1
|
Liu Q, Wang HB, Lin JT, Jiao XH, Liu YP, Li TZ, Xie Z, Zhou CH, Wu YQ, Miao HH. Role of brain-derived neurotrophic factor in dysfunction of short-term to long-term memory transformation after surgery and anaesthesia in older mice. Br J Anaesth 2025; 134:1134-1145. [PMID: 39909796 PMCID: PMC11947570 DOI: 10.1016/j.bja.2024.11.045] [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: 05/09/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Memory decline is one of the main manifestations in perioperative neurocognitive disorder. Short-term memory (STM) to long-term memory (LTM) transformation is one aspect of memory consolidation. Early-phase long-term potentiation (E-LTP) to late-phase long-term potentiation (L-LTP) is the molecular correlate of STM to LTM transformation. We examined whether the STM to LTM transformation was impaired after anaesthesia and surgery in older mice. METHODS Optogenetics and chemogenetics were used to confirm the role of Vglut1+ glutamatergic neurones in the STM to LTM transformation in older mice. Synaptosomes were isolated to analyse expression of brain-derived neurotrophic factor (BDNF). Golgi-Cox staining and hippocampal field potential recordings were also used to measure synaptic plasticity. RESULTS We found that the STM to LTM and E-LTP to L-LTP transformations were impaired after anaesthesia and surgery in older mice, and Vglut1+ excitatory neurone activity in the hippocampal CA1 region was reduced. BDNF expression decreased in the postsynaptic fraction, especially in Vglut1+ neurones, whereas cell-type specific overexpression of BDNF in Vglut1+ neurones reversed postoperative STM to LTM transformation dysfunction in older mice. CONCLUSIONS Reduced BDNF expression was involved in anaesthesia and surgery-induced impairment of the STM to LTM transition involving glutamatergic neurones in the hippocampal CA1 region of older mice. This provides a potential target that might be helpful for understanding and developing treatments for postoperative neurocognitive dysfunction.
Collapse
Affiliation(s)
- Qiang Liu
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China; Department of Anaesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Hai-Bi Wang
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China
| | - Jia-Tao Lin
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China
| | - Xin-Hao Jiao
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China
| | - Yan-Ping Liu
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China
| | - Tian-Zuo Li
- Department of Anaesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, PR China
| | - Zhongcong Xie
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, PR China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anaesthesiology/NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, PR China.
| | - Hui-Hui Miao
- Department of Anaesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
2
|
Huang S, Liu X, Li Z, Si Y, Yang L, Deng J, Luo Y, Xue YX, Lu L. Memory Reconsolidation Updating in Substance Addiction: Applications, Mechanisms, and Future Prospects for Clinical Therapeutics. Neurosci Bull 2025; 41:289-304. [PMID: 39264570 PMCID: PMC11794923 DOI: 10.1007/s12264-024-01294-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: 01/06/2024] [Accepted: 05/09/2024] [Indexed: 09/13/2024] Open
Abstract
Persistent and maladaptive drug-related memories represent a key component in drug addiction. Converging evidence from both preclinical and clinical studies has demonstrated the potential efficacy of the memory reconsolidation updating procedure (MRUP), a non-pharmacological strategy intertwining two distinct memory processes: reconsolidation and extinction-alternatively termed "the memory retrieval-extinction procedure". This procedure presents a promising approach to attenuate, if not erase, entrenched drug memories and prevent relapse. The present review delineates the applications, molecular underpinnings, and operational boundaries of MRUP in the context of various forms of substance dependence. Furthermore, we critically examine the methodological limitations of MRUP, postulating potential refinement to optimize its therapeutic efficacy. In addition, we also look at the potential integration of MRUP and neurostimulation treatments in the domain of substance addiction. Overall, existing studies underscore the significant potential of MRUP, suggesting that interventions predicated on it could herald a promising avenue to enhance clinical outcomes in substance addiction therapy.
Collapse
Affiliation(s)
- Shihao Huang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China
| | - Zhonghao Li
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yue Si
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Liping Yang
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jiahui Deng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China
| | - Yixiao Luo
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Yan-Xue Xue
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
| | - Lin Lu
- Department of Neurobiology, School of Basic Medical Sciences, National Institute on Drug Dependence, Peking University, Beijing, 100191, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, 100191, China.
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Sharbafshaaer M, Cirillo G, Esposito F, Tedeschi G, Trojsi F. Harnessing Brain Plasticity: The Therapeutic Power of Repetitive Transcranial Magnetic Stimulation (rTMS) and Theta Burst Stimulation (TBS) in Neurotransmitter Modulation, Receptor Dynamics, and Neuroimaging for Neurological Innovations. Biomedicines 2024; 12:2506. [PMID: 39595072 PMCID: PMC11592033 DOI: 10.3390/biomedicines12112506] [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: 08/31/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Transcranial magnetic stimulation (TMS) methods have become exciting techniques for altering brain activity and improving synaptic plasticity, earning recognition as valuable non-medicine treatments for a wide range of neurological disorders. Among these methods, repetitive TMS (rTMS) and theta-burst stimulation (TBS) show significant promise in improving outcomes for adults with complex neurological and neurodegenerative conditions, such as Alzheimer's disease, stroke, Parkinson's disease, etc. However, optimizing their effects remains a challenge due to variability in how patients respond and a limited understanding of how these techniques interact with crucial neurotransmitter systems. This narrative review explores the mechanisms of rTMS and TBS, which enhance neuroplasticity and functional improvement. We specifically focus on their effects on GABAergic and glutamatergic pathways and how they interact with key receptors like N-Methyl-D-Aspartate (NMDA) and AMPA receptors, which play essential roles in processes like long-term potentiation (LTP) and long-term depression (LTD). Additionally, we investigate how rTMS and TBS impact neuroplasticity and functional connectivity, particularly concerning brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase receptor type B (TrkB). Here, we highlight the significant potential of this research to expand our understanding of neuroplasticity and better treatment outcomes for patients. Through clarifying the neurobiology mechanisms behind rTMS and TBS with neuroimaging findings, we aim to develop more effective, personalized treatment plans that effectively address the challenges posed by neurological disorders and ultimately enhance the quality of neurorehabilitation services and provide future directions for patients' care.
Collapse
Affiliation(s)
- Minoo Sharbafshaaer
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Giovanni Cirillo
- Division of Human Anatomy, Neuronal Networks Morphology & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy;
| | - Fabrizio Esposito
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Gioacchino Tedeschi
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Francesca Trojsi
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| |
Collapse
|
4
|
Naseem M, Khan H, Parvez S. TrkB-BDNF Signalling and Arc/Arg3.1 Immediate Early Genes in the Anterior Cingulate Cortex and Hippocampus: Insights into Novel Memory Milestones Through Behavioural Tagging. Mol Neurobiol 2024; 61:8307-8319. [PMID: 38485841 DOI: 10.1007/s12035-024-04071-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/26/2024] [Indexed: 09/21/2024]
Abstract
In recent years, there has been a surge in interest in investigating the mechanisms underlying memory consolidation. However, our understanding of the behavioural tagging (BT) model and its establishment in diverse brain regions remains limited. This study elucidates the contributions of the anterior cingulate cortex (ACC) and hippocampus in the formation of long-term memory (LTM) employing behaviour tagging as a model for studying the underlying mechanism of LTM formation in rats. Existing knowledge highlights a protein synthesis-dependent phase as imperative for LTM. Brain-derived neurotrophic factor (BDNF) stands as a pivotal plasticity-related protein (PRP) in mediating molecular alterations crucial for long-term synaptic plasticity and memory consolidation. Our study offers evidence suggesting that tropomyosin receptor kinase B (TrkB), the receptor of BDNF, may act as a combined "behavioural tag/PRP". Interfering with the expression of these molecules resulted in impaired LTM after 24 h. Furthermore, augmenting BDNF expression led to an elevation in Arc protein levels in both the ACC and hippocampus regions. Introducing novelty around weak inhibitory avoidance (IA) training resulted in heightened step-down latencies and expression of these molecules, respectively. We also demonstrate that the increase in Arc expression relies on BDNF synthesis, which is vital for the memory consolidation process. Additionally, inhibiting BDNF using an anti-BDNF function-blocking antibody impacted Arc expression in both the ACC and hippocampus regions, disrupting the transformations from labile to robust memory. These findings mark the initial identification of a "behavioural tag/PRP" combination and underscore the involvement of the TrkB-BDNF-Arc cascade in the behavioural tagging model of learning and memory.
Collapse
Affiliation(s)
- Mehar Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
5
|
Bin Ibrahim MZ, Wang Z, Sajikumar S. Synapses tagged, memories kept: synaptic tagging and capture hypothesis in brain health and disease. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230237. [PMID: 38853570 PMCID: PMC11343274 DOI: 10.1098/rstb.2023.0237] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 06/11/2024] Open
Abstract
The synaptic tagging and capture (STC) hypothesis lays the framework on the synapse-specific mechanism of protein synthesis-dependent long-term plasticity upon synaptic induction. Activated synapses will display a transient tag that will capture plasticity-related products (PRPs). These two events, tag setting and PRP synthesis, can be teased apart and have been studied extensively-from their electrophysiological and pharmacological properties to the molecular events involved. Consequently, the hypothesis also permits interactions of synaptic populations that encode different memories within the same neuronal population-hence, it gives rise to the associativity of plasticity. In this review, the recent advances and progress since the experimental debut of the STC hypothesis will be shared. This includes the role of neuromodulation in PRP synthesis and tag integrity, behavioural correlates of the hypothesis and modelling in silico. STC, as a more sensitive assay for synaptic health, can also assess neuronal aberrations. We will also expound how synaptic plasticity and associativity are altered in ageing-related decline and pathological conditions such as juvenile stress, cancer, sleep deprivation and Alzheimer's disease. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Mohammad Zaki Bin Ibrahim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
| | - Zijun Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
| |
Collapse
|
6
|
Shen YJ, Ji MY, Huang Q, Hsueh CY, Du HD, Zhang M, Zhou L. Nicotine downregulates miR-375-3p via neurotrophic tyrosine receptor kinase 2 to enhance the malignant behaviors of laryngopharyngeal squamous epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 274:116215. [PMID: 38489902 DOI: 10.1016/j.ecoenv.2024.116215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Nicotine exposure from smoking constitutes a significant global public health concern. Furthermore, smoking represents a pivotal risk factor for head and neck squamous cell carcinoma (HNSCC). However, the influence of nicotine on HNSCC remains relatively underexplored. Our aim was to unravel the molecular mechanisms that underlie the effect of nicotine on the metastatic cascade of HNSCC. In this study, we discovered a significant association between smoking and HNSCC metastasis and prognosis. Nicotine significantly enhanced HNSCC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro. Analysis of TCGA-HNSCC and FDEENT-HNSCC cohorts revealed reduced miR-375-3p levels in HNSCC tumor tissues, particularly among current smokers. Additionally, miR-375-3p level was strongly correlated with both lymph node metastasis and tumor stage. By downregulating miR-375-3p, nicotine promotes HNSCC cell metastasis in vitro and hematogenous metastatic capacity in vivo. Utilizing transcriptomic sequencing, molecular docking, dual-luciferase reporter assay, and fluorescence in situ hybridization (FISH), we demonstrated that miR-375-3p specifically binds to 3' untranslated region (3'UTR) of NTRK2 mRNA. Thus, this study uncovers a novel nicotine-induced mechanism involving miR-375-3p-mediated NTRK2 targeting, which promotes HNSCC metastasis. These findings have implications for improving the prognosis of patients with HNSCC, especially in smokers.
Collapse
Affiliation(s)
- Yu-Jie Shen
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China
| | - Meng-You Ji
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China
| | - Qiang Huang
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China
| | - Chi-Yao Hsueh
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China.
| | - Huai-Dong Du
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China.
| | - Ming Zhang
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China.
| | - Liang Zhou
- Department of Otorhinolaryngology- Head and Neck Surgery, Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, China.
| |
Collapse
|
7
|
Schroeder MN, Fullio CL, Ballarini F, Moncada D. Modulation of memory reconsolidation by adjacent novel tasks: timing defines the nature of change. Commun Biol 2023; 6:1288. [PMID: 38114781 PMCID: PMC10730840 DOI: 10.1038/s42003-023-05666-5] [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: 07/03/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023] Open
Abstract
Reconsolidation turns memories into a responsive state that allows their modulation until they stabilize again. This phenomenon attracted remarkable attention due to its potential impact on therapeutics and education. Recent evidence revealed that different memories undergo reconsolidation via a behavioral tagging process. Thus, their re-stabilization involves setting "reconsolidation-tags" and synthesizing plasticity-related proteins for their capture at the tagged sites. Here, we studied the possibility of affecting these fundamental mechanisms to modulate reconsolidation. Our findings, in laboratory rats, indicate that exploring a novel environment 60 min before or after memory reactivation improves spatial object recognition memory by promoting protein synthesis. Conversely, experiencing novelty immediately after reactivation impairs the reconsolidation by affecting the tags. Similar effects, but with a different optimal time window for improvement, occur in inhibitory avoidance memory. These results highlight the possibility of modulating existing memories using non-invasive interventions that selectively affect the fundamental mechanisms of behavioral tagging during their reconsolidation.
Collapse
Affiliation(s)
- Matías Nicolás Schroeder
- Laboratorio de Neurofisiología de la Memoria, Instituto de Biología Celular y Neurociencia, Facultad de Medicina (UBA/CONICET) - Instituto Tecnológico de Buenos Aires (ITBA), Ciudad Autónoma de Buenos Aires, Argentina
| | - Camila L Fullio
- Laboratorio de Neurofisiología de la Memoria, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fabricio Ballarini
- Laboratorio de neurociencia translacional, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires, Av. Madero 399, Ciudad Autónoma de Buenos Aires, Argentina
| | - Diego Moncada
- Laboratorio de Neurofisiología de la Memoria, Instituto de Biología Celular y Neurociencia, Facultad de Medicina (UBA/CONICET) - Instituto Tecnológico de Buenos Aires (ITBA), Ciudad Autónoma de Buenos Aires, Argentina.
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile.
| |
Collapse
|
8
|
Jeanneteau F. Stress and the risk of Alzheimer dementia: Can deconstructed engrams be rebuilt? J Neuroendocrinol 2023; 35:e13235. [PMID: 36775895 DOI: 10.1111/jne.13235] [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: 11/28/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023]
Abstract
The exact neuropathological mechanism by which the dementia process unfolds is under intense scrutiny. The disease affects about 38 million people worldwide, 70% of which are clinically diagnosed with Alzheimer's disease (AD). If the destruction of synapses essential for learning, planning and decision-making is part of the problem, must the restoration of previously lost synapses be part of the solution? It is plausible that neuronal capacity to restitute information corresponds with the adaptive capacity of its connectivity reserve. A challenge will be to promote the functional connectivity that can compensate for the lost one. This will require better clarification of the remodeling of functional connectivity during the progression of AD dementia and its reversal upon experimental treatment. A major difficulty is to promote the neural pathways that are atrophied in AD dementia while suppressing others that are bolstered. Therapeutic strategies should aim at scaling functional connectivity to a just balance between the atrophic and hypertrophic systems. However, the exact factors that can help reach this objective are still unclear. Similarities between the effects of chronic stress and some neuropathological mechanisms underlying AD dementia support the idea that common components deserve prime attention as therapeutic targets.
Collapse
Affiliation(s)
- Freddy Jeanneteau
- Institut de génomique fonctionnelle, Université de Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
9
|
Budriesi P, Tintorelli R, Correa J, Villar ME, Marchal P, Giurfa M, Viola H. A behavioral tagging account of kinase contribution to memory formation after spaced aversive training. iScience 2023; 26:107278. [PMID: 37520708 PMCID: PMC10372744 DOI: 10.1016/j.isci.2023.107278] [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] [Received: 08/28/2022] [Revised: 09/14/2022] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Long-term memory (LTM) can be induced by repeated spaced training trials. Using the weak inhibitory avoidance (wIA) task, we showed that one wIA session does not lead to a 24-h LTM, whereas two identical wIA sessions spaced by 15 min to 6 h induce a 24-h LTM. This LTM promotion depends both on hippocampal protein synthesis and the activity of several kinases. In agreement with the behavioral tagging (BT) hypothesis, our results suggest that the two training sessions induce transient learning tags and lead, via a cooperative effect, to the synthesis of plasticity-related proteins (PRPs) that become available and captured by the tag from the second session. Although ERKs1/2 are needed for PRPs synthesis and CaMKs are required for tag setting, PKA participates in both processes. We conclude that the BT mechanism accounts for the molecular constraints underlying the classic effect of spaced learning on LTM formation.
Collapse
Affiliation(s)
- Pablo Budriesi
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ramiro Tintorelli
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julieta Correa
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maria Eugenia Villar
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Biología y Geología, Física y Química Inorgánica, Área de Biodiversidad y Conservación, Universidad Rey Juan Carlos, Madrid, Spain
| | - Paul Marchal
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
- Poe Lab, Integrative Biology and Physiology department, University of California Los Angeles, Los Angeles, CA, USA
| | - Martin Giurfa
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse cedex 9, France
- Institut Universitaire de France (IUF), Paris, France
| | - Haydee Viola
- Instituto de Biología Celular y Neurociencia “Prof. E. De Robertis” (IBCN), Facultad de Medicina, UBA-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular “Dr. Héctor Maldonado” (FBMC), Facultad de Ciencias Exactas y Naturales, UBA, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
10
|
Kastellakis G, Tasciotti S, Pandi I, Poirazi P. The dendritic engram. Front Behav Neurosci 2023; 17:1212139. [PMID: 37576932 PMCID: PMC10412934 DOI: 10.3389/fnbeh.2023.1212139] [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: 04/25/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Accumulating evidence from a wide range of studies, including behavioral, cellular, molecular and computational findings, support a key role of dendrites in the encoding and recall of new memories. Dendrites can integrate synaptic inputs in non-linear ways, provide the substrate for local protein synthesis and facilitate the orchestration of signaling pathways that regulate local synaptic plasticity. These capabilities allow them to act as a second layer of computation within the neuron and serve as the fundamental unit of plasticity. As such, dendrites are integral parts of the memory engram, namely the physical representation of memories in the brain and are increasingly studied during learning tasks. Here, we review experimental and computational studies that support a novel, dendritic view of the memory engram that is centered on non-linear dendritic branches as elementary memory units. We highlight the potential implications of dendritic engrams for the learning and memory field and discuss future research directions.
Collapse
Affiliation(s)
- George Kastellakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| | - Simone Tasciotti
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Ioanna Pandi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| |
Collapse
|
11
|
Broadbelt T, Mutlu-Smith M, Carnicero-Senabre D, Saido TC, Saito T, Wang SH. Impairment in novelty-promoted memory via behavioral tagging and capture before apparent memory loss in a knock-in model of Alzheimer's disease. Sci Rep 2022; 12:22298. [PMID: 36566248 PMCID: PMC9789965 DOI: 10.1038/s41598-022-26113-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is associated with cognitive impairments and age-dependent memory deficits which have been studied using genetic models of AD. Whether the processes for modulating memory persistence are more vulnerable to the influence of amyloid pathology than the encoding and consolidation of the memory remains unclear. Here, we investigated whether early amyloid pathology would affect peri-learning novelty in promoting memory, through a process called behavioral tagging and capture (BTC). AppNL-G-F/NL-G-F mice and wild-type littermates were trained in an appetitive delayed matching-to-place (ADMP) task which allows for the assessment of peri-learning novelty in facilitating memory. The results show that novelty enabled intermediate-term memory in wild-type mice, but not in AppNL-G-F/NL-G-F mice in adulthood. This effect preceded spatial memory impairment in the ADMP task seen in middle age. Other memory tests in the Barnes maze, Y-maze, novel object or location recognition tasks remained intact. Together, memory modulation through BTC is impaired before apparent deficits in learning and memory. Relevant biological mechanisms underlying BTC and the implication in AD are discussed.
Collapse
Affiliation(s)
- Tabitha Broadbelt
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Menekse Mutlu-Smith
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Daniel Carnicero-Senabre
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,grid.5515.40000000119578126Present Address: Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Takaomi C. Saido
- grid.474690.8Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, 351-0198 Japan
| | - Takashi Saito
- grid.260433.00000 0001 0728 1069Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, 467-8601 Japan
| | - Szu-Han Wang
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| |
Collapse
|
12
|
Hong J, Jeong Y, Heo WD. The Neurotrophic Receptor Tyrosine Kinase in MEC-mPFC Neurons Contributes to Remote Memory Consolidation. J Neurosci 2022; 42:6605-6619. [PMID: 35863892 PMCID: PMC9410758 DOI: 10.1523/jneurosci.2433-21.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022] Open
Abstract
The PFC is thought to be the region where remote memory is recalled. However, the neurotrophic receptors that underlie the remote memory remain largely unknown. Here, we benefited from auto-assembly split Cre to accomplish the neural projection-specific recombinase activity without spontaneous leakage. Deletion of tropomyosin receptor kinase B (TrkB) in neurons projecting from the medial entorhinal cortex to the mPFC displayed reduced remote memory recall from the male mice, but the recent recall was intact. We found that the TrkB deletion attenuates the participation of mPFC cells in the remote fear memory recall. The disruption of remote recall was attributed to reduced reactivation of cells in the mPFC. Notably, TrkB deletion seriously inhibited experience-dependent maturation of oligodendroglia in the PFC, resulting in defects in remote recall that were rescued by clemastine administration. Together, our data suggest that TrkB in intercortical circuits functions in remote memory consolidation.SIGNIFICANCE STATEMENT Retrieving the past experiences or events is essential for the ones to lead life. The investigations performed in the rodent model have disclosed that the systems consolidation of memory accompanying changes of cortical circuits and transcriptome is required for maintaining the memory for a long time. In this study, the split Cre with TrkBflox/flox mice were subjected to discover that TrkB in the neurons plays a role in remote memory consolidation. We evaluated the contextual fear memory and labeled cells, which revealed deletion of TrkB interrupts newborn oligodendrocyte and reactivation of cells in mPFC at remote recall. Our data provide the implication that remote memory is relevant to neurotrophic receptor signaling as well as its influence on non-neuronal cells.
Collapse
Affiliation(s)
- Jongryul Hong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
- Korea Advanced Institute of Science and Technology Institute for the BioCentury, Daejeon, 34141, Republic of Korea
| |
Collapse
|
13
|
Raghuraman R, Manakkadan A, Richter-Levin G, Sajikumar S. Inhibitory Metaplasticity in Juvenile Stressed Rats Restores Associative Memory in Adulthood by Regulating Epigenetic Complex G9a/GLP. Int J Neuropsychopharmacol 2022; 25:576-589. [PMID: 35089327 PMCID: PMC9352179 DOI: 10.1093/ijnp/pyac008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/23/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Exposure to juvenile stress was found to have long-term effects on the plasticity and quality of associative memory in adulthood, but the underlying mechanisms are still poorly understood. METHODS Three- to four week-old male Wistar rats were subjected to a 3-day juvenile stress paradigm. Their electrophysiological correlates of memory using the adult hippocampal slice were inspected to detect alterations in long-term potentiation and synaptic tagging and capture model of associativity. These cellular alterations were tied in with the behavioral outcome by subjecting the rats to a step-down inhibitory avoidance paradigm to measure strength in their memory. Given the role of epigenetic response in altering plasticity as a repercussion of juvenile stress, we aimed to chart out the possible epigenetic marker and its regulation in the long-term memory mechanisms using quantitative reverse transcription polymerase chain reaction. RESULTS We demonstrate that even long after the elimination of actual stressors, an inhibitory metaplastic state is evident, which promotes synaptic competition over synaptic cooperation and decline in latency of associative memory in the behavioral paradigm despite the exposure to novelty. Mechanistically, juvenile stress led to a heightened expression of the epigenetic marker G9a/GLP complex, which is thus far ascribed to transcriptional silencing and goal-directed behavior. CONCLUSIONS The blockade of the G9a/GLP complex was found to alleviate deficits in long-term plasticity and associative memory during the adulthood of animals exposed to juvenile stress. Our data provide insights on the long-term effects of juvenile stress that involve epigenetic mechanisms, which directly impact long-term plasticity, synaptic tagging and capture, and associative memory.
Collapse
Affiliation(s)
- Radha Raghuraman
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Anoop Manakkadan
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Gal Richter-Levin
- Sagol department of Neurobiology, Department of Psychology, University of Haifa, Haifa, Israel
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, Israel
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore
| |
Collapse
|
14
|
Kaushik M, Kaushik P, Parvez S. Memory related molecular signatures: The pivots for memory consolidation and Alzheimer's related memory decline. Ageing Res Rev 2022; 76:101577. [PMID: 35104629 DOI: 10.1016/j.arr.2022.101577] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/23/2021] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Age-related cognitive decline is the major cause of concern due to its 70% more incidence than dementia cases worldwide. Moreover, aging is also the major risk factor of Alzheimer's disease (AD), associated with progressive memory loss. Approx. 13 million people will have Alzheimer-related memory decline by 2050. Learning and memory is the fundamental process of brain functions. However, the mechanism for the same is still under investigation. Thus, it is critical to understand the process of memory consolidation in the brain and extrapolate its understanding to the memory decline mechanism. Research on learning and memory has identified several molecular signatures such as Protein kinase M zeta (PKMζ), Calcium/calmodulin-dependent protein kinase II (CaMKII), Brain-derived neurotrophic factor (BDNF), cAMP-response element binding protein (CREB) and Activity-regulated cytoskeleton-associated protein (Arc) crucial for the maintenance and stabilization of long-term memory in the brain. Interestingly, memory decline in AD has also been linked to the abnormality in expressing these memory-related molecular signatures. Hence, in the present consolidated review, we explored the role of these memory-related molecular signatures in long-term memory consolidation. Additionally, the effect of amyloid-beta toxicity on these molecular signatures is discussed in detail.
Collapse
Affiliation(s)
- Medha Kaushik
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
15
|
Correa J, Tintorelli R, Budriesi P, Viola H. Persistence of spatial memory induced by spaced training involves a behavioral-tagging process. Neuroscience 2022; 497:215-227. [PMID: 35276307 DOI: 10.1016/j.neuroscience.2022.02.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/04/2022] [Accepted: 02/26/2022] [Indexed: 11/27/2022]
Abstract
Spaced training, which involves long inter-trial intervals, has positive effects on memories. One of the main attributes of long-term memories (LTM) is persistence. Here, to identify the process that promotes LTM persistence by spaced learning, we used the spatial object recognition (SOR) task. The protocol consisted of a first strong training session that induced LTM formation (tested 1 day after training), but not LTM persistence (tested 7 or 14 days after training); and a second weak training session that promoted memory persistence when applied 1 day, but not 7 days, after the first training. We propose that the promotion of memory persistence is based on the Behavioral Tagging (BT) mechanism operating when the memory trace is retrieved. BT involves the setting of a tag induced by learning which gives rise to input selectivity, and the use of plasticity-related proteins (PRPs) to establish the mnemonic trace. We postulate that retraining will mainly retag the sites initially activated by the original learning, where the PRPs needed for memory expression and/or induced by retrieval would be used to maintain a persistent mnemonic trace. Our results suggest that the mechanism of memory expression, but not those of memory reinforcement or reconsolidation, is necessary to promote memory persistence after retraining. The molecular mechanisms involve ERKs1/2 activity to set the SOR learning tag, and the availability of GluA2-containing AMPA receptor. In conclusion, both the synthesis of PRPs and the setting of a learning tag are key processes triggered by retraining that allow SOR memory persistence.
Collapse
Affiliation(s)
- J Correa
- Facultad de Medicina. Universidad de Buenos Aires. Buenos Aires, Argentina; Laboratorio de Memoria, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - R Tintorelli
- Facultad de Medicina. Universidad de Buenos Aires. Buenos Aires, Argentina; Laboratorio de Memoria, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - P Budriesi
- Facultad de Medicina. Universidad de Buenos Aires. Buenos Aires, Argentina; Laboratorio de Memoria, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina
| | - H Viola
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (FBMC), Facultad de Ciencias Exactas y Naturales, UBA, Buenos Aires, Argentina; Laboratorio de Memoria, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, UBA-CONICET, Buenos Aires, Argentina; Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina.
| |
Collapse
|
16
|
Fading memories in aging and neurodegeneration: Is p75 neurotrophin receptor a culprit? Ageing Res Rev 2022; 75:101567. [PMID: 35051645 DOI: 10.1016/j.arr.2022.101567] [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: 09/14/2021] [Revised: 12/12/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
Aging and age-related neurodegenerative diseases have become one of the major concerns in modern times as cognitive abilities tend to decline when we get older. It is well known that the main cause of this age-related cognitive deficit is due to aberrant changes in cellular, molecular circuitry and signaling pathways underlying synaptic plasticity and neuronal connections. The p75 neurotrophin receptor (p75NTR) is one of the important mediators regulating the fate of the neurons in the nervous system. Its importance in neuronal apoptosis is well documented. However, the mechanisms involving the regulation of p75NTR in synaptic plasticity and cognitive function remain obscure, although cognitive impairment has been associated with a higher expression of p75NTR in neurons. In this review, we discuss the current understanding of how neurons are influenced by p75NTR function to maintain normal neuronal synaptic strength and connectivity, particularly to support learning and memory in the hippocampus. We then discuss the age-associated alterations in neurophysiological mechanisms of synaptic plasticity and cognitive function. Furthermore, we also describe current evidence that has begun to elucidate how p75NTR regulates synaptic changes in aging and age-related neurodegenerative diseases, focusing on the hippocampus. Elucidating the role that p75NTR signaling plays in regulating synaptic plasticity will contribute to a better understanding of cognitive processes and pathological conditions. This will in turn provide novel approaches to improve therapies for the treatment of neurological diseases in which p75NTR dysfunction has been demonstrated.
Collapse
|
17
|
Distinct roles of astroglia and neurons in synaptic plasticity and memory. Mol Psychiatry 2022; 27:873-885. [PMID: 34642458 DOI: 10.1038/s41380-021-01332-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/17/2021] [Accepted: 09/29/2021] [Indexed: 12/17/2022]
Abstract
Long-term potentiation (LTP) in the hippocampus is the most studied form of synaptic plasticity. Temporal integration of synaptic inputs is essential in synaptic plasticity and is assumed to be achieved through Ca2+ signaling in neurons and astroglia. However, whether these two cell types play different roles in LTP remain unknown. Here, we found that through the integration of synaptic inputs, astrocyte inositol triphosphate (IP3) receptor type 2 (IP3R2)-dependent Ca2+ signaling was critical for late-phase LTP (L-LTP) but not early-phase LTP (E-LTP). Moreover, this process was mediated by astrocyte-derived brain-derived neurotrophic factor (BDNF). In contrast, neuron-derived BDNF was critical for both E-LTP and L-LTP. Importantly, the dynamic differences in BDNF secretion play a role in modulating distinct forms of LTP. Moreover, astrocyte- and neuron-derived BDNF exhibited different roles in memory. These observations enriched our knowledge of LTP and memory at the cellular level and implied distinct roles of astrocytes and neurons in information integration.
Collapse
|
18
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
19
|
Vignoli B, Sansevero G, Sasi M, Rimondini R, Blum R, Bonaldo V, Biasini E, Santi S, Berardi N, Lu B, Canossa M. Astrocytic microdomains from mouse cortex gain molecular control over long-term information storage and memory retention. Commun Biol 2021; 4:1152. [PMID: 34611268 PMCID: PMC8492720 DOI: 10.1038/s42003-021-02678-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 09/15/2021] [Indexed: 12/23/2022] Open
Abstract
Memory consolidation requires astrocytic microdomains for protein recycling; but whether this lays a mechanistic foundation for long-term information storage remains enigmatic. Here we demonstrate that persistent synaptic strengthening invited astrocytic microdomains to convert initially internalized (pro)-brain-derived neurotrophic factor (proBDNF) into active prodomain (BDNFpro) and mature BDNF (mBDNF) for synaptic re-use. While mBDNF activates TrkB, we uncovered a previously unsuspected function for the cleaved BDNFpro, which increases TrkB/SorCS2 receptor complex at post-synaptic sites. Astrocytic BDNFpro release reinforced TrkB phosphorylation to sustain long-term synaptic potentiation and to retain memory in the novel object recognition behavioral test. Thus, the switch from one inactive state to a multi-functional one of the proBDNF provides post-synaptic changes that survive the initial activation. This molecular asset confines local information storage in astrocytic microdomains to selectively support memory circuits. Beatrice Vignoli et al. examine potential molecular mechanisms of long-term storage information in mice. Their results suggest that astrocytes may help convert neuronal BDNF precursor into active prodomain and mature forms to enhance post-synaptic signaling and memory, providing further insight into the development of memory circuits.
Collapse
Affiliation(s)
- Beatrice Vignoli
- Department of Physics, University of Trento, 38123, Povo (TN), Italy. .,Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy.
| | - Gabriele Sansevero
- Neuroscience Institute, National Research Council (IN-CNR), 56100, Pisa, Italy
| | - Manju Sasi
- Institute of Clinical Neurobiology and Department of Neurology, University Hospital Würzburg, 97078, Würzburg, Germany
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126, Bologna, Italy
| | - Robert Blum
- Institute of Clinical Neurobiology and Department of Neurology, University Hospital Würzburg, 97078, Würzburg, Germany
| | - Valerio Bonaldo
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy
| | - Emiliano Biasini
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy
| | - Spartaco Santi
- Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", National Research Council of Italy, 40136, Bologna, Italy.,IRCCS, Istituto Ortopedico Rizzoli, 40136, Bologna, Italy
| | - Nicoletta Berardi
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, 50100, Florence, Italy
| | - Bai Lu
- School of Pharmaceutical Sciences, Tsinghua University, 100084, Beijing, China
| | - Marco Canossa
- Department of Cellular Computational and Integrative Biology (CIBIO), University of Trento, 38123, Povo (TN), Italy.
| |
Collapse
|
20
|
Lian WW, Zhou W, Zhang BY, Jia H, Xu LJ, Liu AL, Du GH. DL0410 ameliorates cognitive disorder in SAMP8 mice by promoting mitochondrial dynamics and the NMDAR-CREB-BDNF pathway. Acta Pharmacol Sin 2021; 42:1055-1068. [PMID: 32868905 DOI: 10.1038/s41401-020-00506-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a worldwide problem and there are no effective drugs for AD treatment. Previous studies show that DL0410 is a multi-target, anti-AD agent. In this study, we investigated the therapeutic effect of DL0410 and its action mechanism in SAMP8 mice. DL0410 (1-10 mg·kg-1·d-1) was orally administered to 8-month-old SAMP mice (SAMP8) for 8 weeks. We showed that DL0410 administration effectively ameliorated the cognitive deficits in the Morris water maze test, novel object recognition test, and nest building test. We revealed that DL0410 dose-dependently increased the expression levels of the mitochondrial proteins (PGC-1α, Mitofusin 2, OPA1, and Drp1), and subsequently ameliorated the processes of mitochondrial biosynthesis, fusion, and fission in the cortex and hippocampus of SAMP8 mice. Furthermore, DL0410 administration promoted the expression of synaptic proteins (synaptophysin and PSD95) in the brain of SAMP8 mice, and upregulated the protein phosphorylation in NMDAR-CAMKII/CAMKIV-CREB pathway responsible for the synaptic plasticity. DL0410 administration dose-dependently increased the expression of BDNF and TrkB, and the neurotrophic effect was mediated via the ERK1/2 and PI3K-AKT-GSK-3β pathways. DL0410 administration upregulated Bcl-2, increased the Bcl-2/Bax ratio and the level of caspase 3 and PARP-1, alleviating neuronal apoptosis. We proposed that the NMDAR-CREB-BDNF pathway might establish a positive feedback loop between synaptic plasticity and neurotrophy, with CREB at the center. In summary, DL0410 promotes synaptic function and neuronal survival, thus ameliorating cognitive deficits in SAMP8 mice via improved mitochondrial dynamics and increased activity of the NMDAR-CREB-BDNF pathway. DL0410 is a promising candidate to treat aging-related AD, and deserves more research and development in future.
Collapse
|
21
|
Lopes da Cunha P, Tintorelli R, Correa J, Budriesi P, Viola H. Behavioral tagging as a mechanism for aversive-memory formation under acute stress. Eur J Neurosci 2021; 55:2651-2665. [PMID: 33914357 DOI: 10.1111/ejn.15249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/01/2021] [Accepted: 04/15/2021] [Indexed: 02/03/2023]
Abstract
The behavioral tagging (BT) hypothesis postulates that a weak learning experience, which only induces short-term memory, may benefit from another event that provides plasticity-related proteins (PRPs) to establish a long-lasting memory. According to BT, the weak experience sets a transient learning tag at specific activated sites, and its temporal and spatial convergence with the PRPs allows the long-term memory (LTM) formation. In this work, rats were subjected to a weak inhibitory avoidance (IAw) training and we observed that acute stress (elevated platform, EP) experienced 1 hr before IAw promoted IA-LTM formation. This effect was dependent on glucocorticoid-receptor activity as well as protein synthesis in the dorsal hippocampus. However, the same stress has negative effects on IA-LTM formation when training is strong, probably by competing for necessary PRPs. Furthermore, our experiments showed that EP immediately after training did not impair the setting of the learning tag and even facilitated IA-LTM formation. These findings reveal different impacts of a given acute stressful experience on the formation of an aversive memory that could be explained by BT processes.
Collapse
Affiliation(s)
- Pamela Lopes da Cunha
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ramiro Tintorelli
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Correa
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Budriesi
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Haydee Viola
- Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular "Dr. Hector Maldonado" (FBMC), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Pareja-Cajiao M, Gransee HM, Sieck GC, Mantilla CB. TrkB signaling contributes to transdiaphragmatic pressure generation in aged mice. J Neurophysiol 2021; 125:1157-1163. [PMID: 33596726 PMCID: PMC8282218 DOI: 10.1152/jn.00004.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/19/2022] Open
Abstract
Ventilatory deficits are common in old age and may result from neuromuscular dysfunction. Signaling via the tropomyosin-related kinase receptor B (TrkB) regulates neuromuscular transmission and, in young mice, is important for the generation of transdiaphragmatic pressure (Pdi). Loss of TrkB signaling worsened neuromuscular transmission failure and reduced maximal Pdi, and these effects are similar to those observed in old age. Administration of TrkB agonists such as 7,8-dihydroxyflavone (7,8-DHF) improves neuromuscular transmission in young and old mice (18 mo; 75% survival). We hypothesized that TrkB signaling contributes to Pdi generation in old mice, particularly during maximal force behaviors. Old male and female TrkBF616A mice, with a mutation that induces 1NMPP1-mediated TrkB kinase inhibition, were randomly assigned to systemic treatment with vehicle, 7,8-DHF, or 1NMPP1 1 h before experiments. Pdi was measured during eupneic breathing (room air), hypoxia-hypercapnia (10% O2/5% CO2), tracheal occlusion, spontaneous deep breaths ("sighs"), and bilateral phrenic nerve stimulation (Pdimax). There were no differences in the Pdi amplitude across treatments during ventilatory behaviors (eupnea, hypoxia-hypercapnia, occlusion, or sigh). As expected, Pdi increased from eupnea and hypoxia-hypercapnia (∼7 cm H2O) to occlusion and sighs (∼25 cm H2O), with no differences across treatments. Pdimax was ∼50 cm H2O in the vehicle and 7,8-DHF groups and ∼40 cm H2O in the 1NMPP1 group (F8,74 = 2; P = 0.02). Our results indicate that TrkB signaling is necessary for generating maximal forces by the diaphragm muscle in old mice and are consistent with aging effects of TrkB signaling on neuromuscular transmission.NEW & NOTEWORTHY TrkB signaling is necessary for generating maximal forces by the diaphragm muscle. In 19- to 21-mo-old TrkBF616A mice susceptible to 1NMPP1-induced inhibition of TrkB kinase activity, maximal Pdi generated by bilateral phrenic nerve stimulation was ∼20% lower after 1NMPP1 compared with vehicle-treated mice. Treatment with the TrkB agonist 7,8-dihydroxyflavone did not affect Pdi generation when compared with age-matched mice. Inhibition of TrkB kinase activity did not affect the forces generated during lower force behaviors in old age.
Collapse
Affiliation(s)
- Miguel Pareja-Cajiao
- Departments of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Heather M Gransee
- Departments of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Gary C Sieck
- Departments of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
- Physiology and Biomedical Engineering, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Carlos B Mantilla
- Departments of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
- Physiology and Biomedical Engineering, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
23
|
Suzuki Y, Yoda Y, Ishikawa Y. Neuropsin-dependent and -independent behavioral tagging. Neuropsychopharmacol Rep 2021; 41:215-222. [PMID: 33773089 PMCID: PMC8340819 DOI: 10.1002/npr2.12177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 11/11/2022] Open
Abstract
Aim The consolidation of short‐term memories into long‐term memories is promoted by associations with novel environmental stimuli. This phenomenon is known as behavioral tagging. Neuropsin, a plasticity‐related serine protease in the hippocampus and amygdala, is involved in memory formation. This study investigated how neuropsin affects associative long‐term memory. Methods Short‐term and long‐term memory were assessed in control and neuropsin‐deficient mice by investigating their performance in inhibitory avoidance and spatial object recognition tasks. The effect of exposure to novelty on the conversion of short‐term memory to associative long‐term memory was also examined. Results The consolidation of task‐related short‐term memories into long‐term memories was facilitated by exposing the animals to a novel environment 1 hour before training. However, this long‐term memory conversion was impaired in neuropsin‐deficient mice performing the inhibitory avoidance task but not the spatial object recognition task. Conclusion Behavioral tagging occurs via neuropsin‐dependent and neuropsin‐independent processes for different behavioral tasks. The consolidation of task‐related short‐term memories into long‐term memories was facilitated by exposing the animals to a novel environment 1 hour before training. However, this long‐term memory conversion was impaired in neuropsin‐deficient mice performing the inhibitory avoidance task but not the spatial object recognition task. Behavioral tagging occurs via neuropsin‐dependent and neuropsin‐independent processes for different behavioral tasks.![]()
Collapse
Affiliation(s)
- Yuka Suzuki
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma, Japan
| | - Yuya Yoda
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma, Japan
| | - Yasuyuki Ishikawa
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma, Japan
| |
Collapse
|
24
|
HUZARD D, RAPPENEAU V, MEIJER OC, TOUMA C, ARANGO-LIEVANO M, GARABEDIAN MJ, JEANNETEAU F. Experience and activity-dependent control of glucocorticoid receptors during the stress response in large-scale brain networks. Stress 2021; 24:130-153. [PMID: 32755268 PMCID: PMC7907260 DOI: 10.1080/10253890.2020.1806226] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The diversity of actions of the glucocorticoid stress hormones among individuals and within organs, tissues and cells is shaped by age, gender, genetics, metabolism, and the quantity of exposure. However, such factors cannot explain the heterogeneity of responses in the brain within cells of the same lineage, or similar tissue environment, or in the same individual. Here, we argue that the stress response is continuously updated by synchronized neural activity on large-scale brain networks. This occurs at the molecular, cellular and behavioral levels by crosstalk communication between activity-dependent and glucocorticoid signaling pathways, which updates the diversity of responses based on prior experience. Such a Bayesian process determines adaptation to the demands of the body and external world. We propose a framework for understanding how the diversity of glucocorticoid actions throughout brain networks is essential for supporting optimal health, while its disruption may contribute to the pathophysiology of stress-related disorders, such as major depression, and resistance to therapeutic treatments.
Collapse
Affiliation(s)
- Damien HUZARD
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | - Virginie RAPPENEAU
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Onno C. MEIJER
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Chadi TOUMA
- Department of Behavioural Biology, University of Osnabrück, Osnabrück, Germany
| | - Margarita ARANGO-LIEVANO
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Freddy JEANNETEAU
- Department of Neuroscience and Physiology, University of Montpellier, CNRS, INSERM, Institut de Génomique Fonctionnelle, Montpellier, France
- Corresponding author:
| |
Collapse
|
25
|
Smolen P, Baxter DA, Byrne JH. Comparing Theories for the Maintenance of Late LTP and Long-Term Memory: Computational Analysis of the Roles of Kinase Feedback Pathways and Synaptic Reactivation. Front Comput Neurosci 2020; 14:569349. [PMID: 33390922 PMCID: PMC7772319 DOI: 10.3389/fncom.2020.569349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/16/2020] [Indexed: 11/26/2022] Open
Abstract
A fundamental neuroscience question is how memories are maintained from days to a lifetime, given turnover of proteins that underlie expression of long-term synaptic potentiation (LTP) or “tag” synapses as eligible for LTP. A likely solution relies on synaptic positive feedback loops, prominently including persistent activation of Ca2+/calmodulin kinase II (CaMKII) and self-activated synthesis of protein kinase M ζ (PKMζ). Data also suggest positive feedback based on recurrent synaptic reactivation within neuron assemblies, or engrams, is necessary to maintain memories. The relative importance of these mechanisms is controversial. To explore the likelihood that each mechanism is necessary or sufficient to maintain memory, we simulated maintenance of LTP with a simplified model incorporating persistent kinase activation, synaptic tagging, and preferential reactivation of strong synapses, and analyzed implications of recent data. We simulated three model variants, each maintaining LTP with one feedback loop: autonomous, self-activated PKMζ synthesis (model variant I); self-activated CamKII (model variant II); and recurrent reactivation of strengthened synapses (model variant III). Variant I predicts that, for successful maintenance of LTP, either 1) PKMζ contributes to synaptic tagging, or 2) a low constitutive tag level persists during maintenance independent of PKMζ, or 3) maintenance of LTP is independent of tagging. Variant II maintains LTP and suggests persistent CaMKII activation could maintain PKMζ activity, a feedforward interaction not previously considered. However, we note data challenging the CaMKII feedback loop. In Variant III synaptic reactivation drives, and thus predicts, recurrent or persistent activation of CamKII and other necessary kinases, plausibly contributing to persistent elevation of PKMζ levels. Reactivation is thus predicted to sustain recurrent rounds of synaptic tagging and incorporation of plasticity-related proteins. We also suggest (model variant IV) that synaptic reactivation and autonomous kinase activation could synergistically maintain LTP. We propose experiments that could discriminate these maintenance mechanisms.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States.,Engineering and Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - John H Byrne
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
26
|
Smolen P, Wood MA, Baxter DA, Byrne JH. Modeling suggests combined-drug treatments for disorders impairing synaptic plasticity via shared signaling pathways. J Comput Neurosci 2020; 49:37-56. [PMID: 33175283 DOI: 10.1007/s10827-020-00771-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/27/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022]
Abstract
Genetic disorders such as Rubinstein-Taybi syndrome (RTS) and Coffin-Lowry syndrome (CLS) cause lifelong cognitive disability, including deficits in learning and memory. Can pharmacological therapies be suggested that improve learning and memory in these disorders? To address this question, we simulated drug effects within a computational model describing induction of late long-term potentiation (L-LTP). Biochemical pathways impaired in these and other disorders converge on a common target, histone acetylation by acetyltransferases such as CREB binding protein (CBP), which facilitates gene induction necessary for L-LTP. We focused on four drug classes: tropomyosin receptor kinase B (TrkB) agonists, cAMP phosphodiesterase inhibitors, histone deacetylase inhibitors, and ampakines. Simulations suggested each drug type alone may rescue deficits in L-LTP. A potential disadvantage, however, was the necessity of simulating strong drug effects (high doses), which could produce adverse side effects. Thus, we investigated the effects of six drug pairs among the four classes described above. These combination treatments normalized impaired L-LTP with substantially smaller individual drug 'doses'. In addition three of these combinations, a TrkB agonist paired with an ampakine and a cAMP phosphodiesterase inhibitor paired with a TrkB agonist or an ampakine, exhibited strong synergism in L-LTP rescue. Therefore, we suggest these drug combinations are promising candidates for further empirical studies in animal models of genetic disorders that impair histone acetylation, L-LTP, and learning.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, 92697, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
27
|
Up-regulation of the p75 neurotrophin receptor is an essential mechanism for HIV-gp120 mediated synaptic loss in the striatum. Brain Behav Immun 2020; 89:371-379. [PMID: 32717404 PMCID: PMC7572812 DOI: 10.1016/j.bbi.2020.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/10/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022] Open
Abstract
Reduced synaptodendritic complexity appears to be a key feature in human immunodeficiency virus (HIV)-associated neurological disorder (HAND). Viral proteins, and in particular the envelope protein gp120, play a role in the pathology of synapses. Gp120 has been shown to increase both in vitro and in vivo the proneurotrophin brain-derived neurotrophic factor, which promotes synaptic simplification through the activation of the p75 neurotrophin receptor (p75NTR). To provide evidence that p75NTR plays a role in gp120-mediated loss of synapses in vivo, we intercrossed gp120tg mice with p75NTR null mice and used molecular, histological and behavioral analyses to establish a link between p75NTR and gp120-mediated synaptic simplification. Synaptosomes obtained from the striatum of gp120tg mice exhibited a significant increase in p75NTR levels concomitantly to a decrease in synaptic markers such as TrkB and PSD95. Analysis of striatal dendritic spines by Golgi staining revealed that gp120tg mice display a reduced proportion of mushroom-type spines in addition to fewer spines overall, when compared to wild type or gp120tg lacking one or two p75NTR alleles. Moreover, removal of one p75NTR allele in gp120 transgenic mice abolished the gp120-driven impairment on a task of striatal-dependent motor learning. These data indicate that p75NTR could be a key player in HIV-mediated synaptic simplification in the striatum.
Collapse
|
28
|
Abstract
Memory reconsolidation occurs when a retrieving event destabilizes transiently a consolidated memory, triggering thereby a new process of restabilization that ensures memory persistence. Although this phenomenon has received wide attention, the effect of new information cooccurring with the reconsolidation process has been less explored. Here we demonstrate that a memory-retrieving event sets a neural tag, which enables the reconsolidation of memory after binding proteins provided by the original or a different contiguous experience. We characterized the specific temporal window during which this association is effective and identified the protein kinase A (PKA) and the extracellular signal-regulated kinase 1 and 2 (ERK 1/2) pathways as the mechanisms related to the setting of the reconsolidation tag and the synthesis of proteins. Our results show, therefore, that memory reconsolidation is mediated by a "behavioral tagging" process, which is common to different memory forms. They represent a significant advance in understanding the fate of memories reconsolidated while being adjacent to other events, and provide a tool for designing noninvasive strategies to attenuate (pathological/traumatic) or improve (education-related) memories.
Collapse
|
29
|
Okuda K, Højgaard K, Privitera L, Bayraktar G, Takeuchi T. Initial memory consolidation and the synaptic tagging and capture hypothesis. Eur J Neurosci 2020; 54:6826-6849. [PMID: 32649022 DOI: 10.1111/ejn.14902] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 01/05/2023]
Abstract
Everyday memories are retained automatically in the hippocampus and then decay very rapidly. Memory retention can be boosted when novel experiences occur shortly before or shortly after the time of memory encoding via a memory stabilization process called "initial memory consolidation." The dopamine release and new protein synthesis in the hippocampus during a novel experience are crucial for this novelty-induced memory boost. The mechanisms underlying initial memory consolidation are not well-understood, but the synaptic tagging and capture (STC) hypothesis provides a conceptual basis of synaptic plasticity events occurring during initial memory consolidation. In this review, we provide an overview of the STC hypothesis and its relevance to dopaminergic signalling, in order to explore the cellular and molecular mechanisms underlying initial memory consolidation in the hippocampus. We summarize electrophysiological STC processes based on the evidence from two-pathway experiments and a behavioural tagging hypothesis, which translates the STC hypothesis into a related behavioural hypothesis. We also discuss the function of two types of molecules, "synaptic tags" and "plasticity-related proteins," which have a crucial role in the STC process and initial memory consolidation. We describe candidate molecules for the roles of synaptic tag and plasticity-related proteins and interpret their candidacy based on evidence from two-pathway experiments ex vivo, behavioural tagging experiments in vivo and recent cutting-edge optical imaging experiments. Lastly, we discuss the direction of future studies to advance our understanding of molecular mechanisms underlying the STC process, which are critical for initial memory consolidation in the hippocampus.
Collapse
Affiliation(s)
- Kosuke Okuda
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark
| | - Kristoffer Højgaard
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus C, Denmark
| | - Lucia Privitera
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,School of Medicine, Ninewells Hospital, University of Dundee, Dundee, UK
| | - Gülberk Bayraktar
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark.,Institut für Klinische Neurobiologie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Tomonori Takeuchi
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
30
|
Pareja-Cajiao M, Gransee HM, Cole NA, Sieck GC, Mantilla CB. Inhibition of TrkB kinase activity impairs transdiaphragmatic pressure generation. J Appl Physiol (1985) 2020; 128:338-344. [PMID: 31944892 PMCID: PMC7052584 DOI: 10.1152/japplphysiol.00564.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/14/2019] [Accepted: 01/15/2020] [Indexed: 12/18/2022] Open
Abstract
Signaling via the tropomyosin-related kinase receptor subtype B (TrkB) regulates neuromuscular transmission, and inhibition of TrkB kinase activity by 1NMPP1 in TrkBF616A mice worsens neuromuscular transmission failure (NMTF). We hypothesized that acute inhibition of TrkB kinase activity will impair the ability of the diaphragm muscle to produce maximal transdiaphragmatic pressure (Pdi) without impacting the ability to generate forces associated with ventilation, consistent with the greater susceptibility to NMTF in motor units responsible for higher-force nonventilatory behaviors. Adult male and female TrkBF616A mice were injected with 1NMPP1 (n = 8) or vehicle (DMSO; n = 8) 1 h before Pdi measurements during eupneic breathing, hypoxia/hypercapnia (10% O2/5% CO2), tracheal occlusion, spontaneous deep breaths ("sighs") and during maximal activation elicited by bilateral phrenic nerve stimulation. In the vehicle-treated group, Pdi increased from ~10 cmH2O during eupnea and hypoxia/hypercapnia, to ~35 cmH2O during sighs and tracheal occlusion, and to ~65 cm H2O during maximal stimulation. There was no effect of acute 1NMPP1 treatment on Pdi generated during most behaviors, except during maximal stimulation (~30% reduction; P < 0.05). This reduction in maximal Pdi is generally similar to the worsening of NMTF previously reported with TrkB kinase inhibition in rodents. Accordingly, impaired TrkB signaling limits the range of motor behaviors accomplished by the diaphragm muscle and may contribute to neuromuscular dysfunction, primarily by impacting fatigable, higher force-generating motor units.NEW & NOTEWORTHY TrkB signaling plays an important role in maintaining neuromuscular function in the diaphragm muscle and may be necessary to accomplish the various motor behaviors ranging from ventilation to expulsive, behaviors requiring near-maximal forces. This study shows that inhibition of TrkB kinase activity impairs maximal pressure generation by the diaphragm muscle, but the ability to generate the lower pressures required for ventilatory behaviors is not impacted.
Collapse
Affiliation(s)
- Miguel Pareja-Cajiao
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Heather M Gransee
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Naomi A Cole
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Gary C Sieck
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| | - Carlos B Mantilla
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
31
|
Lin PY, Kavalali ET, Monteggia LM. Genetic Dissection of Presynaptic and Postsynaptic BDNF-TrkB Signaling in Synaptic Efficacy of CA3-CA1 Synapses. Cell Rep 2020; 24:1550-1561. [PMID: 30089265 PMCID: PMC7176480 DOI: 10.1016/j.celrep.2018.07.020] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/31/2018] [Accepted: 07/05/2018] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its high-affinity receptor, tropomyosin receptor kinase B (TrkB), regulate long-term potentiation (LTP) in the hippocampus, although the sites of BDNF-TrkB receptors in this process are controversial. We used a viral-mediated approach to delete BDNF or TrkB specifically in CA1 and CA3 regions of the Schaffer collateral pathway. Deletion of BDNF in CA3 or CA1 revealed that presynaptic BDNF is involved in LTP induction, while postsynaptic BDNF contributes to LTP maintenance. Similarly, loss of presynaptic or postsynaptic TrkB receptors leads to distinct LTP deficits, with presynaptic TrkB required to maintain LTP, while postsynaptic TrkB is essential for LTP formation. In addition, loss of TrkB in CA3 significantly diminishes release probability, uncovering a role for presynaptic TrkB receptors in basal neurotransmission. Taken together, this direct comparison of presynaptic and postsynaptic BDNF-TrkB reveals insight into BDNF release and TrkB activation sites in hippocampal LTP. Lin et al. directly compare a role for presynaptic and postsynaptic BDNF and TrkB receptors in hippocampal LTP. They find that LTP induction is mediated by anterograde BDNF-TrkB signaling, while both anterograde and retrograde BDNFTrkB signaling persists presynaptically and postsynaptically for LTP maintenance.
Collapse
Affiliation(s)
- Pei-Yi Lin
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ege T Kavalali
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Lisa M Monteggia
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
32
|
Hong J, Heo WD. Optogenetic Modulation of TrkB Signaling in the Mouse Brain. J Mol Biol 2020; 432:815-827. [PMID: 31962123 DOI: 10.1016/j.jmb.2020.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/22/2019] [Accepted: 01/07/2020] [Indexed: 12/17/2022]
Abstract
Optogenetic activation of receptors has advantages compared with chemical or ligand treatment because of its high spatial and temporal precision. Especially in the brain, the use of a genetically encoded light-tunable receptor is superior to direct infusion or systemic drug treatment. We applied light-activatable TrkB receptors in the mouse brain with reduced basal activity by incorporating Cry2PHR mutant, Opto-cytTrkB(E281A). Upon AAV mediated gene delivery, this form was expressed at sufficient levels in the mouse hippocampus (HPC) and medial entorhinal cortex (MEC) retaining normal canonical signal transduction by the blue light stimulus, even by delivery of noninvasive LED light on the mouse head. Within target cells, where its expression was driven by a cell type-specific promoter, Opto-cytTrkB(E281A)-mediated TrkB signaling could be controlled by adjusting light-stimulating conditions. We further demonstrated that Opto-cytTrkB(E281A) could locally induce TrkB signaling in axon terminals in the MEC-HPC. In summary, Opto-cytTrkB(E281A) will be useful for elucidating time- and region-specific roles of TrkB signaling ranging from cellular function to neural circuit mechanisms.
Collapse
Affiliation(s)
- Jongryul Hong
- Department of Biological Science, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - Won Do Heo
- Department of Biological Science, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea; Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, 305-701, Republic of Korea; KAIST Institute for the BioCentury, KAIST, Daejeon, 305-701, Republic of Korea.
| |
Collapse
|
33
|
Wong LW, Tann JY, Ibanez CF, Sajikumar S. The p75 Neurotrophin Receptor Is an Essential Mediator of Impairments in Hippocampal-Dependent Associative Plasticity and Memory Induced by Sleep Deprivation. J Neurosci 2019; 39:5452-5465. [PMID: 31085607 PMCID: PMC6616296 DOI: 10.1523/jneurosci.2876-18.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 01/22/2023] Open
Abstract
Sleep deprivation (SD) interferes with hippocampal structural and functional plasticity, formation of long-term memory and cognitive function. The molecular mechanisms underlying these effects are incompletely understood. Here, we show that SD impaired synaptic tagging and capture and behavioral tagging, two major mechanisms of associative learning and memory. Strikingly, mutant male mice lacking the p75 neurotrophin receptor (p75NTR) were resistant to the detrimental effects of SD on hippocampal plasticity at both cellular and behavioral levels. Mechanistically, SD increased p75NTR expression and its interaction with phosphodiesterase. p75NTR deletion preserved hippocampal structural and functional plasticity by preventing SD-mediated effects on hippocampal cAMP-CREB-BDNF, cAMP-PKA-LIMK1-cofilin, and RhoA-ROCK2 pathways. Our study identifies p75NTR as an important mediator of hippocampal structural and functional changes associated with SD, and suggests that targeting p75NTR could be a promising strategy to limit the memory and cognitive deficits that accompany sleep loss.SIGNIFICANCE STATEMENT The lack of sufficient sleep is a major health concern in today's world. Sleep deprivation (SD) affects cognitive functions such as memory. We have investigated how associative memory mechanisms, synaptic tagging and capture (STC), was impaired in SD mice at cellular and behavioral level. Interestingly, mutant male mice that lacked the p75 neurotrophin receptor (p75NTR) were seen to be resistant to the SD-induced impairments in hippocampal synaptic plasticity and STC. Additionally, we elucidated the molecular pathways responsible for this rescue of plasticity in the mutant mice. Our study has thus identified p75NTR as a promising target to limit the cognitive deficits associated with SD.
Collapse
Affiliation(s)
- Lik-Wei Wong
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Jason Y Tann
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| | - Carlos F Ibanez
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm S-17177, Sweden
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore 117597, Singapore,
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore 117456, and
| |
Collapse
|
34
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
35
|
Baltaci SB, Mogulkoc R, Baltaci AK. Molecular Mechanisms of Early and Late LTP. Neurochem Res 2019; 44:281-296. [PMID: 30523578 DOI: 10.1007/s11064-018-2695-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/31/2018] [Accepted: 12/04/2018] [Indexed: 12/01/2022]
Abstract
LTP is the most intensively studied cellular model of the memory and generally divided at least two distinct phases as early and late. E-LTP requires activation of CaMKII that initiates biochemical events and trafficking of proteins, which eventually potentiate synaptic transmission, and is independent of de novo protein synthesis. In contrast, L-LTP requires gene expression and local protein synthesis regulated via TrkB receptor- and functional prions CPEB2-3-mediated translation. Maintenance of LTP for longer periods depends on constitutively active PKMζ. Throughout this review, current knowledge about early and late phases of LTP will be reviewed.
Collapse
Affiliation(s)
- Saltuk Bugra Baltaci
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | - Rasim Mogulkoc
- Faculty of Medicine, Department of Physiology, Selcuk University, 42031, Konya, Turkey
| | | |
Collapse
|
36
|
Lopes da Cunha P, Villar ME, Ballarini F, Tintorelli R, Ana María Viola H. Spatial object recognition memory formation under acute stress. Hippocampus 2018; 29:491-499. [PMID: 30295349 DOI: 10.1002/hipo.23037] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 09/12/2018] [Accepted: 09/17/2018] [Indexed: 01/30/2023]
Abstract
Stress is known to have a critical impact on memory processes. In the present work, we focus on the effects of an acute stress event closely associated to an unrelated learning task. Here, we show that acute stress (elevated platform [EP] session) experienced 1 hr after a weak spatial object recognition (SOR) training, which only induces a short-term memory (STM), promoted the formation of SOR-long term memory (SOR-LTM) in rats. The effect induced by stress was dependent on the activation of glucocorticoid- and mineralocorticoid-receptors, brain-derived neurotrophic factor (BDNF) and protein synthesis in the dorsal hippocampus. In contrast, EP after a strong SOR impaired SOR-LTM probably by interfering with the use of necessary resources. Moreover, we show that the EP session before training induced anterograde interference, which it was not reversed by a subsequent exposure to an open field. Our findings provide novel insights into the impact of stress on LTM formation in rodents and they are discussed under the behavioral analogue of the synaptic tagging and capture hypothesis.
Collapse
Affiliation(s)
- Pamela Lopes da Cunha
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Eugenia Villar
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fabricio Ballarini
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ramiro Tintorelli
- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Haydée Ana María Viola
- Instituto de Biología Celular y Neurociencias "Dr Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular "Dr. Hector Maldonado" (FBMC), Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| |
Collapse
|
37
|
Reyes-Garcia SZ, de Almeida ACG, Ortiz-Villatoro NN, Scorza FA, Cavalheiro EA, Scorza CA. Robust Network Inhibition and Decay of Early-Phase LTP in the Hippocampal CA1 Subfield of the Amazon Rodent Proechimys. Front Neural Circuits 2018; 12:81. [PMID: 30337859 PMCID: PMC6180286 DOI: 10.3389/fncir.2018.00081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 09/13/2018] [Indexed: 01/28/2023] Open
Abstract
Background: Diverse forms of long-term potentiation (LTP) have been described, but one of the most investigated is encountered in the glutamatergic synapses of the hippocampal cornu Ammonis (CA1) subfield. However, little is known about synaptic plasticity in wildlife populations. Laboratory animals are extremely inbred populations that have been disconnected from their natural environment and so their essential ecological aspects are entirely absent. Proechimys are small rodents from Brazil’s Amazon rainforest and their nervous systems have evolved to carry out specific tasks of their unique ecological environment. It has also been shown that long-term memory duration did not persist for 24-h in Proechimys, in contrast to Wistar rats, when both animal species were assessed by the plus-maze discrimination avoidance task and object recognition test. Methods: In this work, different protocols, such as theta burst, single tetanic burst or multiple trains of high frequency stimulation (HFS), were used to induce LTP in hippocampal brain slices of Proechimys and Wistar rats. Results: A protocol-independent fast decay of early-phase LTP at glutamatergic synapses of the CA1 subfield was encountered in Proechimys. Long-term depression (LTD) and baseline paired-pulse facilitation (PPF) were investigated but no differences were found between animal species. Input/output (I/O) relationships suggested lower excitability in Proechimys in comparison to Wistar rats. Bath application of d-(-)-2-amino-5-phosphonopentanoicacid (D-AP5) and CNQX prevented the induction of LTP in both Proechimys and Wistar. However, in marked contrast to Wistar rats, LTP induction was not facilitated by the GABAA antagonist in the Amazon rodents, even higher concentrations failed to facilitate LTP in Proechimys. Next, the effects of GABAA inhibition on spontaneous activity as well as evoked field potentials (FPs) were evaluated in CA1 pyramidal cells. Likewise, much lower activity was detected in Proechimys brain slices in comparison to those of the Wistar rats. Conclusions: These findings suggest a possible high inhibitory tone in the CA1 network mediated by GABAA receptors in the Amazon rodents. Currently, neuroscience research still seeks to reveal molecular pathways that control learning and memory processes, Proechimys may prove useful in identifying such mechanisms in complement to traditional animal models.
Collapse
Affiliation(s)
- Selvin Z Reyes-Garcia
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Ciencias Morfológicas, Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Antônio-Carlos Guimarães de Almeida
- Laboratório de Neurociência Experimental e Computacional, Departamento de Engenharia de Biossistemas, Universidade Federal de São João del-Rei, São João del-Rei, Brazil
| | - Nancy N Ortiz-Villatoro
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fulvio A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Esper A Cavalheiro
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Yun D, Zhuang Y, Kreutz MR, Behnisch T. The role of 19S proteasome associated deubiquitinases in activity-dependent hippocampal synaptic plasticity. Neuropharmacology 2018; 133:354-365. [DOI: 10.1016/j.neuropharm.2018.01.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/24/2017] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
|
39
|
Sharma M, Razali NB, Sajikumar S. Inhibition of G9a/GLP Complex Promotes Long-Term Potentiation and Synaptic Tagging/Capture in Hippocampal CA1 Pyramidal Neurons. Cereb Cortex 2018; 27:3161-3171. [PMID: 27252354 DOI: 10.1093/cercor/bhw170] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epigenetic regulations play an important role in regulating the learning and memory processes. G9a/G9a-like protein (GLP) lysine dimethyltransferase complex controls a prominent histone H3 lysine9 dimethylation (H3K9me2) that results in transcriptional silencing of the chromatin. Here, we report that the inhibition of G9a/GLP complex by either of the substrate competitive inhibitors UNC 0638 or BIX 01294 reinforces protein synthesis-independent long-term potentiation (early-LTP) to protein synthesis-dependent long-term potentiation (late-LTP). The reinforcement effect was observed if the inhibitors were present during the induction of early-LTP and in addition when G9a/GLP complex inhibition was carried out by priming of synapses within an interval of 30 min before or after the induction of early-LTP. Surprisingly, the reinforced LTP by G9a/GLP complex inhibition was able to associate with a weak plasticity event from nearby independent synaptic populations, resulting in synaptic tagging/capture (STC). We have identified brain-derived neurotrophic factor (BDNF) as a critical plasticity protein that maintains G9a/GLP complex inhibition-mediated LTP facilitation and its STC. Our study reveals an epigenetic mechanism for promoting plasticity and associativity by G9a/GLP complex inhibition, and it may engender a promising epigenetic target for enhancing memory in neural networks.
Collapse
Affiliation(s)
- Mahima Sharma
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| | - Nuralyah Bte Razali
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117 597, Singapore.,Neurobiology/Aging Program, #04-44, 28 Medical Drive, Life Sciences Institute (LSI), National University of Singapore, Singapore 117 456, Singapore
| |
Collapse
|
40
|
Abstract
Behavioral tagging is the transformation of a short-term memory induced by a weak experience into a long-term memory through temporal association with a novel experience. This phenomenon was discovered to recapitulate synaptic tagging and capture at the behavioral level. Significant progress has been made in determining the molecular machinery associated with synaptic tagging and capture and behavioral tagging theories. However, the tag setting and recruitment of plasticity-related proteins that occur within the spatiotemporally constrained cell ensemble at the network level (cellular tagging) in the brain where multimodal sensory information is input are just beginning to be understood. Here, we review the evidence for behavioral tagging and the mechanism underlying memory allocation at the network level leading to the overlap of cell ensembles. We also discuss the functional significance of overlapping cell ensembles in association of standard Pavlovian conditioning and distinct memories. Finally, we describe the role of neuronal ensemble overlap in behavioral tagging.
Collapse
Affiliation(s)
- Masanori Nomoto
- Department of Biochemistry, Faculty of Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kaoru Inokuchi
- Department of Biochemistry, Faculty of Medicine, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
41
|
Guimarães Marques MJ, Reyes-Garcia SZ, Marques-Carneiro JE, Lopes-Silva LB, Andersen ML, Cavalheiro EA, Scorza FA, Scorza CA. Long-term Potentiation Decay and Poor Long-lasting Memory Process in the Wild Rodents Proechimys from Brazil's Amazon Rainforest. Front Behav Neurosci 2018; 12:2. [PMID: 29410617 PMCID: PMC5787059 DOI: 10.3389/fnbeh.2018.00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/09/2018] [Indexed: 11/17/2022] Open
Abstract
Proechimys are small terrestrial rodents from Amazon rainforest. Each animal species is adapted to a specific environment in which the animal evolved therefore without comparative approaches unique characteristics of distinct species cannot be fully recognized. Laboratory rodents are exceedingly inbred strains dissociated from their native habitats and their fundamental ecological aspects are abstracted. Thus, the employment of exotic non-model species can be informative and complement conventional animal models. With the aim of promoting comparative studies between the exotic wildlife populations in the laboratory and traditional rodent model, we surveyed a type of synaptic plasticity intimately related to memory encoding in animals. Using theta-burst paradigm, in vitro long-term potentiation (LTP) in the CA1 subfield of hippocampal slices was assessed in the Amazon rodents Proechimys and Wistar rats. Memory, learning and anxiety were investigated through the plus-maze discriminative avoidance task (PM-DAT) and object recognition test. In PM-DAT, both animal species were submitted to two test sessions (3-h and 24-h) after the conditioning training. Proechimys exhibited higher anxiety-like behavior in the training session but during test sessions both species exhibited similar patterns of anxiety-related behavior. After 3-h of the training, Proechimys and Wistar spent significantly less time in the aversive enclosed arm than in the non-aversive arm. But, at 24-h after training, Wistar rats remained less time in the aversive closed arm in comparison with the non-aversive one, while Proechimys rodents spent the same amount of time in both enclosed arms. In the object recognition test, both species were evaluated at 24-h after the acquisition session and similar findings than those of the PM-DAT (24-h) were obtained, suggesting that long-term memory duration did not persist for 24-h in the Amazon rodent. Field excitatory post-synaptic potentials recordings revealed that LTP decays rapidly over time reaching basal levels at 90 min after theta-burst stimulation in Proechimys, contrasting to the stable LTP found in the Wistar rats which was observed throughout 3-h recording period. These findings suggest a link between the LTP decay and the lack of 24-h long-lasting memory process in Proechimys. Nevertheless, why early-phase LTP in Proechimys decays very rapidly remains to be elucidated.
Collapse
Affiliation(s)
- Marcia J Guimarães Marques
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Selvin Z Reyes-Garcia
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Ciencias Morfológicas, Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - José E Marques-Carneiro
- Université de Strasbourg-INSERM U-1114-Neuropsychologie Cognitive, Physiopathologie de la Schizophrénie, Strasbourg, France
| | - Leonardo B Lopes-Silva
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica L Andersen
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Esper A Cavalheiro
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fulvio A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carla A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Dasgupta A, Kim J, Manakkadan A, Arumugam TV, Sajikumar S. Intermittent fasting promotes prolonged associative interactions during synaptic tagging/capture by altering the metaplastic properties of the CA1 hippocampal neurons. Neurobiol Learn Mem 2017; 154:70-77. [PMID: 29277679 DOI: 10.1016/j.nlm.2017.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 01/18/2023]
Abstract
Metaplasticity is the inherent property of a neuron or neuronal population to undergo activity-dependent changes in neural function that modulate subsequent synaptic plasticity. Here we studied the effect of intermittent fasting (IF) in governing the interactions of associative plasticity mechanisms in the pyramidal neurons of rat hippocampal area CA1. Late long-term potentiation and its associative mechanisms such as synaptic tagging and capture at an interval of 120 min were evaluated in four groups of animals, AL (Ad libitum), IF12 (daily IF for 12 h), IF16 (daily IF for 16 h) and EOD (every other day IF for 24 h). IF had no visible effect on the early or late plasticity but it manifested a critical role in prolonging the associative interactions between weak and strong synapses at an interval of 120 min in IF16 and EOD animals. However, both IF12 and AL did not show associativity at 120 min. Plasticity genes such as Bdnf and Prkcz, which are well known for their expressions in late plasticity and synaptic tagging and capture, were significantly upregulated in IF16 and EOD in comparison to AL. Specific inhibition of brain derived neurotropic factor (BDNF) prevented the prolonged associativity expressed in EOD. Thus, daily IF for 16 h or more can be considered to enhance the metaplastic properties of synapses by improving their associative interactions that might translate into animprovedmemoryformation.
Collapse
Affiliation(s)
- Ananya Dasgupta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore
| | - Joonki Kim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Natural Products Research Center, Korea Institute of Science and Technology, Gangneung, Gangwon-do, Republic of Korea
| | - Anoop Manakkadan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology/Aging Program, Life Sciences Institute (LSI), National University of Singapore, #04-44, 28 Medical Drive, Singapore 117 456, Singapore.
| |
Collapse
|
43
|
Locus Coeruleus and Dopamine-Dependent Memory Consolidation. Neural Plast 2017; 2017:8602690. [PMID: 29123927 PMCID: PMC5662828 DOI: 10.1155/2017/8602690] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 06/06/2017] [Accepted: 06/18/2017] [Indexed: 12/12/2022] Open
Abstract
Most everyday memories including many episodic-like memories that we may form automatically in the hippocampus (HPC) are forgotten, while some of them are retained for a long time by a memory stabilization process, called initial memory consolidation. Specifically, the retention of everyday memory is enhanced, in humans and animals, when something novel happens shortly before or after the time of encoding. Converging evidence has indicated that dopamine (DA) signaling via D1/D5 receptors in HPC is required for persistence of synaptic plasticity and memory, thereby playing an important role in the novelty-associated memory enhancement. In this review paper, we aim to provide an overview of the key findings related to D1/D5 receptor-dependent persistence of synaptic plasticity and memory in HPC, especially focusing on the emerging evidence for a role of the locus coeruleus (LC) in DA-dependent memory consolidation. We then refer to candidate brain areas and circuits that might be responsible for detection and transmission of the environmental novelty signal and molecular and anatomical evidence for the LC-DA system. We also discuss molecular mechanisms that might mediate the environmental novelty-associated memory enhancement, including plasticity-related proteins that are involved in initial memory consolidation processes in HPC.
Collapse
|
44
|
Shivarama Shetty M, Sajikumar S. 'Tagging' along memories in aging: Synaptic tagging and capture mechanisms in the aged hippocampus. Ageing Res Rev 2017; 35:22-35. [PMID: 28065806 DOI: 10.1016/j.arr.2016.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/12/2016] [Accepted: 12/30/2016] [Indexed: 02/06/2023]
Abstract
Aging is accompanied by a general decline in the physiological functions of the body with the deteriorating organ systems. Brain is no exception to this and deficits in cognitive functions are quite common in advanced aging. Though a variety of age-related alterations are observed in the structure and function throughout the brain, certain regions show selective vulnerability. Medial temporal lobe, especially the hippocampus, is one such preferentially vulnerable region and is a crucial structure involved in the learning and long-term memory functions. Hippocampal synaptic plasticity, such as long-term potentiation (LTP) and depression (LTD), are candidate cellular correlates of learning and memory and alterations in these properties have been well documented in aging. A related phenomenon called synaptic tagging and capture (STC) has been proposed as a mechanism for cellular memory consolidation and to account for temporal association of memories. Mounting evidences from behavioral settings suggest that STC could be a physiological phenomenon. In this article, we review the recent data concerning STC and provide a framework for how alterations in STC-related mechanisms could contribute to the age-associated memory impairments. The enormity of impairment in learning and memory functions demands an understanding of age-associated memory deficits at the fundamental level given its impact in the everyday tasks, thereby in the quality of life. Such an understanding is also crucial for designing interventions and preventive measures for successful brain aging.
Collapse
|
45
|
Sasi M, Vignoli B, Canossa M, Blum R. Neurobiology of local and intercellular BDNF signaling. Pflugers Arch 2017; 469:593-610. [PMID: 28280960 PMCID: PMC5438432 DOI: 10.1007/s00424-017-1964-4] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 01/07/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of secreted proteins. Signaling cascades induced by BDNF and its receptor, the receptor tyrosine kinase TrkB, link neuronal growth and differentiation with synaptic plasticity. For this reason, interference with BDNF signaling has emerged as a promising strategy for potential treatments in psychiatric and neurological disorders. In many brain circuits, synaptically released BDNF is essential for structural and functional long-term potentiation, two prototypical cellular models of learning and memory formation. Recent studies have revealed an unexpected complexity in the synaptic communication of mature BDNF and its precursor proBDNF, not only between local pre- and postsynaptic neuronal targets but also with participation of glial cells. Here, we consider recent findings on local actions of the BDNF family of ligands at the synapse and discuss converging lines of evidence which emerge from per se conflicting results.
Collapse
Affiliation(s)
- Manju Sasi
- Institute of Clinical Neurobiology, University Hospital, University of Würzburg, 97078, Würzburg, Germany
| | - Beatrice Vignoli
- Centre for Integrative Biology (CIBIO), University of Trento, 38123, Povo, TN, Italy
| | - Marco Canossa
- Centre for Integrative Biology (CIBIO), University of Trento, 38123, Povo, TN, Italy.,European Brain Research Institute (EBRI) "Rita Levi-Montalcini", 00143, Rome, Italy
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital, University of Würzburg, 97078, Würzburg, Germany.
| |
Collapse
|
46
|
Shetty MS, Sharma M, Sajikumar S. Chelation of hippocampal zinc enhances long-term potentiation and synaptic tagging/capture in CA1 pyramidal neurons of aged rats: implications to aging and memory. Aging Cell 2017; 16:136-148. [PMID: 27633878 PMCID: PMC5242293 DOI: 10.1111/acel.12537] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2016] [Indexed: 01/08/2023] Open
Abstract
Aging is associated with decline in cognitive functions, prominently in the memory consolidation and association capabilities. Hippocampus plays a crucial role in the formation and maintenance of long‐term associative memories, and a significant body of evidence shows that impairments in hippocampal function correlate with aging‐related memory loss. A number of studies have implicated alterations in hippocampal synaptic plasticity, such as long‐term potentiation (LTP), in age‐related cognitive decline although exact mechanisms underlying are not completely clear. Zinc deficiency and the resultant adverse effects on cognition have been well studied. However, the role of excess of zinc in synaptic plasticity, especially in aging, is not addressed well. Here, we have investigated the hippocampal zinc levels and the impairments in synaptic plasticity, such as LTP and synaptic tagging and capture (STC), in the CA1 region of acute hippocampal slices from 82‐ to 84‐week‐old male Wistar rats. We report increased zinc levels in the hippocampus of aged rats and also deficits in the tetani‐induced and dopaminergic agonist‐induced late‐LTP and STC. The observed deficits in synaptic plasticity were restored upon chelation of zinc using a cell‐permeable chelator. These data suggest that functional plasticity and associativity can be successfully established in aged neural networks by chelating zinc with cell‐permeable chelating agents.
Collapse
Affiliation(s)
- Mahesh Shivarama Shetty
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| | - Mahima Sharma
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| | - Sreedharan Sajikumar
- Department of Physiology; Yong Loo Lin School of Medicine; National University of Singapore; Block MD9, 2 Medical Drive Singapore 117 597 Singapore
- Neurobiology/Aging Program; Life Sciences Institute (LSI); National University of Singapore; #04-44, 28 Medical Drive Singapore 117 456 Singapore
| |
Collapse
|
47
|
HAP1 Is Required for Endocytosis and Signalling of BDNF and Its Receptors in Neurons. Mol Neurobiol 2017; 55:1815-1830. [PMID: 28083816 DOI: 10.1007/s12035-016-0379-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022]
Abstract
When BDNF binds to its receptors, TrkB and p75NTR, the BDNF-receptor complex is endocytosed and trafficked to the cell body for downstream signal transduction, which plays a critical role in neuronal functions. Huntingtin-associated protein 1 (HAP1) is involved in trafficking of vesicles intracellularly and also interacts with several membrane proteins including TrkB. Although it has been known that HAP1 has functions in vesicular trafficking and receptor stabilisation, it is not yet established whether HAP1 has a role in BDNF and its receptor endocytosis. In the present study, we found that HAP1 is in an interacting complex with p75NTR, TrkB and BDNF, especially newly endocytosed BDNF. BDNF and TrkB internalisation is abolished in HAP1 knock-out (KO) cortical neurons. TrkB downstream signalling pathways such as ERK, Akt and PLCγ-1 are also impaired in HAP1 KO cortical neurons upon BDNF stimulation. Proliferation of cerebellar granule cells is also impaired in cell culture and cerebellum of HAP1 KO mice. Our findings suggest that HAP1 may play a key role in BDNF and its receptor endocytosis and may promote neuronal survival and proliferation.
Collapse
|
48
|
Mitre M, Mariga A, Chao MV. Neurotrophin signalling: novel insights into mechanisms and pathophysiology. Clin Sci (Lond) 2017; 131:13-23. [PMID: 27908981 PMCID: PMC5295469 DOI: 10.1042/cs20160044] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 07/18/2016] [Accepted: 08/09/2016] [Indexed: 12/29/2022]
Abstract
Neurotrophins, such as brain-derived neurotrophic factor (BDNF), are prominent regulators of neuronal survival, growth and differentiation during development. While trophic factors are viewed as well-understood but not innovative molecules, there are many lines of evidence indicating that BDNF plays an important role in the pathophysiology of many neurodegenerative disorders, depression, anxiety and other psychiatric disorders. In particular, lower levels of BDNF are associated with the aetiology of Alzheimer's and Huntington's diseases. A major challenge is to explain how neurotrophins are able to induce plasticity, improve learning and memory and prevent age-dependent cognitive decline through receptor signalling. This article will review the mechanism of action of neurotrophins and how BDNF/tropomyosin receptor kinase B (TrkB) receptor signaling can dictate trophic responses and change brain plasticity through activity-dependent stimulation. Alternative approaches for modulating BDNF/TrkB signalling to deliver relevant clinical outcomes in neurodegenerative and neuropsychiatric disorders will also be described.
Collapse
Affiliation(s)
- Mariela Mitre
- Neuroscience and Physiology and Psychiatry, New York University School of Medicine, New York, NY 10016, U.S.A.
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, U.S.A
| | - Abigail Mariga
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, U.S.A
- Departments of Cell Biology, New York University School of Medicine, New York, NY 10016, U.S.A
| | - Moses V Chao
- Neuroscience and Physiology and Psychiatry, New York University School of Medicine, New York, NY 10016, U.S.A
- Skirball Institute for Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, U.S.A
- Departments of Cell Biology, New York University School of Medicine, New York, NY 10016, U.S.A
| |
Collapse
|
49
|
Glerup S, Bolcho U, Mølgaard S, Bøggild S, Vaegter CB, Smith AH, Nieto-Gonzalez JL, Ovesen PL, Pedersen LF, Fjorback AN, Kjolby M, Login H, Holm MM, Andersen OM, Nyengaard JR, Willnow TE, Jensen K, Nykjaer A. SorCS2 is required for BDNF-dependent plasticity in the hippocampus. Mol Psychiatry 2016; 21:1740-1751. [PMID: 27457814 DOI: 10.1038/mp.2016.108] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 04/06/2016] [Accepted: 04/18/2016] [Indexed: 12/16/2022]
Abstract
SorCS2 is a member of the Vps10p-domain receptor gene family receptors with critical roles in the control of neuronal viability and function. Several genetic studies have suggested SORCS2 to confer risk of bipolar disorder, schizophrenia and attention deficit-hyperactivity disorder. Here we report that hippocampal N-methyl-d-aspartate receptor-dependent synaptic plasticity is eliminated in SorCS2-deficient mice. This defect was traced to the ability of SorCS2 to form complexes with the neurotrophin receptor p75NTR, required for pro-brain-derived neurotrophic factor (BDNF) to induce long-term depression, and with the BDNF receptor tyrosine kinase TrkB to elicit long-term potentiation. Although the interaction with p75NTR was static, SorCS2 bound to TrkB in an activity-dependent manner to facilitate its translocation to postsynaptic densities for synaptic tagging and maintenance of synaptic potentiation. Neurons lacking SorCS2 failed to respond to BDNF by TrkB autophosphorylation, and activation of downstream signaling cascades, impacting neurite outgrowth and spine formation. Accordingly, Sorcs2-/- mice displayed impaired formation of long-term memory, increased risk taking and stimulus seeking behavior, enhanced susceptibility to stress and impaired prepulse inhibition. Our results identify SorCS2 as an indispensable coreceptor for p75NTR and TrkB in hippocampal neurons and suggest SORCS2 as the link between proBDNF/BDNF signaling and mental disorders.
Collapse
Affiliation(s)
- S Glerup
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - U Bolcho
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - S Mølgaard
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - S Bøggild
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - C B Vaegter
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - A H Smith
- Yale School of Medicine, Interdepartmental Neuroscience Program and Medical Scientist Training Program, New Haven, CT, USA
- Department of Psychiatry, VAT CT Healthcare Center, and Yale School of Medicine, New Haven, CT, USA
| | | | - P L Ovesen
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - L F Pedersen
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - A N Fjorback
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - M Kjolby
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - H Login
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - M M Holm
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - O M Andersen
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - J R Nyengaard
- MIND Center, Stereology and Electron Microscopy Laboratory, Aarhus University, Aarhus C, Denmark
| | - T E Willnow
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - K Jensen
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - A Nykjaer
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE- Nordic EMBL Partnership for Molecular Medicine, Aarhus C, Denmark
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
50
|
Abstract
Brain-derived neurotrophic factor (BDNF) belongs to a family of small secreted proteins that also include nerve growth factor, neurotrophin 3, and neurotrophin 4. BDNF stands out among all neurotrophins by its high expression levels in the brain and its potent effects at synapses. Several aspects of BDNF biology such as transcription, processing, and secretion are regulated by synaptic activity. Such observations prompted the suggestion that BDNF may regulate activity-dependent forms of synaptic plasticity such as long-term potentiation (LTP), a sustained enhancement of excitatory synaptic efficacy thought to underlie learning and memory. Here, we will review the evidence pointing to a fundamental role of this neurotrophin in LTP, especially within the hippocampus. Prominent questions in the field, including the release and action sites of BDNF during LTP, as well as the signaling and molecular mechanisms involved, will also be addressed. The diverse effects of BDNF at excitatory synapses are determined by the activation of TrkB receptors and downstream signaling pathways, and the functions, typically opposing in nature, of its immature form (proBDNF). The activation of p75NTR receptors by proBDNF and the implications for long-term depression will also be addressed. Finally, we discuss the synergy between TrkB and glucocorticoid receptor signaling to determine cellular responses to stress.
Collapse
Affiliation(s)
- G Leal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - C R Bramham
- K.G. Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
| | - C B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; University of Coimbra, Coimbra, Portugal.
| |
Collapse
|