1
|
Liu L, Jia P, Liu T, Liang J, Dang Y, Rastegar-Kashkooli Y, Li Q, Liu J, Man J, Zhao T, Xing N, Wang F, Chen X, Zhang J, Jiang C, Zille M, Zhang Z, Fan X, Wang J, Wang J. AGEING RESEARCH REVIEWS (ARR-D-24-01334R1) Metabolic dysfunction contributes to mood disorders after traumatic brain injury. Ageing Res Rev 2024:102652. [PMID: 39746403 DOI: 10.1016/j.arr.2024.102652] [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: 09/05/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) presents significant risks concerning mortality and morbidity. Individuals who suffer from TBI may exhibit mood disorders, including anxiety and depression. Both preclinical and clinical research have established correlations between TBI and disturbances in the metabolism of amino acids, lipids, iron, zinc, and copper, which are implicated in the emergence of mood disorders post-TBI. The purpose of this review is to elucidate the impact of metabolic dysfunction on mood disorders following TBI and to explore potential strategies for mitigating anxiety and depression symptoms. We researched the PubMed and Web of Science databases to delineate the mechanisms by which metabolic dysfunction contributes to mood disorders in the context of TBI. Particular emphasis was placed on the roles of glutamate, kynurenine, lipids, iron, zinc, and copper metabolism. Metabolic dysfunction is linked to mood disorders post-TBI through multiple pathways, encompassing the glutamatergic system, the kynurenine pathway, endocannabinoids, iron deposition, iron-related ferroptosis, zinc deficiency, and copper dysregulation. Furthermore, this review addresses the influence of metabolic dysfunction on mood disorders in the elderly demographic following TBI. Targeting metabolic dysfunction for therapeutic intervention appears promising in alleviating symptoms of anxiety and depression that arise after TBI. While further investigation is warranted to delineate the underlying pathophysiologic mechanisms of mood disorders post-TBI, current evidence underscores the potential contribution of metabolic dysfunction to these conditions. Therefore, rectifying metabolic dysfunction represents a viable and strategic approach to addressing mood disorders following TBI.
Collapse
Affiliation(s)
- Lang Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Tongzhou Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Jiaxin Liang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Yijia Dang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; School of International Education, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| | - Jingqi Liu
- Guangzhou Yucai Middle School, Guangzhou 510050, Guangdong, China.
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Ting Zhao
- Department of Neurology, Henan Provincial People's Hospital, The People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China.
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China.
| | - Xuemei Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China.
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, The People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China.
| | - Chao Jiang
- Department of Neurology, Henan Provincial People's Hospital, The People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China.
| | - Marrietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, 1090 Vienna, Austria.
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
2
|
Leana-Sandoval G, Kolli AV, Sandoval MA, Saavedra E, Li KH, Chen LY, Burlingame AL, Ramírez-Franco J, Díaz-Alonso J. The VGCC auxiliary subunit α2δ1 is an extracellular GluA1 interactor and regulates LTP, spatial memory, and seizure susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626379. [PMID: 39677598 PMCID: PMC11642997 DOI: 10.1101/2024.12.02.626379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Activity-dependent synaptic accumulation of AMPA receptors (AMPARs) and subsequent long-term synaptic strengthening underlie different forms of learning and memory. The AMPAR subunit GluA1 amino-terminal domain is essential for synaptic docking of AMPAR during LTP, but the precise mechanisms involved are not fully understood. Using unbiased proteomics, we identified the epilepsy and intellectual disability-associated VGCC auxiliary subunit α2δ1 as a candidate extracellular AMPAR slot. Presynaptic α2δ1 deletion in CA3 affects synaptic AMPAR incorporation during long-term potentiation, but not basal synaptic transmission, at CA1 synapses. Consistently, mice lacking α2δ1 in CA3 display a specific impairment in CA1-dependent spatial memory, but not in memory tests involving other cortical regions. Decreased seizure susceptibility in mice lacking α2δ1 in CA3 suggests a regulation of circuit excitability by α2δ1/AMPAR interactions. Our study sheds light on the regulation of activity-dependent AMPAR trafficking, and highlights the synaptic organizing roles of α2δ1.
Collapse
Affiliation(s)
- Gerardo Leana-Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Ananth V. Kolli
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Matthew A. Sandoval
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Emily Saavedra
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Kathy H. Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lulu Y. Chen
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| | - Alma L. Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jorge Ramírez-Franco
- Institut de Neurosciences de la Timone, Aix-Marseille Université (AMU) & CNRS, UMR7289, 13005, Marseille, France
| | - Javier Díaz-Alonso
- Department of Anatomy & Neurobiology, University of California at Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California at Irvine, CA, USA
| |
Collapse
|
3
|
Salvi V, Tripodi B, Cerveri G, Migliarese G, Bertoni L, Nibbio G, Barlati S, Vita A, Mencacci C. Insulin-resistance as a modifiable pathway to cognitive dysfunction in schizophrenia: A systematic review. Schizophr Res 2024; 274:78-89. [PMID: 39265262 DOI: 10.1016/j.schres.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/21/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Cognitive deficits are difficult to treat and negatively influence quality of life and functional outcomes of persons with schizophrenia. In the last twenty years, extensive literature demonstrated that persons with diabetes and insulin resistance (IR) also display cognitive deficits. Being type 2 diabetes (T2DM) and IR highly frequent in persons with schizophrenia, it is plausible to hypothesize that these conditions might play a role in determining dyscognition. If that is the case, acting on glucose dysmetabolism may eventually improve cognitive functioning. This review aims at: 1. evaluating the association between IR or T2DM and cognitive dysfunction in schizophrenia; 2. reviewing the evidence that pharmacological treatment of IR or T2DM may improve dyscognition in schizophrenia. METHODS Two systematic searches were conducted in PubMed, PsycInfo, and Scopus. We followed the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. RESULTS From the first search we included 17 studies, 8 on the effects of T2DM and 9 on the effects of IR-other prediabetes measures on cognition in persons with schizophrenia. From the second search we included 12 studies investigating the effect on cognition of glucose (4 studies), insulin (2 studies), metformin (2 studies), PPAR-γ agonists (2 studies), GLP-1 agonist (1 study), bromocriptine (1 study). CONCLUSIONS T2DM was associated with worse cognitive function in persons with schizophrenia, while IR was less strongly associated with cognitive dysfunction. Evidence regarding the efficacy of glucose-lowering medications on cognition in schizophrenia is inconclusive, yet methodological issues likely contribute to explain conflicting results.
Collapse
Affiliation(s)
- Virginio Salvi
- Department of Mental Health and Addiction, ASST Crema, L.go Ugo Dossena 2, 26013 Crema, CR, Italy.
| | - Beniamino Tripodi
- Department of Mental Health and Addiction, ASST Crema, L.go Ugo Dossena 2, 26013 Crema, CR, Italy
| | - Giancarlo Cerveri
- Department of Mental Health and Addiction, ASST Lodi, Via Mosè Bianchi 26, 26900 Lodi, Italy
| | - Giovanni Migliarese
- Department of Mental Health and Addiction, ASST Pavia, C.so Milano 19, 27029 Vigevano, PV, Italy
| | - Lorenzo Bertoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Gabriele Nibbio
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Claudio Mencacci
- Director Emeritus, Department of Neurosciences-Mental Health, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
4
|
Lee Y, Jeon J, Son SR, Cho E, Moon S, Park AY, Chae HJ, Bae HJ, Moon M, Jeon SJ, Jang DS, Kim DH. 3,4,5-trimethoxycinnamic acid methyl ester isolated from Polygala tenuifolia enhances hippocampal LTP through PKA and calcium-permeable AMPA receptor. Biochem Pharmacol 2024; 230:116622. [PMID: 39542183 DOI: 10.1016/j.bcp.2024.116622] [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: 06/26/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disorder characterized by progressive cognitive decline and neuronal death due to extracellular deposition of amyloid β (Aβ) and intracellular deposition of tau proteins. Recently approved antibody drugs targeting Aβ have been shown to slow the progression of the disease, but they have minimal effects on cognitive improvement. Therefore, there is a need to develop drugs with cognitive-enhancing effects that can be used in conjunction with these antibody treatments. In this study, we investigated whether Polygala tenuifolia (PT), traditionally known for its cognitive-enhancing effects, can improve synaptic plasticity and identified its active components and mechanisms. PT demonstrated a dose-dependent effect in enhancing long-term potentiation (LTP), and among its components, 3,4,5-trimethoxycinnamic acid methyl ester (TMCA) showed a similar LTP-enhancing effect. TMCA increased the phosphorylation of the GluA1 subunit of the α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors and increased the amount of GluA1 on the synapse without affecting the amount of GluA2. Additionally, the increase in GluA1 induced by TMCA was inhibited by a PKA inhibitor. Consistent with these results, the enhancement of LTP by TMCA was inhibited by a GluA1 antagonist and a PKA inhibitor. In silico molecular docking experiments confirmed that TMCA binds to PKA. Finally, we confirmed the LTP-enhancing effect of TMCA in hippocampal slices from 5XFAD mice. These results suggest that PT and its active component, TMCA, can interact with PKA to enhance LTP, indicating the potential for improving cognitive function in AD patients.
Collapse
Affiliation(s)
- Yehee Lee
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Jieun Jeon
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - So-Ri Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Eunbi Cho
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Somin Moon
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - A Young Park
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Hye Ji Chae
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Ho Jung Bae
- Agriculture and Life Science Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea.
| | - Se Jin Jeon
- Department of Pharmacology, College of Medicine, Hallym University, Gangwon, Republic of Korea.
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Dong Hyun Kim
- Department of Advanced Translational Medicine, Graduate School, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; Department of Pharmacology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea; Institute of Biomedical Sciences & Technology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
5
|
Zborowski VA, Martins CC, Marques LS, Heck SO, Nogueira CW. A chloro substituted organoselenium mitigates stress-associated memory impairment and hippocampal glutamatergic function in a repeated Forced Swim Stress Model. Neuroscience 2024; 563:110-116. [PMID: 39521324 DOI: 10.1016/j.neuroscience.2024.11.007] [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: 07/01/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Stress is triggered by a threatening event that alters the regulation of emotion, behavior, and cognition. The effects of stress on memory in animal models are well-documented. Firstly, this study aimed to determine whether the repeated forced swim stress (FSS) protocol induces memory impairment comparable to single prolonged stress (SPS) in the Y-maze test. The second objective was to evaluate whether (p-ClPhSe)2 pretreatment mitigates stress-associated memory impairment and hippocampal glutamatergic neurotransmission in FSS-exposed mice. Mice subjected to FSS and SPS protocols reduced time spent in the novel arm of the Y-maze test compared to the control group, with no observed changes in locomotor or exploratory behavior. (p-ClPhSe)2 was administered to mice at a dose of 5 mg/kg, 30 min before the first forced swimming session on days 1 and 2. Mice underwent a Y-maze test, after which they were euthanized, and hippocampal samples were collected. (p-ClPhSe)2 pretreatment protected against the reduction in time spent in the novel arm by mice subjected to FSS. Repeated FSS exposure increased hippocampal protein levels of NMDAR subunits 2A, 2B, and EAAT1 compared to controls. (p-ClPhSe)2 pretreatment prevented this increase. In conclusion, (p-ClPhSe)2 mitigated stress-induced memory impairment in FSS-exposed mice, normalizing hippocampal NMDAR 2A, 2B, and EAAT1 protein levels.
Collapse
Affiliation(s)
- Vanessa A Zborowski
- Laboratory of Synthesis Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS Zip Code:97105-900, Brazil.
| | - Carolina C Martins
- Laboratory of Synthesis Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS Zip Code:97105-900, Brazil
| | - Luiza S Marques
- Laboratory of Synthesis Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS Zip Code:97105-900, Brazil
| | - Suélen O Heck
- Laboratory of Synthesis Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS Zip Code:97105-900, Brazil
| | - Cristina W Nogueira
- Laboratory of Synthesis Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS Zip Code:97105-900, Brazil
| |
Collapse
|
6
|
Caruso V, Rigoli L. Beyond Wolfram Syndrome 1: The WFS1 Gene's Role in Alzheimer's Disease and Sleep Disorders. Biomolecules 2024; 14:1389. [PMID: 39595565 PMCID: PMC11591713 DOI: 10.3390/biom14111389] [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: 09/27/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The WFS1 gene was first identified in Wolfram Syndrome 1 (WS1), a rare autosomal recessive genetic disorder characterized by severe and progressive neurodegenerative changes. WFS1's role in various cellular mechanisms, particularly in calcium homeostasis and the modulation of endoplasmic reticulum (ER) stress, suggests its potential involvement in the pathogenesis of Alzheimer's disease (AD) and sleep disorders. Because it is involved in maintaining ER balance, calcium signaling, and stress responses, WFS1 plays a multifaceted role in neuronal health. Numerous studies have shown that the absence or improper expression of WFS1 disrupts these cellular processes, leading to neurodegeneration and making neurons more vulnerable. In AD, WFS1 dysfunction is thought to contribute to the accumulation of amyloid-β (Aβ) plaques and tau tangles, thereby accelerating disease progression. Additionally, WFS1 plays an essential role in sleep regulation by influencing neuronal excitability and neurotransmitter release, which may explain the sleep disturbances frequently observed in neurodegenerative diseases. Due to its involvement in the pathological mechanisms of AD and sleep disorders, WFS1 is regarded as a potential early diagnostic marker for these diseases. Further research is required to fully elucidate WFS1's role in the cellular pathway, perhaps facilitating the development of new therapeutic strategies for Alzheimer's disease and sleep disorders.
Collapse
Affiliation(s)
- Valerio Caruso
- Department of Neuroscience, Psychiatric Section, Azienda Ospedaliera Universitaria Pisana (A.U.O.P.), 56126 Pisa, Italy;
| | - Luciana Rigoli
- Department of Human Pathology of Adulthood and Childhood G. Barresi, University of Messina, 98125 Messina, Italy
| |
Collapse
|
7
|
Chen L, Li Y, Liu M, Lan Z, Zhang X, Yang X, Zhao Q, Wang S, Xu L, Zhou Y, Kuang Y, Suzuki T, Tabuchi K, Takahashi E, Zhou M, Chen CD, Xu T, Li W. Desipramine reverses remote memory deficits by activating calmodulin-CaMKII pathway in a UTX knockout mouse model of Kabuki syndrome. Gen Psychiatr 2024; 37:e101430. [PMID: 39493372 PMCID: PMC11529476 DOI: 10.1136/gpsych-2023-101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 06/12/2024] [Indexed: 11/05/2024] Open
Abstract
Background Kabuki syndrome (KS) is a rare developmental disorder characterised by multiple congenital anomalies and intellectual disability. UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome), which encodes a histone demethylase, is one of the two major pathogenic risk genes for KS. Although intellectual disability is a key phenotype of KS, the role of UTX in cognitive function remains unclear. Currently, no targeted therapies are available for KS. Aims This study aimed to investigate how UTX regulates cognition, to explore the mechanisms underlying UTX dysfunction and to identify potential molecular targets for treatment. Methods We generated UTX conditional knockout mice and found that UTX deletion downregulated calmodulin transcription by disrupting H3K27me3 (trimethylated histone H3 at lysine 27) demethylation. Results UTX-knockout mice showed decreased phosphorylation of calcium / calmodulin-dependent protein kinase II, impaired long-term potentiation and deficit in remote contextual fear memory. These effects were reversed by an Food and Drug Administration-approved drug desipramine. Conclusions Our results reveal an epigenetic mechanism underlying the important role of UTX in synaptic plasticity and cognitive function, and suggest that desipramine could be a potential treatment for KS.
Collapse
Affiliation(s)
- Lei Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Minggang Liu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Mental Health and Drug Discovery, Oujiang Laboratory, Wenzhou, Zhejiang, China
| | - Zhaohui Lan
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiujuan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Shuai Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Longyong Xu
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yifang Kuang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Tatsuo Suzuki
- Shinshu University School of Medicine, Nagano, Japan
| | | | - Eiki Takahashi
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Miou Zhou
- Western University of Health Sciences, Pomona, California, USA
| | - Charlie Degui Chen
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tianle Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
- WLA Laboratories, World Laureates Association, Shanghai, China
| |
Collapse
|
8
|
Slater CR. Neuromuscular Transmission in a Biological Context. Compr Physiol 2024; 14:5641-5702. [PMID: 39382166 DOI: 10.1002/cphy.c240001] [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: 10/10/2024]
Abstract
Neuromuscular transmission is the process by which motor neurons activate muscle contraction and thus plays an essential role in generating the purposeful body movements that aid survival. While many features of this process are common throughout the Animal Kingdom, such as the release of transmitter in multimolecular "quanta," and the response to it by opening ligand-gated postsynaptic ion channels, there is also much diversity between and within species. Much of this diversity is associated with specialization for either slow, sustained movements such as maintain posture or fast but brief movements used during escape or prey capture. In invertebrates, with hydrostatic and exoskeletons, most motor neurons evoke graded depolarizations of the muscle which cause graded muscle contractions. By contrast, vertebrate motor neurons trigger action potentials in the muscle fibers which give rise to all-or-none contractions. The properties of neuromuscular transmission, in particular the intensity and persistence of transmitter release, reflect these differences. Neuromuscular transmission varies both between and within individual animals, which often have distinct tonic and phasic subsystems. Adaptive plasticity of neuromuscular transmission, on a range of time scales, occurs in many species. This article describes the main steps in neuromuscular transmission and how they vary in a number of "model" species, including C. elegans , Drosophila , zebrafish, mice, and humans. © 2024 American Physiological Society. Compr Physiol 14:5641-5702, 2024.
Collapse
|
9
|
Baghcheghi Y, Razazpour F, Seyedi F, Arefinia N, Hedayati-Moghadam M. Exploring the molecular mechanisms of PPARγ agonists in modulating memory impairment in neurodegenerative disorders. Mol Biol Rep 2024; 51:945. [PMID: 39215798 DOI: 10.1007/s11033-024-09850-6] [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: 05/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Neurodegenerative diseases are characterized by progressive memory impairment and cognitive decline. This review aims to unravel the molecular mechanisms involved in the enhancement of memory function and mitigation of memory impairment through the activation of PPARγ agonists in neurodegenerative diseases. The findings suggest that PPARγ agonists modulate various molecular pathways involved in memory formation and maintenance. Activation of PPARγ enhances synaptic plasticity, promotes neuroprotection, suppresses neuroinflammation, attenuates oxidative stress, and regulates amyloid-beta metabolism. The comprehensive understanding of these molecular mechanisms would facilitate the development of novel therapeutic approaches targeting PPARγ to improve memory function and ultimately to alleviate the burden of neurodegenerative diseases. Further research, including clinical trials, is warranted to explore the efficacy, safety, and optimal use of specific PPARγ agonists as potential therapeutic agents in the treatment of memory impairments associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fateme Razazpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fatemeh Seyedi
- Department of Anatomical Sciences, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
10
|
Grippe T, Shamli-Oghli Y, Darmani G, Nankoo JF, Raies N, Sarica C, Arora T, Gunraj C, Ding MYR, Rinchon C, DiLuca DG, Pichardo S, Cardoso F, Lozano AM, Chen R. Plasticity-Induced Effects of Theta Burst Transcranial Ultrasound Stimulation in Parkinson's Disease. Mov Disord 2024; 39:1364-1374. [PMID: 38787806 DOI: 10.1002/mds.29836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Low-intensity transcranial ultrasound stimulation (TUS) is a noninvasive brain stimulation (NIBS) technique with high spatial specificity. Previous studies showed that TUS delivered in a theta burst pattern (tbTUS) increased motor cortex (MI) excitability up to 30 minutes due to long-term potentiation (LTP)-like plasticity. Studies using other forms of NIBS suggested that cortical plasticity may be impaired in patients with Parkinson's disease (PD). OBJECTIVE The aim was to investigate the neurophysiological effects of tbTUS in PD patients off and on dopaminergic medications compared to healthy controls. METHODS We studied 20 moderately affected PD patients in on and off dopaminergic medication states (7 with and 13 without dyskinesia) and 17 age-matched healthy controls in a case-controlled study. tbTUS was applied for 80 seconds to the MI. Motor-evoked potentials (MEP), short-interval intracortical inhibition (SICI), and short-interval intracortical facilitation (SICF) were recorded at baseline, and at 5 minutes (T5), T30, and T60 after tbTUS. Motor Unified Parkinson's Disease Rating Scale (mUPDRS) was measured at baseline and T60. RESULTS tbTUS significantly increased MEP amplitude at T30 compared to baseline in controls and in PD patients on but not in PD patients off medications. SICI was reduced in PD off medications compared to controls. tbTUS did not change in SICI or SICF. The bradykinesia subscore of mUPDRS was reduced at T60 compared to baseline in PD on but not in the off medication state. The presence of dyskinesia did not affect tbTUS-induced plasticity. CONCLUSIONS tbTUS-induced LTP plasticity is impaired in PD patients off medications and is restored by dopaminergic medications. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Talyta Grippe
- Department of Neurology, Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, Canada
- Neuroscience Graduate Program, Federal University of Minas Gerais, Belo Horizonte, Brazil
- University Health Network, Toronto, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada
| | | | | | | | | | - Can Sarica
- University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Tarun Arora
- University Health Network, Toronto, Canada
- Division of Clinical Neuroscience, Department of Neurology, Oslo University Hospital, Oslo, Norway
| | | | - Mandy Yi Rong Ding
- University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Cricia Rinchon
- University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Daniel G DiLuca
- Department of Neurology, Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, Canada
- University Health Network, Toronto, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Samuel Pichardo
- Cumming School of Medicine, Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Francisco Cardoso
- Neuroscience Graduate Program, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andres M Lozano
- University Health Network, Toronto, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Robert Chen
- Department of Neurology, Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, Canada
- University Health Network, Toronto, Canada
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
Tullis JE, Bayer KU. Induction of LTP mechanisms in dually innervated dendritic spines. Sci Rep 2024; 14:15855. [PMID: 38982271 PMCID: PMC11233660 DOI: 10.1038/s41598-024-66871-8] [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: 03/05/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024] Open
Abstract
Dendritic spines are the postsynaptic compartments of excitatory synapses, however, a substantial subset of spines additionally receives inhibitory input. In such dually innervated spines (DiSs), excitatory long-term potentiation (LTP) mechanisms are suppressed, but can be enabled by blocking tonic inhibitory GABAB receptor signaling. Here we show that LTP mechanisms at DiSs are also enabled by two other excitatory LTP stimuli. In hippocampal neurons, these chemical LTP (cLTP) stimuli induced robust movement of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) to DiSs. Such synaptic CaMKII accumulation is an essential LTP mechanism at singly innervated spines (SiSs). Indeed, CaMKII accumulation at DiSs was also accompanied by other readouts for successful LTP induction: spine growth and surface insertion of GluA1. Thus, DiSs are capable of the same LTP mechanisms as SiSs, although induction of these mechanism additionally requires either reduced inhibitory signaling or increased excitatory stimulation. This additional regulation may provide further computational control.
Collapse
Affiliation(s)
- Jonathan E Tullis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Program in Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
12
|
Stockwell I, Watson JF, Greger IH. Tuning synaptic strength by regulation of AMPA glutamate receptor localization. Bioessays 2024; 46:e2400006. [PMID: 38693811 PMCID: PMC7616278 DOI: 10.1002/bies.202400006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Long-term potentiation (LTP) of excitatory synapses is a leading model to explain the concept of information storage in the brain. Multiple mechanisms contribute to LTP, but central amongst them is an increased sensitivity of the postsynaptic membrane to neurotransmitter release. This sensitivity is predominantly determined by the abundance and localization of AMPA-type glutamate receptors (AMPARs). A combination of AMPAR structural data, super-resolution imaging of excitatory synapses, and an abundance of electrophysiological studies are providing an ever-clearer picture of how AMPARs are recruited and organized at synaptic junctions. Here, we review the latest insights into this process, and discuss how both cytoplasmic and extracellular receptor elements cooperate to tune the AMPAR response at the hippocampal CA1 synapse.
Collapse
Affiliation(s)
- Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jake F. Watson
- Institute of Science and Technology, Technology (IST) Austria, Klosterneuburg, Austria
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
13
|
Feng H, Zhang Z, Lyu W, Kong X, Li J, Zhou H, Wei P. The Effects of Appropriate Perioperative Exercise on Perioperative Neurocognitive Disorders: a Narrative Review. Mol Neurobiol 2024; 61:4663-4676. [PMID: 38110646 PMCID: PMC11236851 DOI: 10.1007/s12035-023-03864-0] [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: 07/12/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
Perioperative neurocognitive disorders (PNDs) are now considered the most common neurological complication in older adult patients undergoing surgical procedures. A significant increase exists in the incidence of post-operative disability and mortality in patients with PNDs. However, no specific treatment is still available for PNDs. Recent studies have shown that exercise may improve cognitive dysfunction-related disorders, including PNDs. Neuroinflammation is a key mechanism underlying exercise-induced neuroprotection in PNDs; others include the regulation of gut microbiota and mitochondrial and synaptic function. Maintaining optimal skeletal muscle mass through preoperative exercise is important to prevent the occurrence of PNDs. This review summarizes current clinical and preclinical evidence and proposes potential molecular mechanisms by which perioperative exercise improves PNDs, providing a new direction for exploring exercise-mediated neuroprotective effects on PNDs. In addition, it intends to provide new strategies for the prevention and treatment of PNDs.
Collapse
Affiliation(s)
- Hao Feng
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Zheng Zhang
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Wenyuan Lyu
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Xiangyi Kong
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Jianjun Li
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China
| | - Haipeng Zhou
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China.
| | - Penghui Wei
- Department of Anesthesiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, People's Republic of China.
| |
Collapse
|
14
|
Franco-Pérez J. Mechanisms Underlying Memory Impairment Induced by Fructose. Neuroscience 2024; 548:27-38. [PMID: 38679409 DOI: 10.1016/j.neuroscience.2024.04.001] [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: 01/03/2024] [Revised: 03/04/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Fructose consumption has increased over the years, especially in adolescents living in urban areas. Growing evidence indicates that daily fructose consumption leads to some pathological conditions, including memory impairment. This review summarizes relevant data describing cognitive deficits after fructose intake and analyzes the underlying neurobiological mechanisms. Preclinical experiments show sex-related deficits in spatial memory; that is, while males exhibit significant imbalances in spatial processing, females seem unaffected by dietary supplementation with fructose. Recognition memory has also been evaluated; however, only female rodents show a significant decline in the novel object recognition test performance. According to mechanistic evidence, fructose intake induces neuroinflammation, mitochondrial dysfunction, and oxidative stress in the short term. Subsequently, these mechanisms can trigger other long-term effects, such as inhibition of neurogenesis, downregulation of trophic factors and receptors, weakening of synaptic plasticity, and long-term potentiation decay. Integrating all these neurobiological mechanisms will help us understand the cellular and molecular processes that trigger the memory impairment induced by fructose.
Collapse
Affiliation(s)
- Javier Franco-Pérez
- Laboratorio Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía, Manuel Velasco Suárez, Insurgentes Sur 3877, Col. La Fama, C.P. 14269, CDMX, México, Mexico.
| |
Collapse
|
15
|
Reed JM, Wolfe BE, Romero LM. Is resilience a unifying concept for the biological sciences? iScience 2024; 27:109478. [PMID: 38660410 PMCID: PMC11039332 DOI: 10.1016/j.isci.2024.109478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
There is increasing interest in applying resilience concepts at different scales of biological organization to address major interdisciplinary challenges from cancer to climate change. It is unclear, however, whether resilience can be a unifying concept consistently applied across the breadth of the biological sciences, or whether there is limited capacity for integration. In this review, we draw on literature from molecular biology to community ecology to ascertain commonalities and shortcomings in how resilience is measured and interpreted. Resilience is studied at all levels of biological organization, although the term is often not used. There is a suite of resilience mechanisms conserved across biological scales, and there are tradeoffs that affect resilience. Resilience is conceptually useful to help diverse researchers think about how biological systems respond to perturbations, but we need a richer lexicon to describe the diversity of perturbations, and we lack widely applicable metrics of resilience.
Collapse
Affiliation(s)
- J. Michael Reed
- Department of Biology, Tufts University, Medford 02155, MA, USA
| | | | | |
Collapse
|
16
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Sheikhi S, Aghazadeh R, Sayyadi H, Pourheydar B, Saboory E, Bagheri M, Derafshpour L. The effects of choline supplementation in mothers with hypothyroidism on the alteration of cognitive-behavioral, long-term potentiation, morphology, and apoptosis in the hippocampus of pre-pubertal offspring rats. Int J Dev Neurosci 2024; 84:109-121. [PMID: 38311365 DOI: 10.1002/jdn.10312] [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: 05/14/2023] [Revised: 12/10/2023] [Accepted: 12/24/2023] [Indexed: 02/10/2024] Open
Abstract
The mother's thyroid hormone status during gestation and the first few months after delivery can play a crucial role in maturation during the brain development of the child. Transient abnormalities in thyroid function at birth indicate developmental and cognitive disorders in adulthood. Choline supplementation during gestation and the perinatal period in rats causes long-lasting memory improvement in the offspring. However, it remains unclear whether choline is able to restore the deficits in rats with maternal hypothyroidism. The aim of this study was to evaluate the effects of choline supplementation on the alteration of cognitive-behavioral function, long-term potentiation (LTP), and morphological changes as well as apoptosis in pre-pubertal offspring rats. To induce hypothyroidism, 6-propyl-2-thiouracil was added to the drinking water from the 6th day of gestation to the 21st postnatal day (PND). Choline treatment was started twice a day on the first day of the gestation until PND 21 via gavage. LTP recording and Morris water maze (MWM) test were conducted at PND 28. Then, the rats were sacrificed to assess their brains. The results revealed that developmental thyroid hormone deficiency impaired spatial learning and memory and reduced LTP (both: P < 0.001). Choline treatment alleviated LTP (P < 0.001), as well as learning and memory deficits (P < 0.01) in both male and female hypothyroid rats. However, no significant changes were observed in the number of caspase-3 stained cells in choline-receiving hypothyroid groups. The results revealed that developmental thyroid hormone deficiency impaired spatial learning and memory and reduced LTP. Choline treatment alleviated LTP, as well as learning and memory deficits in both male and female hypothyroid rats.
Collapse
Affiliation(s)
- Siamak Sheikhi
- Department of Psychiatry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Razieh Aghazadeh
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hojjat Sayyadi
- Non-Communicable Diseases Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Bagher Pourheydar
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Saboory
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Morteza Bagheri
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
18
|
Aktar S, Ferdousi F, Kondo S, Kagawa T, Isoda H. Transcriptomics and biochemical evidence of trigonelline ameliorating learning and memory decline in the senescence-accelerated mouse prone 8 (SAMP8) model by suppressing proinflammatory cytokines and elevating neurotransmitter release. GeroScience 2024; 46:1671-1691. [PMID: 37721682 PMCID: PMC10828270 DOI: 10.1007/s11357-023-00919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
In recent years, exploring natural compounds with functional properties to ameliorate aging-associated cognitive decline has become a research priority to ensure healthy aging. In the present study, we investigated the effects of Trigonelline (TG), a plant alkaloid, on memory and spatial learning in 16-week-old senescence-accelerated mouse model SAMP8 using an integrated approach for cognitive and molecular biology aspects. After 30 days of oral administration of TG at the dose of 5 mg/kg/day, the mice were trained in Morris Water Maze task. TG-treated SAMP8 mice exhibited significant improvement in the parameters of escape latency, distance moved, and annulus crossing index. Next, we performed a whole-genome transcriptome profiling of the mouse hippocampus using microarrays. Gene ontology analyses showed that a wide range of biological processes, including nervous system development, mitochondrial function, ATP synthesis, and several signaling pathways related to inflammation, autophagy, and neurotransmitter release, were significantly enriched in TG-treated SAMP8 compared to nontreated. Further, a nonlinear dimensionality reduction technique, Uniform Manifold Approximation and Projection (UMAP), was applied to identify clusters of functions that revealed TG primarily regulated pathways related to inflammation, followed by those involved in neurotransmitter release. In addition, a protein-protein interaction network analysis indicated that TG may exert its biological effects through negatively modulating Traf6-mediated NF-κB activation. Finally, ELISA test showed that TG treatment significantly decreased proinflammatory cytokines- TNFα and IL6 and increased neurotransmitters- dopamine, noradrenaline, and serotonin in mouse hippocampus. Altogether, our integrated bio-cognitive approach highlights the potential of TG in alleviating age-related memory and spatial impairment.
Collapse
Affiliation(s)
- Sharmin Aktar
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - Farhana Ferdousi
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Shinji Kondo
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | | | - Hiroko Isoda
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibarak, 305-8572, Japan.
| |
Collapse
|
19
|
Ogelman R, Gomez Wulschner LE, Hoelscher VM, Hwang IW, Chang VN, Oh WC. Serotonin modulates excitatory synapse maturation in the developing prefrontal cortex. Nat Commun 2024; 15:1368. [PMID: 38365905 PMCID: PMC10873381 DOI: 10.1038/s41467-024-45734-w] [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: 04/21/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Serotonin (5-HT) imbalances in the developing prefrontal cortex (PFC) are linked to long-term behavioral deficits. However, the synaptic mechanisms underlying 5-HT-mediated PFC development are unknown. We found that chemogenetic suppression and enhancement of 5-HT release in the PFC during the first two postnatal weeks decreased and increased the density and strength of excitatory spine synapses, respectively, on prefrontal layer 2/3 pyramidal neurons in mice. 5-HT release on single spines induced structural and functional long-term potentiation (LTP), requiring both 5-HT2A and 5-HT7 receptor signals, in a glutamatergic activity-independent manner. Notably, LTP-inducing 5-HT stimuli increased the long-term survival of newly formed spines ( ≥ 6 h) via 5-HT7 Gαs activation. Chronic treatment of mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first two weeks, but not the third week of postnatal development, increased the density and strength of excitatory synapses. The effect of fluoxetine on PFC synaptic alterations in vivo was abolished by 5-HT2A and 5-HT7 receptor antagonists. Our data describe a molecular basis of 5-HT-dependent excitatory synaptic plasticity at the level of single spines in the PFC during early postnatal development.
Collapse
Affiliation(s)
- Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria M Hoelscher
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria N Chang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
20
|
Gao J, Liu W, Liu J, Hao N, Pei J, Zhang L. The Role of Acetylation and Methylation of Rat Hippocampal Histone H3 in the Mechanism of Aluminum-Induced Neurotoxicity. Neurochem Res 2024; 49:441-452. [PMID: 37897558 DOI: 10.1007/s11064-023-04045-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/30/2023]
Abstract
Aluminum is a known neurotoxin and a major environmental contributor to neurodegenerative diseases such as Alzheimer's disease (AD). We uesd a subchronic aluminum chloride exposure model in offspring rats by continuously treating them with AlCl3 solution from the date of birth until day 90 in this research. Then evaluated the neurobehavioral changes in rats, observed the ultrastructural changes of hippocampal synapses and neurons, and examined the level of hippocampal acetylated histone H3 (H3ac), the activity and protein expression of hippocampal HAT1 and G9a, and the protein expression level of H3K9 dimethylation (H3K9me2). The findings demonstrated that aluminum-treated offspring rats had impaired learning and memory abilities as well as ultrastructural alterations in hippocampal synapses and neurons. The level of histone H3ac was decreased along with decreased protein expression and activity of HAT1, while level of H3K9me2 was increased along with increased protein expression and activity of G9a.
Collapse
Affiliation(s)
- Jie Gao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China
| | - Wei Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China
| | - Jiaqi Liu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China
| | - Niping Hao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China
| | - Jing Pei
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China
| | - Lifeng Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Shenyang Medical College, Shenyang, 110034, Liaoning Province, P. R. China.
| |
Collapse
|
21
|
Yin Y, Wu X, Zhu Y, Liu J, Fan Q, Zhao S, Wang J, Liu Y, Li Y, Lu W. Protective effect of Baoyuan Jieyu formula on long-term spaceflight composite stress-induced depressive-like behavior and memory deficits through regulation of Ca 2+ channel currents. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:135-142. [PMID: 38245338 DOI: 10.1016/j.lssr.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/18/2023] [Accepted: 07/19/2023] [Indexed: 01/22/2024]
Abstract
Long-term spaceflight composite stress (LSCS) can cause adverse effects on human systems, especially the central nervous system. This study aimed to identify the underlying mechanisms of the protective effect of Baoyuan Jieyu Formula (BYJYF) on LSCS-induced depressive-like behavior and memory deficits. In this experiment, we simulated the real space station environment for a period of 42 days. Novel object recognition test and forced swimming test were used to assess the memory abilities and depression level of rats as well as test the therapeutic effects of BYJYF treatment. Results showed LSCS could induce depressive-like behavior and damage short-term memory in the behavioral level, and BYJYF could enhance the ability to resist LSCS. Meanwhile, LSCS increased the levels of CRH, ACTH, and CORT and induced HPA axis hyperactivity, which can be relieved by BYJYF. Further, we predicted and verified the potential signaling pathways of BYJYF. Results showed BYJYF may reverse the inhibition of LSCS on Ca2+ channel currents. And we also found that BYJYF may exert its medicinal effects via four main active components including saikosaponin A. Overall, BYJYF exhibited protective effects against LSCS-induced depressive-like behavior and memory deficits, which might be ascribed to the regulation of Ca2+ channel currents and four active components. And it might become a promising candidate medicine for diseases induced by LSCS.
Collapse
Affiliation(s)
- Yishu Yin
- School of Medicine and Health, Harbin Institute of Technology, Harbin, 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin, 150001, China
| | - Xiaorui Wu
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yuanbing Zhu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin, 150001, China
| | - Junlian Liu
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Quanchun Fan
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shuang Zhao
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Jiaping Wang
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yu Liu
- China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin, 150001, China.
| |
Collapse
|
22
|
Legutko D, Kuźniewska B, Kalita K, Yasuda R, Kaczmarek L, Michaluk P. BDNF signaling requires Matrix Metalloproteinase-9 during structural synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.08.569797. [PMID: 38106209 PMCID: PMC10723398 DOI: 10.1101/2023.12.08.569797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Synaptic plasticity underlies learning and memory processes as well as contributes, in its aberrant form, to neuropsychiatric disorders. One of its major forms is structural long-term potentiation (sLTP), an activity-dependent growth of dendritic spines that harbor excitatory synapses. The process depends on the release of brain-derived neurotrophic factor (BDNF), and activation of its receptor, TrkB. Matrix metalloproteinase-9 (MMP-9), an extracellular protease is essential for many forms of neuronal plasticity engaged in physiological as well as pathological processes. Here, we utilized two-photon microscopy and two-photon glutamate uncaging to demonstrate that MMP-9 activity is essential for sLTP and is rapidly (~seconds) released from dendritic spines in response to synaptic stimulation. Moreover, we show that either chemical or genetic inhibition of MMP-9 impairs TrkB activation, as measured by fluorescence lifetime imaging microscopy of FRET sensor. Furthermore, we provide evidence for a cell-free cleavage of proBDNF into mature BDNF by MMP-9. Our findings point to the autocrine mechanism of action of MMP-9 through BDNF maturation and TrkB activation during sLTP.
Collapse
Affiliation(s)
- Diana Legutko
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, Florida 33458, USA
| | - Bożena Kuźniewska
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
- Current address: Department of Animal Physiology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Katarzyna Kalita
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, Florida 33458, USA
| | - Leszek Kaczmarek
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| | - Piotr Michaluk
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| |
Collapse
|
23
|
Wu H, Chen X, Shen Z, Li H, Liang S, Lu Y, Zhang M. Phosphorylation-dependent membraneless organelle fusion and fission illustrated by postsynaptic density assemblies. Mol Cell 2024; 84:309-326.e7. [PMID: 38096828 DOI: 10.1016/j.molcel.2023.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 09/10/2023] [Accepted: 11/13/2023] [Indexed: 01/21/2024]
Abstract
Membraneless organelles formed by phase separation of proteins and nucleic acids play diverse cellular functions. Whether and, if yes, how membraneless organelles in ways analogous to membrane-based organelles also undergo regulated fusion and fission is unknown. Here, using a partially reconstituted mammalian postsynaptic density (PSD) condensate as a paradigm, we show that membraneless organelles can undergo phosphorylation-dependent fusion and fission. Without phosphorylation of the SAPAP guanylate kinase domain-binding repeats, the upper and lower layers of PSD protein mixtures form two immiscible sub-compartments in a phase-in-phase organization. Phosphorylation of SAPAP leads to fusion of the two sub-compartments into one condensate accompanied with an increased Stargazin density in the condensate. Dephosphorylation of SAPAP can reverse this event. Preventing SAPAP phosphorylation in vivo leads to increased separation of proteins from the lower and upper layers of PSD sub-compartments. Thus, analogous to membrane-based organelles, membraneless organelles can also undergo regulated fusion and fission.
Collapse
Affiliation(s)
- Haowei Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xudong Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zeyu Shen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hao Li
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiqi Liang
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Youming Lu
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingjie Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen 518036, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
24
|
Goncalves-Garcia M, Hamilton DA. Unraveling the complex relationship between prenatal alcohol exposure, hippocampal LTP, and learning and memory. Front Mol Neurosci 2024; 16:1326089. [PMID: 38283699 PMCID: PMC10811250 DOI: 10.3389/fnmol.2023.1326089] [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: 10/22/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Prenatal alcohol exposure (PAE) has been extensively studied for its profound impact on neurodevelopment, synaptic plasticity, and cognitive outcomes. While PAE, particularly at moderate levels, has long-lasting cognitive implications for the exposed individuals, there remains a substantial gap in our understanding of the precise mechanisms underlying these deficits. This review provides a framework for comprehending the neurobiological basis of learning and memory processes that are negatively impacted by PAE. Sex differences, diverse PAE protocols, and the timing of exposure are explored as potential variables influencing the diverse outcomes of PAE on long-term potentiation (LTP). Additionally, potential interventions, both pharmacological and non-pharmacological, are reviewed, offering promising avenues for mitigating the detrimental effects of PAE on cognitive processes. While significant progress has been made, further research is required to enhance our understanding of how prenatal alcohol exposure affects neural plasticity and cognitive functions and to develop effective therapeutic interventions for those impacted. Ultimately, this work aims to advance the comprehension of the consequences of PAE on the brain and cognitive functions.
Collapse
|
25
|
Tan JZA, Jang SE, Batallas-Borja A, Bhembre N, Chandra M, Zhang L, Guo H, Ringuet MT, Widagdo J, Collins BM, Anggono V. Copine-6 is a Ca 2+ sensor for activity-induced AMPA receptor exocytosis. Cell Rep 2023; 42:113460. [PMID: 37979168 DOI: 10.1016/j.celrep.2023.113460] [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: 04/13/2023] [Revised: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
The recruitment of synaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors underlies the strengthening of neuronal connectivity during learning and memory. This process is triggered by N-methyl-D-aspartate (NMDA) receptor-dependent postsynaptic Ca2+ influx. Synaptotagmin (Syt)-1 and -7 have been proposed as Ca2+ sensors for AMPA receptor exocytosis but are functionally redundant. Here, we identify a cytosolic C2 domain-containing Ca2+-binding protein, Copine-6, that forms a complex with AMPA receptors. Loss of Copine-6 expression impairs activity-induced exocytosis of AMPA receptors in primary neurons, which is rescued by wild-type Copine-6 but not Ca2+-binding mutants. In contrast, Copine-6 loss of function does not affect steady-state expression or tetrodotoxin-induced synaptic upscaling of surface AMPA receptors. Loss of Syt-1/Syt-7 significantly reduces Copine-6 protein expression. Interestingly, overexpression of wild-type Copine-6, but not the Ca2+-binding mutants, restores activity-dependent exocytosis of AMPA receptors in Syt-1/Syt-7 double-knockdown neurons. We conclude that Copine-6 is a postsynaptic Ca2+ sensor that mediates AMPA receptor exocytosis during synaptic potentiation.
Collapse
Affiliation(s)
- Jing Zhi Anson Tan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Se Eun Jang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ana Batallas-Borja
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nishita Bhembre
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mintu Chandra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Lingrui Zhang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Huimin Guo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Mitchell T Ringuet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Brett M Collins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
26
|
Yan T, Chen J, Wang Y, Wang Y, Zhang Y, Zhao Y. Deficiency of aldehyde dehydrogenase 2 aggravates ethanol-induced cytotoxicity in N2a cells via CaMKII/Drp1-mediated mitophagy. Food Chem Toxicol 2023; 182:114129. [PMID: 37967785 DOI: 10.1016/j.fct.2023.114129] [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: 08/04/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Chronic alcohol abuse causes brain damage and has been associated with an increased risk of Alzheimer's disease. The toxic metabolite of alcohol, acetaldehyde, which is converted to acetate by aldehyde dehydrogenase 2 (ALDH2), has been shown to induce excessive mitochondrial fragmentation and dysfunction leading to neurotoxicity. However, it is still unclear how alcohol affects mitochondrial function in ALDH2-deficient cells. The present study investigated the association between abnormal mitochondrial dynamics, mitophagy and cytotoxicity in ALDH2-deficient N2a cells treated with ethanol. It was found that ethanol induced dynamin-related protein 1 (Drp1)-mediated mitochondrial fragmentation and impaired mitochondrial function, causing excessive mitophagy and cytotoxicity in ALDH2-deficient N2a cells while inducing Ca2+ influx and activating Ca2+/calmodulin-dependent protein kinase II (CaMKII). Inhibition of Ca2+ overload or CaMKII activation prevented Drp1 phosphorylation and ameliorated ethanol-induced mitophagy and cytotoxicity, indicating that Ca2+-dependent CaMKII activation was critical for mediating Drp1-dependent excessive mitochondrial fission and mitophagy in ALDH2-deficient N2a cells. The results of the present study suggested that prevention of intracellular Ca2+ overload might be beneficial for preventing neurotoxicity associated with alcohol abuse in individuals with defective ALDH2.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Jiyang Chen
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yalin Wang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yinuo Wang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yuanqingzhi Zhang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China.
| |
Collapse
|
27
|
Keifer J. Synaptic Mechanisms of Delay Eyeblink Classical Conditioning: AMPAR Trafficking and Gene Regulation in an In Vitro Model. Mol Neurobiol 2023; 60:7088-7103. [PMID: 37531025 DOI: 10.1007/s12035-023-03528-z] [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: 03/14/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023]
Abstract
An in vitro model of delay eyeblink classical conditioning was developed to investigate synaptic plasticity mechanisms underlying acquisition of associative learning. This was achieved by replacing real stimuli, such as an airpuff and tone, with patterned stimulation of the cranial nerves using an isolated brainstem preparation from turtle. Here, our primary findings regarding cellular and molecular mechanisms for learning acquisition using this unique approach are reviewed. The neural correlate of the in vitro eyeblink response is a replica of the actual behavior, and features of conditioned responses (CRs) resemble those observed in behavioral studies. Importantly, it was shown that acquisition of CRs did not require the intact cerebellum, but the appropriate timing did. Studies of synaptic mechanisms indicate that conditioning involves two stages of AMPA receptor (AMPAR) trafficking. Initially, GluA1-containing AMPARs are targeted to synapses followed later by replacement by GluA4 subunits that support CR expression. This two-stage process is regulated by specific signal transduction cascades involving PKA and PKC and is guided by distinct protein chaperones. The expression of the brain-derived neurotrophic factor (BDNF) protein is central to AMPAR trafficking and conditioning. BDNF gene expression is regulated by coordinated epigenetic mechanisms involving DNA methylation/demethylation and chromatin modifications that control access of promoters to transcription factors. Finally, a hypothesis is proposed that learning genes like BDNF are poised by dual chromatin features that allow rapid activation or repression in response to environmental stimuli. These in vitro studies have advanced our understanding of the cellular and molecular mechanisms that underlie associative learning.
Collapse
Affiliation(s)
- Joyce Keifer
- Neuroscience Group, Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
28
|
Keimasi M, Salehifard K, Mirshah Jafar Esfahani N, Esmaeili F, Farghadani A, Amirsadri M, Keimasi M, Noorbakhshnia M, Moradmand M, Mofid MR. The synergic effects of presynaptic calcium channel antagonists purified from spiders on memory elimination of glutamate-induced excitotoxicity in the rat hippocampus trisynaptic circuit. Front Mol Biosci 2023; 10:1243976. [PMID: 38099194 PMCID: PMC10720730 DOI: 10.3389/fmolb.2023.1243976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
The hippocampus is a complex area of the mammalian brain and is responsible for learning and memory. The trisynaptic circuit engages with explicit memory. Hippocampal neurons express two types of presynaptic voltage-gated calcium channels (VGCCs) comprising N and P/Q-types. These VGCCs play a vital role in the release of neurotransmitters from presynaptic neurons. The chief excitatory neurotransmitter at these synapses is glutamate. Glutamate has an essential function in learning and memory under normal conditions. The release of neurotransmitters depends on the activity of presynaptic VGCCs. Excessive glutamate activity, due to either excessive release or insufficient uptake from the synapse, leads to a condition called excitotoxicity. This pathological state is common among all neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. Under these conditions, glutamate adversely affects the trisynaptic circuitry, leading to synaptic destruction and loss of memory and learning performance. This study attempts to clarify the role of presynaptic VGCCs in memory performance and reveals that modulating the activity of presynaptic calcium channels in the trisynaptic pathway can regulate the excitotoxic state and consequently prevent the elimination of neurons and synaptic degradation. All of these can lead to an improvement in learning and memory function. In the current study, two calcium channel blockers-omega-agatoxin-Aa2a and omega-Lsp-IA-were extracted, purified, and identified from spiders (Agelena orientalis and Hogna radiata) and used to modulate N and P/Q VGCCs. The effect of omega-agatoxin-Aa2a and omega-Lsp-IA on glutamate-induced excitotoxicity in rats was evaluated using the Morris water maze task as a behavioral test. The local expression of synaptophysin (SYN) was visualized for synaptic quantification using an immunofluorescence assay. The electrophysiological amplitudes of the field excitatory postsynaptic potentials (fEPSPs) in the input-output and LTP curves of the mossy fiber and Schaffer collateral circuits were recorded. The results of our study demonstrated that N and P/Q VGCC modulation in the hippocampus trisynaptic circuit of rats with glutamate-induced excitotoxicity dysfunction could prevent the destructive consequences of excitotoxicity in synapses and improve memory function and performance.
Collapse
Affiliation(s)
- Mohammad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Mirshah Jafar Esfahani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Arman Farghadani
- Department of Biology, Faculty of Biological Sciences, University Duisburg-Essen, Essen, Germany
| | - Mohammadreza Amirsadri
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadjavad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Maryam Noorbakhshnia
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Moradmand
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Reza Mofid
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
29
|
O’Day DH. Protein Biomarkers Shared by Multiple Neurodegenerative Diseases Are Calmodulin-Binding Proteins Offering Novel and Potentially Universal Therapeutic Targets. J Clin Med 2023; 12:7045. [PMID: 38002659 PMCID: PMC10672630 DOI: 10.3390/jcm12227045] [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: 10/13/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Seven major neurodegenerative diseases and their variants share many overlapping biomarkers that are calmodulin-binding proteins: Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), frontotemporal lobar dementia (FTD), Huntington's disease (HD), Lewy body disease (LBD), multiple sclerosis (MS), and Parkinson's disease (PD). Calcium dysregulation is an early and persistent event in each of these diseases, with calmodulin serving as an initial and primary target of increased cytosolic calcium. Considering the central role of calcium dysregulation and its downstream impact on calcium signaling, calmodulin has gained interest as a major regulator of neurodegenerative events. Here, we show that calmodulin serves a critical role in neurodegenerative diseases via binding to and regulating an abundance of biomarkers, many of which are involved in multiple neurodegenerative diseases. Of special interest are the shared functions of calmodulin in the generation of protein biomarker aggregates in AD, HD, LBD, and PD, where calmodulin not only binds to amyloid beta, pTau, alpha-synuclein, and mutant huntingtin but also, via its regulation of transglutaminase 2, converts them into toxic protein aggregates. It is suggested that several calmodulin binding proteins could immediately serve as primary drug targets, while combinations of calmodulin binding proteins could provide simultaneous insight into the onset and progression of multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
30
|
Xu L, Qu C, Liu Y, Liu H. The environmental enrichment ameliorates chronic cerebral hypoperfusion-induced cognitive impairment by activating autophagy signaling pathway and improving synaptic function in hippocampus. Brain Res Bull 2023; 204:110798. [PMID: 37890595 DOI: 10.1016/j.brainresbull.2023.110798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 10/01/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a frequently observed underlying pathology of both Alzheimer's disease (AD) and vascular dementia (VD), which is a common consequence of cerebral blood flow (CBF) dysregulation. Synaptic damage has been proven as a crucial causative factor for CCH-related cognitive impairment. This study aimed to investigate the neuroprotective impact of environmental enrichment (EE) intervention on CCH-induced synaptic destruction and the consequent cognitive impairment. Furthermore, the underlying mechanism of this neuroprotective effect was explored to provide new insights into therapeutic interventions for individuals suffering from AD or VD. METHODS In this experiment, all rats were initially acclimatized to a standard environment (SE) for a period of one week. On the seventh day, rats underwent either bilateral common carotid artery occlusion (2VO) surgery or sham surgery (Sham) before being subjected to a four-week procedure of exposure to an EE, except for the control group. During the EE or SE procedure, intraperitoneal injection of chloroquine (CQ) into rats was performed once daily for four weeks. Following this, cognitive function was assessed using the Morris water maze (MWM) test. The synapse ultrastructure was subsequently observed using transmission electron microscopy. Expression levels of autophagy-related proteins (LC3, LAMP1, and P62) and synapse-related proteins (Synapsin I and PSD-95) were detected through Western blotting. Finally, immunofluorescence was used to examine the expression levels of Synapsin I and PSD-95 and the colocalization of LAMP-1 and LC3 in the hippocampus. RESULTS After undergoing 2VO, rats exposed to SE exhibited cognitive impairment, autophagic dysfunction, and synapse damage. The synapse damage was evidenced by ultrastructural damage and degradation of synapse-related proteins. However, these effects were significantly mitigated by exposure to an EE intervention. Moreover, the intervention led to an improvement in autophagic dysfunction. CONCLUSION The study found that EE had a positive impact on CCH-induced synaptic damage. Specifically, EE was found to increase synaptic plasticity-associated proteins and postsynaptic density thickness, while decreasing synaptic space. This multifaceted effect resulted in an amelioration of CCH-induced cognitive impairment. It was shown that this beneficial outcome was mediated via the activation of the autophagy-lysosomal pathway. Overall, the findings suggest that EE may have a therapeutic potential for cognitive impairments associated with CCH through autophagy-mediated synaptic improvement.
Collapse
Affiliation(s)
- Linling Xu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China; Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan 430071, Hubei, China
| | - Changhua Qu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Department of Neurology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Yan Liu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China
| | - Hua Liu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China.
| |
Collapse
|
31
|
Keimasi M, Salehifard K, Hoseini SJ, Esmaeili F, Esfahani NMJ, Amirsadri M, Keimasi M, Moradmand M, Mofid MR. Purified Native Protein Extracted from the Venom of Agelena orientalis Attenuates Memory Defects in the Rat Model of Glutamate-Induced Excitotoxicity. Protein J 2023; 42:586-595. [PMID: 37531037 DOI: 10.1007/s10930-023-10140-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/03/2023]
Abstract
Spider venom contains various peptides and proteins, which can be used for pharmacological applications. Finding novel therapeutic strategies against neurodegenerative diseases with the use of purified peptides and proteins, extracted from spiders can be greatly precious. Neurodegenerative diseases are rapidly developing and expanding all over the world. Excitotoxicity is a frequent condition amongst neuro-degenerative disorders. This harmful process is usually induced through hyper-activation of N-Methyl-D-Aspartate (NMDA) receptor, and P/Q-type voltage-gated calcium channels (VGCCs). The omega-agatoxin-Aa4b is a selective and strong VGCCblocker. This study aimed to investigate the effects of this blocker on the NMDA-induced memory and learning defect in rats. For this purpose, nineteen spiders of the funnel-weaver Agelena orientalis species were collected. The extracted venom was lyophilized andpurified through gel-filtration chromatography, and capillary electrophoresis techniques. Subsequently, mass spectrometry (HPLC-ESI-MS) was used for identification of this bio-active small protein. Afterward, the effect of the omega-agatoxin-Aa4b (2 μg, intra-cornu ammonis-3 of the hippocampus) on the NMDA-induced learning and memory deficits in rats was evaluated. Learning and memory performances were evaluated by the use of passive avoidance test. For synaptic quantification and memory function the amount of calcium/calmodulin-dependent protein kinase ІІ (CaCdPKІІ) gene expression was measured using the Real-time PCR technique. To compare the experimental groups, hematoxylin and eosin (H&E) staining of hippocampus tissues was performed. Our results rendered that the omega-Agatoxin-Aa4b treatment can ameliorate and reverse the learning and memory impairment caused by NMDA-induced excitotoxicity in rat hippocampus.
Collapse
Affiliation(s)
- Mohammad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayyed Jafar Hoseini
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Noushin Mirshah Jafar Esfahani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Amirsadri
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadjavad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Moradmand
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran.
| | - Mohammad Reza Mofid
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
32
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
33
|
Chakrabarti N, Sarkar PK, Ray AK, Martin JV. Unveiling the nongenomic actions of thyroid hormones in adult mammalian brain: The legacy of Mary B. Dratman. Front Endocrinol (Lausanne) 2023; 14:1240265. [PMID: 37842308 PMCID: PMC10570802 DOI: 10.3389/fendo.2023.1240265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
A comprehensive review was conducted to compile the contributions of Mary B. Dratman and studies by other researchers in the field of nongenomic actions of thyroid hormones in adult mammalian brain. Dratman and her collaborators authored roughly half of the papers in this area. It has been almost fifty years since Dratman introduced the novel concept of thyroid hormones as neurotransmitters for the first time. The characterization of unique brain-region specific accumulation of thyroid hormones within the nerve terminals in adult mammals was a remarkable contribution by Dratman. It suggested a neurotransmitter- or neuromodulator-like role of thyroid hormone and/or its derivative, 3-iodothyronamine within adrenergic systems in adult mammalian brain. Several studies by other researchers using synaptosomes as a model system, have contributed to the concept of direct nongenomic actions of thyroid hormones at synaptic regions by establishing that thyroid hormones or their derivatives can bind to synaptosomal membranes, alter membrane functions including enzymatic activities and ion transport, elicit Ca2+/NO-dependent signaling pathways and induce substrate-protein phosphorylation. Such findings can help to explain the physiological and pathophysiological roles of thyroid hormone in psychobehavioral control in adult mammalian brain. However, the exact mode of nongenomic actions of thyroid hormones at nerve terminals in adult mammalian brain awaits further study.
Collapse
Affiliation(s)
- Nilkanta Chakrabarti
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
- (CPEPA-UGC) Centre for Electro-Physiological and Neuroimaging studies including Mathematical Modelling, University of Calcutta, Kolkata, West Bengal, India
| | - Pradip K. Sarkar
- Department of Basic Sciences, Parker University, Dallas, TX, United States
| | - Arun K. Ray
- Department of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII-M, Kolkata, India
| | - Joseph V. Martin
- Biology Department, Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, United States
| |
Collapse
|
34
|
Fan Y, McMath AL, Donovan SM. Review on the Impact of Milk Oligosaccharides on the Brain and Neurocognitive Development in Early Life. Nutrients 2023; 15:3743. [PMID: 37686775 PMCID: PMC10490528 DOI: 10.3390/nu15173743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Milk Oligosaccharides (MOS), a group of complex carbohydrates found in human and bovine milk, have emerged as potential modulators of optimal brain development for early life. This review provides a comprehensive investigation of the impact of milk oligosaccharides on brain and neurocognitive development of early life by synthesizing current literature from preclinical models and human observational studies. The literature search was conducted in the PubMed search engine, and the inclusion eligibility was evaluated by three reviewers. Overall, we identified 26 articles for analysis. While the literature supports the crucial roles of fucosylated and sialylated milk oligosaccharides in learning, memory, executive functioning, and brain structural development, limitations were identified. In preclinical models, the supplementation of only the most abundant MOS might overlook the complexity of naturally occurring MOS compositions. Similarly, accurately quantifying MOS intake in human studies is challenging due to potential confounding effects such as formula feeding. Mechanistically, MOS is thought to impact neurodevelopment through modulation of the microbiota and enhancement of neuronal signaling. However, further advancement in our understanding necessitates clinical randomized-controlled trials to elucidate the specific mechanisms and long-term implications of milk oligosaccharides exposure. Understanding the interplay between milk oligosaccharides and cognition may contribute to early nutrition strategies for optimal cognitive outcomes in children.
Collapse
Affiliation(s)
- Yuting Fan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
| | - Arden L. McMath
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA;
| |
Collapse
|
35
|
O’Day DH. Alzheimer's Disease beyond Calcium Dysregulation: The Complex Interplay between Calmodulin, Calmodulin-Binding Proteins and Amyloid Beta from Disease Onset through Progression. Curr Issues Mol Biol 2023; 45:6246-6261. [PMID: 37623212 PMCID: PMC10453589 DOI: 10.3390/cimb45080393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
A multifactorial syndrome, Alzheimer's disease is the main cause of dementia, but there is no existing therapy to prevent it or stop its progression. One of the earliest events of Alzheimer's disease is the disruption of calcium homeostasis but that is just a prelude to the disease's devastating impact. Calcium does not work alone but must interact with downstream cellular components of which the small regulatory protein calmodulin is central, if not primary. This review supports the idea that, due to calcium dyshomeostasis, calmodulin is a dominant regulatory protein that functions in all stages of Alzheimer's disease, and these regulatory events are impacted by amyloid beta. Amyloid beta not only binds to and regulates calmodulin but also multiple calmodulin-binding proteins involved in Alzheimer's. Together, they act on the regulation of calcium dyshomeostasis, neuroinflammation, amyloidogenesis, memory formation, neuronal plasticity and more. The complex interactions between calmodulin, its binding proteins and amyloid beta may explain why many therapies have failed or are doomed to failure unless they are considered.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
36
|
Therapeutic potential of natural molecules against Alzheimer's disease via SIRT1 modulation. Biomed Pharmacother 2023; 161:114474. [PMID: 36878051 DOI: 10.1016/j.biopha.2023.114474] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease mainly characterized by progressive cognitive dysfunction and memory impairment. Recent studies have shown that regulating silent information regulator 1 (SIRT1) expression has a significant neuroprotective effect, and SIRT1 may become a new therapeutic target for AD. Natural molecules are an important source of drug development for use in AD therapy and may regulate a wide range of biological events by regulating SIRT1 as well as other SIRT1-mediated signaling pathways. This review aims to summarize the correlation between SIRT1 and AD and to identify in vivo and in vitro studies investigating the anti-AD properties of natural molecules as modulators of SIRT1 and SIRT1-mediated signaling pathways. A literature search was conducted for studies published between January 2000 and October 2022 using various literature databases, including Web of Science, PubMed, Google Scholar, Science Direct, and EMBASE. Natural molecules, such as resveratrol, quercetin, icariin, bisdemethoxycurcumin, dihydromyricetin, salidroside, patchouli, sesamin, rhein, ligustilide, tetramethoxyflavanone, 1-theanine, schisandrin, curcumin, betaine, pterostilbene, ampelopsin, schisanhenol, and eriodictyol, have the potential to modulate SIRT1 and SIRT1 signaling pathways, thereby combating AD. The natural molecules modulating SIRT1 discussed in this review provide a potentially novel multi-mechanistic therapeutic strategy for AD. However, future clinical trials need to be conducted to further investigate their beneficial properties and to determine the safety and efficacy of SIRT1 natural activators against AD.
Collapse
|
37
|
Inhibition of hippocampal palmitoyl acyltransferase activity impairs spatial learning and memory consolidation. Neurobiol Learn Mem 2023; 200:107733. [PMID: 36804592 DOI: 10.1016/j.nlm.2023.107733] [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: 10/14/2022] [Revised: 01/12/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Protein palmitoylation regulates trafficking, mobilization, localization, interaction, and distribution of proteins through the palmitoyl acyltransferases (PATs) enzymes. Protein palmitoylation controls rapid and dynamic changes of the synaptic architecture that modifies the efficiency and strength of synaptic connections, a fundamental mechanism to generate stable and long-lasting memory traces. Although protein palmitoylation in functional synaptic plasticity has been widely described, its role in learning and memory processes is poorly understood. In this work, we found that PATs inhibition into the hippocampus before and after the training of Morris water maze (MWM) and object location memory (OLM) impaired spatial learning. However, we demonstrated that PATs inhibition during the retrieval does not affect the expression of spatial memory in both MWM and OLM. Accordingly, long-term potentiation induction is impaired by inhibiting PATs into the hippocampus before high-frequency electrical stimulation but not after. These findings suggest that PATs activity is necessary to modify neural plasticity, a mechanism required for memory acquisition and consolidation. Like phosphorylation, active palmitoylation is required to regulate the function of already existing proteins that change synaptic strength in the hippocampus to acquire and later consolidate spatial memories.
Collapse
|
38
|
Keimasi M, Salehifard K, Keimasi M, Amirsadri M, Esfahani NMJ, Moradmand M, Esmaeili F, Mofid MR. Alleviation of cognitive deficits in a rat model of glutamate-induced excitotoxicity, using an N-type voltage-gated calcium channel ligand, extracted from Agelena labyrinthica crude venom. Front Mol Neurosci 2023; 16:1123343. [PMID: 36873105 PMCID: PMC9981952 DOI: 10.3389/fnmol.2023.1123343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Excitotoxicity is a common pathological process in Alzheimer's disease (AD) which is caused by the over-activity of N-Methyl-D-Aspartate receptors (NMDARs). The release of neurotransmitters depends on the activity of voltage-gated calcium channels (VGCCs). Hyper-stimulation of NMDARs can enhance the releasement of neurotransmitters through the VGCCs. This malfunction of channels can be blocked by selective and potent N-type VGCCs ligand. Under excitotoxicity condition, glutamate has negative effects on the pyramidal cells of the hippocampus, which ends in synaptic loss and elimination of these cells. These events leads to learning and memory elimination through the hippocampus circuit's dysfunction. A suitable ligand has a high affinity to receptor or channel and is selective for its target. The bioactive small proteins of venom have these characteristics. Therefore, peptides and small proteins of animal venom are precious sources for pharmacological applications. The omega-agatoxin-Aa2a was purified, and identified from Agelena labyrinthica specimens, as an N-type VGCCs ligand for this study. The effect of the omega-agatoxin-Aa2a on the glutamate-induced excitotoxicity in rats was evaluated through behavioral tests including Morris Water Maze, and Passive avoidance. The syntaxin1A (SY1A), synaptotagmin1 (SYT1), and synaptophysin (SYN) genes expression were measured via Real-Time PCR. The local expression of synaptosomal-associated protein, 25 k Da (SNAP-25) was visualized using an immunofluorescence assay for synaptic quantification. Electrophysiological amplitude of field excitatory postsynaptic potentials (fEPSPs) in the input-output and LTP curves of mossy fiber were recorded. The cresyl violet staining of hippocampus sections was performed for the groups. Our results demonstrated that the omega-agatoxin-Aa2a treatment could recover the learning, and memory impairment caused by NMDA-induced excitotoxicity in rat hippocampus.
Collapse
Affiliation(s)
- Mohammad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammadjavad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Amirsadri
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Mirshah Jafar Esfahani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Majid Moradmand
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Reza Mofid
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
39
|
Hirai H, Sakaba T, Hashimotodani Y. Subcortical glutamatergic inputs exhibit a Hebbian form of long-term potentiation in the dentate gyrus. Cell Rep 2022; 41:111871. [PMID: 36577371 DOI: 10.1016/j.celrep.2022.111871] [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: 06/16/2022] [Revised: 09/19/2022] [Accepted: 12/01/2022] [Indexed: 12/28/2022] Open
Abstract
The hippocampus receives glutamatergic and GABAergic inputs from subcortical regions. Despite the important roles of these subcortical inputs in the regulation of hippocampal circuit, it has not been explored whether associative activation of the subcorticohippocampal pathway induces Hebbian plasticity of subcortical inputs. Here, we demonstrate that the hypothalamic supramammillary nucleus (SuM) to the dentate granule cell (GC) synapses, which co-release glutamate and GABA, undergo associative long-term potentiation (LTP) of glutamatergic, but not GABAergic, co-transmission. This LTP is induced by pairing of SuM inputs with GC spikes. We found that this Hebbian LTP is input-specific, requires NMDA receptors and CaMKII activation, and is expressed postsynaptically. By the net increase in excitatory drive of SuM inputs following LTP induction, associative inputs of SuM and the perforant path effectively discharge GCs. Our results highlight the important role of associative plasticity at SuM-GC synapses in the regulation of dentate gyrus activity and for the encoding of SuM-related information.
Collapse
Affiliation(s)
- Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Yuki Hashimotodani
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan.
| |
Collapse
|
40
|
Offline neuronal activity and synaptic plasticity during sleep and memory consolidation. Neurosci Res 2022; 189:29-36. [PMID: 36584924 DOI: 10.1016/j.neures.2022.12.021] [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: 08/22/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
After initial formation during learning, memories are further processed in the brain during subsequent days for long-term consolidation, with sleep playing a key role in this process. Studies have shown that neuronal activity patterns during the awake period are repeated in the hippocampus during sleep, which may coordinate brain-wide reactivation leading to memory consolidation. Consistently, perturbation of this activity blocks the formation of long-term memory. This 'replay' of activity during sleep likely triggers plastic changes in synaptic transmission, a cellular substrate of memory, in multiple brain regions, which likely plays a critical role in long-term memory. Two forms of synaptic plasticity, potentiation and depression of synaptic transmission, are induced in parallel during sleep and is termed "offline synaptic plasticity", as opposed to the "online synaptic plasticity" that occurs immediately following a memory event.
Collapse
|
41
|
Keimasi M, Salehifard K, Shahidi M, Esmaeili F, Mirshah Jafar Esfahani N, Beheshti S, Amirsadri M, Naseri F, Keimasi M, Ghorbani N, Mofid MR, Moradmand M. Ameliorative effects of omega-lycotoxin-Gsp2671e purified from the spider venom of Lycosa praegrandis on memory deficits of glutamate-induced excitotoxicity rat model. Front Pharmacol 2022; 13:1048563. [PMID: 36588719 PMCID: PMC9800828 DOI: 10.3389/fphar.2022.1048563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Memory impairment is one of the main complications of Alzheimer's disease (AD). This condition can be induced by hyper-stimulation of N-Methyl-D-aspartate receptors (NMDARs) of glutamate in the hippocampus, which ends up to pyramidal neurons determination. The release of neurotransmitters relies on voltage-gated calcium channels (VGCCs) such as P/Q-types. Omega-lycotoxin-Gsp2671e (OLG1e) is a P/Q-type VGCC modulator with high affinity and selectivity. This bio-active small protein was purified and identified from the Lycosa praegrandis venom. The effect of this state-dependent low molecular weight P/Q-type calcium modulator on rats was investigated via glutamate-induced excitotoxicity by N-Methyl-D-aspartate. Also, Electrophysiological amplitude of field excitatory postsynaptic potentials (fEPSPs) in the input-output and Long-term potentiation (LTP) curves were recorded in mossy fiber and the amount of synaptophysin (SYN), synaptosomal-associated protein, 25 kDa (SNAP-25), and synaptotagmin 1(SYT1) genes expression were measured using Real-time PCR technique for synaptic quantification. The outcomes of the current study suggest that OLG1e as a P/Q-type VGCC modulator has an ameliorative effect on excitotoxicity-induced memory defects and prevents the impairment of pyramidal neurons in the rat hippocampus.
Collapse
Affiliation(s)
- Mohammad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran,*Correspondence: Majid Moradmand, ; Mohammad Reza Mofid, ; Mohammad Keimasi,
| | - Kowsar Salehifard
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Shahidi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Noushin Mirshah Jafar Esfahani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Siamak Beheshti
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Amirsadri
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Faezeh Naseri
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadjavad Keimasi
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Najmeh Ghorbani
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Reza Mofid
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran,*Correspondence: Majid Moradmand, ; Mohammad Reza Mofid, ; Mohammad Keimasi,
| | - Majid Moradmand
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran,*Correspondence: Majid Moradmand, ; Mohammad Reza Mofid, ; Mohammad Keimasi,
| |
Collapse
|
42
|
Sugisaki E, Fukushima Y, Nakajima N, Aihara T. The dependence of acetylcholine on dynamic changes in the membrane potential and an action potential during spike timing-dependent plasticity induction in the hippocampus. Eur J Neurosci 2022; 56:5972-5986. [PMID: 36164804 DOI: 10.1111/ejn.15832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 12/29/2022]
Abstract
The hippocampus is an important area for memory encoding and retrieval and is the location of spike timing-dependent plasticity (STDP), a basic phenomenon of learning and memory. STDP is facilitated if acetylcholine (ACh) is released from cholinergic neurons during attentional processes. However, it is unclear how ACh influences postsynaptic changes during STDP induction and determines the STDP magnitude. To address these issues, we obtained patch clamp recordings from CA1 pyramidal neurons to evaluate the postsynaptic changes during stimuli injection in Schaffer collaterals by quantifying baseline amplitudes (i.e., the lowest values elicited by paired pulses comprising STDP stimuli) and action potentials. The results showed that baseline amplitudes were elevated if eserine was applied in the presence of picrotoxin. In addition, muscarinic ACh receptors (mAChRs) contributed more to the baseline amplitude elevation than nicotinic AChRs (nAChRs). Moreover, the magnitude of the STDP depended on the magnitude of the baseline amplitude. However, in the absence of picrotoxin, baseline amplitudes were balanced, regardless of the ACh concentration, resulting in a similar magnitude of the STDP, except under the nAChR alone-activated condition, which showed a larger STDP and lower baseline amplitude induction. This was due to broadened widths of action potentials. These results suggest that activation of mAChRs and nAChRs, which are effective for baseline amplitudes and action potentials, respectively, plays an important role in postsynaptic changes during memory consolidation.
Collapse
Affiliation(s)
- Eriko Sugisaki
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| | - Yasuhiro Fukushima
- Brain Science Institute, Tamagawa University, Tokyo, Japan.,Kawasaki University of Medical Welfare, Okayama, Japan
| | - Naoki Nakajima
- Graduated School of Engineering, Tamagawa University, Tokyo, Japan
| | - Takeshi Aihara
- Brain Science Institute, Tamagawa University, Tokyo, Japan
| |
Collapse
|
43
|
Brain single cell transcriptomic profiles in episodic memory phenotypes associated with temporal lobe epilepsy. NPJ Genom Med 2022; 7:69. [PMID: 36446800 PMCID: PMC9709106 DOI: 10.1038/s41525-022-00339-4] [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: 06/14/2022] [Accepted: 11/08/2022] [Indexed: 12/02/2022] Open
Abstract
Memory dysfunction is prevalent in temporal lobe epilepsy (TLE), but little is known about the underlying molecular etiologies. Single-nucleus RNA sequencing technology was used to examine differences in cellular heterogeneity among left (language-dominant) temporal neocortical tissues from patients with TLE with (n = 4) or without (n = 2) impairment in verbal episodic memory. We observed marked cell heterogeneity between memory phenotypes and identified numerous differentially expressed genes across all brain cell types. The most notable differences were observed in glutamatergic (excitatory) and GABAergic (inhibitory) neurons with an overrepresentation of genes associated with long-term potentiation, long-term depression, and MAPK signaling, processes known to be essential for episodic memory formation.
Collapse
|
44
|
Harley HE, Fellner W, Frances C, Thomas A, Losch B, Newton K, Feuerbach D. Information-seeking across auditory scenes by an echolocating dolphin. Anim Cogn 2022; 25:1109-1131. [PMID: 36018473 DOI: 10.1007/s10071-022-01679-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022]
Abstract
Dolphins gain information through echolocation, a publicly accessible sensory system in which dolphins produce clicks and process returning echoes, thereby both investigating and contributing to auditory scenes. How their knowledge of these scenes contributes to their echoic information-seeking is unclear. Here, we investigate their top-down cognitive processes in an echoic matching-to-sample task in which targets and auditory scenes vary in their decipherability and shift from being completely unfamiliar to familiar. A blind-folded adult male dolphin investigated a target sample positioned in front of a hydrophone to allow recording of clicks, a measure of information-seeking and effort; the dolphin received fish for choosing an object identical to the sample from 3 alternatives. We presented 20 three-object sets, unfamiliar in the first five 18-trial sessions with each set. Performance accuracy and click counts varied widely across sets. Click counts of the four lowest-performance-accuracy/low-discriminability sets (X = 41%) and the four highest-performance-accuracy/high-discriminability sets (X = 91%) were similar at the first sessions' starts and then decreased for both kinds of scenes, although the decrease was substantially greater for low-discriminability sets. In four challenging-but-doable sets, number of clicks remained relatively steady across the 5 sessions. Reduced echoic effort with low-discriminability sets was not due to overall motivation: the differential relationship between click number and object-set discriminability was maintained when difficult and easy trials were interleaved and when objects from originally difficult scenes were grouped with more discriminable objects. These data suggest that dolphins calibrate their echoic information-seeking effort based on their knowledge and expectations of auditory scenes.
Collapse
Affiliation(s)
- Heidi E Harley
- Division of Social Sciences, New College of Florida, 5800 Bay Shore Road, Sarasota, FL, 34243, USA.
- The Seas, Epcot®, Walt Disney World® Resorts , Lake Buena Vista, FL, USA.
| | - Wendi Fellner
- The Seas, Epcot®, Walt Disney World® Resorts , Lake Buena Vista, FL, USA
| | - Candice Frances
- Division of Social Sciences, New College of Florida, 5800 Bay Shore Road, Sarasota, FL, 34243, USA
- Basque Center on Cognition, Brain and Language, Donostia, Spain
| | - Amber Thomas
- Division of Social Sciences, New College of Florida, 5800 Bay Shore Road, Sarasota, FL, 34243, USA
- The Seas, Epcot®, Walt Disney World® Resorts , Lake Buena Vista, FL, USA
| | - Barbara Losch
- The Seas, Epcot®, Walt Disney World® Resorts , Lake Buena Vista, FL, USA
| | - Katherine Newton
- Division of Social Sciences, New College of Florida, 5800 Bay Shore Road, Sarasota, FL, 34243, USA
- Department of Fisheries and Wildlife, Oregon State University, Corvallis, USA
| | - David Feuerbach
- The Seas, Epcot®, Walt Disney World® Resorts , Lake Buena Vista, FL, USA
| |
Collapse
|
45
|
Carles A, Schlernitzauer A, Vignes M, Cros G, Magous R, Maurice T, Oiry C. Heptafluoroisobutyronitrile (C 4F 7N), a gas used for insulating and arc quenching in electrical switchgear, is neurotoxic in the mouse brain. Toxicology 2022; 480:153319. [PMID: 36100137 DOI: 10.1016/j.tox.2022.153319] [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: 07/22/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
Fluoronitrile gas (C4F7N, CAS number 42532-60-5) is one of the most promising candidates as insulating and/or breaking medium in high and medium voltage electrical equipment. Besides its promising properties, C4F7N gas is however not devoid of acute toxicity when used pure or in gas mixtures. The toxicity was not extensively analyzed and reported. The aim of the present study was to analyze in mice the consequences of a single exposure to C4F7N gas, at different concentrations and different timepoints after exposure. Male and female Swiss mice were exposed to breathable air or C4F7N gas, at 800 ppmv or 1500 ppmv, for 4 h on day 0. Behavioral tests (spontaneous alternation in the Y-maze and object recognition) were performed on days 1, 7 and 14 to assess memory alterations. The animals were then sacrificed and their brains dissected for biochemical analyses or fixed with paraformaldehyde for histology and immunohistochemistry. Results showed behavioral impairments and memory deficits, with impairments of alternation at days 1 and 7 and object recognition at day 14. Histological alterations of pyramidal neuronal layer in the hippocampus, neuroinflammatory astroglial reaction, and microglial alterations were observed, more marked in female than male mice. Moreover, the biochemical analyses done in the brain of 1500 ppmv exposed female mice showed a reductive stress with decreased lipid peroxidation and release of cytochrome c, leading to apoptosis with increases in caspase-9 cleavage and γ-H2AX/H2AX ratio. Finally, electrophysiological analyses using a multi-electrode array allowed the measure of the extracellular activity of pyramidal neurons in the CA2 area and revealed that exposure to the gas not only prevented the induction of long-term potentiation but even provoked an epileptoid-like activity in some neurons suggesting major alterations of synaptic plasticity. This study therefore showed that an acute exposure of mice to C4F7N gas provoked, particularly in female animals, memory alterations and brain toxicity characterized by a reductive stress, microglial toxicity, loss of synaptic plasticity and apoptosis. Its use in industrial installations must be done with extreme caution.
Collapse
Affiliation(s)
- Allison Carles
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Michel Vignes
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Gérard Cros
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Richard Magous
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Tangui Maurice
- ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| | - Catherine Oiry
- ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| |
Collapse
|
46
|
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: 5] [Impact Index Per Article: 1.7] [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
|
47
|
Liu Y, Li Z, Wu W, Wang Y, Zhao G, Liu Y, Liu J, Song Z. UVR Promotes Keratinocyte Phagocytosis and Skin Pigmentation Through TRPA1 Channels. CLINICAL, COSMETIC AND INVESTIGATIONAL DERMATOLOGY 2022; 15:1183-1193. [PMID: 35784271 PMCID: PMC9249384 DOI: 10.2147/ccid.s365682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/15/2022] [Indexed: 11/23/2022]
Affiliation(s)
- Ying Liu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Zhou Li
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Wei Wu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yupeng Wang
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Guangming Zhao
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Yuejian Liu
- Central Laboratory, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Jing Liu
- Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
| | - Zhiqi Song
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, People’s Republic of China
- Correspondence: Zhiqi Song, Department of Dermatology, First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, People’s Republic of China, Email
| |
Collapse
|
48
|
Identification and characterization of long non-coding RNA Carip in modulating spatial learning and memory. Cell Rep 2022; 38:110398. [PMID: 35196493 DOI: 10.1016/j.celrep.2022.110398] [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: 06/16/2021] [Revised: 11/18/2021] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
CaMKII has long been known to be a key effector for synaptic plasticity. Recent studies have shown that a variety of modulators interact with the subunits of CaMKII to regulate the long-term potentiation (LTP) of hippocampal neurons. However, whether long non-coding RNAs modulate the activity of CaMKII and affect synaptic plasticity is still elusive. Here, we identify a previously uncharacterized long non-coding RNA Carip that functions as a scaffold, specifically interacts with CaMKIIβ, and regulates the phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptor subunits in the hippocampus. The absence of Carip causes dysfunction of synaptic transmission and attenuates LTP in hippocampal CA3-CA1 synapses, which further leads to impairment of spatial learning and memory. In summary, our findings demonstrate that Carip modulates long-term synaptic plasticity by changing AMPA receptor and NMDA receptor activities, thereby affecting spatial learning and memory in mice.
Collapse
|
49
|
Hayashi-Takagi A. Constructive Understanding of Multi-scale Dynamism of Psychiatric Disorders. Neurosci Res 2022; 175:1-2. [PMID: 35038499 DOI: 10.1016/j.neures.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Akiko Hayashi-Takagi
- Laboratory for Multi-scale Biological Psychiatry, Center for Brain Science, RIKEN, 2-1 Hirosawa, Wako-city, Saitama, 351-0198, Japan.
| |
Collapse
|
50
|
Flores-Clemente C, Nicolás-Vázquez MI, Mera Jiménez E, Hernández-Rodríguez M. Inhibition of Astrocytic Histamine N-Methyltransferase as a Possible Target for the Treatment of Alzheimer's Disease. Biomolecules 2021; 11:1408. [PMID: 34680041 PMCID: PMC8533269 DOI: 10.3390/biom11101408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer's disease (AD) represents the principal cause of dementia among the elderly. Great efforts have been established to understand the physiopathology of AD. Changes in neurotransmitter systems in patients with AD, including cholinergic, GABAergic, serotoninergic, noradrenergic, and histaminergic changes have been reported. Interestingly, changes in the histaminergic system have been related to cognitive impairment in AD patients. The principal pathological changes in the brains of AD patients, related to the histaminergic system, are neurofibrillary degeneration of the tuberomammillary nucleus, the main source of histamine in the brain, low histamine levels, and altered signaling of its receptors. The increase of histamine levels can be achieved by inhibiting its degrading enzyme, histamine N-methyltransferase (HNMT), a cytoplasmatic enzyme located in astrocytes. Thus, increasing histamine levels could be employed in AD patients as co-therapy due to their effects on cognitive functions, neuroplasticity, neuronal survival, neurogenesis, and the degradation of amyloid beta (Aβ) peptides. In this sense, the evaluation of the impact of HNMT inhibitors on animal models of AD would be interesting, consequently highlighting its relevance.
Collapse
Affiliation(s)
- Cecilia Flores-Clemente
- Laboratorio de Cultivo Celular, Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (C.F.-C.); (E.M.J.)
| | - María Inés Nicolás-Vázquez
- Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán Campo 1, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54714, Mexico;
| | - Elvia Mera Jiménez
- Laboratorio de Cultivo Celular, Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (C.F.-C.); (E.M.J.)
| | - Maricarmen Hernández-Rodríguez
- Laboratorio de Cultivo Celular, Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico; (C.F.-C.); (E.M.J.)
| |
Collapse
|