1
|
Stephenson KA, Peters P, Rae MG, O'Malley D. Astrocyte proliferation in the hippocampal dentate gyrus is suppressed across the lifespan of dystrophin-deficient mdx mice. Exp Physiol 2025. [PMID: 39792584 DOI: 10.1113/ep092150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
Absence of the structural protein, dystrophin, results in the neuromuscular disorder Duchenne Muscular Dystrophy (DMD). In addition to progressive skeletal muscle dysfunction, this multisystemic disorder can also result in cognitive deficits and behavioural changes that are likely to be consequences of dystrophin loss from central neurons and astrocytes. Dystrophin-deficient mdx mice exhibit decreases in grey matter volume in the hippocampus, the brain region that encodes and consolidates memories, and this is exacerbated with ageing. To understand changes in cellular composition that might underpin these age-related developments, we have compared neurogenesis and the prevalence of immunofluorescently identified newly born and mature neurons, astrocytes and microglia in the dentate gyrus of mdx and wild-type mice at 2, 4, 8 and 16 months of age. The number of adult-born neurons was suppressed in the dentate gyrus subgranular zone of 2-month-old mdx mice. However, the numbers of granule cells and GABAA receptor, alpha 1-expressing cells were similar in wild-type and mdx mice at all ages. Strikingly, the numbers of astrocytes, particularly in the dentate gyrus molecular layer, were suppressed in mdx mice at all time points. Thus, dystrophin loss was associated with reduced hippocampal neurogenesis in early life but did not impact the prevalence of mature neurons across the lifespan of mdx mice. In contrast, normal age-related dentate gyrus astrocyte proliferation was suppressed in dystrophic mice. Astrocytes are the most abundant cell type in the brain and are crucial in supporting neuronal function, such that loss of these cells is likely to contribute to hippocampal dysfunction reported in mdx mice.
Collapse
Affiliation(s)
| | - Polly Peters
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - Mark G Rae
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Lei B, Kang B, Hao Y, Yang H, Zhong Z, Zhai Z, Zhong Y. Reconstructing a new hippocampal engram for systems reconsolidation and remote memory updating. Neuron 2024:S0896-6273(24)00835-3. [PMID: 39689709 DOI: 10.1016/j.neuron.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/23/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Recalling systems-consolidated neocortex-dependent remote memories re-engages the hippocampus in a process called systems reconsolidation. However, underlying mechanisms, particularly for the origin of the reinstated hippocampal memory engram, remain elusive. By developing a triple-event labeling tool and employing two-photon imaging, we trace hippocampal engram ensembles from memory acquisition to systems reconsolidation and find that remote recall recruits a new engram ensemble in the hippocampus for subsequent memory retrieval. Consistently, recruiting new engrams is supported by adult hippocampal neurogenesis-mediated silencing of original engrams. This new engram ensemble receives currently experienced contextual information, incorporates new information into the remote memory, and supports remote memory updating. Such a reconstructed hippocampal memory is then integrated with the valence of remote memory via medial prefrontal cortex projection-mediated activity coordination between the hippocampus and amygdala. Thus, the reconstruction of new memory engrams underlies systems reconsolidation, which explains how remote memories are updated with new information.
Collapse
Affiliation(s)
- Bo Lei
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Beijing Academy of Artificial Intelligence, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China.
| | - Bilin Kang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China
| | - Yuejun Hao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China
| | - Haoyu Yang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China
| | - Zihan Zhong
- Eight-Year Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P.R. China
| | - Zihan Zhai
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; IDG/McGovern Institute of Brain Research, Tsinghua University, Beijing 100084, P.R. China.
| |
Collapse
|
3
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially resolved transcriptomic signatures of hippocampal subregions and Arc-expressing ensembles in active place avoidance memory. Front Mol Neurosci 2024; 17:1386239. [PMID: 39544521 PMCID: PMC11560897 DOI: 10.3389/fnmol.2024.1386239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/20/2024] [Indexed: 11/17/2024] Open
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc + and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc + and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Shwetha Phatarpekar
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Victoria Sook Keng Tung
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Alejandro Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| | - Oleg V. Evgrafov
- Institute for Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- School of Graduate Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, United States
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, United States
| |
Collapse
|
4
|
Herpich ME, de Oliveira Guarnieri L, de Oliveira ACP, Moraes MFD. Bacterial Lipopolysaccharide Post-Conditioning in The kainic acid animal model of Temporal Lobe epilepsy. Epilepsy Behav 2024; 161:110076. [PMID: 39467457 DOI: 10.1016/j.yebeh.2024.110076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
This study used intra-hippocampal injections of Kainic Acid (KA) in Wistar rats to induce spontaneous recurrent seizures (SRS) after a 9-day latent period. A post-conditioning protocol with LPS, injected at the same site 72 h after the initial KA insult, was employed to trigger secondary competing processes. To evaluate the post-conditioning effect of LPS, 25 animals were divided into four groups: SAL-SAL (n = 6), KA-SAL (n = 6), SAL-LPS (n = 7), and KA-LPS (n = 6). SRS occurrence and seizure duration were quantified through video monitoring from days 9 to 17, along with other ictal behaviors, such as tail-chasing and wet-dog-shakes. Behavioral assessments revealed that the KA-LPS group had preserved sucrose preference and intact long-term memory in the object recognition test, indicating reduced depressive-like behavior and cognitive preservation compared to the KA-SAL group. The forced swim test showed increased depressive-like behavior in the SAL-LPS group, with LPS mitigating these effects in the KA group. The marble-burying test showed no significant differences among groups. Animals were euthanized on day 26, and hippocampal slices were analyzed using fluoro-jade staining for cell death and immunofluorescence staining for Iba-1 (microglia) and GFAP (astrocyte) labeling. The results support the hypothesis that epileptogenesis involves a cascade of plastic changes in neural networks and that precise, timely interventions can potentially interfere with this process.
Collapse
Affiliation(s)
- Mateus Eduardo Herpich
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo de Oliveira Guarnieri
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas - Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
5
|
Hernández-Frausto M, Vivar C. Entorhinal cortex-hippocampal circuit connectivity in health and disease. Front Hum Neurosci 2024; 18:1448791. [PMID: 39372192 PMCID: PMC11449717 DOI: 10.3389/fnhum.2024.1448791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
The entorhinal cortex (EC) and hippocampal (HC) connectivity is the main source of episodic memory formation and consolidation. The entorhinal-hippocampal (EC-HC) connection is classified as canonically glutamatergic and, more recently, has been characterized as a non-canonical GABAergic connection. Recent evidence shows that both EC and HC receive inputs from dopaminergic, cholinergic, and noradrenergic projections that modulate the mnemonic processes linked to the encoding and consolidation of memories. In the present review, we address the latest findings on the EC-HC connectivity and the role of neuromodulations during the mnemonic mechanisms of encoding and consolidation of memories and highlight the value of the cross-species approach to unravel the underlying cellular mechanisms known. Furthermore, we discuss how EC-HC connectivity early neurodegeneration may contribute to the dysfunction of episodic memories observed in aging and Alzheimer's disease (AD). Finally, we described how exercise may be a fundamental tool to prevent or decrease neurodegeneration.
Collapse
Affiliation(s)
- Melissa Hernández-Frausto
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York University Langone Medical Center, New York, NY, United States
| | - Carmen Vivar
- Laboratory of Neurogenesis and Neuroplasticity, Department of Physiology, Biophysics and Neuroscience, Centro de Investigacion y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
6
|
Berry JA, Guhle DC, Davis RL. Active forgetting and neuropsychiatric diseases. Mol Psychiatry 2024; 29:2810-2820. [PMID: 38532011 PMCID: PMC11420092 DOI: 10.1038/s41380-024-02521-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
Recent and pioneering animal research has revealed the brain utilizes a variety of molecular, cellular, and network-level mechanisms used to forget memories in a process referred to as "active forgetting". Active forgetting increases behavioral flexibility and removes irrelevant information. Individuals with impaired active forgetting mechanisms can experience intrusive memories, distressing thoughts, and unwanted impulses that occur in neuropsychiatric diseases. The current evidence indicates that active forgetting mechanisms degrade, or mask, molecular and cellular memory traces created in synaptic connections of "engram cells" that are specific for a given memory. Combined molecular genetic/behavioral studies using Drosophila have uncovered a complex system of cellular active-forgetting pathways within engram cells that is regulated by dopamine neurons and involves dopamine-nitric oxide co-transmission and reception, endoplasmic reticulum Ca2+ signaling, and cytoskeletal remodeling machinery regulated by small GTPases. Some of these molecular cellular mechanisms have already been found to be conserved in mammals. Interestingly, some pathways independently regulate forgetting of distinct memory types and temporal phases, suggesting a multi-layering organization of forgetting systems. In mammals, active forgetting also involves modulation of memory trace synaptic strength by altering AMPA receptor trafficking. Furthermore, active-forgetting employs network level mechanisms wherein non-engram neurons, newly born-engram neurons, and glial cells regulate engram synapses in a state and experience dependent manner. Remarkably, there is evidence for potential coordination between the network and cellular level forgetting mechanisms. Finally, subjects with several neuropsychiatric diseases have been tested and shown to be impaired in active forgetting. Insights obtained from research on active forgetting in animal models will continue to enrich our understanding of the brain dysfunctions that occur in neuropsychiatric diseases.
Collapse
Affiliation(s)
- Jacob A Berry
- Department of Biological Sciences, University of Alberta, Edmonton, AL, T6G 2E9, Canada.
| | - Dana C Guhle
- Department of Biological Sciences, University of Alberta, Edmonton, AL, T6G 2E9, Canada
| | - Ronald L Davis
- Department of Neuroscience, UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA.
| |
Collapse
|
7
|
Caruso MG, Nicolas S, Lucassen PJ, Mul JD, O’Leary OF, Nolan YM. Ageing, Cognitive Decline, and Effects of Physical Exercise: Complexities, and Considerations from Animal Models. Brain Plast 2024; 9:43-73. [PMID: 38993577 PMCID: PMC11234681 DOI: 10.3233/bpl-230157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 07/13/2024] Open
Abstract
In our ageing global population, the cognitive decline associated with dementia and neurodegenerative diseases represents a major healthcare problem. To date, there are no effective treatments for age-related cognitive impairment, thus preventative strategies are urgently required. Physical exercise is gaining traction as a non-pharmacological approach to promote brain health. Adult hippocampal neurogenesis (AHN), a unique form of brain plasticity which is necessary for certain cognitive functions declines with age and is enhanced in response to exercise. Accumulating evidence from research in rodents suggests that physical exercise has beneficial effects on cognition through its proneurogenic capabilities. Given ethical and technical limitations in human studies, preclinical research in rodents is crucial for a better understanding of such exercise-induced brain and behavioural changes. In this review, exercise paradigms used in preclinical research are compared. We provide an overview of the effects of different exercise paradigms on age-related cognitive decline from middle-age until older-age. We discuss the relationship between the age-related decrease in AHN and the potential impact of exercise on mitigating this decline. We highlight the emerging literature on the impact of exercise on gut microbiota during ageing and consider the role of the gut-brain axis as a future possible strategy to optimize exercise-enhanced cognitive function. Finally, we propose a guideline for designing optimal exercise protocols in rodent studies, which would inform clinical research and contribute to developing preventative strategies for age-related cognitive decline.
Collapse
Affiliation(s)
- Maria Giovanna Caruso
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Sarah Nicolas
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Paul J. Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Joram D. Mul
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
- Center for Urban Mental Health, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivia F. O’Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| | - Yvonne M. Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
8
|
Ma J, Cao H, Hou D, Wang W, Liu T. Investigation of high-dose radiotherapy's effect on brain structure aggravated cognitive impairment and deteriorated patient psychological status in brain tumor treatment. Sci Rep 2024; 14:10149. [PMID: 38698048 PMCID: PMC11066031 DOI: 10.1038/s41598-024-59694-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 04/15/2024] [Indexed: 05/05/2024] Open
Abstract
This study aims to investigate the potential impact of high-dose radiotherapy (RT) on brain structure, cognitive impairment, and the psychological status of patients undergoing brain tumor treatment. We recruited and grouped 144 RT-treated patients with brain tumors into the Low dose group (N = 72) and the High dose group (N = 72) according to the RT dose applied. Patient data were collected by using the HADS and QLQ-BN20 system for subsequent analysis and comparison. Our analysis showed no significant correlation between the RT doses and the clinicopathological characteristics. We found that a high dose of RT could aggravate cognitive impairment and deteriorate patient role functioning, indicated by a higher MMSE and worsened role functioning in the High dose group. However, the depression status, social functioning, and global health status were comparable between the High dose group and the Low dose group at Month 0 and Month 1, while being worsened in the High dose group at Month 3, indicating the potential long-term deterioration of depression status in brain tumor patients induced by high-dose RT. By comparing patient data at Month 0, Month 1, Month 3, Month 6, and Month 9 after RT, we found that during RT treatment, RT at a high dose could aggravate cognitive impairment in the short term and lead to worsened patient role functioning, and even deteriorate the overall psychological health status of patients in the long term.
Collapse
Affiliation(s)
- Jianpeng Ma
- Department of Magnetic Resonance Imaging, Dingbian County People's Hospital, Dingbian, Yulin, 718600, Shaanxi, China
| | - Hetao Cao
- Department of Medical Imaging, Affiliated Hospital of Nantong University, No.20 Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Dongmei Hou
- Department of Medical Imaging, Affiliated Hospital of Nantong University, No.20 Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China
| | - Weiqi Wang
- School of Pharmacy, Nantong University, Nantong, 226019, Jiangsu, China
| | - Tingting Liu
- Department of Medical Imaging, Affiliated Hospital of Nantong University, No.20 Xisi Road, Chongchuan District, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Peternel M, Jenko A, Peterlin P, Petrovič L, Strojan P, Plavc G. Comparison of conventional and hippocampus-sparing radiotherapy in nasopharyngeal carcinoma: In silico study and systematic review. Clin Transl Radiat Oncol 2024; 46:100751. [PMID: 38425692 PMCID: PMC10900111 DOI: 10.1016/j.ctro.2024.100751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
Background and purpose Radiation-induced damage to the hippocampi can cause cognitive decline. International recommendations for nasopharyngeal cancer (NPC) radiotherapy (RT) lack specific guidelines for protecting the hippocampi. Our study evaluates if hippocampi-sparing (HS) RT in NPC ensures target coverage and meets recommended dose limits for other at-risk organs. Materials and methods In a systematic literature review, we compared hippocampal D40% in conventional and HS RT plans. In an in silico dosimetric study, conventional and HS-VMAT plans were created for each patient, following international recommendations for OAR delineation, dose prioritization and acceptance criteria. We assessed the impact on neurocognitive function using a previously published normal tissue complication probability (NTCP) model. Results In four previous studies (n = 79), researchers reduced D40% hippocampal radiation doses in HS plans compared to conventional RT on average from 24.9 Gy to 12.6 Gy.Among 12 NPC patients included in this in silico study, statistically significant differences between HS and conventional VMAT plans were observed in hippocampal EQD2 Dmax (23.8 vs. 46.4 Gy), Dmin (3.8 vs. 4.6 Gy), Dmean (8.1 vs. 15.1 Gy), and D40% (8.3 vs. 15.8 Gy). PTV coverage and OAR doses were similar, with less homogeneous PTV coverage in HS plans (p = 0.038). This translated to a lower probability of memory decline in HS plans (interquartile range 15.8-29.6 %) compared to conventional plans (33.8-81.1 %) based on the NTCP model (p = 0.002). Conclusion Sparing the hippocampus in NPC RT is safe and feasible. Given the life expectancy of many NPC patients, their cognitive well-being must be paramount in radiotherapy planning.
Collapse
Affiliation(s)
- Monika Peternel
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
| | - Aljaša Jenko
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
| | - Primož Peterlin
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
| | - Larisa Petrovič
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
| | - Primož Strojan
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Gaber Plavc
- Institute of Oncology, Department of Radiotherapy, Zaloška cesta 2, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| |
Collapse
|
10
|
Dey J, Chandra S, Gupta J, Tripathi PP. Hippocampal neurodegeneration induces transient endogenous regeneration and long-term exhaustion of the neurogenic niche. J Cell Physiol 2024; 239:e31249. [PMID: 38501376 DOI: 10.1002/jcp.31249] [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: 09/17/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
The hippocampal dentate gyrus, responds to diverse pathological stimuli through neurogenesis. This phenomenon, observed following brain injury or neurodegeneration, is postulated to contribute to neuronal repair and functional recovery, thereby presenting an avenue for endogenous neuronal restoration. This study investigated the extent of regenerative response in hippocampal neurogenesis by leveraging the well-established kainic acid-induced status epilepticus model in vivo. In our study, we observed the activation and proliferation of neuronal progenitors or neural stem cell (NSC) and their subsequent migration to the injury sites following the seizure. At the injury sites, new neurons (Tuj1+BrdU+ and NeuN+BrdU+) have been generated indicating regenerative and reparative roles of the progenitor cells. We further detected whether this transient neurogenic burst, which might be a response towards an attempt to repair the brain, is associated with persistent long-term exhaustion of the dentate progenitor cells and impairment of adult neurogenesis marked by downregulation of Ki67, HoPX, and Sox2 with BrdU+ cell in the later part of life. Our studies suggest that the adult brain has the constitutive endogenous regenerative potential for brain repair to restore the damaged neurons, meanwhile, in the long term, it accelerates the depletion of the finite NSC pool in the hippocampal neurogenic niche by changing its proliferative and neurogenic capacity. A thorough understanding of the impact of modulating adult neurogenesis will eventually be required to design novel therapeutics to stimulate or assist brain repair while simultaneously preventing the adverse effects of early robust neurogenesis on the proliferative potential of endogenous neuronal progenitors.
Collapse
Affiliation(s)
- Jhilik Dey
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sreyashi Chandra
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jalaj Gupta
- Stem Cell Research Centre, Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, India
| | - Prem Prakash Tripathi
- Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
11
|
Lopez MR, Wasberg SMH, Gagliardi CM, Normandin ME, Muzzio IA. Mystery of the memory engram: History, current knowledge, and unanswered questions. Neurosci Biobehav Rev 2024; 159:105574. [PMID: 38331127 DOI: 10.1016/j.neubiorev.2024.105574] [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: 09/18/2023] [Revised: 12/22/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024]
Abstract
The quest to understand the memory engram has intrigued humans for centuries. Recent technological advances, including genetic labelling, imaging, optogenetic and chemogenetic techniques, have propelled the field of memory research forward. These tools have enabled researchers to create and erase memory components. While these innovative techniques have yielded invaluable insights, they often focus on specific elements of the memory trace. Genetic labelling may rely on a particular immediate early gene as a marker of activity, optogenetics may activate or inhibit one specific type of neuron, and imaging may capture activity snapshots in a given brain region at specific times. Yet, memories are multifaceted, involving diverse arrays of neuronal subpopulations, circuits, and regions that work in concert to create, store, and retrieve information. Consideration of contributions of both excitatory and inhibitory neurons, micro and macro circuits across brain regions, the dynamic nature of active ensembles, and representational drift is crucial for a comprehensive understanding of the complex nature of memory.
Collapse
Affiliation(s)
- M R Lopez
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - S M H Wasberg
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - C M Gagliardi
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - M E Normandin
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - I A Muzzio
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
12
|
Naffaa MM. Significance of the anterior cingulate cortex in neurogenesis plasticity: Connections, functions, and disorders across postnatal and adult stages. Bioessays 2024; 46:e2300160. [PMID: 38135889 DOI: 10.1002/bies.202300160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
The anterior cingulate cortex (ACC) is a complex and continually evolving brain region that remains a primary focus of research due to its multifaceted functions. Various studies and analyses have significantly advanced our understanding of how the ACC participates in a wide spectrum of memory and cognitive processes. However, despite its strong connections to brain areas associated with hippocampal and olfactory neurogenesis, the functions of the ACC in regulating postnatal and adult neurogenesis in these regions are still insufficiently explored. Investigating the intricate involvement of the ACC in neurogenesis could enhance our comprehension of essential aspects of brain plasticity. This involvement stems from its complex circuitry with other relevant brain regions, thereby exerting both direct and indirect impacts on the neurogenesis process. This review sheds light on the promising significance of the ACC in orchestrating postnatal and adult neurogenesis in conditions related to memory, cognitive behavior, and associated disorders.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
13
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially Resolved Transcriptomic Signatures of Hippocampal Subregions and Arc-Expressing Ensembles in Active Place Avoidance Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573225. [PMID: 38260257 PMCID: PMC10802250 DOI: 10.1101/2023.12.30.573225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc+ and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc+ and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Shwetha Phatarpekar
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Victoria Sook Keng Tung
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - A. Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Oleg V. Evgrafov
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
14
|
Yokose J, Marks WD, Kitamura T. Visuotactile integration facilitates mirror-induced self-directed behavior through activation of hippocampal neuronal ensembles in mice. Neuron 2024; 112:306-318.e8. [PMID: 38056456 DOI: 10.1016/j.neuron.2023.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 08/28/2023] [Accepted: 10/17/2023] [Indexed: 12/08/2023]
Abstract
Remembering the visual features of oneself is critical for self-recognition. However, the neural mechanisms of how the visual self-image is developed remain unknown because of the limited availability of behavioral paradigms in experimental animals. Here, we demonstrate a mirror-induced self-directed behavior (MSB) in mice, resembling visual self-recognition. Mice displayed increased mark-directed grooming to remove ink placed on their heads when an ink-induced visual-tactile stimulus contingency occurred. MSB required mirror habituation and social experience. The chemogenetic inhibition of dorsal or ventral hippocampal CA1 (vCA1) neurons attenuated MSB. Especially, a subset of vCA1 neurons activated during the mirror exposure was significantly reactivated during re-exposure to the mirror and was necessary for MSB. The self-responding vCA1 neurons were also reactivated when mice were exposed to a conspecific of the same strain. These results suggest that visual self-image may be developed through social experience and mirror habituation and stored in a subset of vCA1 neurons.
Collapse
Affiliation(s)
- Jun Yokose
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - William D Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Martínez-Canabal A, López-Oropeza G, Sotres-Bayón F. Hippocampal neurogenesis facilitates cognitive flexibility in a fear discrimination task. Front Behav Neurosci 2024; 17:1331928. [PMID: 38282713 PMCID: PMC10813213 DOI: 10.3389/fnbeh.2023.1331928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 01/30/2024] Open
Abstract
Hippocampal neurogenesis, the continuous creation of new neurons in the adult brain, influences memory, regulates the expression of defensive responses to threat (fear), and cognitive processes like pattern separation and behavioral flexibility. One hypothesis proposes that neurogenesis promotes cognitive flexibility by degrading established memories and promoting relearning. Yet, empirical evidence on its role in fear discrimination tasks is scarce. In this study, male rats were initially trained to differentiate between two similar environments, one associated with a threat. Subsequently, we enhanced neurogenesis through environmental enrichment and memantine treatments. We then reversed the emotional valence of these contexts. In both cases, neurogenesis improved the rats' ability to relearn the aversive context. Interestingly, we observed increased hippocampal activity, and decreased activity in the prelimbic cortex and lateral habenula, while the infralimbic cortex remained unchanged, suggesting neurogenesis-induced plasticity changes in this brain network. Moreover, when we pharmacologically inhibited the increased neurogenesis with Methotrexate, rats struggled to relearn context discrimination, confirming the crucial role of neurogenesis in this cognitive process. Overall, our findings highlight neurogenesis's capacity to facilitate changes in fear discrimination and emphasize the involvement of a prefrontal-hippocampal-habenula mechanism in this process. This study emphasizes the intricate relationship between hippocampal neurogenesis, cognitive flexibility, and the modulation of fear-related memories.
Collapse
Affiliation(s)
- Alonso Martínez-Canabal
- Department of Cell Biology, Faculty of Sciences, National Autonomous University of Mexico (UNAM), México City, Mexico
- Cell Physiology Institute - Neuroscience, National Autonomous University of Mexico (UNAM), México City, Mexico
| | - Grecia López-Oropeza
- Department of Cell Biology, Faculty of Sciences, National Autonomous University of Mexico (UNAM), México City, Mexico
- Cell Physiology Institute - Neuroscience, National Autonomous University of Mexico (UNAM), México City, Mexico
- Graduate Program in Biological Sciences, National Autonomous University of Mexico (UNAM), México City, Mexico
| | - Francisco Sotres-Bayón
- Cell Physiology Institute - Neuroscience, National Autonomous University of Mexico (UNAM), México City, Mexico
| |
Collapse
|
16
|
Kitamura T, Ramesh K, Terranova JI. Understanding Others' Distress Through Past Experiences: The Role of Memory Engram Cells in Observational Fear. ADVANCES IN NEUROBIOLOGY 2024; 38:215-234. [PMID: 39008018 DOI: 10.1007/978-3-031-62983-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
For individuals to survive and function in society, it is essential that they recognize, interact with, and learn from other conspecifics. Observational fear (OF) is the well-conserved empathic ability of individuals to understand the other's aversive situation. While it is widely known that factors such as prior similar aversive experience and social familiarity with the demonstrator facilitate OF, the neural circuit mechanisms that explicitly regulate experience-dependent OF (Exp OF) were unclear. In this review, we examine the neural circuit mechanisms that regulate OF, with an emphasis on rodent models, and then discuss emerging evidence for the role of fear memory engram cells in the regulation of Exp OF. First, we examine the neural circuit mechanisms that underlie Naive OF, which is when an observer lacks prior experiences relevant to OF. In particular, the anterior cingulate cortex to basolateral amygdala (BLA) neural circuit is essential for Naive OF. Next, we discuss a recent study that developed a behavioral paradigm in mice to examine the neural circuit mechanisms that underlie Exp OF. This study found that fear memory engram cells in the BLA of observers, which form during a prior similar aversive experience with shock, are reactivated by ventral hippocampal neurons in response to shock delivery to the familiar demonstrator to elicit Exp OF. Finally, we discuss the implications of fear memory engram cells in Exp OF and directions of future research that are of both translational and basic interest.
Collapse
Affiliation(s)
- Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Kritika Ramesh
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
17
|
Chang WL, Hen R. Adult Neurogenesis, Context Encoding, and Pattern Separation: A Pathway for Treating Overgeneralization. ADVANCES IN NEUROBIOLOGY 2024; 38:163-193. [PMID: 39008016 DOI: 10.1007/978-3-031-62983-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In mammals, the subgranular zone of the dentate gyrus is one of two brain regions (with the subventricular zone of the olfactory bulb) that continues to generate new neurons throughout adulthood, a phenomenon known as adult hippocampal neurogenesis (AHN) (Eriksson et al., Nat Med 4:1313-1317, 1998; García-Verdugo et al., J Neurobiol 36:234-248, 1998). The integration of these new neurons into the dentate gyrus (DG) has implications for memory encoding, with unique firing and wiring properties of immature neurons that affect how the hippocampal network encodes and stores attributes of memory. In this chapter, we will describe the process of AHN and properties of adult-born cells as they integrate into the hippocampal circuit and mature. Then, we will discuss some methodological considerations before we review evidence for the role of AHN in two major processes supporting memory that are performed by the DG. First, we will discuss encoding of contextual information for episodic memories and how this is facilitated by AHN. Second, will discuss pattern separation, a major role of the DG that reduces interference for the formation of new memories. Finally, we will review clinical and translational considerations, suggesting that stimulation of AHN may help decrease overgeneralization-a common endophenotype of mood, anxiety, trauma-related, and age-related disorders.
Collapse
Affiliation(s)
- Wei-Li Chang
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA
| | - Rene Hen
- Departments of Psychiatry and Neuroscience, Columbia University, New York, NY, USA.
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
18
|
Jazaeri SZ, Taghizadeh G, Babaei JF, Goudarzi S, Saadatmand P, Joghataei MT, Khanahmadi Z. Aquaporin 4 beyond a water channel; participation in motor, sensory, cognitive and psychological performances, a comprehensive review. Physiol Behav 2023; 271:114353. [PMID: 37714320 DOI: 10.1016/j.physbeh.2023.114353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
Aquaporin 4 (AQP4) is a protein highly expressed in the central nervous system (CNS) and peripheral nervous system (PNS) as well as various other organs, whose different sites of action indicate its importance in various functions. AQP4 has a variety of essential roles beyond water homeostasis. In this article, we have for the first time summarized different roles of AQP4 in motor and sensory functions, besides cognitive and psychological performances, and most importantly, possible physiological mechanisms by which AQP4 can exert its effects. Furthermore, we demonstrated that AQP4 participates in pathology of different neurological disorders, various effects depending on the disease type. Since neurological diseases involve a spectrum of dysfunctions and due to the difficulty of obtaining a treatment that can simultaneously affect these deficits, it is therefore suggested that future studies consider the role of this protein in different functional impairments related to neurological disorders simultaneously or separately by targeting AQP4 expression and/or polarity modulation.
Collapse
Affiliation(s)
- Seyede Zohreh Jazaeri
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of medical Sciences, Tehran, Iran
| | - Pegah Saadatmand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Innovation in Medical Education, Faculty of Medicine, Ottawa University, Ottawa, Canada.
| | - Zohreh Khanahmadi
- Department of Occupational Therapy, School of Rehabilitation Services, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Wang W, Li Y, Ma F, Sheng X, Chen K, Zhuo R, Wang C, Zheng H, Zhang YW, Bu G, Chen XF, Zhong L. Microglial repopulation reverses cognitive and synaptic deficits in an Alzheimer's disease model by restoring BDNF signaling. Brain Behav Immun 2023; 113:275-288. [PMID: 37482204 DOI: 10.1016/j.bbi.2023.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023] Open
Abstract
Over the past decade, compelling genetic evidence has highlighted the crucial role of microglial dysregulation in the development of Alzheimer's disease (AD). As resident immune cells in the brain, microglia undergo dystrophy and senescence during the chronic progression of AD. To explore the potential therapeutic benefits of replenishing the brain with new microglia in AD, we utilized the CSF1R inhibitor PLX3397 to deplete existing microglia and induce repopulation after inhibitor withdrawal in 5xFAD transgenic mice. Our findings revealed the remarkable benefits of microglial repopulation in ameliorating AD-associated cognitive deficits, accompanied by a notable elevation in synaptic proteins and an enhancement of hippocampal long-term potentiation (LTP). Additionally, we observed the profound restoration of microglial morphology and synaptic engulfment following their self-renewal. The impact of microglial repopulation on amyloid pathology is dependent on the duration of repopulation. Transcriptome analysis revealed a high resemblance between the gene expression profiles of repopulated microglia from 5xFAD mice and those of microglia from WT mice. Importantly, the dysregulated neurotrophic signaling pathway and hippocampal neurogenesis in the AD brain are restored following microglial replenishment. Lastly, we demonstrated that the repopulation restores the expression of brain-derived neurotrophic factor (BDNF) in microglia, thereby contributing to synaptic plasticity. In conclusion, our findings provide compelling evidence to support the notion that microglial self-renewal confers substantial benefits to the AD brain by restoring the BDNF neurotrophic signaling pathway. Thus, targeted microglial repopulation emerges as a highly promising and novel therapeutic strategy for alleviating cognitive impairment in AD.
Collapse
Affiliation(s)
- Wanbing Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yanzhong Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Fangling Ma
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xuan Sheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Kai Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Rengong Zhuo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Chen Wang
- Department of Neurology, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China; Shenzhen Research Institute of Xiamen University, Shenzhen 518063, China.
| | - Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
20
|
Silvas-Baltazar M, López-Oropeza G, Durán P, Martínez-Canabal A. Olfactory neurogenesis and its role in fear memory modulation. Front Behav Neurosci 2023; 17:1278324. [PMID: 37840547 PMCID: PMC10569173 DOI: 10.3389/fnbeh.2023.1278324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Olfaction is a critical sense that allows animals to navigate and understand their environment. In mammals, the critical brain structure to receive and process olfactory information is the olfactory bulb, a structure characterized by a laminated pattern with different types of neurons, some of which project to distant telencephalic structures, like the piriform cortex, the amygdala, and the hippocampal formation. Therefore, the olfactory bulb is the first structure of a complex cognitive network that relates olfaction to different types of memory, including episodic memories. The olfactory bulb continuously adds inhibitory newborn neurons throughout life; these cells locate both in the granule and glomerular layers and integrate into the olfactory circuits, inhibiting projection neurons. However, the roles of these cells modulating olfactory memories are unclear, particularly their role in fear memories. We consider that olfactory neurogenesis might modulate olfactory fear memories by a plastic process occurring in the olfactory bulb.
Collapse
Affiliation(s)
- Monserrat Silvas-Baltazar
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Grecia López-Oropeza
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Pilar Durán
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Alonso Martínez-Canabal
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Posgrado en Ciencias Biológicas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
21
|
Refaeli R, Kreisel T, Groysman M, Adamsky A, Goshen I. Engram stability and maturation during systems consolidation. Curr Biol 2023; 33:3942-3950.e3. [PMID: 37586373 PMCID: PMC10524918 DOI: 10.1016/j.cub.2023.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/27/2023] [Accepted: 07/20/2023] [Indexed: 08/18/2023]
Abstract
Remote memories play an important role in how we perceive the world, and they are rooted throughout the brain in "engrams": ensembles of cells that are formed during acquisition. Upon their reactivation, a specific memory can be recalled.1,2,3,4,5,6,7,8,9,10,11,12 Many studies have focused on the ensembles in CA1 of the hippocampus and the anterior cingulate cortex (ACC). However, the evolution of these components during systems' consolidation has not yet been comprehensively addressed.13,14,15,16 By applying transgenic approaches for ensemble identification, CLARITY, retro-AAV, and pseudo-rabies virus for circuit mapping, and chemogenetics for functional interrogation, we addressed the dynamics of recent and remote CA1 ensembles. We expected both stability (as they represent the same memory) and maturation (over time). Indeed, we found that CA1 engrams remain stable between recent and remote recalls, and the inhibition of engrams for recent recall during remote recall functionally impairs memory. We also found that new cells in the remote recall engram in the CA1 are not added randomly during maturation but differ according to their connections. First, we show in two ways that the anterograde CA1 → ACC engram cell projection grows larger. Finally, in the retrograde projections, the ACC reduces input to CA1 engram cells, whereas input from the entorhinal cortex and paraventricular nucleus of the thalamus increases. Our results shine fresh light on systems' consolidation by providing a deeper understanding of engram stability and maturation in the transition from recent to remote memory.
Collapse
Affiliation(s)
- Ron Refaeli
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Tirzah Kreisel
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Maya Groysman
- ELSC Vector Core Facility, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Adar Adamsky
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| |
Collapse
|
22
|
Osanai H, Yamamoto J, Kitamura T. Extracting electromyographic signals from multi-channel LFPs using independent component analysis without direct muscular recording. CELL REPORTS METHODS 2023; 3:100482. [PMID: 37426755 PMCID: PMC10326347 DOI: 10.1016/j.crmeth.2023.100482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/12/2023] [Accepted: 04/25/2023] [Indexed: 07/11/2023]
Abstract
Electromyography (EMG) has been commonly used for the precise identification of animal behavior. However, it is often not recorded together with in vivo electrophysiology due to the need for additional surgeries and setups and the high risk of mechanical wire disconnection. While independent component analysis (ICA) has been used to reduce noise from field potential data, there has been no attempt to proactively use the removed "noise," of which EMG signals are thought to be one of the major sources. Here, we demonstrate that EMG signals can be reconstructed without direct EMG recording using the "noise" ICA component from local field potentials. The extracted component is highly correlated with directly measured EMG, termed IC-EMG. IC-EMG is useful for measuring an animal's sleep/wake, freezing response, and non-rapid eye movement (NREM)/REM sleep states consistently with actual EMG. Our method has advantages in precise and long-term behavioral measurement in wide-ranging in vivo electrophysiology experiments.
Collapse
Affiliation(s)
- Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Yamamoto
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
23
|
Khan M, Baussan Y, Hebert-Chatelain E. Connecting Dots between Mitochondrial Dysfunction and Depression. Biomolecules 2023; 13:695. [PMID: 37189442 PMCID: PMC10135685 DOI: 10.3390/biom13040695] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Mitochondria are the prime source of cellular energy, and are also responsible for important processes such as oxidative stress, apoptosis and Ca2+ homeostasis. Depression is a psychiatric disease characterized by alteration in the metabolism, neurotransmission and neuroplasticity. In this manuscript, we summarize the recent evidence linking mitochondrial dysfunction to the pathophysiology of depression. Impaired expression of mitochondria-related genes, damage to mitochondrial membrane proteins and lipids, disruption of the electron transport chain, higher oxidative stress, neuroinflammation and apoptosis are all observed in preclinical models of depression and most of these parameters can be altered in the brain of patients with depression. A deeper knowledge of the depression pathophysiology and the identification of phenotypes and biomarkers with respect to mitochondrial dysfunction are needed to help early diagnosis and the development of new treatment strategies for this devastating disorder.
Collapse
Affiliation(s)
- Mehtab Khan
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| | - Yann Baussan
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| | - Etienne Hebert-Chatelain
- Department of Biology, University of Moncton, Moncton, NB E1A 3E9, Canada
- Mitochondrial Signaling and Pathophysiology, University of Moncton, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
24
|
Simkin DR. Post-Traumatic Stress Disorder/Developmental Trauma Disorder/Complex Post-Traumatic Stress Disorder and Complementary and Integrative Medicine/Functional Medicine. Child Adolesc Psychiatr Clin N Am 2023; 32:317-365. [PMID: 37147042 DOI: 10.1016/j.chc.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Response to PTSD treatments differ based on the age the abuse occurred, the type of abuse, and the chronicity of the abuse. Even when modifications to treatment are made based on the developmental age when the abuse occurred, therapies may be insufficient. In addition, when diagnostic criteria are modified to identify more children, some children continue to escape detection. Developmental Trauma Disorder, (akin to the RDoC), may be more suitable to identify epigenetic and inflammatory effects of early abuse that may be responsible for the nonresponsive to treatment. Complementary and Integrative Medicine interventions (meditation, EFT, EMDR, PUFAs, etc.) may reverse these effects.
Collapse
Affiliation(s)
- Deborah R Simkin
- Department of Psychiatry, Emory University School of Medicine, 8955 Highway 98 West, Suite 204, Miramar Beach, FL 32550, USA.
| |
Collapse
|
25
|
Inactivation of the dorsal CA1 hippocampus impairs the consolidation of discriminative avoidance memory by modulating the intrinsic and extrinsic hippocampal circuitry. J Chem Neuroanat 2023; 128:102209. [PMID: 36496001 DOI: 10.1016/j.jchemneu.2022.102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Despite progress in understanding the role of the dorsal hippocampus in the acquisition, consolidation and retrieval of episodic-like memory, plastic changes within the intra- and extrahippocampal circuits for aversive memory formation and anxiety-like behaviours must still be identified since both processes contribute to multiple aspects of flexible decision-making. Here, we investigated the effect of reversible inactivation induced by a muscimol microinfusion into the dorsal CA1 subfield (dCA1) either prior to acquisition or to retrieval testing of a discriminative avoidance task performed in a plus-maze apparatus (PM-DAT). Differential cAMP-response-element-binding protein 1 (CREB-1) expression in the dorsal and ventral CA1 and CA3 of the hippocampus (dCA1, dCA3, vCA1, and vCA3), dorsal dentate gyrus (dDG), and infralimbic (IL) and prelimbic (PrL) regions of the medial prefrontal cortex was also assessed to investigate the molecular changes associated with the consolidation or retrieval of episodic-like memory and anxiety. Adult male Wistar rats were assigned to two control groups, learning (no surgery/no microinfusion, n = 7) and sham-operated (sham surgery/no microinfusion, n = 6) groups, or four experimental groups, in which the vehicle (0.5 µl per side, n = 8/per group) or a GABAA receptor agonist (0.5 µg/0.5 µl muscimol/per side) was bilaterally microinfused in the dCA1 30 min prior to training (n = 9) or prior to testing sessions (n = 6) with a 24 h intertrial interval. Memory was evaluated using the percentage of time spent in the nonaversive enclosed arms, whereas anxiety was measured by calculating the percentages of time spent and entries into open arms and the percentage of time spent self-grooming. Our findings corroborated previous data showing that the dCA1 is required for discriminative avoidance consolidation. Furthermore, additional information indicated that impaired long-term memory was associated with downregulated CREB-1 expression in the dDG and vCA3. Moreover, memory retrieval was not impaired by dCA1 inactivation prior to the testing session, which was associated with the upregulation of CREB-1 in the dCA3 and vCA1 and downregulation in the dCA1 and vCA3. Differential expression of CREB was not identified in the IL or PrL areas. These results improve our understanding of how the hippocampal circuitry mediates the acquisition and retrieval of aversive memory and anxiety.
Collapse
|
26
|
Prothymosin α Plays Role as a Brain Guardian through Ecto-F 1 ATPase-P2Y 12 Complex and TLR4/MD2. Cells 2023; 12:cells12030496. [PMID: 36766838 PMCID: PMC9914670 DOI: 10.3390/cells12030496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Prothymosin alpha (ProTα) was discovered to be a necrosis inhibitor from the conditioned medium of a primary culture of rat cortical neurons under starved conditions. This protein carries out a neuronal cell-death-mode switch from necrosis to apoptosis, which is, in turn, suppressed by a variety of neurotrophic factors (NTFs). This type of NTF-assisted survival action of ProTα is reproduced in cerebral and retinal ischemia-reperfusion models. Further studies that used a retinal ischemia-reperfusion model revealed that ProTα protects retinal cells via ecto-F1 ATPase coupled with the Gi-coupled P2Y12 receptor and Toll-like receptor 4 (TLR4)/MD2 coupled with a Toll-IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF). In cerebral ischemia-reperfusion models, ProTα has additional survival mechanisms via an inhibition of matrix metalloproteases in microglia and vascular endothelial cells. Heterozygous or conditional ProTα knockout mice show phenotypes of anxiety, memory learning impairment, and a loss of neurogenesis. There are many reports that ProTα has multiple intracellular functions for cell survival and proliferation through a variety of protein-protein interactions. Overall, it is suggested that ProTα plays a key role as a brain guardian against ischemia stress through a cell-death-mode switch assisted by NTFs and a role of neurogenesis.
Collapse
|
27
|
Wu Y, Chen L, Zhong F, Zhou K, Lu C, Cheng X, Wang S. Cognitive impairment in patients with heart failure: molecular mechanism and therapy. Heart Fail Rev 2023:10.1007/s10741-022-10289-9. [PMID: 36593370 DOI: 10.1007/s10741-022-10289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
Heart failure (HF) is associated with multiple organ dysfunction and many comorbidities. Its incidence is high among the elderly and is a major health burden worldwide. Cognitive impairment (CI) is highly prevalent in older patients with HF, which is an abnormality in one or more of the items of cognition, attention, memory, language, psychomotor function, and visual spatial acuity. Studies have shown that the incidence of CI in HF patients is between 13 and 54%, and patients with both conditions have poor self-care ability and prognosis, as well as increased mortality rates. However, the mechanisms of CI development in HF patients are still unclear. In this review, we describe the epidemiology and risk factors as well as measures of improving CI in HF patients. We update the latest pathophysiological mechanisms related to the neurocognitive changes in HF patients, expounding on the mechanisms associated with the development of CI in HF patients.
Collapse
Affiliation(s)
- Yanan Wu
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Liwen Chen
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Feng Zhong
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Kaiyi Zhou
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Chao Lu
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao Cheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sheng Wang
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| |
Collapse
|
28
|
Fölsz O, Trouche S, Croset V. Adult-born neurons add flexibility to hippocampal memories. Front Neurosci 2023; 17:1128623. [PMID: 36875670 PMCID: PMC9975346 DOI: 10.3389/fnins.2023.1128623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Although most neurons are generated embryonically, neurogenesis is maintained at low rates in specific brain areas throughout adulthood, including the dentate gyrus of the mammalian hippocampus. Episodic-like memories encoded in the hippocampus require the dentate gyrus to decorrelate similar experiences by generating distinct neuronal representations from overlapping inputs (pattern separation). Adult-born neurons integrating into the dentate gyrus circuit compete with resident mature cells for neuronal inputs and outputs, and recruit inhibitory circuits to limit hippocampal activity. They display transient hyperexcitability and hyperplasticity during maturation, making them more likely to be recruited by any given experience. Behavioral evidence suggests that adult-born neurons support pattern separation in the rodent dentate gyrus during encoding, and they have been proposed to provide a temporal stamp to memories encoded in close succession. The constant addition of neurons gradually degrades old connections, promoting generalization and ultimately forgetting of remote memories in the hippocampus. This makes space for new memories, preventing saturation and interference. Overall, a small population of adult-born neurons appears to make a unique contribution to hippocampal information encoding and removal. Although several inconsistencies regarding the functional relevance of neurogenesis remain, in this review we argue that immature neurons confer a unique form of transience on the dentate gyrus that complements synaptic plasticity to help animals flexibly adapt to changing environments.
Collapse
Affiliation(s)
- Orsolya Fölsz
- Department of Biosciences, Durham University, Durham, United Kingdom.,MSc in Neuroscience Programme, University of Oxford, Oxford, United Kingdom
| | - Stéphanie Trouche
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Croset
- Department of Biosciences, Durham University, Durham, United Kingdom
| |
Collapse
|
29
|
FOXG1 Contributes Adult Hippocampal Neurogenesis in Mice. Int J Mol Sci 2022; 23:ijms232314979. [PMID: 36499306 PMCID: PMC9735854 DOI: 10.3390/ijms232314979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Strategies to enhance hippocampal precursor cells efficiently differentiate into neurons could be crucial for structural repair after neurodegenerative damage. FOXG1 has been shown to play an important role in pattern formation, cell proliferation, and cell specification during embryonic and early postnatal neurogenesis. Thus far, the role of FOXG1 in adult hippocampal neurogenesis is largely unknown. Utilizing CAG-loxp-stop-loxp-Foxg1-IRES-EGFP (Foxg1fl/fl), a specific mouse line combined with CreAAV infusion, we successfully forced FOXG1 overexpressed in the hippocampal dentate gyrus (DG) of the genotype mice. Thereafter, we explored the function of FOXG1 on neuronal lineage progression and hippocampal neurogenesis in adult mice. By inhibiting p21cip1 expression, FOXG1-regulated activities enable the expansion of the precursor cell population. Besides, FOXG1 induced quiescent radial-glia like type I neural progenitor, giving rise to intermediate progenitor cells, neuroblasts in the hippocampal DG. Through increasing the length of G1 phase, FOXG1 promoted lineage-committed cells to exit the cell cycle and differentiate into mature neurons. The present results suggest that FOXG1 likely promotes neuronal lineage progression and thereby contributes to adult hippocampal neurogenesis. Elevating FOXG1 levels either pharmacologically or through other means could present a therapeutic strategy for disease related with neuronal loss.
Collapse
|
30
|
Anand SK, Ahmad MH, Sahu MR, Subba R, Mondal AC. Detrimental Effects of Alcohol-Induced Inflammation on Brain Health: From Neurogenesis to Neurodegeneration. Cell Mol Neurobiol 2022:10.1007/s10571-022-01308-2. [DOI: 10.1007/s10571-022-01308-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/11/2022] [Indexed: 11/28/2022]
|
31
|
Lei B, Kang B, Lin W, Chen H, Hao Y, Ma J, Shi S, Zhong Y. Adult newborn granule cells confer emotional state-dependent adaptability in memory retrieval. SCIENCE ADVANCES 2022; 8:eabn2136. [PMID: 36367932 PMCID: PMC9651853 DOI: 10.1126/sciadv.abn2136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
Achieving optimal behavior requires animals to flexibly retrieve prior knowledge. Here, we show that adult newborn granule cells (anbGCs) mediate emotional state-dependent adaptability of memory retrieval. We find that acute social reward (aSR) enhances memory retrieval by increasing the reactivation of engram cells, while acute social stress (aSS) weakens retrieval and reduces the reactivation. Such bidirectional regulation relies on the activation of distinct populations of anbGCs by aSR and aSS, triggering opposing modifications of dDG activity, which is sufficient to regulate and predict the performance of memory retrieval. Concordantly, in emotional disorder models, aSR-dependent memory adaptability is impaired, while the effect of aSS remains intact. Together, our data revealed that anbGCs mediate adaptability of memory retrieval, allowing animals to flexibly retrieve memory according to the current emotional state, and suggested the essential roles of anbGCs in translating emotional information to the regulation of memory expression.
Collapse
Affiliation(s)
- Bo Lei
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Bilin Kang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Wantong Lin
- Department of Computer Science, Brandeis University, Waltham, MA 02453, USA
| | - Haichao Chen
- School of Medicine, Tsinghua University, Beijing 100084, P.R. China
| | - Yuejun Hao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Jian Ma
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Songhai Shi
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
| | - Yi Zhong
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, P.R. China
- MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
32
|
Bradley-Garcia M, Winocur G, Sekeres MJ. Episodic Memory and Recollection Network Disruptions Following Chemotherapy Treatment in Breast Cancer Survivors: A Review of Neuroimaging Findings. Cancers (Basel) 2022; 14:4752. [PMID: 36230678 PMCID: PMC9563268 DOI: 10.3390/cancers14194752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term memory disturbances are amongst the most common and disruptive cognitive symptoms experienced by breast cancer survivors following chemotherapy. To date, most clinical assessments of long-term memory dysfunction in breast cancer survivors have utilized basic verbal and visual memory tasks that do not capture the complexities of everyday event memories. Complex event memories, including episodic memory and autobiographical memory, critically rely on hippocampal processing for encoding and retrieval. Systemic chemotherapy treatments used in breast cancer commonly cause neurotoxicity within the hippocampus, thereby creating a vulnerability to memory impairment. We review structural and functional neuroimaging studies that have identified disruptions in the recollection network and related episodic memory impairments in chemotherapy-treated breast cancer survivors, and argue for the need to better characterize hippocampally mediated memory dysfunction following chemotherapy treatments. Given the importance of autobiographical memory for a person's sense of identity, ability to plan for the future, and general functioning, under-appreciation of how this type of memory is impacted by cancer treatment can lead to overlooking or minimizing the negative experiences of breast cancer survivors, and neglecting a cognitive domain that may benefit from intervention strategies.
Collapse
Affiliation(s)
| | - Gordon Winocur
- Rotman Research Institute, Baycrest Centre, Toronto, ON M6A 2E1, Canada
- Department of Psychology, Department of Psychiatry, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| | - Melanie J Sekeres
- School of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
33
|
Chang YS, Lin CL, Lee CW, Lin HC, Wu YT, Shih YH. Exercise Normalized the Hippocampal Renin-Angiotensin System and Restored Spatial Memory Function, Neurogenesis, and Blood-Brain Barrier Permeability in the 2K1C-Hypertensive Mouse. Int J Mol Sci 2022; 23:ijms23105531. [PMID: 35628344 PMCID: PMC9146761 DOI: 10.3390/ijms23105531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/14/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Hypertension is associated with blood-brain barrier alteration and brain function decline. Previously, we established the 2-kidney,1-clip (2K1C) hypertensive mice model by renin-angiotensin system (RAS) stimulating. We found that 2K1C-induced hypertension would impair hippocampus-related memory function and decrease adult hippocampal neurogenesis. Even though large studies have investigated the mechanism of hypertension affecting brain function, there remains a lack of efficient ways to halt this vicious effect. The previous study indicated that running exercise ameliorates neurogenesis and spatial memory function in aging mice. Moreover, studies showed that exercise could normalize RAS activity, which might be associated with neurogenesis impairment. Thus, we hypothesize that running exercise could ameliorate neurogenesis and spatial memory function impairment in the 2K1C-hypertension mice. In this study, we performed 2K1C surgery on eight-weeks-old C57BL/6 mice and put them on treadmill exercise one month after the surgery. The results indicate that running exercise improves the spatial memory and neurogenesis impairment of the 2K1C-mice. Moreover, running exercise normalized the activated RAS and blood-brain barrier leakage of the hippocampus, although the blood pressure was not decreased. In conclusion, running exercise could halt hypertension-induced brain impairment through RAS normalization.
Collapse
Affiliation(s)
- Ying-Shuang Chang
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Sanmin District, Kaohsiung 80708, Taiwan; (Y.-S.C.); (H.-C.L.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Sanmin District, Kaohsiung 80708, Taiwan;
| | - Chih-Lung Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Sanmin District, Kaohsiung 80708, Taiwan;
- Department of Neurosurgery, Kaohsiung Medical University Hospital, 100, Tzyou 1st Road, Sanmin District, Kaohsiung 80756, Taiwan
| | - Chu-Wan Lee
- Department of Nursing, National Tainan Junior College of Nursing, 78, Section 2, Minzu Road, West Central District, Tainan 70043, Taiwan;
| | - Han-Chen Lin
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Sanmin District, Kaohsiung 80708, Taiwan; (Y.-S.C.); (H.-C.L.)
- Department of Medical Research, Kaohsiung Medical University Hospital, 100, Tzyou 1st Road, Sanmin District, Kaohsiung 80756, Taiwan
| | - Yi-Ting Wu
- Department of Nursing, Tzu Hui Institute of Technology, Pingtung County 92641, Taiwan;
| | - Yao-Hsiang Shih
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Sanmin District, Kaohsiung 80708, Taiwan; (Y.-S.C.); (H.-C.L.)
- Department of Medical Research, Kaohsiung Medical University Hospital, 100, Tzyou 1st Road, Sanmin District, Kaohsiung 80756, Taiwan
- Correspondence: ; Tel.: +886-7-3121101 (ext. 2144)
| |
Collapse
|
34
|
Cao S, Li M, Sun Y, Wu P, Yang W, Dai H, Guo Y, Ye Y, Wang Z, Xie X, Chen X, Liang W. Intermittent fasting enhances hippocampal NPY expression to promote neurogenesis after traumatic brain injury. Nutrition 2022; 97:111621. [PMID: 35255397 DOI: 10.1016/j.nut.2022.111621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVES Interventions for preventing cognitive dysfunction after traumatic brain injury (TBI) are limited. Given that adult hippocampal neurogenesis after brain injury contributes to cognitive recovery, and hippocampal neurogenesis is potentially affected by nutritional factors, the aim of this study was to examine whether fasting could promote hippocampal neurogenesis and thus ameliorate the cognitive defects after TBI. METHODS The present study used 8- to 10-wk-old C57 BL/6 N mice weighing 23 g, half males and half females. The mice were randomly assigned to each group, with 10 to 18 mice per group. All mice were housed in an approved animal facility with a 12-h light/dark cycle. In the metabolic study (food intake, body weight, blood glucose, triacylglycerol, total cholesterol, and β-hydroxybutyric acid ), 54 mice (male:female = 1:1) were randomized to the ad libitum (AL) group (n = 18) and the intermittent fasting (IF) group (n = 36). In the neurogenesis study, 45 mice (male:female = 1:1) were randomized to AL (n = 18), IF (n = 9), IF + scramble (n = 9), and the IF + neuropeptide Y (NPY)_siRNA (n = 9) groups. In the Morris water maze test, 48 mice (male:female = 1:1) were randomized to AL (n = 12), IF (n = 12), IF + scramble (n = 12), and the IF + NPY_siRNA (n = 12) groups. RESULTS We showed that a 1-mo-long IF regimen enhanced the proliferation of neural stem cells in the subgranular zone of the hippocampus 3 d after TBI, in addition to improving the cognitive performance in the Morris water maze test. Furthermore, an increase in the hippocampal NPY expression was detected in the IF group after the injury, compared with the mice fed AL, and local knockdown of NPY in vivo attenuated the effects of IF on TBI. CONCLUSIONS These findings suggest that IF promotes hippocampal neurogenesis after TBI by a mechanism that involves enhancement of NPY expression, to alleviate cognitive dysfunction caused by injury.
Collapse
Affiliation(s)
- Shuqiang Cao
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Manrui Li
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuwen Sun
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Peiyan Wu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Wenjie Yang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Hao Dai
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yadong Guo
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Yi Ye
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zheng Wang
- Institute of Forensic Medicine, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoqi Xie
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| | - Weibo Liang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
35
|
Sex Differences in the Spatial Behavior Functions of Adult-Born Neurons in Rats. eNeuro 2022; 9:ENEURO.0054-22.2022. [PMID: 35473765 PMCID: PMC9116935 DOI: 10.1523/eneuro.0054-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023] Open
Abstract
Adult neurogenesis modifies hippocampal circuits and behavior, but removing newborn neurons does not consistently alter spatial processing, a core function of the hippocampus. Additionally, little is known about sex differences in neurogenesis since few studies have compared males and females. Since adult-born neurons regulate the stress response, we hypothesized that spatial functions may be more prominent under aversive conditions and may differ between males and females given sex differences in stress responding. We therefore trained intact and neurogenesis-deficient rats in the spatial water maze at temperatures that vary in their degree of aversiveness. In the standard water maze, ablating neurogenesis did not alter spatial learning in either sex. However, in cold water, ablating neurogenesis had divergent sex-dependent effects: relative to intact rats, male neurogenesis-deficient rats were slower to escape the maze and female neurogenesis-deficient rats were faster. Neurogenesis promoted temperature-related changes in search strategy in females, but it promoted search strategy stability in males. Females displayed greater recruitment (Fos expression) of the dorsal hippocampus than males, particularly in cold water. However, blocking neurogenesis did not alter Fos expression in either sex. Finally, morphologic analyses revealed greater experience-dependent plasticity in males. Adult-born neurons in males and females had similar morphology at baseline but training increased spine density and reduced presynaptic terminal size, specifically in males. Collectively, these findings indicate that adult-born neurons contribute to spatial learning in stressful conditions and they provide new evidence for sex differences in their behavioral functions.
Collapse
|
36
|
Terranova JI, Yokose J, Osanai H, Marks WD, Yamamoto J, Ogawa SK, Kitamura T. Hippocampal-amygdala memory circuits govern experience-dependent observational fear. Neuron 2022; 110:1416-1431.e13. [PMID: 35139362 PMCID: PMC9035063 DOI: 10.1016/j.neuron.2022.01.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/10/2021] [Accepted: 01/13/2022] [Indexed: 11/28/2022]
Abstract
The empathic ability to vicariously experience the other's fearful situation, a process called observational fear (OF), is critical to survive in nature and function in society. OF can be facilitated by both prior similar fear experience in the observer and social familiarity with the demonstrator. However, the neural circuit mechanisms of experience-dependent OF (Exp OF) remain unknown. Here, we demonstrate that hippocampal-basolateral amygdala (HPC-BLA) circuits in mice without involving the anterior cingulate cortex, considered a center of OF, mediate Exp OF. Dorsal HPC neurons generate fear memory engram cells in BLA encoding prior similar fear experiences, which are essential for Exp OF. On the other hand, ventral HPC neurons respond to the familiar demonstrator's aversive situation during Exp OF, which reactivates the fear memory engram cells in BLA to elicit Exp OF. Our study provides new insights into the memory engram-dependent perception-action coupling that underlies empathic behaviors like Exp OF.
Collapse
Affiliation(s)
- Joseph I Terranova
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Yokose
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hisayuki Osanai
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - William D Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Yamamoto
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sachie K Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
37
|
Marks WD, Yokose J, Kitamura T, Ogawa SK. Neuronal Ensembles Organize Activity to Generate Contextual Memory. Front Behav Neurosci 2022; 16:805132. [PMID: 35368306 PMCID: PMC8965349 DOI: 10.3389/fnbeh.2022.805132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Contextual learning is a critical component of episodic memory and important for living in any environment. Context can be described as the attributes of a location that are not the location itself. This includes a variety of non-spatial information that can be derived from sensory systems (sounds, smells, lighting, etc.) and internal state. In this review, we first address the behavioral underpinnings of contextual memory and the development of context memory theory, with a particular focus on the contextual fear conditioning paradigm as a means of assessing contextual learning and the underlying processes contributing to it. We then present the various neural centers that play roles in contextual learning. We continue with a discussion of the current knowledge of the neural circuitry and physiological processes that underlie contextual representations in the Entorhinal cortex-Hippocampal (EC-HPC) circuit, as the most well studied contributor to contextual memory, focusing on the role of ensemble activity as a representation of context with a description of remapping, and pattern separation and completion in the processing of contextual information. We then discuss other critical regions involved in contextual memory formation and retrieval. We finally consider the engram assembly as an indicator of stored contextual memories and discuss its potential contribution to contextual memory.
Collapse
Affiliation(s)
- William D. Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jun Yokose
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sachie K. Ogawa
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
38
|
Chen QN, Ding XL, Guo XX, Zhou G, Guan JS. Suv39h1 regulates memory stability by inhibiting the expression of Shank1 in hippocampal newborn neurons. Eur J Neurosci 2022; 55:1424-1441. [PMID: 35181969 DOI: 10.1111/ejn.15626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/11/2022] [Accepted: 02/15/2022] [Indexed: 11/28/2022]
Abstract
Adult newborn neurons are involved in memory encoding and extinction, but the neural mechanism is unclear. We found the adult newborn neurons at 4 weeks are recruited by learning and subjected to epigenetic regulations, consequently reducing their ability to be re-recruited later. After removal of the epigenetic blockage, Suv39h1 KO mice showed an increased recruiting number of aged newborn neurons and enhanced flexibility in learning tasks. Besides NRXN1, we found SHANK1, the synaptic scaffold protein, is one of the major targets of Suv39h1, regulating memory stability. Expression of Shank1 is transiently engaged to enhance synaptogenesis during learning and is strongly suppressed by Suv39h1 from 5 hours after learning. Exogenously overexpression of Shank1 in dentate gyrus increased the density of mushroom spines and decreased the persistency of old memories. Our study indicated the activity-regulated epigenetic modification in newly matured newborn neurons in hippocampus insulates temporally distinct experiences and stabilizes old memories.
Collapse
Affiliation(s)
- Qi-Nan Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xin-Lu Ding
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiu-Xian Guo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Gang Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ji-Song Guan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
39
|
Ihunwo AO, Perego J, Martino G, Vicenzi E, Panina-Bordignon P. Neurogenesis and Viral Infection. Front Immunol 2022; 13:826091. [PMID: 35251006 PMCID: PMC8891128 DOI: 10.3389/fimmu.2022.826091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Neural stem cells (NSCs) are multipotent stem cells that reside in the fetal and adult mammalian brain, which can self-renew and differentiate into neurons and supporting cells. Intrinsic and extrinsic cues, from cells in the local niche and from distant sites, stringently orchestrates the self-renewal and differentiation competence of NSCs. Ample evidence supports the important role of NSCs in neuroplasticity, aging, disease, and repair of the nervous system. Indeed, activation of NSCs or their transplantation into injured areas of the central nervous system can lead to regeneration in animal models. Viral invasion of NSCs can negatively affect neurogenesis and synaptogenesis, with consequent cell death, impairment of cell cycle progression, early differentiation, which cause neural progenitors depletion in the cortical layer of the brain. Herein, we will review the current understanding of Zika virus (ZIKV) infection of the fetal brain and the NSCs, which are the preferential population targeted by ZIKV. Furthermore, the potential neurotropic properties of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which may cause direct neurological damage, will be discussed.
Collapse
Affiliation(s)
- Amadi Ogonda Ihunwo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jessica Perego
- Neuroimmunology Unit, Division of Neuroscience, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Division of Neuroscience, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, Division of Immunology, Transplantation and Infectious Disesases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Paola Panina-Bordignon
- Neuroimmunology Unit, Division of Neuroscience, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
40
|
A53T α-synuclein induces neurogenesis impairment and cognitive dysfunction in line M83 transgenic mice and reduces the proliferation of embryonic neural stem cells. Brain Res Bull 2022; 182:118-129. [PMID: 35182691 DOI: 10.1016/j.brainresbull.2022.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 12/26/2022]
Abstract
Dementia with Lewy body (DLB) is the second most common degenerative dementia after Alzheimer's disease. There is no therapeutic drug for DLB currently. It's urgent for us to understand the pathological mechanism of dementia mediated by α-synuclein, as the main component of Lewy body. Here, we found that the A53T α-synuclein transgenic mice showed decreased nesting behavior starting from the age of 1 month. The results in Morris water maze test suggested that the 6-month-old mice had learning memory deficits. Golgi staining indicated that the apical neuronal dendritic spines of hippocampal CA1 neurons were significantly reduced in 6-month-old homozygotes and heterozygotes, although MAP2 protein expression revealed no significant difference in the hippocampus among wild-type mice, homozygotes and heterozygotes. In vitro, we proved mutant A53T α-synuclein decreased the dendritic branches and dendrite spines on the embryonic mice hippocampal neurons. Furthermore, Ki67 immunofluorescence staining identified that the Ki67-positive cells of the hippocampal dentate gyrus and subventricular zone were significantly reduced in 6-month-old homozygotes and heterozygotes, compared with age-matched wild-type mice. Similarly, when 6-month-old mice were injected with BrdU for one day, the immunostaining results also confirmed that BrdU-positive cells were significantly reduced in homozygous and heterozygous mice. Lastly, we transfected primary embryonic hippocampal neural stem cells with lentivirus vector expressing A53T α-synuclein in vitro. Both BrdU staining and Western blotting showed that A53T α-synuclein significantly decreased the proliferation of embryonic neural stem cells. Taken together, these data suggest that A53T α-synuclein can induce adult neurogenesis impairment and cognitive dysfunction. The A53T α-synuclein transgenic mice may be used as an animal model for DLB. Promoting adult neurogenesis may be a promising approach to treat DLB pathogenesis.
Collapse
|
41
|
Hernández-Mercado K, Zepeda A. Morris Water Maze and Contextual Fear Conditioning Tasks to Evaluate Cognitive Functions Associated With Adult Hippocampal Neurogenesis. Front Neurosci 2022; 15:782947. [PMID: 35046769 PMCID: PMC8761726 DOI: 10.3389/fnins.2021.782947] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
New neurons are continuously generated and functionally integrated into the dentate gyrus (DG) network during the adult lifespan of most mammals. The hippocampus is a crucial structure for spatial learning and memory, and the addition of new neurons into the DG circuitry of rodents seems to be a key element for these processes to occur. The Morris water maze (MWM) and contextual fear conditioning (CFC) are among the most commonly used hippocampus-dependent behavioral tasks to study episodic-like learning and memory in rodents. While the functional contribution of adult hippocampal neurogenesis (AHN) through these paradigms has been widely addressed, results have generated controversial findings. In this review, we analyze and discuss possible factors in the experimental methods that could explain the inconsistent results among AHN studies; moreover, we provide specific suggestions for the design of more sensitive protocols to assess AHN-mediated learning and memory functions.
Collapse
Affiliation(s)
- Karina Hernández-Mercado
- Departamento de Medicina Genómica y Toxicológia Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicológia Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
42
|
Ryan TJ, Frankland PW. Forgetting as a form of adaptive engram cell plasticity. Nat Rev Neurosci 2022; 23:173-186. [PMID: 35027710 DOI: 10.1038/s41583-021-00548-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2021] [Indexed: 12/30/2022]
Abstract
One leading hypothesis suggests that memories are stored in ensembles of neurons (or 'engram cells') and that successful recall involves reactivation of these ensembles. A logical extension of this idea is that forgetting occurs when engram cells cannot be reactivated. Forms of 'natural forgetting' vary considerably in terms of their underlying mechanisms, time course and reversibility. However, we suggest that all forms of forgetting involve circuit remodelling that switches engram cells from an accessible state (where they can be reactivated by natural recall cues) to an inaccessible state (where they cannot). In many cases, forgetting rates are modulated by environmental conditions and we therefore propose that forgetting is a form of neuroplasticity that alters engram cell accessibility in a manner that is sensitive to mismatches between expectations and the environment. Moreover, we hypothesize that disease states associated with forgetting may hijack natural forgetting mechanisms, resulting in reduced engram cell accessibility and memory loss.
Collapse
Affiliation(s)
- Tomás J Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland. .,Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland. .,Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, Victoria, Australia. .,Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| | - Paul W Frankland
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada. .,Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada. .,Department of Psychology, University of Toronto, Toronto, Ontario, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, Canada. .,Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Role of Bioactive Compounds in the Regulation of Mitochondrial Dysfunctions in Brain and Age-Related Neurodegenerative Diseases. Cells 2022; 11:cells11020257. [PMID: 35053373 PMCID: PMC8773907 DOI: 10.3390/cells11020257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondria are multifunctional organelles that participate in a wide range of metabolic processes, including energy production and biomolecule synthesis. The morphology and distribution of intracellular mitochondria change dynamically, reflecting a cell’s metabolic activity. Oxidative stress is defined as a mismatch between the body’s ability to neutralise and eliminate reactive oxygen and nitrogen species (ROS and RNS). A determination of mitochondria failure in increasing oxidative stress, as well as its implications in neurodegenerative illnesses and apoptosis, is a significant developmental process of focus in this review. The neuroprotective effects of bioactive compounds linked to neuronal regulation, as well as related neuronal development abnormalities, will be investigated. In conclusion, the study of secondary components and the use of mitochondrial features in the analysis of various neurodevelopmental diseases has enabled the development of a new class of mitochondrial-targeted pharmaceuticals capable of alleviating neurodegenerative disease states and enabling longevity and healthy ageing for the vast majority of people.
Collapse
|
44
|
Surget A, Belzung C. Adult hippocampal neurogenesis shapes adaptation and improves stress response: a mechanistic and integrative perspective. Mol Psychiatry 2022; 27:403-421. [PMID: 33990771 PMCID: PMC8960391 DOI: 10.1038/s41380-021-01136-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 02/03/2023]
Abstract
Adult hippocampal neurogenesis (AHN) represents a remarkable form of neuroplasticity that has increasingly been linked to the stress response in recent years. However, the hippocampus does not itself support the expression of the different dimensions of the stress response. Moreover, the main hippocampal functions are essentially preserved under AHN depletion and adult-born immature neurons (abGNs) have no extrahippocampal projections, which questions the mechanisms by which abGNs influence functions supported by brain areas far from the hippocampus. Within this framework, we propose that through its computational influences AHN is pivotal in shaping adaption to environmental demands, underlying its role in stress response. The hippocampus with its high input convergence and output divergence represents a computational hub, ideally positioned in the brain (1) to detect cues and contexts linked to past, current and predicted stressful experiences, and (2) to supervise the expression of the stress response at the cognitive, affective, behavioral, and physiological levels. AHN appears to bias hippocampal computations toward enhanced conjunctive encoding and pattern separation, promoting contextual discrimination and cognitive flexibility, reducing proactive interference and generalization of stressful experiences to safe contexts. These effects result in gating downstream brain areas with more accurate and contextualized information, enabling the different dimensions of the stress response to be more appropriately set with specific contexts. Here, we first provide an integrative perspective of the functional involvement of AHN in the hippocampus and a phenomenological overview of the stress response. We then examine the mechanistic underpinning of the role of AHN in the stress response and describe its potential implications in the different dimensions accompanying this response.
Collapse
Affiliation(s)
- A Surget
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | - C Belzung
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| |
Collapse
|
45
|
Valenti D, Stagni F, Emili M, Guidi S, Bartesaghi R, Vacca RA. Impaired Brain Mitochondrial Bioenergetics in the Ts65Dn Mouse Model of Down Syndrome Is Restored by Neonatal Treatment with the Polyphenol 7,8-Dihydroxyflavone. Antioxidants (Basel) 2021; 11:antiox11010062. [PMID: 35052567 PMCID: PMC8773005 DOI: 10.3390/antiox11010062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/18/2022] Open
Abstract
Down syndrome (DS), a major genetic cause of intellectual disability, is characterized by numerous neurodevelopmental defects. Previous in vitro studies highlighted a relationship between bioenergetic dysfunction and reduced neurogenesis in progenitor cells from the Ts65Dn mouse model of DS, suggesting a critical role of mitochondrial dysfunction in neurodevelopmental alterations in DS. Recent in vivo studies in Ts65Dn mice showed that neonatal supplementation (Days P3–P15) with the polyphenol 7,8-dihydroxyflavone (7,8-DHF) fully restored hippocampal neurogenesis. The current study was aimed to establish whether brain mitochondrial bioenergetic defects are already present in Ts65Dn pups and whether early treatment with 7,8-DHF positively impacts on mitochondrial function. In the brain and cerebellum of P3 and P15 Ts65Dn pups we found a strong impairment in the oxidative phosphorylation apparatus, resulting in a deficit in mitochondrial ATP production and ATP content. Administration of 7,8-DHF (dose: 5 mg/kg/day) during Days P3–P15 fully restored bioenergetic dysfunction in Ts65Dn mice, reduced the levels of oxygen radicals and reinstated the hippocampal levels of PGC-1α. No pharmacotherapy is available for DS. From current findings, 7,8-DHF emerges as a treatment with a good translational potential for improving mitochondrial bioenergetics and, thus, mitochondria-linked neurodevelopmental alterations in DS.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
- Correspondence: (D.V.); (R.B.)
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy;
| | - Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (M.E.); (S.G.)
- Correspondence: (D.V.); (R.B.)
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), 70126 Bari, Italy;
| |
Collapse
|
46
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
47
|
Çelik K, Bilim P, Garip G, Durmaz B, Yildirim Sözmen E, Baka M. Acute hypoxia exposure following prenatal stress impairs hippocampus and novelty-seeking behavior in adolescent rats. Int J Dev Neurosci 2021; 82:85-95. [PMID: 34850973 DOI: 10.1002/jdn.10162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/13/2021] [Accepted: 11/23/2021] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES The present study aimed to investigate the effects of acute hypoxia exposure following prenatal stress on the novelty-seeking behavior and hippocampus of adolescent rats. METHODS The offspring were divided into prenatal stress (PS) and non-stress (NS) groups. Both groups were exposed to hypoxia on postnatal day 10 (P10) while control groups were undisturbed. Novel object recognition task was performed in each group. Next, brains were collected to examine hippocampus via immunohistochemical and biochemical studies on postnatal day 35 (P35). RESULTS PS decreased novelty discrimination and synaptophysin (SYN) expressions in both CA1 and CA3 of the hypoxia group prominently (p < 0.05). Nestin-expressing cells were reduced while vascular endothelial growth factor (VEGF) expression was enhanced in the subgranular zone (SGZ) of PS-hypoxia group (p < 0.05). VEGF enhancement triggered angiogenesis in the CA1 and CA3 significantly (p < 0.05). PS also increased thiobarbituric acid reactive substances (TBARS) levels in the hypoxia group as a result of oxidative stress (p < 0.05). CONCLUSION These findings demonstrated that PS exacerbates neurodevelopmental deficits in the hippocampus of acute hypoxia-induced offspring in adolescence.
Collapse
Affiliation(s)
- Kübra Çelik
- Department of Neuroscience, Institute of Health Sciences, Ege University, Izmir, Turkey.,Department of Electroneurophysiology, Vocational School of Health Care Services, Istinye University, Istanbul, Turkey
| | - Petek Bilim
- Department of Neuroscience, Institute of Health Sciences, Ege University, Izmir, Turkey.,Department of Psychology, Faculty of Economics, Business Administration and Social Sciences, Toros University, Mersin, Turkey
| | - Gurur Garip
- Department of Histology and Embryology, School of Medicine, Ege University, Izmir, Turkey
| | - Burak Durmaz
- Department of Medical Biochemistry, School of Medicine, Ege University, Izmir, Turkey
| | - Eser Yildirim Sözmen
- Department of Medical Biochemistry, School of Medicine, Ege University, Izmir, Turkey
| | - Meral Baka
- Department of Histology and Embryology, School of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
48
|
Sekeres MJ, Bradley-Garcia M, Martinez-Canabal A, Winocur G. Chemotherapy-Induced Cognitive Impairment and Hippocampal Neurogenesis: A Review of Physiological Mechanisms and Interventions. Int J Mol Sci 2021; 22:12697. [PMID: 34884513 PMCID: PMC8657487 DOI: 10.3390/ijms222312697] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/15/2021] [Accepted: 11/20/2021] [Indexed: 12/16/2022] Open
Abstract
A wide range of cognitive deficits, including memory loss associated with hippocampal dysfunction, have been widely reported in cancer survivors who received chemotherapy. Changes in both white matter and gray matter volume have been observed following chemotherapy treatment, with reduced volume in the medial temporal lobe thought to be due in part to reductions in hippocampal neurogenesis. Pre-clinical rodent models confirm that common chemotherapeutic agents used to treat various forms of non-CNS cancers reduce rates of hippocampal neurogenesis and impair performance on hippocampally-mediated learning and memory tasks. We review the pre-clinical rodent literature to identify how various chemotherapeutic drugs affect hippocampal neurogenesis and induce cognitive impairment. We also review factors such as physical exercise and environmental stimulation that may protect against chemotherapy-induced neurogenic suppression and hippocampal neurotoxicity. Finally, we review pharmacological interventions that target the hippocampus and are designed to prevent or reduce the cognitive and neurotoxic side effects of chemotherapy.
Collapse
Affiliation(s)
| | | | - Alonso Martinez-Canabal
- Cell Biology Department, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Gordon Winocur
- Rotman Research Institute, Baycrest Center, Toronto, ON M6A 2E1, Canada;
- Department of Psychology, Department of Psychiatry, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Psychology, Trent University, Peterborough, ON K9J 7B8, Canada
| |
Collapse
|
49
|
Marks WD, Yamamoto N, Kitamura T. Complementary roles of differential medial entorhinal cortex inputs to the hippocampus for the formation and integration of temporal and contextual memory (Systems Neuroscience). Eur J Neurosci 2021; 54:6762-6779. [PMID: 32277786 PMCID: PMC8187108 DOI: 10.1111/ejn.14737] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/28/2020] [Accepted: 03/30/2020] [Indexed: 11/29/2022]
Abstract
In humans and rodents, the entorhinal cortical (EC)-hippocampal (HPC) circuit is crucial for the formation and recall of memory, preserving both spatial information and temporal information about the occurrence of past events. Both modeling and experimental studies have revealed circuits within this network that play crucial roles in encoding space and context. However, our understanding about the time-related aspects of memory is just beginning to be understood. In this review, we first describe updates regarding recent anatomical discoveries for the EC-HPC network, as several important neural circuits critical for memory formation have been discovered by newly developed neural tracing technologies. Second, we examine the complementary roles of multiple medial entorhinal cortical inputs, including newly discovered circuits, into the hippocampus for the temporal and spatial aspects of memory. Finally, we will discuss how temporal and contextual memory information is integrated in HPC cornu ammonis 1 cells. We provide new insights into the neural circuit mechanisms for anatomical and functional segregation and integration of the temporal and spatial aspects of memory encoding in the EC-HPC networks.
Collapse
Affiliation(s)
- William D. Marks
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Naoki Yamamoto
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Takashi Kitamura
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| |
Collapse
|
50
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|