1
|
Blanco FA, Saifullah MAB, Cheng JX, Abella C, Scala F, Firozi K, Niu S, Park J, Chin J, Tolias KF. Targeting Tiam1 Enhances Hippocampal-Dependent Learning and Memory in the Adult Brain and Promotes NMDA Receptor-Mediated Synaptic Plasticity and Function. J Neurosci 2025; 45:e0298242024. [PMID: 39725519 PMCID: PMC11800756 DOI: 10.1523/jneurosci.0298-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 10/18/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Excitatory synapses and the actin-rich dendritic spines on which they reside are indispensable for information processing and storage in the brain. In the adult hippocampus, excitatory synapses must balance plasticity and stability to support learning and memory. However, the mechanisms governing this balance remain poorly understood. Tiam1 is an actin cytoskeleton regulator prominently expressed in the dentate gyrus (DG) throughout life. Previously, we showed that Tiam1 promotes dentate granule cell synapse and spine stabilization during development, but its role in the adult hippocampus remains unclear. Here, we deleted Tiam1 from adult forebrain excitatory neurons (Tiam1fKO ) and assessed the effects on hippocampal-dependent behaviors. Adult male and female Tiam1fKO mice displayed enhanced contextual fear memory, fear extinction, and spatial discrimination. Investigation into underlying mechanisms revealed that dentate granule cells from Tiam1fKO brain slices exhibited augmented synaptic plasticity and N-methyl-D-aspartate-type glutamate receptor (NMDAR) function. Additionally, Tiam1 loss in primary hippocampal neurons blocked agonist-induced NMDAR internalization, reduced filamentous actin levels, and promoted activity-dependent spine remodeling. Notably, strong NMDAR activation in wild-type hippocampal neurons triggered Tiam1 loss from spines. Our results suggest that Tiam1 normally constrains hippocampal-dependent learning and memory in the adult brain by restricting NMDAR-mediated synaptic plasticity in the DG. We propose that Tiam1 achieves this by limiting NMDAR availability at synaptic membranes and stabilizing spine actin cytoskeleton and that these constraints can be alleviated by activity-dependent degradation of Tiam1. These findings reveal a previously unknown mechanism restricting hippocampal synaptic plasticity and highlight Tiam1 as a therapeutic target for enhancing cognitive function.
Collapse
Affiliation(s)
- Francisco A Blanco
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | | | - Jinxuan X Cheng
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Carlota Abella
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Federico Scala
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Sanyong Niu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Jin Park
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Memory & Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
2
|
Nam S, Kang D, Jeon SP, Nam D, Jo JW, Park SJ, Lee J, Kim MG, Ha TJ, Park SK, Kim YH. Contact-Engineered Oxide Memtransistors for Homeostasis-Based High-Linearity and Precision Neuromorphic Computing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409510. [PMID: 39757564 DOI: 10.1002/smll.202409510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Homeostasis is essential in biological neural networks, optimizing information processing and experience-dependent learning by maintaining the balance of neuronal activity. However, conventional two-terminal memristors have limitations in implementing homeostatic functions due to the absence of global regulation ability. Here, three-terminal oxide memtransistor-based homeostatic synapses are demonstrated to perform highly linear synaptic weight update and enhanced accuracy in neuromorphic computing. Particularly, by leveraging the gate control of contact-engineered indium-gallium-zinc-oxide (IGZO) memtransistor, synaptic weight scaling is enabled for high-linearity and precision neuromorphic computing. Moreover, sinusoidal control of gate voltage is demonstrated, possibly enabling the emulation of higher-order synaptic functions. The device structure of IGZO memtransistor is optimized regarding the source/drain electrode materials and an interfacial layer inserted between the IGZO channel and source electrode. As a result, memtransistors exhibiting high current switching ratio of >104 and reliable endurance characteristics are obtained. Furthermore, through the adaptation of synaptic scaling, emulating the homeostasis, non-linearity values of 0.01 and -0.01 are achieved for potentiation and depression, respectively, exhibiting a recognition accuracy of 91.77% for digit images. It is envisioned that the contact-engineered IGZO memtransistors hold significant promise for implementing the homeostasis in neuromorphic computing for high linearity and high efficiency.
Collapse
Affiliation(s)
- San Nam
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Donghyun Kang
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seong-Pil Jeon
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dayul Nam
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jeong-Wan Jo
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ, United Kingdom
| | - Sang-Joon Park
- Department of Electronic Materials Engineering, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Jiyong Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Myung-Gil Kim
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Tae-Jun Ha
- Department of Electronic Materials Engineering, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Sung Kyu Park
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yong-Hoon Kim
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
3
|
Li J, Ye J. Chronic intermittent hypoxia induces cognitive impairment in Alzheimer's disease mouse model via postsynaptic mechanisms. Sleep Breath 2024; 28:1197-1205. [PMID: 38267641 DOI: 10.1007/s11325-023-02970-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/26/2024]
Abstract
PURPOSE Obstructive sleep apnea (OSA) is highly comorbid with Alzheimer's disease (AD) and may represent a risk factor for inducing or accelerating cognitive impairment in AD. Chronic intermittent hypoxia (CIH) has been considered to be a predictor of developing cognitive decline and AD. However, the precise underlying mechanisms by which CIH contributes to cognitive impairment remain unknown. In the present study, we examined the effects of CIH on cognition and hippocampal function in APP/PS1 mice, an animal model of AD. METHODS Wild-type (WT) and APP/PS1 mice were subjected to one of the following conditions for 2 weeks: (1) sham condition (continuous room air) or (2) CIH condition. The oxygen concentration of the CIH condition transitioned from 5 to 21%. Behavioral tests, electrophysiological recording, real-time polymerase chain reaction, and Western blot were used to assess the effect of CIH on cognitive performance and synaptic plasticity. RESULTS CIH exposure did not affect motor coordination, general locomotor activity, anxiety, or willingness to explore. However, behavioral test results indicated that APP/PS1-CIH mice showed more spatial learning and memory deficits. CIH induced long-term potentiation (LTP) dysfunction of the hippocampus in WT mice. These effects were aggravated in APP/PS1 mice. The N-methyl-D-aspartic acid receptor (NMDAR) NR1 subunit and postsynaptic density 95 (PSD95) in the hippocampus of WT and APP/PS1 mice were downregulated. CONCLUSIONS These findings showed that a postsynaptic mechanism was involved in the effect of CIH on cognitive impairment.
Collapse
Affiliation(s)
- Juan Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Jingying Ye
- Department of Otorhinolaryngology-Head and Neck Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
- Institute for Precision Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
4
|
Wu C, Tu T, Xie M, Wang Y, Yan B, Gong Y, Zhang J, Zhou X, Xie Z. Spatially resolved transcriptome of the aging mouse brain. Aging Cell 2024; 23:e14109. [PMID: 38372175 PMCID: PMC11113349 DOI: 10.1111/acel.14109] [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: 09/28/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024] Open
Abstract
Brain aging is associated with cognitive decline, memory loss and many neurodegenerative disorders. The mammalian brain has distinct structural regions that perform specific functions. However, our understanding in gene expression and cell types within the context of the spatial organization of the mammalian aging brain is limited. Here we generated spatial transcriptomic maps of young and old mouse brains. We identified 27 distinguished brain spatial domains, including layer-specific subregions that are difficult to dissect individually. We comprehensively characterized spatial-specific changes in gene expression in the aging brain, particularly for isocortex, the hippocampal formation, brainstem and fiber tracts, and validated some gene expression differences by qPCR and immunohistochemistry. We identified aging-related genes and pathways that vary in a coordinated manner across spatial regions and parsed the spatial features of aging-related signals, providing important clues to understand genes with specific functions in different brain regions during aging. Combined with single-cell transcriptomics data, we characterized the spatial distribution of brain cell types. The proportion of immature neurons decreased in the DG region with aging, indicating that the formation of new neurons is blocked. Finally, we detected changes in information interactions between regions and found specific pathways were deregulated with aging, including classic signaling WNT and layer-specific signaling COLLAGEN. In summary, we established a spatial molecular atlas of the aging mouse brain (http://sysbio.gzzoc.com/Mouse-Brain-Aging/), which provides important resources and novel insights into the molecular mechanism of brain aging.
Collapse
Affiliation(s)
- Cheng Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Tianxiang Tu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Mingzhe Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yiting Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesInstitutes of Brain Science, Institute for Medical and Engineering Innovation, Department of Ophthalmology, Eye & ENT Hospital, Fudan UniversityShanghaiChina
| | - Biao Yan
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesInstitutes of Brain Science, Institute for Medical and Engineering Innovation, Department of Ophthalmology, Eye & ENT Hospital, Fudan UniversityShanghaiChina
| | - Yajun Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Jiayi Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory DiseasesInstitutes of Brain Science, Institute for Medical and Engineering Innovation, Department of Ophthalmology, Eye & ENT Hospital, Fudan UniversityShanghaiChina
| | - Xiaolai Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
5
|
Savarimuthu A, Ponniah RJ. Receive, Retain and Retrieve: Psychological and Neurobiological Perspectives on Memory Retrieval. Integr Psychol Behav Sci 2024; 58:303-318. [PMID: 36738400 DOI: 10.1007/s12124-023-09752-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
Memory and learning are interdependent processes that involve encoding, storage, and retrieval. Especially memory retrieval is a fundamental cognitive ability to recall memory traces and update stored memory with new information. For effective memory retrieval and learning, the memory must be stabilized from short-term memory to long-term memory. Hence, it is necessary to understand the process of memory retention and retrieval that enhances the process of learning. Though previous cognitive neuroscience research has focused on memory acquisition and storage, the neurobiological mechanisms underlying memory retrieval and its role in learning are less understood. Therefore, this article offers the viewpoint that memory retrieval is essential for selecting, reactivating, stabilizing, and storing information in long-term memory. In arguing how memories are retrieved, consolidated, transmitted, and strengthened for the long term, the article will examine the psychological and neurobiological aspects of memory and learning with synaptic plasticity, long-term potentiation, genetic transcription, and theta oscillation in the brain.
Collapse
Affiliation(s)
- Anisha Savarimuthu
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India
| | - R Joseph Ponniah
- Department of Humanities and Social Sciences, National Institute of Technology, Tiruchirappalli, India.
| |
Collapse
|
6
|
Nguyen TAS, Castro N, Vitevitch MS, Harding A, Teng R, Arciuli J, Leyton CE, Piguet O, Ballard KJ. Do age and language impairment affect speed of recognition for words with high and low closeness centrality within the phonological network? INTERNATIONAL JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2023; 25:915-928. [PMID: 36416187 DOI: 10.1080/17549507.2022.2141323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
PURPOSE Speed and accuracy of lexical access change with healthy ageing and neurodegeneration. While a word's immediate phonological neighbourhood density (i.e. words differing by a single phoneme) influences access, connectivity to all words in the phonological network (i.e. closeness centrality) may influence processing. This study aimed to investigate the effect of closeness centrality on speed and accuracy of lexical processing pre- and post- a single word-training session in healthy younger and older adults, and adults with logopenic primary progressive aphasia (lvPPA), which affects phonological processing. METHOD Participants included 29 young and 17 older healthy controls, and 10 adults with lvPPA. Participants received one session of word-training on words with high or low closeness centrality, using a picture-word verification task. Changes in lexical decision reaction times (RT) and accuracy were measured. RESULT Baseline RT was unaffected by age and accuracy was at ceiling for controls. Post-training, only young adults' RT were significantly faster. Adults with lvPPA were slower and less accurate than controls at baseline, with no training effect. Closeness centrality did not influence performance. CONCLUSION Absence of training effect for older adults suggests higher threshold to induce priming, possibly associated with insufficient dosage or fatigue. Implications for word-finding interventions with older adults are discussed.
Collapse
Affiliation(s)
| | - Nichol Castro
- Department of Communicative Disorders and Sciences, University at Buffalo, Buffalo, NY, USA
| | | | - Annabel Harding
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Renata Teng
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Joanne Arciuli
- College of Nursing and Health Sciences, Flinders University, Adelaide, SA, Australia
| | - Cristian E Leyton
- School of Psychology and the Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Olivier Piguet
- School of Psychology and the Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Kirrie J Ballard
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- School of Psychology and the Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
7
|
Custers E, Franco A, Kiliaan AJ. Bariatric Surgery and Gut-Brain-Axis Driven Alterations in Cognition and Inflammation. J Inflamm Res 2023; 16:5495-5514. [PMID: 38026245 PMCID: PMC10676679 DOI: 10.2147/jir.s437156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Obesity is associated with systemic inflammation, comorbidities like diabetes, cardiovascular disease and several cancers, cognitive decline and structural and functional brain changes. To treat, or potentially prevent these related comorbidities, individuals with obesity must achieve long-term sustainable weight loss. Often life style interventions, such as dieting and increased physical activity are not successful in achieving long-term weight loss. Meanwhile bariatric surgery has emerged as a safe and effective procedure to treat obesity. Bariatric surgery causes changes in physiological processes, but it is still not fully understood which exact mechanisms are involved. The successful weight loss after bariatric surgery might depend on changes in various energy regulating hormones, such as ghrelin, glucagon-like peptide-1 and peptide YY. Moreover, changes in microbiota composition and white adipose tissue functionality might play a role. Here, we review the effect of obesity on neuroendocrine effects, microbiota composition and adipose tissue and how these may affect inflammation, brain structure and cognition. Finally, we will discuss how these obesity-related changes may improve after bariatric surgery.
Collapse
Affiliation(s)
- Emma Custers
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Ayla Franco
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| | - Amanda Johanne Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain Cognition and Behaviour, Nijmegen, the Netherlands
| |
Collapse
|
8
|
Xu S, Deng Y, Luo J, He E, Liu Y, Zhang K, Yang Y, Xu S, Sha L, Song Y, Xu Q, Cai X. High-Throughput PEDOT:PSS/PtNPs-Modified Microelectrode Array for Simultaneous Recording and Stimulation of Hippocampal Neuronal Networks in Gradual Learning Process. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15736-15746. [PMID: 35294190 DOI: 10.1021/acsami.1c23170] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
When it comes to mechanisms of brain functions such as learning and memory mediated by neural networks, existing multichannel electrophysiological detection and regulation technology at the cellular level does not suffice. To address this challenge, a 128-channel microelectrode array (MEA) was fabricated for electrical stimulation (ES) training and electrophysiological recording of the hippocampal neurons in vitro. The PEDOT:PSS/PtNPs-coated microelectrodes dramatically promote the recording and electrical stimulation performance. The MEA exhibited low impedance (10.94 ± 0.49 kohm), small phase delay (-12.54 ± 0.51°), high charge storage capacity (14.84 ± 2.72 mC/cm2), and high maximum safe injection charge density (4.37 ± 0.22 mC/cm2), meeting the specific requirements for training neural networks in vitro. A series of ESs at various frequencies was applied to the neuronal cultures in vitro, seeking the optimum training mode that enables the neuron to display the most obvious plasticity, and 1 Hz ES was determined. The network learning process, including three consecutive trainings, affected the original random spontaneous activity. Along with that, the firing pattern gradually changed to burst and the correlation and synchrony of the neuronal activity in the network have progressively improved, increasing by 314% and 240%, respectively. The neurons remembered these changes for at least 4 h. Collectively, ES activates the learning and memory functions of neurons, which is manifested in transformations in the discharge pattern and the improvement of network correlation and synchrony. This study offers a high-performance MEA revealing the underlying learning and memory functions of the brain and therefore serves as a useful tool for the development of brain functions in the future.
Collapse
Affiliation(s)
- Shihong Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Deng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Enhui He
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaoyao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kui Zhang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shengwei Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Longze Sha
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Fluid intelligence and the locus coeruleus-norepinephrine system. Proc Natl Acad Sci U S A 2021; 118:2110630118. [PMID: 34764223 DOI: 10.1073/pnas.2110630118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 11/18/2022] Open
Abstract
The last decade has seen significant progress identifying genetic and brain differences related to intelligence. However, there remain considerable gaps in our understanding of how cognitive mechanisms that underpin intelligence map onto various brain functions. In this article, we argue that the locus coeruleus-norepinephrine system is essential for understanding the biological basis of intelligence. We review evidence suggesting that the locus coeruleus-norepinephrine system plays a central role at all levels of brain function, from metabolic processes to the organization of large-scale brain networks. We connect this evidence with our executive attention view of working-memory capacity and fluid intelligence and present analyses on baseline pupil size, an indicator of locus coeruleus activity. Using a latent variable approach, our analyses showed that a common executive attention factor predicted baseline pupil size. Additionally, the executive attention function of disengagement--not maintenance--uniquely predicted baseline pupil size. These findings suggest that the ability to control attention may be important for understanding how cognitive mechanisms of fluid intelligence map onto the locus coeruleus-norepinephrine system. We discuss how further research is needed to better understand the relationships between fluid intelligence, the locus coeruleus-norepinephrine system, and functionally organized brain networks.
Collapse
|
10
|
El Ahdab N, Haque M, Madogwe E, Koski KG, Scott ME. Maternal nematode infection upregulates expression of Th2/Treg and diapedesis related genes in the neonatal brain. Sci Rep 2021; 11:22082. [PMID: 34764345 PMCID: PMC8585879 DOI: 10.1038/s41598-021-01510-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/26/2021] [Indexed: 11/28/2022] Open
Abstract
Intestinal nematode infections common during pregnancy have recently been shown to have impacts that extend to their uninfected offspring including altered brain gene expression. If maternal immune signals reach the neonatal brain, they might alter neuroimmune development. We explored expression of genes associated with four distinct types of T cells (Th1, Th2, Th17, Treg) and with leukocyte transendothelial migration and endocytosis transport across the blood–brain barrier (BBB) in the postnatal brain of offspring of nematode-infected mice, through secondary analysis of a whole brain gene expression database. Th1/Th17 expression was lowered by maternal infection as evidenced by down-regulated expression of IL1β, Th1 receptors and related proteins, and of IL22 and several Th17 genes associated with immunopathology. In contrast, Th2/Treg related pathways were upregulated as shown by higher expression of IL4 and TGF-β family genes. Maternal infection also upregulated expression of pathways and integrin genes involved in transport of leukocytes in between endothelial cells but downregulated endosome vesicle formation related genes that are necessary for endocytosis of immunoglobulins across the BBB. Taken together, pup brain gene expression indicates that maternal nematode infection enhanced movement of leukocytes across the neonatal BBB and promoted a Th2/Treg environment that presumably minimizes the proinflammatory Th1 response in the pup brain.
Collapse
Affiliation(s)
- Nawal El Ahdab
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Ejimedo Madogwe
- Department of Animal Science, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
11
|
Lewis V, Laberge F, Heyland A. Transcriptomic signature of extinction learning in the brain of the fire-bellied toad, Bombina orientalis. Neurobiol Learn Mem 2021; 184:107502. [PMID: 34391934 DOI: 10.1016/j.nlm.2021.107502] [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: 04/19/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 11/26/2022]
Abstract
Insight into the molecular and cellular mechanisms of learning and memory from a diverse array of taxa contributes to our understanding of the evolution of these processes. The fire-bellied toad, Bombina orientalis, is a basal anuran amphibian model species who could help us describe shared and divergent characteristics of learning and memory mechanisms between amphibians and other vertebrates, and hence answer questions about the evolution of learning. Utilizing next generation sequencing techniques, we profiled gene expression patterns associated with the extinction of prey-catching conditioning in the brain of the fire-bellied toad. For this purpose, gene expression was at first compared between toads sacrificed after acquisition and extinction of the conditioned response. A second comparison was done between toads submitted to extinction following either short or long acquisition training, which results in toads displaying response extinction or resistance to extinction, respectively. We analyzed brain tissue transcription profiles common to both acquisition and extinction learning, or unique to extinction learning and resistance to extinction, and found significant overlap in gene expression related to molecular pathways involving neuronal plasticity (e.g. structural modification, transcription). However, extinction learning induced a unique GABAergic transcriptomic signal, which may be responsible for suppression of the original response memory. Further, when comparing extinction learning in short- and long-trained groups, short training engaged many pathways related to neuronal plasticity, as expected, but long training engaged molecular pathways related to the suppression of learning through epigenetic mediated transcriptional suppression and inhibitory neurotransmission. Overall, gene expression patterns associated with extinction learning in the fire-bellied toad were similar to those found in mammals submitted to extinction, although some divergent profiles highlighted potential differences in the mechanisms of learning and memory among tetrapods.
Collapse
Affiliation(s)
- Vern Lewis
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Frédéric Laberge
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Andreas Heyland
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
12
|
Calleja-Felipe M, Wojtas MN, Diaz-González M, Ciceri D, Escribano R, Ouro A, Morales M, Knafo S. FORTIS: a live-cell assay to monitor AMPA receptors using pH-sensitive fluorescence tags. Transl Psychiatry 2021; 11:324. [PMID: 34045447 PMCID: PMC8160262 DOI: 10.1038/s41398-021-01457-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/09/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
The real-time live fluorescent monitoring of surface AMPA receptors (AMPARs) could open new opportunities for drug discovery and phenotypic screening concerning neuropsychiatric disorders. We have developed FORTIS, a tool based on pH sensitivity capable of detecting subtle changes in surface AMPARs at a neuronal population level. The expression of SEP-GluA1 or pHuji-GluA1 recombinant AMPAR subunits in mammalian neurons cultured in 96-well plates enables surface AMPARs to be monitored with a microplate reader. Thus, FORTIS can register rapid changes in surface AMPARs induced by drugs or genetic modifications without having to rely on conventional electrophysiology or imaging. By combining FORTIS with pharmacological manipulations, basal surface AMPARs, and plasticity-like changes can be monitored. We expect that employing FORTIS to screen for changes in surface AMPARs will accelerate both neuroscience research and drug discovery.
Collapse
Affiliation(s)
- María Calleja-Felipe
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Magdalena Natalia Wojtas
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Marta Diaz-González
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dalila Ciceri
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Raúl Escribano
- Fundación Biofísica Bizkaia/Biofisika Bizkaia Fundazioa (FBB), Barrio Sarriena s/n, Leioa, E-48940, Spain
| | - Alberto Ouro
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Miguel Morales
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Shira Knafo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain.
| |
Collapse
|
13
|
Chang HM, Lin HC, Cheng HL, Liao CK, Tseng TJ, Renn TY, Lan CT, Chen LY. Melatonin Successfully Rescues the Hippocampal Molecular Machinery and Enhances Anti-oxidative Activity Following Early-Life Sleep Deprivation Injury. Antioxidants (Basel) 2021; 10:antiox10050774. [PMID: 34068192 PMCID: PMC8153000 DOI: 10.3390/antiox10050774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Early-life sleep deprivation (ESD) is a serious condition with severe cognitive sequelae. Considering hippocampus plays an essential role in cognitive regulation, the present study aims to determine whether melatonin, a neuroendocrine beard with significant anti-oxidative activity, would greatly depress the hippocampal oxidative stress, improves the molecular machinery, and consequently exerts the neuro-protective effects following ESD. Male weanling Wistar rats (postnatal day 21) were subjected to ESD for three weeks. During this period, the animals were administered normal saline or melatonin (10 mg/kg) via intraperitoneal injection between 09:00 and 09:30 daily. After three cycles of ESD, the animals were kept under normal sleep/wake cycle until they reached adulthood and were sacrificed. The results indicated that ESD causes long-term effects, such as impairment of ionic distribution, interruption of the expressions of neurotransmitters and receptors, decreases in the levels of several antioxidant enzymes, and impairment of several signaling pathways, which contribute to neuronal death in hippocampal regions. Melatonin administration during ESD prevented these effects. Quantitative evaluation of cells also revealed a higher number of neurons in the melatonin-treated animals when compared with the saline-treated animals. As the hippocampus is critical to cognitive activity, preserving or even improving the hippocampal molecular machinery by melatonin during ESD not only helps us to better understand the underlying mechanisms of ESD-induced neuronal dysfunction, but also the therapeutic use of melatonin to counteract ESD-induced neuronal deficiency.
Collapse
Affiliation(s)
- Hung-Ming Chang
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-M.C.); (T.-Y.R.)
| | - Hsing-Chun Lin
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan; (H.-C.L.); (H.-L.C.)
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Hsin-Lin Cheng
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan; (H.-C.L.); (H.-L.C.)
| | - Chih-Kai Liao
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-K.L.); (T.-J.T.); (C.-T.L.)
| | - To-Jung Tseng
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-K.L.); (T.-J.T.); (C.-T.L.)
| | - Ting-Yi Renn
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-M.C.); (T.-Y.R.)
| | - Chyn-Tair Lan
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-K.L.); (T.-J.T.); (C.-T.L.)
| | - Li-You Chen
- Department of Anatomy, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (C.-K.L.); (T.-J.T.); (C.-T.L.)
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: ; Tel.: +886-4-2473-0022 (ext. 11602)
| |
Collapse
|
14
|
Avchalumov Y, Mandyam CD. Plasticity in the Hippocampus, Neurogenesis and Drugs of Abuse. Brain Sci 2021; 11:404. [PMID: 33810204 PMCID: PMC8004884 DOI: 10.3390/brainsci11030404] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
Synaptic plasticity in the hippocampus assists with consolidation and storage of long-lasting memories. Decades of research has provided substantial information on the cellular and molecular mechanisms underlying synaptic plasticity in the hippocampus, and this review discusses these mechanisms in brief. Addiction is a chronic relapsing disorder with loss of control over drug taking and drug seeking that is caused by long-lasting memories of drug experience. Relapse to drug use is caused by exposure to context and cues associated with the drug experience, and is a major clinical problem that contributes to the persistence of addiction. This review also briefly discusses some evidence that drugs of abuse alter plasticity in the hippocampus, and that development of novel treatment strategies that reverse or prevent drug-induced synaptic alterations in the hippocampus may reduce relapse behaviors associated with addiction.
Collapse
Affiliation(s)
| | - Chitra D. Mandyam
- VA San Diego Healthcare System, San Diego, CA 92161, USA;
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| |
Collapse
|
15
|
Jiang CH, Wei M, Zhang C, Shi YS. The amino-terminal domain of GluA1 mediates LTP maintenance via interaction with neuroplastin-65. Proc Natl Acad Sci U S A 2021; 118:e2019194118. [PMID: 33627404 PMCID: PMC7936340 DOI: 10.1073/pnas.2019194118] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Long-term potentiation (LTP) has long been considered as an important cellular mechanism for learning and memory. LTP expression involves NMDA receptor-dependent synaptic insertion of AMPA receptors (AMPARs). However, how AMPARs are recruited and anchored at the postsynaptic membrane during LTP remains largely unknown. In this study, using CRISPR/Cas9 to delete the endogenous AMPARs and replace them with the mutant forms in single neurons, we have found that the amino-terminal domain (ATD) of GluA1 is required for LTP maintenance. Moreover, we show that GluA1 ATD directly interacts with the cell adhesion molecule neuroplastin-65 (Np65). Neurons lacking Np65 exhibit severely impaired LTP maintenance, and Np65 deletion prevents GluA1 from rescuing LTP in AMPARs-deleted neurons. Thus, our study reveals an essential role for GluA1/Np65 binding in anchoring AMPARs at the postsynaptic membrane during LTP.
Collapse
Affiliation(s)
- Chao-Hua Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, 210032 Nanjing, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 210032 Nanjing, China
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China;
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing University, 210032 Nanjing, China;
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 210032 Nanjing, China
- Institute for Brain Sciences, Nanjing University, 210032 Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, 210032 Nanjing, China
| |
Collapse
|
16
|
Duran J, Gruart A, López-Ramos JC, Delgado-García JM, Guinovart JJ. Glycogen in Astrocytes and Neurons: Physiological and Pathological Aspects. ADVANCES IN NEUROBIOLOGY 2019; 23:311-329. [PMID: 31667813 DOI: 10.1007/978-3-030-27480-1_10] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain glycogen is stored mainly in astrocytes, although neurons also have an active glycogen metabolism. Glycogen has gained relevance as a key player in brain function. In this regard, genetically modified animals have allowed researchers to unravel new roles of this polysaccharide in the brain. Remarkably, mice in which glycogen synthase is abolished in the brain, and thus devoid of brain glycogen, are viable, thereby indicating that the polysaccharide in this organ is not a requirement for survival. While there was growing evidence supporting a role of glycogen in learning and memory, these animals have now confirmed that glycogen participates in these two processes.The association of epilepsy with brain glycogen has also attracted attention. Analysis of genetically modified mice indicates that the relation between brain glycogen and epilepsy is complex. While the formation of glycogen aggregates clearly underlies epilepsy, as in Lafora Disease (LD), the absence of glycogen also favors the occurrence of seizures.LD is a rare genetic condition that affects children. It is characterized by epileptic seizures and neurodegeneration, and it develops rapidly until finally causing death. Research into this disease has unveiled new aspects of glycogen metabolism. Animal models of LD accumulate polyglucosan bodies formed by aberrant glycogen aggregates, called Lafora bodies (LBs). The abolition of glycogen synthase (GS) prevents the formation of LBs and the development of LD, thereby indicating that glycogen accumulation underlies this disease and the associated symptoms, and thus establishing a clear relation between the accumulation of glycogen aggregates and the incidence of seizures.Although it was initially accepted that LBs were essentially neuronal, it is now evident that astrocytes also accumulate polyglucosan aggregates in LD. However, the appearance and composition of these deposits differs from that observed in neurons. Of note, the astrocytic aggregates in LD models show remarkable similarities with corpora amylacea (CA), a type of polyglucosan aggregate observed in the brains of aged mice and humans. The abolition of GS in mice also impedes the formation of CA with age and at the same time prevents the formation of a number of protein aggregates associated with aging. Therefore CA may play a role in age-related neurological decline.
Collapse
Affiliation(s)
- Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, Seville, Spain
| | | | | | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
17
|
Rathour RK, Narayanan R. Degeneracy in hippocampal physiology and plasticity. Hippocampus 2019; 29:980-1022. [PMID: 31301166 PMCID: PMC6771840 DOI: 10.1002/hipo.23139] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/27/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022]
Abstract
Degeneracy, defined as the ability of structurally disparate elements to perform analogous function, has largely been assessed from the perspective of maintaining robustness of physiology or plasticity. How does the framework of degeneracy assimilate into an encoding system where the ability to change is an essential ingredient for storing new incoming information? Could degeneracy maintain the balance between the apparently contradictory goals of the need to change for encoding and the need to resist change towards maintaining homeostasis? In this review, we explore these fundamental questions with the mammalian hippocampus as an example encoding system. We systematically catalog lines of evidence, spanning multiple scales of analysis that point to the expression of degeneracy in hippocampal physiology and plasticity. We assess the potential of degeneracy as a framework to achieve the conjoint goals of encoding and homeostasis without cross-interferences. We postulate that biological complexity, involving interactions among the numerous parameters spanning different scales of analysis, could establish disparate routes towards accomplishing these conjoint goals. These disparate routes then provide several degrees of freedom to the encoding-homeostasis system in accomplishing its tasks in an input- and state-dependent manner. Finally, the expression of degeneracy spanning multiple scales offers an ideal reconciliation to several outstanding controversies, through the recognition that the seemingly contradictory disparate observations are merely alternate routes that the system might recruit towards accomplishment of its goals.
Collapse
Affiliation(s)
- Rahul K. Rathour
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| | - Rishikesh Narayanan
- Cellular Neurophysiology LaboratoryMolecular Biophysics Unit, Indian Institute of ScienceBangaloreIndia
| |
Collapse
|
18
|
Leal-Galicia P, Romo-Parra H, Rodríguez-Serrano LM, Buenrostro-Jáuregui M. Regulation of adult hippocampal neurogenesis exerted by sexual, cognitive and physical activity: An update. J Chem Neuroanat 2019; 101:101667. [PMID: 31421204 DOI: 10.1016/j.jchemneu.2019.101667] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/29/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
In 1962, Joseph Altman described that the brain generates neurons after the postnatal period, and this continues throughout your life (Altman, 1962). This was a breakthrough in the neuroscience field because before this the accepted paradigm was that the brain only generated neurons during the embryonal development. This discovery has been controversial ever since, especially since one of the areas of the brain with neurogenic properties is the hippocampus, which is the area involved in memory storage and neurodegenerative processes. The adult hippocampal neurogenesis modulates in response to different environmental factors. In this article, we review how exercise and cognitive and sexual activity can regulate the generation of new neurons in the hippocampal in an adult brain and the impact of these new neurons in the brain circuitry.
Collapse
Affiliation(s)
- P Leal-Galicia
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, México.
| | - H Romo-Parra
- Facultad de Psicología, Universidad Anáhuac, Mexico City, Mexico
| | - L M Rodríguez-Serrano
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, México
| | - M Buenrostro-Jáuregui
- Laboratorio de Neurociencias, Departamento de Psicología, Universidad Iberoamericana Ciudad de México, México.
| |
Collapse
|
19
|
Wang X, Liu D, Huang HZ, Wang ZH, Hou TY, Yang X, Pang P, Wei N, Zhou YF, Dupras MJ, Calon F, Wang YT, Man HY, Chen JG, Wang JZ, Hébert SS, Lu Y, Zhu LQ. A Novel MicroRNA-124/PTPN1 Signal Pathway Mediates Synaptic and Memory Deficits in Alzheimer's Disease. Biol Psychiatry 2018; 83:395-405. [PMID: 28965984 DOI: 10.1016/j.biopsych.2017.07.023] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Synaptic loss is an early pathological event in Alzheimer's disease (AD), but its underlying molecular mechanisms remain largely unknown. Recently, microRNAs (miRNAs) have emerged as important modulators of synaptic function and memory. METHODS We used miRNA array and quantitative polymerase chain reaction to examine the alteration of miRNAs in AD mice and patients as well as the Morris water maze to evaluate learning and memory in the mice. We also used adeno-associated virus or lentivirus to introduce tyrosine-protein phosphatase non-receptor type 1 (PTPN1) expression of silencing RNAs. Long-term potentiation and Golgi staining were used to evaluate the synaptic function and structure. We designed a peptide to interrupt miR-124/PTPN1 interaction. RESULTS Here we report that neuronal miR-124 is dramatically increased in the hippocampus of Tg2576 mice, a recognized AD mouse model. Similar changes were observed in specific brain regions of affected AD individuals. We further identified PTPN1 as a direct target of miR-124. Overexpression of miR-124 or knockdown of PTPN1 recapitulated AD-like phenotypes in mice, including deficits in synaptic transmission and plasticity as well as memory by impairing the glutamate receptor 2 membrane insertion. Most importantly, rebuilding the miR-124/PTPN1 pathway by suppression of miR-124, overexpression of PTPN1, or application of a peptide that disrupts the miR-124/PTPN1 interaction could restore synaptic failure and memory deficits. CONCLUSIONS Taken together, these results identified the miR-124/PTPN1 pathway as a critical mediator of synaptic dysfunction and memory loss in AD, and the miR-124/PTPN1 pathway could be considered as a promising novel therapeutic target for AD patients.
Collapse
Affiliation(s)
- Xiong Wang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Dan Liu
- Department of Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - He-Zhou Huang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Zhi-Hao Wang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Tong-Yao Hou
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Xin Yang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Pei Pang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Na Wei
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ya-Fan Zhou
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Marie-Josée Dupras
- Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada
| | - Frédéric Calon
- Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada
| | - Yu-Tian Wang
- Brain Research Centre and Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, Massachusetts
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Sébastien S Hébert
- Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Department of Psychiatry and Neuroscience, Université Laval, Québec City, Québec, Canada; Centre de recherche du CHU de Québec, Axe Neurosciences, Québec City, Québec, Canada
| | - Youming Lu
- Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Sino-Canada Collaborative Platform on Molecular Biology of Neurological Disease, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China; Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
20
|
Galaj E, Seepersad N, Dakmak Z, Ranaldi R. Blockade of NMDA receptors blocks the acquisition of cocaine conditioned approach in rats. Eur J Pharmacol 2017; 818:480-485. [PMID: 29157983 DOI: 10.1016/j.ejphar.2017.11.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 11/13/2017] [Accepted: 11/16/2017] [Indexed: 10/18/2022]
Abstract
Conditioned stimuli (CSs) exert motivational effects on both adaptive and pathological reward-related behaviors, including drug taking and seeking. We developed a paradigm that allows us to investigate the neuropharmacology by which previously neutral stimuli acquire the capacity to function as CSs and elicit (intravenous) cocaine conditioned approach and used this paradigm to test the role of NMDA receptor stimulation in the acquisition of cocaine conditioned approach. Rats were injected systemically with the NMDA receptor antagonist, MK-801, before the start of 4 consecutive conditioning sessions, each of which consisted of 20 randomly presented light/tone (CS) presentations paired with cocaine infusion contingent upon nose pokes. Rats later were subjected to a CS-only test. To test the role of NMDA receptor stimulation in the already established conditioned approach, rats were injected with MK-801 prior to the CS-only test that occurred after 18 CS-cocaine conditioning sessions. Blockade of NMDA receptors significantly impaired the acquisition of cocaine-conditioned approach as indicated by the emission of significantly fewer nose pokes and significantly longer latencies to nose poke during CS presentations. When MK-801 treatment was applied after the acquisition of conditioned approach responding it had no effect on these measures. These results suggest that NMDA receptor stimulation plays an important role in the acquisition of reward-related conditioned responses driven by intravenous cocaine-associated CSs.
Collapse
Affiliation(s)
- Ewa Galaj
- Neuropsychology Doctoral Program, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Neal Seepersad
- Department of Psychology, Queens College, City University of New York, Flushing, NY 11367, USA
| | - Zena Dakmak
- Department of Psychology, Queens College, City University of New York, Flushing, NY 11367, USA
| | - Robert Ranaldi
- Neuropsychology Doctoral Program, The Graduate Center of the City University of New York, New York, NY 10016, USA; Department of Psychology, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
21
|
Parent AS, Pinson A, Woods N, Chatzi C, Vaaga CE, Bensen A, Gérard A, Thome JP, Bourguignon JP, Westbrook GL. Early exposure to Aroclor 1254 in vivo disrupts the functional synaptic development of newborn hippocampal granule cells. Eur J Neurosci 2016; 44:3001-3010. [PMID: 27740705 DOI: 10.1111/ejn.13437] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 02/05/2023]
Abstract
Neurogenesis in the dentate gyrus is sensitive to endogenous and exogenous factors that influence hippocampal function. Ongoing neurogenesis and the integration of these new neurons throughout life thus may provide a sensitive indicator of environmental stress. We examined the effects of Aroclor 1254 (A1254), a mixture of polychlorinated biphenyls (PCBs), on the development and function of newly generated dentate granule cells. Early exposure to A1254 has been associated with learning impairment in children, suggesting potential impact on the development of hippocampus and/or cortical circuits. Oral A1254 (from the 6th day of gestation to postnatal day 21) produced the expected increase in PCB levels in brain at postnatal day 21, which persisted at lower levels into adulthood. A1254 did not affect the proliferation or survival of newborn neurons in immature animals nor did it cause overt changes in neuronal morphology. However, A1254 occluded the normal developmental increase in sEPSC frequency in the third post-mitotic week without altering the average sEPSC amplitude. Our results suggest that early exposure to PCBs can disrupt excitatory synaptic function during a period of active synaptogenesis, and thus could contribute to the cognitive effects noted in children exposed to PCBs.
Collapse
Affiliation(s)
- A S Parent
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - A Pinson
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - N Woods
- Vollum Institute, Portland, OR, USA
| | - C Chatzi
- Vollum Institute, Portland, OR, USA
| | | | - A Bensen
- Vollum Institute, Portland, OR, USA
| | - A Gérard
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | - J P Thome
- Laboratory of Animal Ecology and Ecotoxicology, University of Liège, Liège, Belgium
| | - J P Bourguignon
- Neuroendocrinology Unit, GIGA-N, University of Liège, CHU Sart Tilman, 4000, Liège, Belgium
| | | |
Collapse
|
22
|
Moon M, Jeong HU, Choi JG, Jeon SG, Song EJ, Hong SP, Oh MS. Memory-enhancing effects of Cuscuta japonica Choisy via enhancement of adult hippocampal neurogenesis in mice. Behav Brain Res 2016; 311:173-182. [DOI: 10.1016/j.bbr.2016.05.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/08/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
|
23
|
Abstract
The central point of this article is that the concept of memory as information storage in the brain is inadequate for and irrelevant to understanding the nervous system. Beginning from the sensorimotor hypothesis that underlies neuroscience—that the entire function of the nervous system is to connect experience to appropriate behavior—the paper defines memories as sequences of events that connect remote experience to present behavior. Their essential components are (a) persistent events that bridge the time from remote experience to present behavior and (b) junctional events in which connections from remote experience and recent experience merge to produce behavior. The sequences comprising even the simplest memories are complex. This is both necessary—to preserve previously learned behaviors—and inevitable—due to secondary activity-driven plasticity. This complexity further highlights the inadequacy of the information storage concept and the importance of extreme simplicity in models used to study memory.
Collapse
Affiliation(s)
- Jonathan R Wolpaw
- Wadsworth Center, New York State Department of Health, Albany, NY 12201-0509, USA.
| |
Collapse
|
24
|
Gonzalez J, Villarreal DM, Morales IS, Derrick BE. Long-term Potentiation at Temporoammonic Path-CA1 Synapses in Freely Moving Rats. Front Neural Circuits 2016; 10:2. [PMID: 26903815 PMCID: PMC4748048 DOI: 10.3389/fncir.2016.00002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/12/2016] [Indexed: 11/23/2022] Open
Abstract
Hippocampal area CA1 receives direct entorhinal layer III input via the temporoammonic path (TAP) and recent studies implicate TAP-CA1 synapses are important for some aspects of hippocampal memory function. Nonetheless, as few studies have examined TAP-CA1 synaptic plasticity in vivo, the induction and longevity of TAP-CA1 long-term potentiation (LTP) has not been fully characterized. We analyzed CA1 responses following stimulation of the medial aspect of the angular bundle and investigated LTP at medial temporoammonic path (mTAP)-CA1 synapses in freely moving rats. We demonstrate monosynaptic mTAP-CA1 responses can be isolated in vivo as evidenced by observations of independent current sinks in the stratum lacunosum moleculare of both areas CA1 and CA3 following angular bundle stimulation. Contrasting prior indications that TAP input rarely elicits CA1 discharge, we observed mTAP-CA1 responses that appeared to contain putative population spikes in 40% of our behaving animals. Theta burst high frequency stimulation of mTAP afferents resulted in an input specific and N-methyl-D-aspartate (NMDA) receptor-dependent LTP of mTAP-CA1 responses in behaving animals. LTP of mTAP-CA1 responses decayed as a function of two exponential decay curves with time constants (τ) of 2.7 and 148 days to decay 63.2% of maximal LTP. In contrast, mTAP-CA1 population spike potentiation longevity demonstrated a τ of 9.6 days. To our knowledge, these studies provide the first description of mTAP-CA1 LTP longevity in vivo. These data indicate TAP input to area CA1 is a physiologically relevant afferent system that displays robust synaptic plasticity.
Collapse
Affiliation(s)
- Jossina Gonzalez
- Department of Biology, University of Texas at San Antonio San Antonio, TX, USA
| | | | - Isaiah S Morales
- Department of Biology, University of Texas at San Antonio San Antonio, TX, USA
| | - Brian E Derrick
- Department of Biology, University of Texas at San AntonioSan Antonio, TX, USA; UTSA Neurosciences Institute, University of Texas at San AntonioSan Antonio, TX, USA
| |
Collapse
|
25
|
Pandey K, Sharma KP, Sharma SK. Histone deacetylase inhibition facilitates massed pattern-induced synaptic plasticity and memory. Learn Mem 2015; 22:514-8. [PMID: 26373830 PMCID: PMC4579358 DOI: 10.1101/lm.039289.115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/09/2015] [Indexed: 11/24/2022]
Abstract
Massed training is less effective for long-term memory formation than the spaced training. The role of acetylation in synaptic plasticity and memory is now well established. However, the role of this important protein modification in synaptic plasticity induced by massed pattern of stimulation or memory induced by massed training is not well understood. Here we show that increasing the level of acetylation enhances long-term potentiation induced by massed pattern of high frequency stimulation. Furthermore, enhancing acetylation level facilitates long-term memory by massed training. Thus, increasing acetylation level facilitates synaptic plasticity and memory by massed patterns.
Collapse
Affiliation(s)
- Kiran Pandey
- National Brain Research Centre, Manesar, Haryana 122051, India
| | | | - Shiv K Sharma
- National Brain Research Centre, Manesar, Haryana 122051, India
| |
Collapse
|
26
|
The modulatory effect of CA1 GABAb receptors on ketamine-induced spatial and non-spatial novelty detection deficits with respect to Ca2+. Neuroscience 2015; 305:157-68. [DOI: 10.1016/j.neuroscience.2015.07.083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 07/24/2015] [Accepted: 07/30/2015] [Indexed: 11/18/2022]
|
27
|
Le Menuet D, Lombès M. The neuronal mineralocorticoid receptor: from cell survival to neurogenesis. Steroids 2014; 91:11-9. [PMID: 24928721 DOI: 10.1016/j.steroids.2014.05.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/18/2014] [Accepted: 05/28/2014] [Indexed: 02/07/2023]
Abstract
Mineralocorticoid receptor (MR), a hormone-activated transcription factor belonging to the nuclear receptor superfamily, exerts widespread actions in many tissues such as tight epithelia, the cardiovascular system, adipose tissues and macrophages. In the mammalian brain, MR is present in the limbic areas where it is highly expressed in neurons of the hippocampus and mostly absent in other regions while the glucocorticoid receptor (GR) expression is ubiquitous. MR binds both aldosterone and glucocorticoids, the latter having a ten-fold higher affinity for MR than for the closely related GR. However, owing to the minimal aldosterone transfer across the blood brain barrier and the absence of neuronal 11β hydroxysteroid dehydrogenase type 2 as an intracellular gate-keeper, neuronal MR appears to be fully occupied even at low physiological glucocorticoid levels while GR activation only occurs at high glucocorticoid concentrations, i.e. at the peak of the circadian rhythm or under stress. This defined a one hormone/two receptors system that works in balance, modulating a large spectrum of actions in the central nervous system. MR and GR are involved in the stress responses, the regulation of neuron excitability, long term potentiation, neuroprotection and neurogenesis in the dentate gyrus. MR thus constitutes a key factor in the arising of higher cognitive functions such as memorization, learning and mood. This review presents an overview of various roles of MR in the central nervous system which are somewhat less studied than that of GR, in the light of recent data obtained using cellular models, animal models and clinical investigations.
Collapse
Affiliation(s)
- Damien Le Menuet
- Inserm U693, Le Kremlin-Bicêtre F-94276, France; Univ Paris-Sud, Faculté de Médecine Paris-Sud, UMR-S693, Le Kremlin-Bicêtre F-94276, France.
| | - Marc Lombès
- Inserm U693, Le Kremlin-Bicêtre F-94276, France; Univ Paris-Sud, Faculté de Médecine Paris-Sud, UMR-S693, Le Kremlin-Bicêtre F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin Bicêtre F-94275, France
| |
Collapse
|
28
|
Jenson D, Yang K, Acevedo-Rodriguez A, Levine A, Broussard JI, Tang J, Dani JA. Dopamine and norepinephrine receptors participate in methylphenidate enhancement of in vivo hippocampal synaptic plasticity. Neuropharmacology 2014; 90:23-32. [PMID: 25445492 DOI: 10.1016/j.neuropharm.2014.10.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 11/30/2022]
Abstract
Attention-deficit hyperactive disorder (ADHD) is the most commonly studied and diagnosed psychiatric disorder in children. Methylphenidate (MPH, e.g., Ritalin) has been used to treat ADHD for over 50 years. It is the most commonly prescribed treatment for ADHD, and in the past decade it was the drug most commonly prescribed to teenagers. In addition, MPH has become one of the most widely abused drugs on college campuses. In this study, we examined the effects of MPH on hippocampal synaptic plasticity, which serves as a measurable quantification of memory mechanisms. Field potentials were recorded with permanently implanted electrodes in freely-moving mice to quantify MPH modulation of perforant path synaptic transmission onto granule cells of the dentate gyrus. Our hypothesis was that MPH affects hippocampal synaptic plasticity underlying learning because MPH boosts catecholamine signaling by blocking the dopamine and norepinephrine transporters (DAT and NET respectively). In vitro hippocampal slice experiments indicated MPH enhances perforant path plasticity, and this MPH enhancement arose from action via D1-type dopamine receptors and β-type adrenergic receptors. Similarly, MPH boosted in vivo initiation of long-term potentiation (LTP). While there was an effect via both dopamine and adrenergic receptors in vivo, LTP induction was more dependent on the MPH-induced action via D1-type dopamine receptors. Under biologically reasonable experimental conditions, MPH enhances hippocampal synaptic plasticity via catecholamine receptors.
Collapse
Affiliation(s)
- Daniel Jenson
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kechun Yang
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Alexandra Acevedo-Rodriguez
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amber Levine
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - John I Broussard
- Department of Neuroscience, Center on Addiction, Learning, Memory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - John A Dani
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Chitty KM, Lagopoulos J, Hickie IB, Hermens DF. The impact of alcohol and tobacco use on in vivo glutathione in youth with bipolar disorder: an exploratory study. J Psychiatr Res 2014; 55:59-67. [PMID: 24755258 DOI: 10.1016/j.jpsychires.2014.03.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 02/08/2023]
Abstract
Risky alcohol consumption and tobacco smoking is highly prevalent in bipolar disorder (BD) and is associated with increased formation of neural reactive oxygen species. Proton magnetic resonance spectroscopy ((1)H-MRS) is an in vivo imaging modality that allows quantification of glutathione (GSH) concentration, the brains primary antioxidant. Sixty-four patients with BD and 49 controls (18-30 years) completed self-report questionnaires regarding alcohol and tobacco use and underwent (1)H-MRS. Levels of GSH in the hippocampus and anterior cingulate cortex (ACC) were determined. Within-group Pearson's correlations were used to explore the relationship between alcohol use and GSH concentration for BD and controls, covarying for age, gender, family history of alcohol dependence and smoking status. Relationships between GSH and presence/severity of alcohol-induced blackouts were determined using Spearman's correlations. In BD, reduced hippocampal-GSH associated with higher alcohol use (R = -0.489, p < 0.021). Reduction of ACC-GSH with increased drinking was non-significant when controlling for tobacco use. Independent samples t-test revealed a significantly decreased ACC-GSH in smokers with BD (t (53) = 4.162, p < 0.001). In controls, alcohol use was not correlated to GSH in either region. In both patients and controls, reduced hippocampal-GSH was associated with blackout presence/severity, supporting a role for the hippocampus in the continuum of alcohol-induced memory impairments. Our preliminary findings suggest that in youth with BD reduced hippocampal-GSH is associated with risky alcohol use and alcohol and tobacco use is associated with reduced ACC-GSH, highlighting the role of these substances as modifiable risk factors for decreased anti-oxidant capacity in BD.
Collapse
Affiliation(s)
- Kate M Chitty
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia.
| | - Jim Lagopoulos
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Ian B Hickie
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Daniel F Hermens
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
30
|
Smarr BL, Jennings KJ, Driscoll JR, Kriegsfeld LJ. A time to remember: the role of circadian clocks in learning and memory. Behav Neurosci 2014; 128:283-303. [PMID: 24708297 PMCID: PMC4385793 DOI: 10.1037/a0035963] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The circadian system has pronounced influence on learning and memory, manifesting as marked changes in memory acquisition and recall across the day. From a mechanistic perspective, the majority of studies have investigated mammalian hippocampal-dependent learning and memory, as this system is highly tractable. The hippocampus plays a major role in learning and memory, and has the potential to integrate circadian information in many ways, including information from local, independent oscillators, and through circadian modulation of neurogenesis, synaptic remodeling, intracellular cascades, and epigenetic regulation of gene expression. These local processes are combined with input from other oscillatory systems to synergistically augment hippocampal rhythmic function. This overview presents an account of the current state of knowledge on circadian interactions with learning and memory circuitry and provides a framework for those interested in further exploring these interactions.
Collapse
Affiliation(s)
- Benjamin L. Smarr
- Department of Psychology, University of California, Berkeley, CA, 94720 USA
| | | | - Joseph R. Driscoll
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA
| | - Lance J. Kriegsfeld
- Department of Psychology, University of California, Berkeley, CA, 94720 USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720 USA
| |
Collapse
|
31
|
Chitty KM, Kaur M, Lagopoulos J, Hickie IB, Hermens DF. Risky alcohol use predicts temporal mismatch negativity impairments in young people with bipolar disorder. Biol Psychol 2014; 99:60-8. [PMID: 24594113 DOI: 10.1016/j.biopsycho.2014.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 01/22/2014] [Accepted: 02/23/2014] [Indexed: 01/08/2023]
Abstract
Alcohol misuse in bipolar disorder (BD) has a negative impact on illness progression. The NMDA/glutamatergic system is implicated in BD pathophysiology and is critically involved in the effects of alcohol on the brain. Mismatch negativity (MMN) is purported to reflect NMDA receptor output, providing a measure for investigating this association. Forty-two patients and 34 controls (16-30 years) were split into low and high-risk drinkers (based on the Alcohol Use Disorders Identification Test) and underwent a two-tone passive auditory oddball, duration deviant MMN paradigm. Multiple regression models revealed risky drinking and BD diagnosis were predictors of impaired temporal MMN. Potentially reflecting an additive effect of alcohol on a perturbed NMDA/glutamatergic system in BD, these findings highlight alcohol as both a modifiable risk factor of neurobiological impairments and as a potential confounder in MMN studies. Given the increasing use of glutamatergic agents for BD treatment, this finding is important clinically.
Collapse
Affiliation(s)
- Kate M Chitty
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia.
| | - Manreena Kaur
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Jim Lagopoulos
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Ian B Hickie
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| | - Daniel F Hermens
- Clinical Research Unit, Brain and Mind Research Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
32
|
Distinct and simultaneously active plasticity mechanisms in mouse hippocampus during different phases of Morris water maze training. Brain Struct Funct 2014; 220:1273-90. [PMID: 24562414 DOI: 10.1007/s00429-014-0722-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/28/2014] [Indexed: 01/30/2023]
Abstract
Although the Morris water maze (MWM) is the most frequently used protocol to examine hippocampus-dependent learning in mice, not much is known about the spatio-temporal dynamics of underlying plasticity processes. Here, we studied molecular and cellular hippocampal plasticity mechanisms during early and late phases of spatial learning in the MWM. Quantitative in situ hybridization for the immediate early genes zif268 and Homer1a (H1a) revealed phase-dependent differences in their expression between areas CA1 and CA3. During the initial learning phase, CA1 expression levels of the molecular plasticity marker H1a, but not of the activity reporter gene zif268, were related to task proficiency; whereas no learning-specific changes could be detected in CA3. Simultaneously, the ratio of surface-expressed NMDAR subunits NR2A and NR2B was downregulated as measured by acute slice biotinylation assay, while the total number of surface NMDARs was unaltered. When intrinsic 'somatic' and synaptic plasticity in the CA1-region of hippocampal slices were examined, we found that early learning promotes intrinsic neuronal plasticity as manifested by a reduction of spike frequency adaptation and postburst afterhyperpolarization. At the synaptic level, however, maintenance of long-term potentiation (LTP) in all learning groups was impaired which is most likely due to 'intrinsic' learning-induced LTP which occluded any further electrically induced LTP. Late learning, in contrast, was characterized by re-normalized H1a, NR2A and NR2B expression and neuronal firing, yet a further strengthening of learning-induced LTP. Together, our data support a precisely timed cascade of complex molecular and subcellular transformations occurring from early to late MWM learning.
Collapse
|
33
|
Headley DB, Paré D. In sync: gamma oscillations and emotional memory. Front Behav Neurosci 2013; 7:170. [PMID: 24319416 PMCID: PMC3836200 DOI: 10.3389/fnbeh.2013.00170] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/03/2013] [Indexed: 11/26/2022] Open
Abstract
Emotional experiences leave vivid memories that can last a lifetime. The emotional facilitation of memory has been attributed to the engagement of diffusely projecting neuromodulatory systems that enhance the consolidation of synaptic plasticity in regions activated by the experience. This process requires the propagation of signals between brain regions, and for those signals to induce long-lasting synaptic plasticity. Both of these demands are met by gamma oscillations, which reflect synchronous population activity on a fast timescale (35-120 Hz). Regions known to participate in the formation of emotional memories, such as the basolateral amygdala, also promote gamma-band activation throughout cortical and subcortical circuits. Recent studies have demonstrated that gamma oscillations are enhanced during emotional situations, coherent between regions engaged by salient stimuli, and predict subsequent memory for cues associated with aversive stimuli. Furthermore, neutral stimuli that come to predict emotional events develop enhanced gamma oscillations, reflecting altered processing in the brain, which may underpin how past emotional experiences color future learning and memory.
Collapse
Affiliation(s)
- Drew B. Headley
- Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New JerseyNewark, NJ, USA
| | | |
Collapse
|
34
|
Effects of CA1 glutamatergic systems upon memory impairments in cholestatic rats. Behav Brain Res 2013; 256:636-45. [PMID: 24050889 DOI: 10.1016/j.bbr.2013.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/05/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Bile duct ligation (BDL) is shown to induce cholestasis-related liver function impairments as well as consequent cognitive dysfunctions (i.e. impaired learning and memory formation). Glutamatergic neurotransmission plays an important role in hippocampal modulation of learning and memory function. The present study aimed to investigate the possible involvement of dorsal hippocampal (CA1) glutamatergic systems upon cholestasis-induced amnesia. METHOD Cholestasis was induced in male Wistar rats through double-ligation of the main bile duct (at two points) and transection of the interposed segment. Step-through passive avoidance test was employed to examine rats' learning and memory function. All drugs were injected into CA1 region of the hippocampus. RESULTS our results indicated a decrease in memory retrieval following cholestasis (11, 17 and 24 days post BDL). Only subthreshold doses of N-methyl-d-aspartate (NMDA; 0.125 and 0.25 μg/μl) but not its effective dose (0.5 μg/μl), restored the cholestasis-induced amnesia in step-through passive avoidance test, 11, 17 and 24 days post BDL. This effect was blocked by the subthreshold dose of D-[1]-2-amino-7-phosphonoheptanoic acid (D-AP7, NMDA receptor antagonist; 0.0625 μg/μl, intra-CA1) at 0.125 μg/μl and 0.25 μg/μl doses of NMDA. Moreover, our data revealed that only effective doses of D-AP7 (0.125 and 0.25 μg/μl, intra-CA1) potentiate memory impairments in 11 days after BDL. It was noted that none of applied drugs/doses exerted an effect on memory acquisition and locomotors activity, 10 and 12 days post laparotomy, respectively. CONCLUSION Our findings suggest the potential involvement of CA1 glutamatergic system(s) in cholestasis-induced memory deficits.
Collapse
|
35
|
Ballesteros KA, Sikorski A, Orfila JE, Martinez JL. Effects of inhaled anesthetic isoflurane on long-term potentiation of CA3 pyramidal cell afferents in vivo. Int J Gen Med 2012. [PMID: 23204857 PMCID: PMC3508563 DOI: 10.2147/ijgm.s30570] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Isoflurane is a preferred anesthetic, due to its properties that allow a precise concentration to be delivered continually during in vivo experimentation. The major mechanism of action of isoflurane is modulation of the γ-amino butyric acid (GABAA) receptor-chloride channel, mediating inhibitory synaptic transmission. Animal studies have shown that isoflurane does not cause cell death, but it does inhibit cell growth and causes long-term hippocampal learning deficits. As there are no studies characterizing the effects of isoflurane on electrophysiological aspects of long-term potentiation (LTP) in the hippocampus, it is important to determine whether isoflurane alters the characteristic responses of hippocampal afferents to cornu ammonis region 3 (CA3). We investigated the effects of isoflurane on adult male rats during in vivo induction of LTP, using the mossy fiber pathway, the lateral perforant pathway, the medial perforant pathway, and the commissural CA3 (cCA3) to CA3, with intracranial administration of Ringer’s solution, naloxone, RS-aminoindan-1, 5-dicarboxylic acid (AIDA), or 3-[(R)-2-carboxypiperazin-4-yl]-propo-2-enyl-1-phosphonic acid (CPP). Then, we compared these responses to published electrophysiological data, using sodium pentobarbital as an anesthetic, under similar experimental conditions. Our results showed that LTP was exhibited in animals anesthetized with isoflurane under vehicle conditions. With the exception of AIDA in the lateral perforant pathway, the defining characteristics of the four pathways appeared to remain intact, except for the observation that LTP was markedly reduced in animals anesthetized with isoflurane compared to those anesthetized with sodium pentobarbital. The results suggest that isoflurane may affect amplitude through activation of GABAA receptors or mechanisms important to LTP in CA3 afferent fibers.
Collapse
|
36
|
Malik R, Chattarji S. Enhanced intrinsic excitability and EPSP-spike coupling accompany enriched environment-induced facilitation of LTP in hippocampal CA1 pyramidal neurons. J Neurophysiol 2012; 107:1366-78. [DOI: 10.1152/jn.01009.2011] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Environmental enrichment (EE) is a well-established paradigm for studying naturally occurring changes in synaptic efficacy in the hippocampus that underlie experience-induced modulation of learning and memory in rodents. Earlier research on the effects of EE on hippocampal plasticity focused on long-term potentiation (LTP). Whereas many of these studies investigated changes in synaptic weight, little is known about potential contributions of neuronal excitability to EE-induced plasticity. Here, using whole-cell recordings in hippocampal slices, we address this gap by analyzing the impact of EE on both synaptic plasticity and intrinsic excitability of hippocampal CA1 pyramidal neurons. Consistent with earlier reports, EE increased contextual fear memory and dendritic spine density on CA1 cells. Furthermore, EE facilitated LTP at Schaffer collateral inputs to CA1 pyramidal neurons. Analysis of the underlying causes for enhanced LTP shows EE to increase the frequency but not amplitude of miniature excitatory postsynaptic currents. However, presynaptic release probability, assayed using paired-pulse ratios and use-dependent block of N-methyl-d-aspartate receptor currents, was not affected. Furthermore, CA1 neurons fired more action potentials (APs) in response to somatic depolarization, as well as during the induction of LTP. EE also reduced spiking threshold and after-hyperpolarization amplitude. Strikingly, this EE-induced increase in excitability caused the same-sized excitatory postsynaptic potential to fire more APs. Together, these findings suggest that EE may enhance the capacity for plasticity in CA1 neurons, not only by strengthening synapses but also by enhancing their efficacy to fire spikes—and the two combine to act as an effective substrate for amplifying LTP.
Collapse
Affiliation(s)
- Ruchi Malik
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
37
|
Network, cellular, and molecular mechanisms underlying long-term memory formation. Curr Top Behav Neurosci 2012; 15:73-115. [PMID: 22976275 DOI: 10.1007/7854_2012_229] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The neural network stores information through activity-dependent synaptic plasticity that occurs in populations of neurons. Persistent forms of synaptic plasticity may account for long-term memory storage, and the most salient forms are the changes in the structure of synapses. The theory proposes that encoding should use a sparse code and evidence suggests that this can be achieved through offline reactivation or by sparse initial recruitment of the network units. This idea implies that in some cases the neurons that underwent structural synaptic plasticity might be a subpopulation of those originally recruited; However, it is not yet clear whether all the neurons recruited during acquisition are the ones that underwent persistent forms of synaptic plasticity and responsible for memory retrieval. To determine which neural units underlie long-term memory storage, we need to characterize which are the persistent forms of synaptic plasticity occurring in these neural ensembles and the best hints so far are the molecular signals underlying structural modifications of the synapses. Structural synaptic plasticity can be achieved by the activity of various signal transduction pathways, including the NMDA-CaMKII and ACh-MAPK. These pathways converge with the Rho family of GTPases and the consequent ERK 1/2 activation, which regulates multiple cellular functions such as protein translation, protein trafficking, and gene transcription. The most detailed explanation may come from models that allow us to determine the contribution of each piece of this fascinating puzzle that is the neuron and the neural network.
Collapse
|
38
|
Wright DJ, Holmes PS, Smith D. Using the Movement-Related Cortical Potential to Study Motor Skill Learning. J Mot Behav 2011; 43:193-201. [DOI: 10.1080/00222895.2011.557751] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
39
|
Wright JW, Harding JW. Contributions of matrix metalloproteinases to neural plasticity, habituation, associative learning and drug addiction. Neural Plast 2010; 2009:579382. [PMID: 20169175 PMCID: PMC2821634 DOI: 10.1155/2009/579382] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 11/22/2009] [Accepted: 12/15/2009] [Indexed: 12/13/2022] Open
Abstract
The premise of this paper is that increased expression of matrix metalloproteinases (MMPs) permits the reconfiguration of synaptic connections (i.e., neural plasticity) by degrading cell adhesion molecules (CAMs) designed to provide stability to those extracellular matrix (ECM) proteins that form scaffolding supporting neurons and glia. It is presumed that while these ECM proteins are weakened, and/or detached, synaptic connections can form resulting in new neural pathways. Tissue inhibitors of metalloproteinases (TIMPs) are designed to deactivate MMPs permitting the reestablishment of CAMs, thus returning the system to a reasonably fixed state. This review considers available findings concerning the roles of MMPs and TIMPs in reorganizing ECM proteins thus facilitating the neural plasticity underlying long-term potentiation (LTP), habituation, and associative learning. We conclude with a consideration of the influence of these phenomena on drug addiction, given that these same processes may be instrumental in the formation of addiction and subsequent relapse. However, our knowledge concerning the precise spatial and temporal relationships among the mechanisms of neural plasticity, habituation, associative learning, and memory consolidation is far from complete and the possibility that these phenomena mediate drug addiction is a new direction of research.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | | |
Collapse
|
40
|
Muhia M, Yee BK, Feldon J, Markopoulos F, Knuesel I. Disruption of hippocampus-regulated behavioural and cognitive processes by heterozygous constitutive deletion of SynGAP. Eur J Neurosci 2010; 31:529-43. [PMID: 20105235 DOI: 10.1111/j.1460-9568.2010.07079.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The brain-specific Ras/Rap-GTPase activating protein (SynGAP) is a prime candidate linking N-methyl-d-aspartate receptors to the regulation of the ERK/MAP kinase signalling cascade, suggested to be essential for experience-dependent synaptic plasticity. Here, we evaluated the behavioural phenotype of SynGAP heterozygous knockout mice (SG(+/-)), expressing roughly half the normal levels of SynGAP. In the cognitive domain, SG(+/-) mice demonstrated severe working and reference memory deficits in the radial arm maze task, a mild impairment early in the transfer test of the water maze task, and a deficiency in spontaneous alternation in an elevated T-maze. In the non-cognitive domain, SG(+/-) mice were hyperactive in the open field and appeared less anxious in the elevated plus maze test. In contrast, object recognition memory performance was not impaired in SG(+/-) mice. The reduction in SynGAP thus resulted in multiple behavioural traits suggestive of aberrant cognitive and non-cognitive processes normally mediated by the hippocampus. Immunohistochemical evaluation further revealed a significant reduction in calbindin-positive interneurons in the hippocampus and doublecortin-positive neurons in the dentate gyrus of adult SG(+/-) mice. Heterozygous constitutive deletion of SynGAP is therefore associated with notable behavioural as well as morphological phenotypes indicative of hippocampal dysfunction. Any suggestion of a possible causal link between them however remains a matter for further investigation.
Collapse
Affiliation(s)
- Mary Muhia
- Laboratory of Behavioural Neurobiology, Swiss Federal Institute of Technology, Schwerzenbach, Switzerland
| | | | | | | | | |
Collapse
|
41
|
Tang J, Dani JA. Dopamine enables in vivo synaptic plasticity associated with the addictive drug nicotine. Neuron 2009; 63:673-82. [PMID: 19755109 DOI: 10.1016/j.neuron.2009.07.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 05/15/2009] [Accepted: 07/30/2009] [Indexed: 11/29/2022]
Abstract
Addictive drugs induce a dopamine signal that contributes to the initiation of addiction, and the dopamine signal influences drug-associated memories that perpetuate drug use. The addiction process shares many commonalities with the synaptic plasticity mechanisms normally attributed to learning and memory. Environmental stimuli repeatedly linked to addictive drugs become learned associations, and those stimuli come to elicit memories or sensations that motivate continued drug use. Applying in vivo recording techniques to freely moving mice, we show that physiologically relevant concentrations of the addictive drug nicotine directly cause in vivo hippocampal synaptic potentiation of the kind that underlies learning and memory. The drug-induced long-term synaptic plasticity required a local hippocampal dopamine signal. Disrupting general dopamine signaling prevented the nicotine-induced synaptic plasticity and conditioned place preference. These results suggest that dopaminergic signaling serves as a functional label of salient events by enabling and scaling synaptic plasticity that underlies drug-induced associative memory.
Collapse
Affiliation(s)
- Jianrong Tang
- Department of Neuroscience, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
42
|
Cao D, Kevala K, Kim J, Moon HS, Jun SB, Lovinger D, Kim HY. Docosahexaenoic acid promotes hippocampal neuronal development and synaptic function. J Neurochem 2009; 111:510-21. [PMID: 19682204 PMCID: PMC2773444 DOI: 10.1111/j.1471-4159.2009.06335.x] [Citation(s) in RCA: 269] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Docosahexaenoic acid (DHA, 22:6n-3), the major polyunsaturated fatty acid accumulated in the brain during development, has been implicated in learning and memory, but underlying cellular mechanisms are not clearly understood. Here, we demonstrate that DHA significantly affects hippocampal neuronal development and synaptic function in developing hippocampi. In embryonic neuronal cultures, DHA supplementation uniquely promoted neurite growth, synapsin puncta formation and synaptic protein expression, particularly synapsins and glutamate receptors. In DHA-supplemented neurons, spontaneous synaptic activity was significantly increased, mostly because of enhanced glutamatergic synaptic activity. Conversely, hippocampal neurons from DHA-depleted fetuses showed inhibited neurite growth and synaptogenesis. Furthermore, n-3 fatty acid deprivation during development resulted in marked decreases of synapsins and glutamate receptor subunits in the hippocampi of 18-day-old pups with concomitant impairment of long-term potentiation, a cellular mechanism underlying learning and memory. While levels of synapsins and NMDA receptor subunit NR2A were decreased in most hippocampal regions, NR2A expression was particularly reduced in CA3, suggesting possible role of DHA in CA3-NMDA receptor-dependent learning and memory processes. The DHA-induced neurite growth, synaptogenesis, synapsin, and glutamate receptor expression, and glutamatergic synaptic function may represent important cellular aspects supporting the hippocampus-related cognitive function improved by DHA.
Collapse
Affiliation(s)
- Dehua Cao
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - Karl Kevala
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - Jeffrey Kim
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - Hyun-Seuk Moon
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - Sang Beom Jun
- Laboratory of Integrative Neuroscience, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - David Lovinger
- Laboratory of Integrative Neuroscience, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, DICBR, NIAAA, NIH, Bethesda, Maryland, USA
| |
Collapse
|
43
|
The facilitating effect of systemic administration of Kv7/M channel blocker XE991 on LTP induction in the hippocampal CA1 area independent of muscarinic activation. Neurosci Lett 2009; 461:25-9. [DOI: 10.1016/j.neulet.2009.05.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 04/30/2009] [Accepted: 05/15/2009] [Indexed: 11/23/2022]
|
44
|
Memory modification as an outcome variable in anxiety disorder treatment. J Anxiety Disord 2009; 23:546-56. [PMID: 19117720 DOI: 10.1016/j.janxdis.2008.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 11/06/2008] [Accepted: 11/06/2008] [Indexed: 01/31/2023]
Abstract
Learning and memory are interdependent processes. Memories are learned, and cumulative learning requires memory. It is generally accepted that learning contributes to psychopathology and consequently to pertinent memory formation. Neuroscience and psychological research have established that memory is an active reconstructive process that is influenced by thoughts, feelings, and behaviors including post-event information. Recent research on the treatment of anxiety disorders using medications (i.e., d-cyclcloserine) to alter neurological systems associated with memory used in conjunction with behavior therapy suggests that memory is part of a central mechanism in the etiology and maintenance of these conditions. The main thesis of this article is that learning-based interventions create new memories that may modify existing ones. This raises the possibility of using such memory modifications to measure intervention outcome. A connectionist context for understanding this phenomenon and informing intervention is provided, with specific reference to post-traumatic stress disorder, obsessive-compulsive disorder, and generalized anxiety disorder. Recommendations for future research examining the role of memory change in treatment outcome are suggested.
Collapse
|
45
|
Smith AM, Chen WJA. Neonatal amphetamine exposure and hippocampus-mediated behaviors. Neurobiol Learn Mem 2009; 91:207-17. [PMID: 19146964 PMCID: PMC2692737 DOI: 10.1016/j.nlm.2008.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2008] [Revised: 12/01/2008] [Accepted: 12/03/2008] [Indexed: 12/12/2022]
Abstract
Previous studies linking amphetamine use during pregnancy to changes in the behavioral development of affected infants have greatly increased society's level of concern regarding amphetamine use by women of reproductive age. The aim of this study was to investigate whether exposure to d-amphetamine sulfate during the brain growth spurt, the most dynamic period of brain development, alters hippocampus-mediated behaviors during both pre-adolescence and young adulthood. Sprague-Dawley rat pups were intragastrically administered a milk formula containing 0, 5, 15 or 25 mg/kg/day of amphetamine from postnatal day (PD) 4-9. Following weaning, the effects of neonatal amphetamine exposure on hippocampus-mediated behaviors were assessed using the open-field, the water maze, and the conditioned taste aversion behavioral tasks. Results from these behavioral tests revealed that while amphetamine exposure during the brain growth spurt alters behaviors in open-field testing, it does not interfere with performance in either the water maze or the conditioned taste aversion paradigm. These results offer speculation that the effects of neonatal amphetamine exposure on hippocampus-mediated behaviors may be related to interactions between the "temporal" (time of drug exposure) and "regional" (different regions of the hippocampus) vulnerability issues.
Collapse
Affiliation(s)
- Andrew M Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, 233 Reynolds Medical Building, College Station, TX 77843-1114, United States
| | | |
Collapse
|
46
|
Speed HE, Dobrunz LE. Developmental changes in short-term facilitation are opposite at temporoammonic synapses compared to Schaffer collateral synapses onto CA1 pyramidal cells. Hippocampus 2009; 19:187-204. [PMID: 18777561 PMCID: PMC2631616 DOI: 10.1002/hipo.20496] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CA1 pyramidal neurons receive two distinct excitatory inputs that are each capable of influencing hippocampal output and learning and memory. The Schaffer collateral (SC) input from CA3 axons onto the more proximal dendrites of CA1 is part of the trisynaptic circuit, which originates in Layer II of the entorhinal cortex (EC). The temporoammonic (TA) pathway to CA1 provides input directly from Layer III of the EC onto the most distal dendrites of CA1 pyramidal cells, and is involved in spatial memory and memory consolidation. We have previously described a developmental decrease in short-term facilitation from juvenile (P13-18) to young adult (P28-42) rats at SC synapses that is due to feedback inhibition via synaptically activated mGluR1 on CA1 interneurons. It is not known how short-term changes in synaptic strength are regulated at TA synapses, nor is it known how short-term plasticity is balanced at SC and TA inputs during development. Here we describe a novel developmental increase in short-term facilitation at TA synapses, which is the opposite of the decrease in facilitation occurring at SC synapses. Although short-term facilitation is much lower at TA synapses when compared with SC synapses in juveniles, short-term plasticity at SC and TA synapses converges at similar levels of paired-pulse facilitation in the young adult rat. However, in young adults CA3-CA1 synapses still exhibit more facilitation than TA-CA1 synapses during physiologically-relevant activity, suggesting that the two pathways are each poised to uniquely modulate CA1 output in an activity-dependent manner. Finally, we show that there is a developmental decrease in the initial release probability at TA synapses that underlies their developmental decrease in facilitation, but no developmental change in release probability at SC synapses. This represents a fundamental difference in the presynaptic function of the two major inputs to CA1, which could alter the flow of information in hippocampus during development.
Collapse
Affiliation(s)
- Haley E Speed
- Department of Neurobiology, Civitan International Research Center and Evelyn F McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
47
|
Zellner MR, Kest K, Ranaldi R. NMDA receptor antagonism in the ventral tegmental area impairs acquisition of reward-related learning. Behav Brain Res 2008; 197:442-9. [PMID: 18983876 DOI: 10.1016/j.bbr.2008.10.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 07/24/2008] [Accepted: 10/08/2008] [Indexed: 11/29/2022]
Abstract
Mechanisms underlying reward-related learning presumably involve neural plasticity integrating signals representing unconditioned and conditioned stimuli in regions mediating reward. The ventral tegmental area (VTA) receives such signals and shows synaptic plasticity which is NMDA receptor-dependent. To test the hypothesis that NMDA receptor stimulation in the VTA is necessary for the acquisition of food-reinforced appetitive learning, Long-Evans male rats were prepared with bilateral VTA cannulae and tested in operant chambers with the opportunity to lever press for food for 10 sessions. Animals received microinjections of AP-5 or vehicle immediately before sessions 1-4 and 10. AP-5 impaired acquisition of lever pressing during sessions 1-4 (but not when injected dorsal to the VTA). All groups increased lever pressing across sessions 5-9. On session 10, lever pressing was not affected regardless of treatment. In separate experiments, AP-5 failed to reduce free feeding, food reward or motor activity, suggesting that impairment in acquisition was not due to reduced food motivation or activity. NMDA transmission in the VTA thus appears to be necessary for the acquisition, but not expression, of reward-related learning.
Collapse
|
48
|
Calabrese EJ. Alzheimer's disease drugs: an application of the hormetic dose-response model. Crit Rev Toxicol 2008; 38:419-51. [PMID: 18568864 DOI: 10.1080/10408440802003991] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This article provides an evaluation of the dose-response features of drugs that are intended to improve memory, some of which have been used in the treatment of Alzheimer's disease (AD). A common feature of these drugs is that they act via an inverted U-shaped dose response, consistent with the hormetic dose response model. This article assesses historical foundations that lead to the development of AD drugs, their dose-response features and how the quantitative features of such dose responses affected drug discovery and development, and the successes and possible failures of such agents in preclinical and clinical settings. This story begins about 150 years ago with the discovery of an active agent in the Calabar bean plant called physostigmine, its unfolding medical applications, and its implications for dose-response relationships, memory enhancement, and improved drug discovery activities. The article also demonstrates the occurrence of U-shaped dose responses for memory with numerous endogenous agonists including neurosteroids, various peptides (e.g., vasopressin, CCK-8, neuropeptide Y), and other agents (e.g., epinephrine, antagonists for platelet activity factor and nicotinic receptors), supporting the generalizability of the hormetic biphasic dose response. Finally, the significance of the U-shaped dose response is critical for successful clinical application, since it defines the therapeutic window.
Collapse
Affiliation(s)
- Edward J Calabrese
- Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| |
Collapse
|
49
|
Zavala AR, Osredkar T, Joyce JN, Neisewander JL. Upregulation of Arc mRNA expression in the prefrontal cortex following cue-induced reinstatement of extinguished cocaine-seeking behavior. Synapse 2008; 62:421-31. [PMID: 18361437 PMCID: PMC2832122 DOI: 10.1002/syn.20502] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cocaine-associated cues acquire incentive motivational effects that manifest as cue-elicited craving in humans and cocaine-seeking behavior in rats. Here we examine the hypothesis that neuronal processes associated with incentive motivational effects of cocaine cues involve increased expression of the plasticity-associated gene, Arc. Rats trained to self-administer cocaine subsequently underwent extinction training, during which cocaine-seeking behavior (i.e., responses without cocaine reinforcement) progressively decreased. Rats were then tested for cocaine-seeking behavior either with or without response-contingent presentations of light/tone cues that had been previously paired with cocaine infusions during self-administration training. Cues elicited reinstatement of cocaine-seeking behavior and were accompanied by increased Arc mRNA levels in the orbitofrontal, prelimbic, and anterior cingulate cortices, suggesting Arc involvement in conditioned plasticity associated with incentive motivational effects of cocaine cues. Additionally, rats with a history of cocaine self-administration and extinction exhibited upregulation of Arc expression in several limbic and cortical regions relative to saline-yoked controls regardless of cue exposure condition, suggesting persistent neuroadaptations involving Arc within these regions.
Collapse
Affiliation(s)
- Arturo R Zavala
- Department of Psychology, Arizona State University, Tempe, Arizona 85287-1104, USA
| | | | | | | |
Collapse
|
50
|
Gilbert ME, Sui L. Developmental exposure to perchlorate alters synaptic transmission in hippocampus of the adult rat. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:752-60. [PMID: 18560531 PMCID: PMC2430231 DOI: 10.1289/ehp.11089] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Accepted: 03/05/2008] [Indexed: 05/07/2023]
Abstract
BACKGROUND Perchlorate is an environmental contaminant that blocks iodine uptake into the thyroid gland and reduces thyroid hormones. This action of perchlorate raises significant concern over its effects on brain development. OBJECTIVES The purpose of this study was to evaluate neurologic function in rats after developmental exposure to perchlorate. METHODS Pregnant rats were exposed to 0, 30, 300, or 1,000 ppm perchlorate in drinking water from gestational day 6 until weaning. Adult male offspring were evaluated on a series of behavioral tasks and neurophysiologic measures of synaptic function in the hippocampus. RESULTS At the highest perchlorate dose, triiodothyronine (T(3)) and thyroxine (T(4)) were reduced in pups on postnatal day 21. T(4) in dams was reduced relative to controls by 16%, 28%, and 60% in the 30-, 300-, and 1,000-ppm dose groups, respectively. Reductions in T(4) were associated with increases in thyroid-stimulating hormone in the high-dose group. No changes were seen in serum T(3). Perchlorate did not impair motor activity, spatial learning, or fear conditioning. However, significant reductions in baseline synaptic transmission were observed in hippocampal field potentials at all dose levels. Reductions in inhibitory function were evident at 300 and 1,000 ppm, and augmentations in long-term potentiation were observed in the population spike measure at the highest dose. CONCLUSIONS Dose-dependent deficits in hippocampal synaptic function were detectable with relatively minor perturbations of the thyroid axis, indicative of an irreversible impairment in synaptic transmission in response to developmental exposure to perchlorate.
Collapse
Affiliation(s)
- Mary E Gilbert
- Neurotoxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA.
| | | |
Collapse
|