and functioning without explicitly modeling the NMDA receptor-mediated intracellular calcium, a local trigger of synaptic plasticity. We embedded the model into a two-compartmental model of a hippocampal CA1 pyramidal cell and validated it against experimental data of spike-timing-dependent synaptic plasticity (STDP), high and low-frequency stimulation. The developed model predicts altered learning rules in synapses formed on the apical dendrites of the detailed compartmental model of CA1 pyramidal neuron in the presence of the GluN2B-NMDA receptor hypofunction and can be used in hippocampal networks to model learning in health and disease.
Collapse
Affiliation(s)
- Justinas J. Dainauskas
- Laboratory of Biophysics and Bioinformatics, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Informatics, Vytautas Magnus University, Kaunas, Lithuania
| | - Hélène Marie
- Université Côte d'Azur, Centre National de la Recherche Scientifique (CNRS) UMR 7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Ausra Saudargiene
- Laboratory of Biophysics and Bioinformatics, Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
- *Correspondence: Ausra Saudargiene
| |
Collapse
|
27
|
Cyclin Y regulates spatial learning and memory flexibility through distinct control of the actin pathway. Mol Psychiatry 2023; 28:1351-1364. [PMID: 36434054 PMCID: PMC10005959 DOI: 10.1038/s41380-022-01877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022]
Abstract
Spatial learning and memory flexibility are known to require long-term potentiation (LTP) and long-term depression (LTD), respectively, on a cellular basis. We previously showed that cyclin Y (CCNY), a synapse-remodeling cyclin, is a novel actin-binding protein and an inhibitory regulator of functional and structural LTP in vitro. In this study, we report that Ccny knockout (KO) mice exhibit enhanced LTP and weak LTD at Schaffer collateral-CA1 synapses in the hippocampus. In accordance with enhanced LTP, Ccny KO mice showed improved spatial learning and memory. However, although previous studies reported that normal LTD is necessary for memory flexibility, Ccny KO mice intriguingly showed improved memory flexibility, suggesting that weak LTD could exert memory flexibility when combined with enhanced LTP. At the molecular level, CCNY modulated spatial learning and memory flexibility by distinctively affecting the cofilin-actin signaling pathway in the hippocampus. Specifically, CCNY inhibited cofilin activation by original learning, but reversed such inhibition by reversal learning. Furthermore, viral-mediated overexpression of a phosphomimetic cofilin-S3E in hippocampal CA1 regions enhanced LTP, weakened LTD, and improved spatial learning and memory flexibility, thus mirroring the phenotype of Ccny KO mice. In contrast, the overexpression of a non-phosphorylatable cofilin-S3A in hippocampal CA1 regions of Ccny KO mice reversed the synaptic plasticity, spatial learning, and memory flexibility phenotypes observed in Ccny KO mice. Altogether, our findings demonstrate that LTP and LTD cooperatively regulate memory flexibility. Moreover, CCNY suppresses LTP while facilitating LTD in the hippocampus and negatively regulates spatial learning and memory flexibility through the control of cofilin-actin signaling, proposing CCNY as a learning regulator modulating both memorizing and forgetting processes.
Collapse
|
28
|
The times they are a-changin': a proposal on how brain flexibility goes beyond the obvious to include the concepts of "upward" and "downward" to neuroplasticity. Mol Psychiatry 2023; 28:977-992. [PMID: 36575306 PMCID: PMC10005965 DOI: 10.1038/s41380-022-01931-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Since the brain was found to be somehow flexible, plastic, researchers worldwide have been trying to comprehend its fundamentals to better understand the brain itself, make predictions, disentangle the neurobiology of brain diseases, and finally propose up-to-date treatments. Neuroplasticity is simple as a concept, but extremely complex when it comes to its mechanisms. This review aims to bring to light an aspect about neuroplasticity that is often not given enough attention as it should, the fact that the brain's ability to change would include its ability to disconnect synapses. So, neuronal shrinkage, decrease in spine density or dendritic complexity should be included within the concept of neuroplasticity as part of its mechanisms, not as an impairment of it. To that end, we extensively describe a variety of studies involving topics such as neurodevelopment, aging, stress, memory and homeostatic plasticity to highlight how the weakening and disconnection of synapses organically permeate the brain in so many ways as a good practice of its intrinsic physiology. Therefore, we propose to break down neuroplasticity into two sub-concepts, "upward neuroplasticity" for changes related to synaptic construction and "downward neuroplasticity" for changes related to synaptic deconstruction. With these sub-concepts, neuroplasticity could be better understood from a bigger landscape as a vector in which both directions could be taken for the brain to flexibly adapt to certain demands. Such a paradigm shift would allow a better understanding of the concept of neuroplasticity to avoid any data interpretation bias, once it makes clear that there is no morality with regard to the organic and physiological changes that involve dynamic biological systems as seen in the brain.
Collapse
|
29
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and neuronal Panx1 support long-term reference memory in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.16.524236. [PMID: 36711845 PMCID: PMC9882221 DOI: 10.1101/2023.01.16.524236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Pannexin 1 (Panx1) are ubiquitously expressed proteins that form plasma membrane channels permeable to anions and moderate sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels have been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.) but knowledge of extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the 8-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral - CA1 synapses without alterations basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
|
30
|
Niloy N, Hediyal TA, Vichitra C, Sonali S, Chidambaram SB, Gorantla VR, Mahalakshmi AM. Effect of Cannabis on Memory Consolidation, Learning and Retrieval and Its Current Legal Status in India: A Review. Biomolecules 2023; 13:biom13010162. [PMID: 36671547 PMCID: PMC9855787 DOI: 10.3390/biom13010162] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Cannabis is one of the oldest crops grown, traditionally held religious attachments in various cultures for its medicinal use much before its introduction to Western medicine. Multiple preclinical and clinical investigations have explored the beneficial effects of cannabis in various neurocognitive and neurodegenerative diseases affecting the cognitive domains. Tetrahydrocannabinol (THC), the major psychoactive component, is responsible for cognition-related deficits, while cannabidiol (CBD), a non-psychoactive phytocannabinoid, has been shown to elicit neuroprotective activity. In the present integrative review, the authors focus on the effects of cannabis on the different cognitive domains, including learning, consolidation, and retrieval. The present study is the first attempt in which significant focus has been imparted on all three aspects of cognition, thus linking to its usage. Furthermore, the investigators have also depicted the current legal position of cannabis in India and the requirement for reforms.
Collapse
Affiliation(s)
- Nandi Niloy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Sharma Sonali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Vasavi Rakesh Gorantla
- Department of Anatomical Science, St. George’s University, University Centre, St. Georges FZ818, Grenada
- Correspondence: (V.R.G.); (A.M.M.)
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Correspondence: (V.R.G.); (A.M.M.)
| |
Collapse
|
31
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
32
|
Obot P, Subah G, Schonwald A, Pan J, Velíšek L, Velíšková J, Stanton PK, Scemes E. Astrocyte and Neuronal Panx1 Support Long-Term Reference Memory in Mice. ASN Neuro 2023; 15:17590914231184712. [PMID: 37365910 DOI: 10.1177/17590914231184712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Pannexin 1 (Panx1) is an ubiquitously expressed protein that forms plasma membrane channels permeable to anions and moderate-sized signaling molecules (e.g., ATP, glutamate). In the nervous system, activation of Panx1 channels has been extensively shown to contribute to distinct neurological disorders (epilepsy, chronic pain, migraine, neuroAIDS, etc.), but knowledge of the extent to which these channels have a physiological role remains restricted to three studies supporting their involvement in hippocampus dependent learning. Given that Panx1 channels may provide an important mechanism for activity-dependent neuron-glia interaction, we used Panx1 transgenic mice with global and cell-type specific deletions of Panx1 to interrogate their participation in working and reference memory. Using the eight-arm radial maze, we show that long-term spatial reference memory, but not spatial working memory, is deficient in Panx1-null mice and that both astrocyte and neuronal Panx1 contribute to the consolidation of long-term spatial memory. Field potential recordings in hippocampal slices of Panx1-null mice revealed an attenuation of both long-term potentiation (LTP) of synaptic strength and long-term depression (LTD) at Schaffer collateral-CA1 synapses without alterations of basal synaptic transmission or pre-synaptic paired-pulse facilitation. Our results implicate both neuronal and astrocyte Panx1 channels as critical players for the development and maintenance of long-term spatial reference memory in mice.
Collapse
Affiliation(s)
- Price Obot
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Emergency Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - Galadu Subah
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Antonia Schonwald
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Jian Pan
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Libor Velíšek
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Pediatrics, Penn State Hershey Medical Center, Hershey, PA, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Jana Velíšková
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Obstetrics and Gynecology, New York Medical College, Valhalla, NY, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
- Department of Neurology, New York Medical College, Valhalla, NY, USA
| | - Eliana Scemes
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
33
|
Hamzé R, Delangre E, Tolu S, Moreau M, Janel N, Bailbé D, Movassat J. Type 2 Diabetes Mellitus and Alzheimer's Disease: Shared Molecular Mechanisms and Potential Common Therapeutic Targets. Int J Mol Sci 2022; 23:ijms232315287. [PMID: 36499613 PMCID: PMC9739879 DOI: 10.3390/ijms232315287] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The global prevalence of diabetes mellitus and Alzheimer's disease is increasing alarmingly with the aging of the population. Numerous epidemiological data suggest that there is a strong association between type 2 diabetes and an increased risk of dementia. These diseases are both degenerative and progressive and share common risk factors. The amyloid cascade plays a key role in the pathophysiology of Alzheimer's disease. The accumulation of amyloid beta peptides gradually leads to the hyperphosphorylation of tau proteins, which then form neurofibrillary tangles, resulting in neurodegeneration and cerebral atrophy. In Alzheimer's disease, apart from these processes, the alteration of glucose metabolism and insulin signaling in the brain seems to induce early neuronal loss and the impairment of synaptic plasticity, years before the clinical manifestation of the disease. The large amount of evidence on the existence of insulin resistance in the brain during Alzheimer's disease has led to the description of this disease as "type 3 diabetes". Available animal models have been valuable in the understanding of the relationships between type 2 diabetes and Alzheimer's disease, but to date, the mechanistical links are poorly understood. In this non-exhaustive review, we describe the main molecular mechanisms that may link these two diseases, with an emphasis on impaired insulin and IGF-1 signaling. We also focus on GSK3β and DYRK1A, markers of Alzheimer's disease, which are also closely associated with pancreatic β-cell dysfunction and type 2 diabetes, and thus may represent common therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Rim Hamzé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Etienne Delangre
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Stefania Tolu
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Manon Moreau
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Nathalie Janel
- Team Degenerative Process, Stress and Aging, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Danielle Bailbé
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
| | - Jamileh Movassat
- Team Biology and Pathology of the Endocrine Pancreas, Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université Paris Cité, F-75013 Paris, France
- Correspondence: ; Tel.: +33-1-57-27-77-82; Fax: +33-1-57-27-77-91
| |
Collapse
|
34
|
Xu C, Xiong Q, Tian X, Liu W, Sun B, Ru Q, Shu X. Alcohol Exposure Induces Depressive and Anxiety-like Behaviors via Activating Ferroptosis in Mice. Int J Mol Sci 2022; 23:ijms232213828. [PMID: 36430312 PMCID: PMC9698590 DOI: 10.3390/ijms232213828] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a global public health problem and is frequently comorbid with mental disorders, including anxiety and depression. Ferroptosis is an iron-dependent cell death, which is involved in the pathological process of various diseases such as neurodegenerative diseases, but the role of ferroptosis in the mediation of AUD and its induced mental disorders is unclear. In this study, we aimed to investigate whether ferroptosis was involved in alcohol-induced depressive and anxiety-like behaviors in mice. Following an 8-week period of intermittent alcohol exposure, the alcohol group showed noticeable depressive and anxiety-like behaviors. In addition, nissl staining revealed that alcohol exposure induced neuron damage in the hippocampus (Hip) and prefrontal cortex (PFC) of mice. The levels of synapse-related proteins were significantly reduced in the alcohol group. Iron staining demonstrated that alcohol increased the number of iron-positive staining cells. The protein expression of the transferrin receptor (TFRC) was increased, and the expression of glutathione peroxidase 4 (GPX4) was decreased, respectively, in the alcohol group. Furthermore, the ferroptosis inhibitor ferrostatin-1 significantly prevented alcohol-induced neuron damage and enhanced the expression of N-methyl-d-aspartic acid (NMDA) receptor 2B (NR2B), α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptor 1 (GluA1) and GPX4 in vitro. These results indicated that alcohol exposure could induce depressive and anxiety-like behaviors, and that this effect may occur via activating ferroptosis.
Collapse
Affiliation(s)
- Congyue Xu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Qi Xiong
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Xiang Tian
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Wei Liu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Binlian Sun
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430000, China
- Correspondence: (Q.R.); (X.S.); Tel.: +86-27-84225807 (X.S.)
| | - Xiji Shu
- Wuhan Institutes of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430000, China
- Correspondence: (Q.R.); (X.S.); Tel.: +86-27-84225807 (X.S.)
| |
Collapse
|
35
|
Zhu DY, Lu J, Xu R, Yang JZ, Meng XR, Ou-Yang XN, Yan QY, Nie RF, Zhao T, Chen YD, Lu Y, Zhang YN, Li WJ, Shen X. FX5, a non-steroidal glucocorticoid receptor antagonist, ameliorates diabetic cognitive impairment in mice. Acta Pharmacol Sin 2022; 43:2495-2510. [PMID: 35260821 PMCID: PMC9525278 DOI: 10.1038/s41401-022-00884-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022]
Abstract
Diabetic cognitive impairment (DCI) is a common diabetic complication characterized by learning and memory deficits. In diabetic patients, hyperactivated hypothalamic-pituitary-adrenal (HPA) axis leads to abnormal increase of glucocorticoids (GCs), which causes the damage of hippocampal neurons and cognitive impairment. In this study we investigated the cognition-improving effects of a non-steroidal glucocorticoid receptor (GR) antagonist 5-chloro-N-[4-chloro-3-(trifluoromethyl) phenyl]thiophene-2-sulfonamide (FX5) in diabetic mice. Four weeks after T1DM or T2DM was induced, the mice were administered FX5 (20, 40 mg·kg-1·d-1, i.g.) for 8 weeks. Cognitive impairment was assessed in open field test, novel object recognition test, Y-maze test, and Morris water maze test. We showed that FX5 administration significantly ameliorated the cognitive impairments in both type 1 and 2 diabetic mice. Similar cognitive improvement was observed in diabetic mice following brain GR-specific knockdown by injecting AAV-si-GR. Moreover, AAV-si-GR injection occluded the cognition-improving effects of FX5, suggesting that FX5 functioning as a non-steroidal GR antagonist. In PA-treated primary neurons (as DCI model in vitro), we demonstrated that FX5 (2, 5, 10 μM) dose-dependently ameliorated synaptic impairment via upregulating GR/BDNF/TrkB/CREB pathway, protected against neuronal apoptosis through repressing GR/PI3K/AKT/GSK3β-mediated tauopathy and subsequent endoplasmic reticulum stress. In LPS-treated primary microglia, FX5 dose-dependently inhibited inflammation through GR/NF-κB/NLRP3/ASC/Caspase-1 pathway. These beneficial effects were also observed in the hippocampus of diabetic mice following FX5 administration. Collectively, we have elucidated the mechanisms underlying the beneficial effects of non-steroidal GR antagonist FX5 on DCI and highlighted the potential of FX5 in the treatment of the disease.
Collapse
Affiliation(s)
- Dan-Yang Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Juan-Zhen Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiang-Rui Meng
- Faculty of Art and Science, Queens University, Kingston, ON, K7L 3N6, Canada
| | - Xing-Nan Ou-Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiu-Ying Yan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui-Fang Nie
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-di Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-Nan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Wen-Jun Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
36
|
Bioactive human Alzheimer brain soluble Aβ: pathophysiology and therapeutic opportunities. Mol Psychiatry 2022; 27:3182-3191. [PMID: 35484241 DOI: 10.1038/s41380-022-01589-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022]
Abstract
The accumulation of amyloid-β protein (Aβ) plays an early role in the pathogenesis of Alzheimer's disease (AD). The precise mechanism of how Aβ accumulation leads to synaptic dysfunction and cognitive impairment remains unclear but is likely due to small soluble oligomers of Aβ (oAβ). Most studies have used chemical synthetic or cell-secreted Aβ oligomers to study their pathogenic mechanisms, but the Aβ derived from human AD brain tissue is less well characterized. Here we review updated knowledge on the extraction and characterization of bioactive human AD brain oAβ and the mechanisms by which they cause hippocampal synaptic dysfunction. Human AD brain-derived oAβ can impair hippocampal long-term potentiation (LTP) and enhance long-term depression (LTD). Many studies suggest that oAβ may directly disrupt neuronal NMDA receptors, AMPA receptors and metabotropic glutamate receptors (mGluRs). oAβ also impairs astrocytic synaptic functions, including glutamate uptake, D-serine release, and NMDA receptor function. We also discuss oAβ-induced neuronal hyperexcitation. These results may suggest a multi-target approach for the treatment of AD, including both oAβ neutralization and reversal of glutamate-mediated excitotoxicity.
Collapse
|
37
|
Cognitive Sequelae and Hippocampal Dysfunction in Chronic Kidney Disease following 5/6 Nephrectomy. Brain Sci 2022; 12:brainsci12070905. [PMID: 35884712 PMCID: PMC9321175 DOI: 10.3390/brainsci12070905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 01/18/2023] Open
Abstract
Neurological disorders are prevalent in patients with chronic kidney disease (CKD). Vascular factors and uremic toxins are involved with cognitive impairment in CKD. In addition, vascular dementia-induced alterations in the structure and function of the hippocampus can lead to deficits in hippocampal synaptic plasticity and cognitive function. However, regardless of this clinical evidence, the pathophysiology of cognitive impairment in patients with CKD is not fully understood. We used male Sprague Dawley rats and performed 5/6 nephrectomy to observe the changes in behavior, field excitatory postsynaptic potential, and immunostaining of the hippocampus following CKD progression. We measured the hippocampus volume on magnetic resonance imaging scans in the controls (n = 34) and end-stage renal disease (ESRD) hemodialysis patients (n = 42). In four cognition-related behavior assays, including novel object recognition, Y-maze, Barnes maze, and classical contextual fear conditioning, we identified deficits in spatial working memory, learning and memory, and contextual memory, as well as the ability to distinguish familiar and new objects, in the rats with CKD. Immunohistochemical staining of Na+/H+ exchanger1 was increased in the hippocampus of the CKD rat models. We performed double immunofluorescent staining for aquaporin-4 and glial fibrillary acidic protein and then verified the high coexpression in the hippocampus of the CKD rat model. Furthermore, results from recoding of the field excitatory postsynaptic potential (fEPSP) in the hippocampus showed the reduced amplitude and slope of fEPSP in the CKD rats. ESRD patients with cognitive impairment showed a significant decrease in the hippocampus volume compared with ESRD patients without cognitive impairment or the controls. Our findings suggest that uremia resulting from decreased kidney function may cause the destruction of the blood–brain barrier and hippocampus-related cognitive impairment in CKD.
Collapse
|
38
|
Deschamps C, Uyttersprot F, Debris M, Marié C, Fouquet G, Marcq I, Vilpoux C, Naassila M, Pierrefiche O. Anti-inflammatory drugs prevent memory and hippocampal plasticity deficits following initial binge-like alcohol exposure in adolescent male rats. Psychopharmacology (Berl) 2022; 239:2245-2262. [PMID: 35314896 DOI: 10.1007/s00213-022-06112-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 03/01/2022] [Indexed: 11/30/2022]
Abstract
RATIONALE Binge drinking during adolescence impairs learning and memory on the long term, and many studies suggest a role of neuroinflammation. However, whether neuroinflammation occurs after the very first exposures to alcohol remains unclear, while initial alcohol exposure impairs learning for several days in male rats. OBJECTIVES To investigate the role of neuroinflammation in the effects of only two binge-like episodes on learning and on neuronal plasticity in adolescent male rat hippocampus. METHODS Animals received two ethanol i.p. injections (3 g/kg) 9 h apart. Forty-eight hours later, we recorded long-term depression (LTD) and potentiation (LTP) in CA1 area of hippocampus slices. In isolated CA1, we measured immunolabelings for microglial activation and Toll-like receptor 4 (TLR4) and mRNA levels for several cytokines. RESULTS Forty-eight hours after the two binges, rats performed worse than control rats in novel object recognition, LTD was reduced, LTP was increased, and excitatory neurotransmission was more sensitive to an antagonist of the GluN2B subunit of the NMDA receptor. Exposure to ethanol with minocycline or indomethacin, two anti-inflammatory drugs, or with a TLR4 antagonist, prevented all effects of ethanol. Immunolabelings at 48 h showed a reduction of neuronal TLR4 that was prevented by minocycline pretreatment, while microglial reactivity was undetected and inflammatory cytokines mRNA levels were unchanged. CONCLUSION Two binge-like ethanol exposures during adolescence in rat involved neuroinflammation leading to changes in TLR4 expression and in GluN2B functioning inducing disturbances in synaptic plasticity and cognitive deficits. Anti-inflammatory drugs are good candidates to prevent brain function and memory deficits induced by few binge-drinking episodes.
Collapse
Affiliation(s)
- Chloé Deschamps
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Floriane Uyttersprot
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Margot Debris
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Constance Marié
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Grégory Fouquet
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Ingrid Marcq
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Catherine Vilpoux
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Mickael Naassila
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France
| | - Olivier Pierrefiche
- UMR1247 INSERM, Groupe de Recherche Sur L'Alcool Et Les Pharmacodépendances, Université de Picardie Jules Verne, Centre Universitaire de Recherche en Santé, Chemin du Thil, 80025, Amiens, France.
| |
Collapse
|
39
|
Contó MB, Pautassi RM, Camarini R. Rewarding and Antidepressant Properties of Ketamine and Ethanol: Effects on the Brain-Derived Neurotrophic Factor and TrkB and p75 NTR Receptors. Neuroscience 2022; 493:1-14. [PMID: 35469972 DOI: 10.1016/j.neuroscience.2022.04.015] [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: 01/02/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022]
Abstract
There is a high level of comorbidity between depression and alcohol use disorder. Subanesthetic doses of ketamine induce short-acting and enduring antidepressant effects after a single or a few administrations. Considering such comorbidity, we assessed, in Swiss male mice, if ketamine-induced antidepressant-like effects would alter ethanol's rewarding effects; and, if ethanol pretreatment would alter the rewarding and antidepressant effects of ketamine. The role of the brain-derived neurotrophic factor (BDNF) and its high and low affinity receptors TrkB and p75NTR, respectively, in both reward and depression-related behaviors is well established. The present study assessed, in outbred Swiss male mice, the expression of these proteins in the prefrontal cortex and hippocampus. Ketamine did not alter the development of ethanol-induced conditioned place preference (CPP), yet ethanol inhibited the expression of CPP induced by 50 mg/kg ketamine. The antidepressant action of 50 mg/kg ketamine was attenuated after repeated treatment (i.e., developed tolerance), an effect blocked by ethanol preexposure; ethanol also inhibited the antidepressant effect of 30 mg/kg ketamine. Ketamine (50 mg/kg) and Ethanol-Ketamine (50 mg/kg) groups showed lower levels of 145 kDa TrkB in the hippocampus than Saline-treated group. Ethanol-Ketamine (50 mg/kg) decreased the hippocampal expression of p75NTR compared to Saline-Saline and Saline-Ethanol groups. Ketamine (50 mg/kg) induced hippocampal downregulation of 145 kDa TrkB may contribute to ketamine-induced antidepressant tolerance. Likewise, a relationship between low hippocampal levels of p75NTR in the Ethanol-Ketamine (50 mg/kg) and ketamine-induced CPP blockade may be considered. The findings underscore potential ethanol-ketamine interactions likely to undermine ketamine putative antidepressant effects.
Collapse
Affiliation(s)
- Marcos Brandão Contó
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brasil.
| | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica M. y M. Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba C.P. 5000, Argentina; Facultad de Psicología, Universidad Nacional de Córdoba, Córdoba C.P. 5000 Argentina
| | - Rosana Camarini
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brasil.
| |
Collapse
|
40
|
Satoh R, Kawakami K, Nakadate K. Effects of Smart Drugs on Cholinergic System and Non-Neuronal Acetylcholine in the Mouse Hippocampus: Histopathological Approach. J Clin Med 2022; 11:jcm11123310. [PMID: 35743382 PMCID: PMC9224974 DOI: 10.3390/jcm11123310] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022] Open
Abstract
In recent years, people in the United States and other countries have been using smart drugs, called nootropic or cognitive enhancers, to improve concentration and memory learning skills. However, these drugs were originally prescribed for attention-deficit hyperactivity disorder and dementia, and their efficacy in healthy people has not yet been established. We focused on acetylcholine in the hippocampus, which is responsible for memory learning, and elucidate the long-term effects of smart drugs on the neural circuits. Smart drugs were administered orally in normal young mice for seven weeks. The hippocampus was sectioned and compared histologically by hematoxylin and eosin (HE) staining, immunohistochemistry for acetylcholine, and immunoelectron microscopy. There were no significant changes in acetylcholinesterase staining. However, in HE, we found perivascular edema, and choline acetyltransferase staining showed increased staining throughout the hippocampus and new signal induction in the perivascular area in the CA3, especially in the aniracetam and α-glyceryl phosphoryl choline group. Additionally, new muscarinic acetylcholine receptor signals were observed in the CA1 due to smart drug intake, suggesting that vasodilation might cause neuronal activation by increasing the influx of nutrients and oxygen. Moreover, these results suggest a possible new mechanism of acetylcholine-mediated neural circuit activation by smart drug intake.
Collapse
|
41
|
Khalife MR, Scott RC, Hernan AE. Mechanisms for Cognitive Impairment in Epilepsy: Moving Beyond Seizures. Front Neurol 2022; 13:878991. [PMID: 35645970 PMCID: PMC9135108 DOI: 10.3389/fneur.2022.878991] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
There has been a major emphasis on defining the role of seizures in the causation of cognitive impairments like memory deficits in epilepsy. Here we focus on an alternative hypothesis behind these deficits, emphasizing the mechanisms of information processing underlying healthy cognition characterized as rate, temporal and population coding. We discuss the role of the underlying etiology of epilepsy in altering neural networks thereby leading to both the propensity for seizures and the associated cognitive impairments. In addition, we address potential treatments that can recover the network function in the context of a diseased brain, thereby improving both seizure and cognitive outcomes simultaneously. This review shows the importance of moving beyond seizures and approaching the deficits from a system-level perspective with the guidance of network neuroscience.
Collapse
Affiliation(s)
- Mohamed R. Khalife
- Division of Neuroscience, Nemours Children's Health, Wilmington, DE, United States
- Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Rod C. Scott
- Division of Neuroscience, Nemours Children's Health, Wilmington, DE, United States
- Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
- Institute of Child Health, Neurosciences Unit University College London, London, United Kingdom
| | - Amanda E. Hernan
- Division of Neuroscience, Nemours Children's Health, Wilmington, DE, United States
- Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
42
|
Sun W, Li J, Li X, Chen X, Mei Y, Yang Y, An L. Aluminium oxide nanoparticles compromise spatial memory performance and proBDNF-mediated neuronal function in the hippocampus of rats. Part Fibre Toxicol 2022; 19:34. [PMID: 35538555 PMCID: PMC9087928 DOI: 10.1186/s12989-022-00477-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
Background Alumina nanoparticles (aluminaNPs), which are widely used in a range of daily and medical fields, have been shown to penetrate blood-brain barrier, and distribute and accumulate in different brain areas. Although oral treatment of aluminaNPs induces hippocampus-dependent learning and memory impairments, characteristic effects and exact mechanisms have not been fully elucidated. Here, male adult rats received a single bilateral infusion of aluminaNPs (10 or 20 µg/kg of body weight) into the hippocampal region, and their behavioral performance and neural function were assessed. Results The results indicated that the intra-hippocampus infusions at both doses of aluminaNPs did not cause spatial learning inability but memory deficit in the water maze task. This impairment was attributed to the effects of aluminaNP on memory consolidation phase through activation of proBDNF/RhoA pathway. Inhibition of the increased proBDNF by hippocampal infusions of p75NTR antagonist could effectively rescue the memory impairment. Incubation of aluminaNPs exaggerated GluN2B-dependent LTD induction with no effects on LTD expression in hippocampal slices. AluminaNP could also depress the amplitude of NMDA-GluN2B EPSCs. Meanwhile, increased reactive oxygen specie production was reduced by blocking proBDNF-p75NTR pathway in the hippocampal homogenates. Furthermore, the neuronal correlate of memory behavior was drastically weakened in the aluminaNP-infused groups. The dysfunction of synaptic and neuronal could be obviously mitigated by blocking proBDNF receptor p75NTR, implying the involvement of proBDNF signaling in aluminaNP-impaired memory process. Conclusions Taken together, our findings provide the first evidence that the accumulation of aluminaNPs in the hippocampus exaggeratedly activates proBDNF signaling, which leads to neural and memory impairments.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Jia Li
- College of Acupuncture and Orthopedics, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xiaoliang Li
- Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Xiao Chen
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China.,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China
| | - Yazi Mei
- Graduate School of Guangzhou, University of Chinese Medicine, Guangzhou, 510006, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Lei An
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Behavioral Neuroscience Laboratory, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China. .,Department of Neurology, Jinan Geriatric/Rehabilitation Hospital, Jinan, 250013, China. .,Graduate School of Guangzhou, University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
43
|
Stacho M, Manahan-Vaughan D. The Intriguing Contribution of Hippocampal Long-Term Depression to Spatial Learning and Long-Term Memory. Front Behav Neurosci 2022; 16:806356. [PMID: 35548697 PMCID: PMC9084281 DOI: 10.3389/fnbeh.2022.806356] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/10/2022] [Indexed: 01/03/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) comprise the principal cellular mechanisms that fulfill established criteria for the physiological correlates of learning and memory. Traditionally LTP, that increases synaptic weights, has been ascribed a prominent role in learning and memory whereas LTD, that decreases them, has often been relegated to the category of "counterpart to LTP" that serves to prevent saturation of synapses. In contradiction of these assumptions, studies over the last several years have provided functional evidence for distinct roles of LTD in specific aspects of hippocampus-dependent associative learning and information encoding. Furthermore, evidence of the experience-dependent "pruning" of excitatory synapses, the majority of which are located on dendritic spines, by means of LTD has been provided. In addition, reports exist of the temporal and physical restriction of LTP in dendritic compartments by means of LTD. Here, we discuss the role of LTD and LTP in experience-dependent information encoding based on empirical evidence derived from conjoint behavioral and electrophysiological studies conducted in behaving rodents. We pinpoint the close interrelation between structural modifications of dendritic spines and the occurrence of LTP and LTD. We report on findings that support that whereas LTP serves to acquire the general scheme of a spatial representation, LTD enables retention of content details. We argue that LTD contributes to learning by engaging in a functional interplay with LTP, rather than serving as its simple counterpart, or negator. We propose that similar spatial experiences that share elements of neuronal representations can be modified by means of LTD to enable pattern separation. Therewith, LTD plays a crucial role in the disambiguation of similar spatial representations and the prevention of generalization.
Collapse
|
44
|
Burjanadze MA, Dashniani MG, Solomonia RO, Beselia GV, Tsverava L, Lagani V, Chkhikvishvili NC, Naneishvili TL, Kruashvili LB, Chighladze MR. Age-related changes in medial septal cholinergic and GABAergic projection neurons and hippocampal neurotransmitter receptors: relationship with memory impairment. Exp Brain Res 2022; 240:1589-1604. [PMID: 35357523 DOI: 10.1007/s00221-022-06354-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
The hippocampus, which provides cognitive functions, has been shown to become highly vulnerable during aging. One important modulator of the hippocampal neural network is the medial septum (MS). The present study attempts to determine how age-related mnemonic dysfunction is associated with neurochemical changes in the septohippocampal (SH) system, using behavioral and immunochemical experiments performed on young-adult, middle-aged and aged rats. According to these behavioral results, the aged and around 52.8% of middle-aged rats (within the "middle-aged-impaired" sub-group) showed both impaired spatial reference memory in the Morris water maze and habituation in the open field. Immunohistochemical studies revealed a significant decrease in the number of MS choline acetyltransferase immunoreactive cells in the aged and all middle-aged rats, in comparison to the young; however the number of gamma-aminobutyric acid-ergic (GABAergic) parvalbumin immunoreactive cells was higher in middle-aged-impaired and older rats compared to young and middle-aged-unimpaired rats. Western Blot analysis moreover showed a decrease in the level of expression of cholinergic, GABAergic and glutamatergic receptors in the hippocampus of middle-aged-impaired and aged rats in contrast to middle-aged-unimpaired and young rats. The present results demonstrate for the first time that a decrease in the expression level of hippocampal receptors in naturally aged rats with impaired cognitive abilities occurs in parallel with an increase in the number of GABAergic neurons in the MS, and it highlights the particular importance of inhibitory signaling in the SH network for memory function.
Collapse
Affiliation(s)
- Maia A Burjanadze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.
| | - Manana G Dashniani
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Revaz O Solomonia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.,Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Gela V Beselia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.,Department of Physiology and Pharmacology, Petre Shotadze Tbilisi Medical Academy, 0144, Tbilisi, Georgia
| | - Lia Tsverava
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.,Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Vincenzo Lagani
- Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Nino C Chkhikvishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Temur L Naneishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Lali B Kruashvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Mariam R Chighladze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| |
Collapse
|
45
|
Dysmetabolism and Neurodegeneration: Trick or Treat? Nutrients 2022; 14:nu14071425. [PMID: 35406040 PMCID: PMC9003269 DOI: 10.3390/nu14071425] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests the existence of a strong link between metabolic syndrome and neurodegeneration. Indeed, epidemiologic studies have described solid associations between metabolic syndrome and neurodegeneration, whereas animal models contributed for the clarification of the mechanistic underlying the complex relationships between these conditions, having the development of an insulin resistance state a pivotal role in this relationship. Herein, we review in a concise manner the association between metabolic syndrome and neurodegeneration. We start by providing concepts regarding the role of insulin and insulin signaling pathways as well as the pathophysiological mechanisms that are in the genesis of metabolic diseases. Then, we focus on the role of insulin in the brain, with special attention to its function in the regulation of brain glucose metabolism, feeding, and cognition. Moreover, we extensively report on the association between neurodegeneration and metabolic diseases, with a particular emphasis on the evidence observed in animal models of dysmetabolism induced by hypercaloric diets. We also debate on strategies to prevent and/or delay neurodegeneration through the normalization of whole-body glucose homeostasis, particularly via the modulation of the carotid bodies, organs known to be key in connecting the periphery with the brain.
Collapse
|
46
|
Bie B, Wu J, Lin F, Naguib M, Xu J. Suppression of hippocampal GABAergic transmission impairs memory in rodent models of Alzheimer's disease. Eur J Pharmacol 2022; 917:174771. [PMID: 35041847 DOI: 10.1016/j.ejphar.2022.174771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/27/2022]
Abstract
Emerging evidence demonstrates the potential involvement of hippocampal GABAergic transmission in the process of memory acquisition and consolidation, while no consistent report is available to address the adaptation of hippocampal GABAergic transmission and its contribution to memory deficiency in the setting of Alzheimer's disease (AD). Brain-derived neurotrophic factor (BDNF) is a key molecule that regulates GABAergic transmission. In the brain, mature BDNF is generated from the proteolytic cleavage of proBDNF, while BDNF and proBDNF have differential effects on central GABAergic transmission. First, the present study reports a remarkable increase of proBDNF/BNDF ratio in the hippocampal CA1 area in rodent models of AD, indicating a potential impaired process of BDNF maturation from proBDNF cleavage. We report a suppressed hippocampal GABAergic strength, potentially resulting from the reduced expression of anion chloride co-transporter KCC2 and subsequent positive shift of GABAergic Cl-equilibrium potential (ECl-), which is attenuated by microinjection of BDNF with proBDNF inhibitor TAT-Pep5. We also show that normalization of proBDNF/BDNF signaling or GABAergic ECl-by intracerebroventricular (i.c.v.) administration of bumetanide remarkably improves the cognitive performance in Morris water maze test and fear conditioning test in rodent models of AD. These results demonstrate a critical role of hippocampal proBDNF/BDNF in regulating GABAergic transmission and contributing to memory dysfunction in rodent models of AD.
Collapse
Affiliation(s)
- Bihua Bie
- Department of Pain Management, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jiang Wu
- Department of Pain Management, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Mohamed Naguib
- Department of General Anesthesiology, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jijun Xu
- Department of Pain Management, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
47
|
Kallergi E, Daskalaki AD, Kolaxi A, Camus C, Ioannou E, Mercaldo V, Haberkant P, Stein F, Sidiropoulou K, Dalezios Y, Savitski MM, Bagni C, Choquet D, Hosy E, Nikoletopoulou V. Dendritic autophagy degrades postsynaptic proteins and is required for long-term synaptic depression in mice. Nat Commun 2022; 13:680. [PMID: 35115539 PMCID: PMC8814153 DOI: 10.1038/s41467-022-28301-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/14/2022] [Indexed: 01/18/2023] Open
Abstract
The pruning of dendritic spines during development requires autophagy. This process is facilitated by long-term depression (LTD)-like mechanisms, which has led to speculation that LTD, a fundamental form of synaptic plasticity, also requires autophagy. Here, we show that the induction of LTD via activation of NMDA receptors or metabotropic glutamate receptors initiates autophagy in the postsynaptic dendrites in mice. Dendritic autophagic vesicles (AVs) act in parallel with the endocytic machinery to remove AMPA receptor subunits from the membrane for degradation. During NMDAR-LTD, key postsynaptic proteins are sequestered for autophagic degradation, as revealed by quantitative proteomic profiling of purified AVs. Pharmacological inhibition of AV biogenesis, or conditional ablation of atg5 in pyramidal neurons abolishes LTD and triggers sustained potentiation in the hippocampus. These deficits in synaptic plasticity are recapitulated by knockdown of atg5 specifically in postsynaptic pyramidal neurons in the CA1 area. Conducive to the role of synaptic plasticity in behavioral flexibility, mice with autophagy deficiency in excitatory neurons exhibit altered response in reversal learning. Therefore, local assembly of the autophagic machinery in dendrites ensures the degradation of postsynaptic components and facilitates LTD expression.
Collapse
Affiliation(s)
- Emmanouela Kallergi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | | | - Angeliki Kolaxi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Come Camus
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | - Evangelia Ioannou
- School of Biological Sciences, University of Crete, Heraklion, 70013, Greece
| | - Valentina Mercaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
| | - Per Haberkant
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Frank Stein
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Yannis Dalezios
- School of Medicine, University of Crete, Heraklion, 71003, Greece
- Institute of Applied and Computational Mathematics (IACM), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Mikhail M Savitski
- Proteomic Core Facility (PCF), European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), University of Rome Tor Vergata, Rome, 00133, Italy
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, 1005, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, F-33000, Bordeaux, France
| | - Eric Hosy
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, F-33000, Bordeaux, France
| | | |
Collapse
|
48
|
Lee Y, Bortolotto ZA, Bradley CA, Sanderson TM, Zhuo M, Kaang BK, Collingridge GL. The GSK-3 Inhibitor CT99021 Enhances the Acquisition of Spatial Learning and the Accuracy of Spatial Memory. Front Mol Neurosci 2022; 14:804130. [PMID: 35153671 PMCID: PMC8829050 DOI: 10.3389/fnmol.2021.804130] [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: 10/28/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a Ser/Thr protein kinase that regulates many cellular processes, including synaptic plasticity. Previously, we reported that inhibition of GSK-3 prevents the induction of one of the major forms of synaptic plasticity, N-methyl-D-aspartate receptor (NMDAR)-dependent long-term depression (LTD), in hippocampal slices. In the present study, we have investigated the effects of inhibiting GSK-3 on learning and memory in healthy naïve animals. Systemic administration of a highly selective GSK-3 inhibitor, CT99021, reversibly blocked NMDAR-dependent LTD in the CA1 region of the hippocampus in anesthetized adult mice. In behavioral tasks, CT99021 had no effect on locomotor activity, anxiety, hippocampus-dependent contextual fear memory, and hippocampus-dependent reversal learning. However, CT99021 facilitated the rate of learning in the Morris water maze (MWM) and T-maze and enhanced the accuracy of long-term spatial memory in the MWM. These findings suggest that GSK-3 regulates the accuracy of spatial memory acquisition and recall.
Collapse
Affiliation(s)
- Yeseul Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Zuner A. Bortolotto
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Clarrisa A. Bradley
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Genes and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Thomas M. Sanderson
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Min Zhuo
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Bong-Kiun Kaang
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- *Correspondence: Bong-Kiun Kaang,
| | - Graham L. Collingridge
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Graham L. Collingridge,
| |
Collapse
|
49
|
Laha K, Zhu M, Gemperline E, Rau V, Li L, Fanselow MS, Lennertz R, Pearce RA. CPP impairs contextual learning at concentrations below those that block pyramidal neuron NMDARs and LTP in the CA1 region of the hippocampus. Neuropharmacology 2022; 202:108846. [PMID: 34687710 PMCID: PMC8627488 DOI: 10.1016/j.neuropharm.2021.108846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/29/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Drugs that block N-methyl-d-aspartate receptors (NMDARs) suppress hippocampus-dependent memory formation; they also block long-term potentiation (LTP), a cellular model of learning and memory. However, the fractional block that is required to achieve these effects is unknown. Here, we measured the dose-dependent suppression of contextual memory in vivo by systemic administration of the competitive antagonist (R,S)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP); in parallel, we measured the concentration-dependent block by CPP of NMDAR-mediated synapses and LTP of excitatory synapses in hippocampal brain slices in vitro. We found that the dose of CPP that suppresses contextual memory in vivo (EC50 = 2.3 mg/kg) corresponds to a free concentration of 53 nM. Surprisingly, applying this concentration of CPP to hippocampal brain slices had no effect on the NMDAR component of evoked field excitatory postsynaptic potentials (fEPSPNMDA), or on LTP. Rather, the IC50 for blocking the fEPSPNMDA was 434 nM, and for blocking LTP was 361 nM - both nearly an order of magnitude higher. We conclude that memory impairment produced by systemically administered CPP is not due primarily to its blockade of NMDARs on hippocampal pyramidal neurons. Rather, systemic CPP suppresses memory formation by actions elsewhere in the memory-encoding circuitry.
Collapse
Affiliation(s)
- Kurt Laha
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Mengwen Zhu
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Erin Gemperline
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| | - Vinuta Rau
- Department of Anesthesiology, University of California-San Francisco, San Francisco, CA, USA.
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
| | - Michael S Fanselow
- Departments of Psychology and Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Richard Lennertz
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Robert A Pearce
- Department of Anesthesiology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
50
|
Age-Dependent Contributions of NMDA Receptors and L-Type Calcium Channels to Long-Term Depression in the Piriform Cortex. Int J Mol Sci 2021; 22:ijms222413551. [PMID: 34948347 PMCID: PMC8706958 DOI: 10.3390/ijms222413551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
In the hippocampus, the contributions of N-methyl-D-aspartate receptors (NMDARs) and L-type calcium channels (LTCCs) to neuronal transmission and synaptic plasticity change with aging, underlying calcium dysregulation and cognitive dysfunction. However, the relative contributions of NMDARs and LTCCs in other learning encoding structures during aging are not known. The piriform cortex (PC) plays a significant role in odor associative memories, and like the hippocampus, exhibits forms of long-term synaptic plasticity. Here, we investigated the expression and contribution of NMDARs and LTCCs in long-term depression (LTD) of the PC associational fiber pathway in three cohorts of Sprague Dawley rats: neonatal (1-2 weeks), young adult (2-3 months) and aged (20-25 months). Using a combination of slice electrophysiology, Western blotting, fluorescent immunohistochemistry and confocal imaging, we observed a shift from an NMDAR to LTCC mediation of LTD in aged rats, despite no difference in the amount of LTD expression. These changes in plasticity are related to age-dependent differential receptor expression in the PC. LTCC Cav1.2 expression relative to postsynaptic density protein 95 is increased in the associational pathway of the aged PC layer Ib. Enhanced LTCC contribution in synaptic depression in the PC may contribute to altered olfactory function and learning with aging.
Collapse
|