1
|
McKiernan EC, Herrera-Valdez MA, Marrone DF. A biophysical minimal model to investigate age-related changes in CA1 pyramidal cell electrical activity. PLoS One 2024; 19:e0308809. [PMID: 39231135 PMCID: PMC11373847 DOI: 10.1371/journal.pone.0308809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024] Open
Abstract
Aging is a physiological process that is still poorly understood, especially with respect to effects on the brain. There are open questions about aging that are difficult to answer with an experimental approach. Underlying challenges include the difficulty of recording in vivo single cell and network activity simultaneously with submillisecond resolution, and brain compensatory mechanisms triggered by genetic, pharmacologic, or behavioral manipulations. Mathematical modeling can help address some of these questions by allowing us to fix parameters that cannot be controlled experimentally and investigate neural activity under different conditions. We present a biophysical minimal model of CA1 pyramidal cells (PCs) based on general expressions for transmembrane ion transport derived from thermodynamical principles. The model allows directly varying the contribution of ion channels by changing their number. By analyzing the dynamics of the model, we find parameter ranges that reproduce the variability in electrical activity seen in PCs. In addition, increasing the L-type Ca2+ channel expression in the model reproduces age-related changes in electrical activity that are qualitatively and quantitatively similar to those observed in PCs from aged animals. We also make predictions about age-related changes in PC bursting activity that, to our knowledge, have not been reported previously. We conclude that the model's biophysical nature, flexibility, and computational simplicity make it a potentially powerful complement to experimental studies of aging.
Collapse
Affiliation(s)
- Erin C McKiernan
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, CDMX, México
| | - Marco A Herrera-Valdez
- Laboratorio de Dinámica, Biofísica y Fisiología de Sistemas, Departamento de Matemáticas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, CDMX, México
| | - Diano F Marrone
- Department of Psychology, Wilfrid Laurier University, Waterloo, ON, Canada
- McKnight Brain Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
2
|
Richardson B, Goedert T, Quraishe S, Deinhardt K, Mudher A. How do neurons age? A focused review on the aging of the microtubular cytoskeleton. Neural Regen Res 2024; 19:1899-1907. [PMID: 38227514 DOI: 10.4103/1673-5374.390974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024] Open
Abstract
Aging is the leading risk factor for Alzheimer's disease and other neurodegenerative diseases. We now understand that a breakdown in the neuronal cytoskeleton, mainly underpinned by protein modifications leading to the destabilization of microtubules, is central to the pathogenesis of Alzheimer's disease. This is accompanied by morphological defects across the somatodendritic compartment, axon, and synapse. However, knowledge of what occurs to the microtubule cytoskeleton and morphology of the neuron during physiological aging is comparatively poor. Several recent studies have suggested that there is an age-related increase in the phosphorylation of the key microtubule stabilizing protein tau, a modification, which is known to destabilize the cytoskeleton in Alzheimer's disease. This indicates that the cytoskeleton and potentially other neuronal structures reliant on the cytoskeleton become functionally compromised during normal physiological aging. The current literature shows age-related reductions in synaptic spine density and shifts in synaptic spine conformation which might explain age-related synaptic functional deficits. However, knowledge of what occurs to the microtubular and actin cytoskeleton, with increasing age is extremely limited. When considering the somatodendritic compartment, a regression in dendrites and loss of dendritic length and volume is reported whilst a reduction in soma volume/size is often seen. However, research into cytoskeletal change is limited to a handful of studies demonstrating reductions in and mislocalizations of microtubule-associated proteins with just one study directly exploring the integrity of the microtubules. In the axon, an increase in axonal diameter and age-related appearance of swellings is reported but like the dendrites, just one study investigates the microtubules directly with others reporting loss or mislocalization of microtubule-associated proteins. Though these are the general trends reported, there are clear disparities between model organisms and brain regions that are worthy of further investigation. Additionally, longitudinal studies of neuronal/cytoskeletal aging should also investigate whether these age-related changes contribute not just to vulnerability to disease but also to the decline in nervous system function and behavioral output that all organisms experience. This will highlight the utility, if any, of cytoskeletal fortification for the promotion of healthy neuronal aging and potential protection against age-related neurodegenerative disease. This review seeks to summarize what is currently known about the physiological aging of the neuron and microtubular cytoskeleton in the hope of uncovering mechanisms underpinning age-related risk to disease.
Collapse
Affiliation(s)
- Brad Richardson
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Thomas Goedert
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Shmma Quraishe
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Katrin Deinhardt
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Amritpal Mudher
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
3
|
Sagheddu C, Stojanovic T, Kouhnavardi S, Savchenko A, Hussein AM, Pistis M, Monje FJ, Plasenzotti R, Aufy M, Studenik CR, Lubec J, Lubec G. Cognitive performance in aged rats is associated with differences in distinctive neuronal populations in the ventral tegmental area and altered synaptic plasticity in the hippocampus. Front Aging Neurosci 2024; 16:1357347. [PMID: 38469164 PMCID: PMC10926450 DOI: 10.3389/fnagi.2024.1357347] [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: 12/17/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Deterioration of cognitive functions is commonly associated with aging, although there is wide variation in the onset and manifestation. Albeit heterogeneity in age-related cognitive decline has been studied at the cellular and molecular level, there is poor evidence for electrophysiological correlates. The aim of the current study was to address the electrophysiological basis of heterogeneity of cognitive functions in cognitively Inferior and Superior old (19-20 months) rats in the ventral tegmental area (VTA) and the hippocampus, having Young (12 weeks) rats as a control. The midbrain VTA operates as a hub amidst affective and cognitive facets, processing sensory inputs related to motivated behaviours and hippocampal memory. Increasing evidence shows direct dopaminergic and non-dopaminergic input from the VTA to the hippocampus. Methods Aged Superior and Inferior male rats were selected from a cohort of 88 animals based on their performance in a spatial learning and memory task. Using in vivo single-cell recording in the VTA, we examined the electrical activity of different neuronal populations (putative dopaminergic, glutamatergic and GABAergic neurons). In the same animals, basal synaptic transmission and synaptic plasticity were examined in hippocampal slices. Results Electrophysiological recordings from the VTA and hippocampus showed alterations associated with aging per se, together with differences specifically linked to the cognitive status of aged animals. In particular, the bursting activity of dopamine neurons was lower, while the firing frequency of glutamatergic neurons was higher in VTA of Inferior old rats. The response to high-frequency stimulation in hippocampal slices also discriminated between Superior and Inferior aged animals. Discussion This study provides new insight into electrophysiological information underlying compromised cerebral ageing. Further understanding of brain senescence, possibly related to neurocognitive decline, will help develop new strategies towards the preservation of a high quality of life.
Collapse
Affiliation(s)
- Claudia Sagheddu
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Shima Kouhnavardi
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Artem Savchenko
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
- Institute of Pharmacology, Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - Ahmed M. Hussein
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Zoology, Faculty of Science, Al-Azhar University, Asyut, Egypt
| | - Marco Pistis
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
- Section of Cagliari, Neuroscience Institute National Research Council of Italy (CNR), Cagliari, Italy
- Unit of Clinical Pharmacology, University Hospital, Cagliari, Italy
| | - Francisco J. Monje
- Center for Physiology and Pharmacology, Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Roberto Plasenzotti
- Division of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Jana Lubec
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| | - Gert Lubec
- Programme for Proteomics, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
4
|
Huffels CFM, van Dijk RE, Karst H, Meye FJ, Hol EM, Middeldorp J. Systemic Injection of Aged Blood Plasma in Adult C57BL/6 Mice Induces Neurophysiological Impairments in the Hippocampal CA1. J Alzheimers Dis 2022; 89:283-297. [PMID: 35871343 DOI: 10.3233/jad-220337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Aging is characterized by systemic alterations and forms an important risk factor for Alzheimer's disease. Recently, it has been indicated that blood-borne factors present in the systemic milieu contribute to the aging process. Exposing young mice to aged blood plasma results in impaired neurogenesis and synaptic plasticity in the dentate gyrus, as well as impaired cognition. Vice versa, treating aged mice with young blood plasma rescues impairments associated with aging. OBJECTIVE Whether blood-borne factors are sufficient to drive impairments outside the dentate gyrus, how they impact neurophysiology, and how the functional outcome compares to impairments found in mouse models for AD is still unclear. METHODS Here, we treated adult mice with blood plasma from aged mice and assessed neurophysiological parameters in the hippocampal CA1. RESULTS Mice treated with aged blood plasma show significantly impaired levels of long-term potentiation (LTP), similar to those present in APP/PS1 mice. These impaired levels of LTP in plasma-treated mice are associated with alterations in basic properties of glutamatergic transmission and the enhanced activity of voltage-gated Ca2 + channels. CONCLUSION Together, the data presented in this study show that blood-borne factors are sufficient to drive neurophysiological impairments in the hippocampal CA1.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Roland E van Dijk
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| |
Collapse
|
5
|
Moretti J, Rodger J. A little goes a long way: Neurobiological effects of low intensity rTMS and implications for mechanisms of rTMS. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100033. [PMID: 36685761 PMCID: PMC9846462 DOI: 10.1016/j.crneur.2022.100033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 01/25/2023] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a widespread technique in neuroscience and medicine, however its mechanisms are not well known. In this review, we consider intensity as a key therapeutic parameter of rTMS, and review the studies that have examined the biological effects of rTMS using magnetic fields that are orders of magnitude lower that those currently used in the clinic. We discuss how extensive characterisation of "low intensity" rTMS has set the stage for translation of new rTMS parameters from a mechanistic evidence base, with potential for innovative and effective therapeutic applications. Low-intensity rTMS demonstrates neurobiological effects across healthy and disease models, which include depression, injury and regeneration, abnormal circuit organisation, tinnitus etc. Various short and long-term changes to metabolism, neurotransmitter release, functional connectivity, genetic changes, cell survival and behaviour have been investigated and we summarise these key changes and the possible mechanisms behind them. Mechanisms at genetic, molecular, cellular and system levels have been identified with evidence that low-intensity rTMS and potentially rTMS in general acts through several key pathways to induce changes in the brain with modulation of internal calcium signalling identified as a major mechanism. We discuss the role that preclinical models can play to inform current clinical research as well as uncover new pathways for investigation.
Collapse
Affiliation(s)
- Jessica Moretti
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia,Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, WA, Australia,Perron Institute for Neurological and Translational Science, Perth, WA, Australia,Corresponding author. School of Biological Sciences M317, The University of Western Australia, 35 Stirling Highway, Crawley WA, 6009, Australia.
| |
Collapse
|
6
|
Tang AD, Bennett W, Bindoff AD, Bolland S, Collins J, Langley RC, Garry MI, Summers JJ, Hinder MR, Rodger J, Canty AJ. Subthreshold repetitive transcranial magnetic stimulation drives structural synaptic plasticity in the young and aged motor cortex. Brain Stimul 2021; 14:1498-1507. [PMID: 34653682 DOI: 10.1016/j.brs.2021.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/27/2021] [Accepted: 10/11/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive tool commonly used to drive neural plasticity in the young adult and aged brain. Recent data from mouse models have shown that even at subthreshold intensities (0.12 T), rTMS can drive neuronal and glial plasticity in the motor cortex. However, the physiological mechanisms underlying subthreshold rTMS induced plasticity and whether these are altered with normal ageing are unclear. OBJECTIVE To assess the effect of subthreshold rTMS, using the intermittent theta burst stimulation (iTBS) protocol on structural synaptic plasticity in the mouse motor cortex of young and aged mice. METHODS Longitudinal in vivo 2-photon microscopy was used to measure changes to the structural plasticity of pyramidal neuron dendritic spines in the motor cortex following a single train of subthreshold rTMS (in young adult and aged animals) or the same rTMS train administered on 4 consecutive days (in young adult animals only). Data were analysed with Bayesian hierarchical generalized linear regression models and interpreted with the aid of Bayes Factors (BF). RESULTS We found strong evidence (BF > 10) that subthreshold rTMS altered the rate of dendritic spine losses and gains, dependent on the number of stimulation sessions and that a single session of subthreshold rTMS was effective in driving structural synaptic plasticity in both young adult and aged mice. CONCLUSION These findings provide further evidence that rTMS drives synaptic plasticity in the brain and uncovers structural synaptic plasticity as a key mechanism of subthreshold rTMS induced plasticity.
Collapse
Affiliation(s)
- Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, 35 Stirling Highway (M317), Crawley, 6009, WA, Australia; Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, 6008, WA, Australia.
| | - William Bennett
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Private Bag 143, Hobart, 7001, TAS, Australia
| | - Aidan D Bindoff
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Private Bag 143, Hobart, 7001, TAS, Australia
| | - Samuel Bolland
- Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, 35 Stirling Highway (M317), Crawley, 6009, WA, Australia; Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, 6008, WA, Australia
| | - Jessica Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Private Bag 143, Hobart, 7001, TAS, Australia
| | - Ross C Langley
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Private Bag 143, Hobart, 7001, TAS, Australia
| | - Michael I Garry
- School of Psychological Sciences, College of Health and Medicine, University of Tasmania, Hobart, Australia. Private Bag 30, Hobart, 7001, TAS, Australia
| | - Jeffery J Summers
- School of Psychological Sciences, College of Health and Medicine, University of Tasmania, Hobart, Australia. Private Bag 30, Hobart, 7001, TAS, Australia; Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Tom Reilly Building, Byrom Street, L3 3AF, Liverpool, United Kingdom
| | - Mark R Hinder
- School of Psychological Sciences, College of Health and Medicine, University of Tasmania, Hobart, Australia. Private Bag 30, Hobart, 7001, TAS, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, 35 Stirling Highway (M317), Crawley, 6009, WA, Australia; Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, 6008, WA, Australia
| | - Alison J Canty
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Private Bag 143, Hobart, 7001, TAS, Australia
| |
Collapse
|
7
|
Ghoweri AO, Gagolewicz P, Frazier HN, Gant JC, Andrew RD, Bennett BM, Thibault O. Neuronal Calcium Imaging, Excitability, and Plasticity Changes in the Aldh2-/- Mouse Model of Sporadic Alzheimer's Disease. J Alzheimers Dis 2021; 77:1623-1637. [PMID: 32925058 PMCID: PMC7683088 DOI: 10.3233/jad-200617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Dysregulated signaling in neurons and astrocytes participates in pathophysiological alterations seen in the Alzheimer’s disease brain, including increases in amyloid-β, hyperphosphorylated tau, inflammation, calcium dysregulation, and oxidative stress. These are often noted prior to the development of behavioral, cognitive, and non-cognitive deficits. However, the extent to which these pathological changes function together or independently is unclear. Objective: Little is known about the temporal relationship between calcium dysregulation and oxidative stress, as some reports suggest that dysregulated calcium promotes increased formation of reactive oxygen species, while others support the opposite. Prior work has quantified several key outcome measures associated with oxidative stress in aldehyde dehydrogenase 2 knockout (Aldh2–/–) mice, a non-transgenic model of sporadic Alzheimer’s disease. Methods: Here, we tested the hypothesis that early oxidative stress can promote calcium dysregulation across aging by measuring calcium-dependent processes using electrophysiological and imaging methods and focusing on the afterhyperpolarization (AHP), synaptic activation, somatic calcium, and long-term potentiation in the Aldh2–/– mouse. Results: Our results show a significant age-related decrease in the AHP along with an increase in the slow AHP amplitude in Aldh2–/– animals. Measures of synaptic excitability were unaltered, although significant reductions in long-term potentiation maintenance were noted in the Aldh2–/– animals compared to wild-type. Conclusion: With so few changes in calcium and calcium-dependent processes in an animal model that shows significant increases in HNE adducts, Aβ, p-tau, and activated caspases across age, the current findings do not support a direct link between neuronal calcium dysregulation and uncontrolled oxidative stress.
Collapse
Affiliation(s)
- Adam O Ghoweri
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - Peter Gagolewicz
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Hilaree N Frazier
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - John C Gant
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - R David Andrew
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Brian M Bennett
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Olivier Thibault
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| |
Collapse
|
8
|
Sikora E, Bielak-Zmijewska A, Dudkowska M, Krzystyniak A, Mosieniak G, Wesierska M, Wlodarczyk J. Cellular Senescence in Brain Aging. Front Aging Neurosci 2021; 13:646924. [PMID: 33732142 PMCID: PMC7959760 DOI: 10.3389/fnagi.2021.646924] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/02/2021] [Indexed: 12/25/2022] Open
Abstract
Aging of the brain can manifest itself as a memory and cognitive decline, which has been shown to frequently coincide with changes in the structural plasticity of dendritic spines. Decreased number and maturity of spines in aged animals and humans, together with changes in synaptic transmission, may reflect aberrant neuronal plasticity directly associated with impaired brain functions. In extreme, a neurodegenerative disease, which completely devastates the basic functions of the brain, may develop. While cellular senescence in peripheral tissues has recently been linked to aging and a number of aging-related disorders, its involvement in brain aging is just beginning to be explored. However, accumulated evidence suggests that cell senescence may play a role in the aging of the brain, as it has been documented in other organs. Senescent cells stop dividing and shift their activity to strengthen the secretory function, which leads to the acquisition of the so called senescence-associated secretory phenotype (SASP). Senescent cells have also other characteristics, such as altered morphology and proteostasis, decreased propensity to undergo apoptosis, autophagy impairment, accumulation of lipid droplets, increased activity of senescence-associated-β-galactosidase (SA-β-gal), and epigenetic alterations, including DNA methylation, chromatin remodeling, and histone post-translational modifications that, in consequence, result in altered gene expression. Proliferation-competent glial cells can undergo senescence both in vitro and in vivo, and they likely participate in neuroinflammation, which is characteristic for the aging brain. However, apart from proliferation-competent glial cells, the brain consists of post-mitotic neurons. Interestingly, it has emerged recently, that non-proliferating neuronal cells present in the brain or cultivated in vitro can also have some hallmarks, including SASP, typical for senescent cells that ceased to divide. It has been documented that so called senolytics, which by definition, eliminate senescent cells, can improve cognitive ability in mice models. In this review, we ask questions about the role of senescent brain cells in brain plasticity and cognitive functions impairments and how senolytics can improve them. We will discuss whether neuronal plasticity, defined as morphological and functional changes at the level of neurons and dendritic spines, can be the hallmark of neuronal senescence susceptible to the effects of senolytics.
Collapse
Affiliation(s)
- Ewa Sikora
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Anna Bielak-Zmijewska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Magdalena Dudkowska
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Adam Krzystyniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Grazyna Mosieniak
- Laboratory of Molecular Bases of Aging, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Malgorzata Wesierska
- Laboratory of Neuropsychology, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, PAS, Warsaw, Poland
| |
Collapse
|
9
|
The aging mouse brain: cognition, connectivity and calcium. Cell Calcium 2021; 94:102358. [PMID: 33517250 DOI: 10.1016/j.ceca.2021.102358] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023]
Abstract
Aging is a complex process that differentially impacts multiple cognitive, sensory, neuronal and molecular processes. Technological innovations now allow for parallel investigation of neuronal circuit function, structure and molecular composition in the brain of awake behaving adult mice. Thus, mice have become a critical tool to better understand how aging impacts the brain. However, a more granular systems-based approach, which considers the impact of age on key features relating to neural processing, is required. Here, we review evidence probing the impact of age on the mouse brain. We focus on a range of processes relating to neuronal function, including cognitive abilities, sensory systems, synaptic plasticity and calcium regulation. Across many systems, we find evidence for prominent age-related dysregulation even before 12 months of age, suggesting that emerging age-related alterations can manifest by late adulthood. However, we also find reports suggesting that some processes are remarkably resilient to aging. The evidence suggests that aging does not drive a parallel, linear dysregulation of all systems, but instead impacts some processes earlier, and more severely, than others. We propose that capturing the more fine-scale emerging features of age-related vulnerability and resilience may provide better opportunities for the rejuvenation of the aged brain.
Collapse
|
10
|
Ghoweri AO, Ouillette L, Frazier HN, Anderson KL, Lin RL, Gant JC, Parent R, Moore S, Murphy GG, Thibault O. Electrophysiological and Imaging Calcium Biomarkers of Aging in Male and Female 5×FAD Mice. J Alzheimers Dis 2020; 78:1419-1438. [PMID: 33164928 PMCID: PMC7836067 DOI: 10.3233/jad-200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND In animal models and tissue preparations, calcium dyshomeostasis is a biomarker of aging and Alzheimer's disease that is associated with synaptic dysfunction, neuritic pruning, and dysregulated cellular processes. It is unclear, however, whether the onset of calcium dysregulation precedes, is concurrent with, or is the product of pathological cellular events (e.g., oxidation, amyloid-β production, and neuroinflammation). Further, neuronal calcium dysregulation is not always present in animal models of amyloidogenesis, questioning its reliability as a disease biomarker. OBJECTIVE Here, we directly tested for the presence of calcium dysregulation in dorsal hippocampal neurons in male and female 5×FAD mice on a C57BL/6 genetic background using sharp electrodes coupled with Oregon-green Bapta-1 imaging. We focused on three ages that coincide with the course of amyloid deposition: 1.5, 4, and 10 months old. METHODS Outcome variables included measures of the afterhyperpolarization, short-term synaptic plasticity, and calcium kinetics during synaptic activation. Quantitative analyses of spatial learning and memory were also conducted using the Morris water maze. Main effects of sex, age, and genotype were identified on measures of electrophysiology and calcium imaging. RESULTS Measures of resting Oregon-green Bapta-1 fluorescence showed significant reductions in the 5×FAD group compared to controls. Deficits in spatial memory, along with increases in Aβ load, were detectable at older ages, allowing us to test for temporal associations with the onset of calcium dysregulation. CONCLUSION Our results provide evidence that reduced, rather than elevated, neuronal calcium is identified in this 5×FAD model and suggests that this surprising result may be a novel biomarker of AD.
Collapse
Affiliation(s)
- Adam O Ghoweri
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lara Ouillette
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hilaree N Frazier
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Katie L Anderson
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ruei-Lung Lin
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - John C Gant
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Rachel Parent
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Shannon Moore
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey G Murphy
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olivier Thibault
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
11
|
Ali A, Shah SA, Zaman N, Uddin MN, Khan W, Ali A, Riaz M, Kamil A. Vitamin D exerts neuroprotection via SIRT1/nrf-2/ NF-kB signaling pathways against D-galactose-induced memory impairment in adult mice. Neurochem Int 2020; 142:104893. [PMID: 33159979 DOI: 10.1016/j.neuint.2020.104893] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/16/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
Vitamin D (Vt. D) is one of the vital hormone having multiple functions in various tissues, including brain. Several evidences reported that Vt. D plays a significant part in memory and cognition as its inadequate amount may accelerate cognitive impairment. This study shows for the first time the antioxidant potential of Vt. D against D-Galactose (D-gal) induced oxidative stress mediated Alzheimer disease (AD) pathology in male adult albino mice. The result reveals that the mice exposed to D-gal (120 mg/kg) for eight weeks have pre-and post-synaptic dysfunction and impaired memory investigated through Morris water maze and Y-maze tests. This is followed by the suppressed Nuclear factor erythroid 2-related factor 2 (NRF2), Heme Oxygenase-1 (HO-1) and elevated expressions of Nuclear Factor kappa B (NF-kB), Tumor Necrosis Factor alpha (TNF-α) and Interleukin 1 beta (IL-1β) proteins in the brain homogenates evaluated through western blotting technique. On the other hand Vt. D (100 μg/kg) administration (three times a week for 4 weeks) activated Silent mating type information regulation 2 homolog 1 (SIRT1) and significantly improved both the neuronal synapse and memory, reduced oxidative stress by upregulating NRF-2 and HO-1 and downregulating NF-kB, TNF-α and IL-1β proteins expression. Most importantly, Vt. D significantly abrogate the amyloidogenic pathway of amyloid beta (Aβ) production against D-gal in the brains of adult male albino mice. These results reveal that Vt. D being an antioxidant agent plays a vital role in reducing the AD pathophysiology in D-gal induced animal model of aging, therefore act as a potential drug candidate in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ammar Ali
- Department of Biotechnology, Abdul Wali Khan University, Mardan, Pakistan
| | - Shahid Ali Shah
- Department of Chemistry, Sarhad University of Information and Technology, Peshawar, Pakistan; Neuro Molecular Medicine Research Center (NMMRC), Ring Road, Peshawar, Pakistan
| | - Nasib Zaman
- Center for Biotechnology and Microbiology, University of Swat, Pakistan
| | | | - Wajid Khan
- Center for Biotechnology and Microbiology, University of Swat, Pakistan
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
| | - Muhammad Riaz
- Department of Biochemistry, Abdul Wali Khan University, Mardan, Pakistan
| | - Atif Kamil
- Department of Biotechnology, Abdul Wali Khan University, Mardan, Pakistan.
| |
Collapse
|
12
|
Ploux E, Freret T, Billard JM. d-serine in physiological and pathological brain aging. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140542. [PMID: 32950692 DOI: 10.1016/j.bbapap.2020.140542] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 01/24/2023]
Abstract
Among aging-induced impairments, those affecting cognitive functions certainly represent one the most major challenge to face to improve elderly quality of life. In last decades, our knowledge on changes in the morphology and function of neuronal networks associated with normal and pathological brain aging has rapidly progressed, initiating the development of different pharmacological and behavioural strategies to alleviate cognitive aging. In particular, experimental evidences have accumulated indicating that the communication between neurons and its plasticity gradually weakens with aging. Because of its pivotal role for brain functional plasticity, the N-Methyl‑d-Aspartate receptor subtype of glutamate receptors (NMDAr) has gathered much of the experimental interest. NMDAr activation is regulated by many mechanisms. Among is the mandatory binding of a co-agonist, such as the amino acid d-serine, in order to activate NMDAr. This mini-review presents the most recent information indicating how d-serine could contribute to mechanisms of physiological cognitive aging and also considers the divergent views relative of the role of the NMDAr co-agonist in Alzheimer's disease.
Collapse
Affiliation(s)
- E Ploux
- Normandie Univ, UNICAEN, INSERM, CYCERON, COMETE, 14000 Caen, France.
| | - T Freret
- Normandie Univ, UNICAEN, INSERM, CYCERON, COMETE, 14000 Caen, France
| | - J-M Billard
- Normandie Univ, UNICAEN, INSERM, CYCERON, COMETE, 14000 Caen, France.
| |
Collapse
|
13
|
Short AK, Baram TZ. Early-life adversity and neurological disease: age-old questions and novel answers. Nat Rev Neurol 2019; 15:657-669. [PMID: 31530940 PMCID: PMC7261498 DOI: 10.1038/s41582-019-0246-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Neurological illnesses, including cognitive impairment, memory decline and dementia, affect over 50 million people worldwide, imposing a substantial burden on individuals and society. These disorders arise from a combination of genetic, environmental and experiential factors, with the latter two factors having the greatest impact during sensitive periods in development. In this Review, we focus on the contribution of adverse early-life experiences to aberrant brain maturation, which might underlie vulnerability to cognitive brain disorders. Specifically, we draw on recent robust discoveries from diverse disciplines, encompassing human studies and experimental models. These discoveries suggest that early-life adversity, especially in the perinatal period, influences the maturation of brain circuits involved in cognition. Importantly, new findings suggest that fragmented and unpredictable environmental and parental signals comprise a novel potent type of adversity, which contributes to subsequent vulnerabilities to cognitive illnesses via mechanisms involving disordered maturation of brain 'wiring'.
Collapse
Affiliation(s)
- Annabel K Short
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy and Neruobiology, University of California-Irvine, Irvine, CA, USA.
- Departments of Pediatrics, University of California-Irvine, Irvine, CA, USA.
- Departments of Neurology, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|
14
|
Temido-Ferreira M, Coelho JE, Pousinha PA, Lopes LV. Novel Players in the Aging Synapse: Impact on Cognition. J Caffeine Adenosine Res 2019; 9:104-127. [PMID: 31559391 PMCID: PMC6761599 DOI: 10.1089/caff.2019.0013] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While neuronal loss has long been considered as the main contributor to age-related cognitive decline, these alterations are currently attributed to gradual synaptic dysfunction driven by calcium dyshomeostasis and alterations in ionotropic/metabotropic receptors. Given the key role of the hippocampus in encoding, storage, and retrieval of memory, the morpho- and electrophysiological alterations that occur in the major synapse of this network-the glutamatergic-deserve special attention. We guide you through the hippocampal anatomy, circuitry, and function in physiological context and focus on alterations in neuronal morphology, calcium dynamics, and plasticity induced by aging and Alzheimer's disease (AD). We provide state-of-the art knowledge on glutamatergic transmission and discuss implications of these novel players for intervention. A link between regular consumption of caffeine-an adenosine receptor blocker-to decreased risk of AD in humans is well established, while the mechanisms responsible have only now been uncovered. We review compelling evidence from humans and animal models that implicate adenosine A2A receptors (A2AR) upsurge as a crucial mediator of age-related synaptic dysfunction. The relevance of this mechanism in patients was very recently demonstrated in the form of a significant association of the A2AR-encoding gene with hippocampal volume (synaptic loss) in mild cognitive impairment and AD. Novel pathways implicate A2AR in the control of mGluR5-dependent NMDAR activation and subsequent Ca2+ dysfunction upon aging. The nature of this receptor makes it particularly suited for long-term therapies, as an alternative for regulating aberrant mGluR5/NMDAR signaling in aging and disease, without disrupting their crucial constitutive activity.
Collapse
Affiliation(s)
- Mariana Temido-Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana E. Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Paula A. Pousinha
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRS UMR7275, Université Côte d'Azur, Valbonne, France
| | - Luísa V. Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
15
|
Kelly SP, Dawson-Scully K. Natural polymorphism in protein kinase G modulates functional senescence in D rosophila melanogaster. J Exp Biol 2019; 222:jeb.199364. [PMID: 30910834 DOI: 10.1242/jeb.199364] [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: 01/08/2019] [Accepted: 03/19/2019] [Indexed: 11/20/2022]
Abstract
The common fruit fly, Drosophila melanogaster, is a well-characterized model for neurological disorders and is widely used to investigate the biology of aging, stress tolerance and pleiotropy. The foraging (for) gene encodes a cGMP-dependent protein kinase (PKG), which has been implicated in several behavioral phenotypes including feeding, sleep, learning and memory, and environmental stress tolerance. We used the well-established Drosophila activity monitor (DAM) to investigate the effects of the conserved NO/cGMP/PKG signaling pathway on functional senescence. Our results show that the polymorphic for gene confers protection during low oxygen stress at the expense of longevity and a decline in locomotor activity with age in D. melanogaster, which suggests a novel role for the PKG pathway in healthy aging and senescence.
Collapse
Affiliation(s)
- Stephanie P Kelly
- Florida Atlantic University, Department of Biological Sciences, Boca Raton, FL 33431, USA
| | - Ken Dawson-Scully
- Florida Atlantic University, Department of Biological Sciences, Boca Raton, FL 33431, USA
| |
Collapse
|
16
|
Tsai SF, Ku NW, Wang TF, Yang YH, Shih YH, Wu SY, Lee CW, Yu M, Yang TT, Kuo YM. Long-Term Moderate Exercise Rescues Age-Related Decline in Hippocampal Neuronal Complexity and Memory. Gerontology 2018; 64:551-561. [DOI: 10.1159/000488589] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/20/2018] [Indexed: 11/19/2022] Open
Abstract
Background: Aging impairs hippocampal neuroplasticity and hippocampus-related learning and memory. In contrast, exercise training is known to improve hippocampal neuronal function. However, whether exercise is capable of restoring memory function in old animals is less clear. Objective: Here, we investigated the effects of exercise on the hippocampal neuroplasticity and memory functions during aging. Methods: Young (3 months), middle-aged (9–12 months), and old (18 months) mice underwent moderate-intensity treadmill running training for 6 weeks, and their hippocampus-related learning and memory, and the plasticity of their CA1 neurons was evaluated. Results: The memory performance (Morris water maze and novel object recognition tests), and dendritic complexity (branch and length) and spine density of their hippocampal CA1 neurons decreased as their age increased. The induction and maintenance of high-frequency stimulation-induced long-term potentiation in the CA1 area and the expressions of neuroplasticity-related proteins were not affected by age. Treadmill running increased CA1 neuron long-term potentiation and dendritic complexity in all three age groups, and it restored the learning and memory ability in middle-aged and old mice. Furthermore, treadmill running upregulated the hippocampal expressions of brain-derived neurotrophic factor and monocarboxylate transporter-4 in middle-aged mice, glutamine synthetase in old mice, and full-length TrkB in middle-aged and old mice. Conclusion: The hippocampus-related memory function declines from middle age, but long-term moderate-intensity running effectively increased hippocampal neuroplasticity and memory in mice of different ages, even when the memory impairment had progressed to an advanced stage. Thus, long-term, moderate intensity exercise training might be a way of delaying and treating aging-related memory decline.
Collapse
|
17
|
McKiernan EC, Marrone DF. CA1 pyramidal cells have diverse biophysical properties, affected by development, experience, and aging. PeerJ 2017; 5:e3836. [PMID: 28948109 PMCID: PMC5609525 DOI: 10.7717/peerj.3836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/31/2017] [Indexed: 12/04/2022] Open
Abstract
Neuron types (e.g., pyramidal cells) within one area of the brain are often considered homogeneous, despite variability in their biophysical properties. Here we review literature demonstrating variability in the electrical activity of CA1 hippocampal pyramidal cells (PCs), including responses to somatic current injection, synaptic stimulation, and spontaneous network-related activity. In addition, we describe how responses of CA1 PCs vary with development, experience, and aging, and some of the underlying ionic currents responsible. Finally, we suggest directions that may be the most impactful in expanding this knowledge, including the use of text and data mining to systematically study cellular heterogeneity in more depth; dynamical systems theory to understand and potentially classify neuron firing patterns; and mathematical modeling to study the interaction between cellular properties and network output. Our goals are to provide a synthesis of the literature for experimentalists studying CA1 PCs, to give theorists an idea of the rich diversity of behaviors models may need to reproduce to accurately represent these cells, and to provide suggestions for future research.
Collapse
Affiliation(s)
- Erin C McKiernan
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Diano F Marrone
- Department of Psychology, Wilfrid Laurier University, Waterloo, Ontario, Canada.,McKnight Brain Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
18
|
Lester AW, Moffat SD, Wiener JM, Barnes CA, Wolbers T. The Aging Navigational System. Neuron 2017; 95:1019-1035. [PMID: 28858613 PMCID: PMC5659315 DOI: 10.1016/j.neuron.2017.06.037] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/17/2022]
Abstract
The discovery of neuronal systems dedicated to computing spatial information, composed of functionally distinct cell types such as place and grid cells, combined with an extensive body of human-based behavioral and neuroimaging research has provided us with a detailed understanding of the brain's navigation circuit. In this review, we discuss emerging evidence from rodents, non-human primates, and humans that demonstrates how cognitive aging affects the navigational computations supported by these systems. Critically, we show 1) that navigational deficits cannot solely be explained by general deficits in learning and memory, 2) that there is no uniform decline across different navigational computations, and 3) that navigational deficits might be sensitive markers for impending pathological decline. Following an introduction to the mechanisms underlying spatial navigation and how they relate to general processes of learning and memory, the review discusses how aging affects the perception and integration of spatial information, the creation and storage of memory traces for spatial information, and the use of spatial information during navigational behavior. The closing section highlights the clinical potential of behavioral and neural markers of spatial navigation, with a particular emphasis on neurodegenerative disorders.
Collapse
Affiliation(s)
- Adam W Lester
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, USA; Division of Neural Systems, Memory and Aging, University of Arizona, Tucson, AZ 85721, USA
| | - Scott D Moffat
- School of Psychology, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Jan M Wiener
- Department of Psychology, Ageing and Dementia Institute, Bournemouth University, Poole BH12 5BB, UK
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, USA; Division of Neural Systems, Memory and Aging, University of Arizona, Tucson, AZ 85721, USA; Departments of Psychology, Neurology, and Neuroscience, University of Arizona, Tucson, AZ 85721, USA
| | - Thomas Wolbers
- German Center for Neurodegenerative Diseases (DZNE), Aging and Cognition Research Group, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39118 Magdeburg, Germany.
| |
Collapse
|
19
|
Peña-Altamira E, Petralla S, Massenzio F, Virgili M, Bolognesi ML, Monti B. Nutritional and Pharmacological Strategies to Regulate Microglial Polarization in Cognitive Aging and Alzheimer's Disease. Front Aging Neurosci 2017. [PMID: 28638339 PMCID: PMC5461295 DOI: 10.3389/fnagi.2017.00175] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The study of microglia, the immune cells of the brain, has experienced a renaissance after the discovery of microglia polarization. In fact, the concept that activated microglia can shift into the M1 pro-inflammatory or M2 neuroprotective phenotypes, depending on brain microenvironment, has completely changed the understanding of microglia in brain aging and neurodegenerative diseases. Microglia polarization is particularly important in aging since an increased inflammatory status of body compartments, including the brain, has been reported in elderly people. In addition, inflammatory markers, mainly derived from activated microglia, are widely present in neurodegenerative diseases. Microglial inflammatory dysfunction, also linked to microglial senescence, has been extensively demonstrated and associated with cognitive impairment in neuropathological conditions related to aging. In fact, microglia polarization is known to influence cognitive function and has therefore become a main player in neurodegenerative diseases leading to dementia. As the life span of human beings increases, so does the prevalence of cognitive dysfunction. Thus, therapeutic strategies aimed to modify microglia polarization are currently being developed. Pharmacological approaches able to shift microglia from M1 pro-inflammatory to M2 neuroprotective phenotype are actually being studied, by acting on many different molecular targets, such as glycogen synthase kinase-3 (GSK3) β, AMP-activated protein kinase (AMPK), histone deacetylases (HDACs), etc. Furthermore, nutritional approaches can also modify microglia polarization and, consequently, impact cognitive function. Several bioactive compounds normally present in foods, such as polyphenols, can have anti-inflammatory effects on microglia. Both pharmacological and nutritional approaches seem to be promising, but still need further development. Here we review recent data on these approaches and propose that their combination could have a synergistic effect to counteract cognitive aging impairment and Alzheimer's disease (AD) through immunomodulation of microglia polarization, i.e., by driving the shift of activated microglia from the pro-inflammatory M1 to the neuroprotective M2 phenotype.
Collapse
Affiliation(s)
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Marco Virgili
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Maria L Bolognesi
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of BolognaBologna, Italy
| |
Collapse
|
20
|
Bettio LEB, Rajendran L, Gil-Mohapel J. The effects of aging in the hippocampus and cognitive decline. Neurosci Biobehav Rev 2017; 79:66-86. [PMID: 28476525 DOI: 10.1016/j.neubiorev.2017.04.030] [Citation(s) in RCA: 404] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/15/2017] [Accepted: 04/10/2017] [Indexed: 02/06/2023]
Abstract
Aging is a natural process that is associated with cognitive decline as well as functional and social impairments. One structure of particular interest when considering aging and cognitive decline is the hippocampus, a brain region known to play an important role in learning and memory consolidation as well as in affective behaviours and mood regulation, and where both functional and structural plasticity (e.g., neurogenesis) occur well into adulthood. Neurobiological alterations seen in the aging hippocampus including increased oxidative stress and neuroinflammation, altered intracellular signalling and gene expression, as well as reduced neurogenesis and synaptic plasticity, are thought to be associated with age-related cognitive decline. Non-invasive strategies such as caloric restriction, physical exercise, and environmental enrichment have been shown to counteract many of the age-induced alterations in hippocampal signalling, structure, and function. Thus, such approaches may have therapeutic value in counteracting the deleterious effects of aging and protecting the brain against age-associated neurodegenerative processes.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Luckshi Rajendran
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; UBC Island Medical program, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
21
|
Passow S, Thurm F, Li SC. Activating Developmental Reserve Capacity Via Cognitive Training or Non-invasive Brain Stimulation: Potentials for Promoting Fronto-Parietal and Hippocampal-Striatal Network Functions in Old Age. Front Aging Neurosci 2017; 9:33. [PMID: 28280465 PMCID: PMC5322263 DOI: 10.3389/fnagi.2017.00033] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/08/2017] [Indexed: 01/06/2023] Open
Abstract
Existing neurocomputational and empirical data link deficient neuromodulation of the fronto-parietal and hippocampal-striatal circuitries with aging-related increase in processing noise and declines in various cognitive functions. Specifically, the theory of aging neuronal gain control postulates that aging-related suboptimal neuromodulation may attenuate neuronal gain control, which yields computational consequences on reducing the signal-to-noise-ratio of synaptic signal transmission and hampering information processing within and between cortical networks. Intervention methods such as cognitive training and non-invasive brain stimulation, e.g., transcranial direct current stimulation (tDCS), have been considered as means to buffer cognitive functions or delay cognitive decline in old age. However, to date the reported effect sizes of immediate training gains and maintenance effects of a variety of cognitive trainings are small to moderate at best; moreover, training-related transfer effects to non-trained but closely related (i.e., near-transfer) or other (i.e., far-transfer) cognitive functions are inconsistent or lacking. Similarly, although applying different tDCS protocols to reduce aging-related cognitive impairments by inducing temporary changes in cortical excitability seem somewhat promising, evidence of effects on short- and long-term plasticity is still equivocal. In this article, we will review and critically discuss existing findings of cognitive training- and stimulation-related behavioral and neural plasticity effects in the context of cognitive aging, focusing specifically on working memory and episodic memory functions, which are subserved by the fronto-parietal and hippocampal-striatal networks, respectively. Furthermore, in line with the theory of aging neuronal gain control we will highlight that developing age-specific brain stimulation protocols and the concurrent applications of tDCS during cognitive training may potentially facilitate short- and long-term cognitive and brain plasticity in old age.
Collapse
Affiliation(s)
- Susanne Passow
- Chair of Lifespan Developmental Neuroscience, Department of Psychology, TU Dresden Dresden, Germany
| | - Franka Thurm
- Chair of Lifespan Developmental Neuroscience, Department of Psychology, TU Dresden Dresden, Germany
| | - Shu-Chen Li
- Chair of Lifespan Developmental Neuroscience, Department of Psychology, TU Dresden Dresden, Germany
| |
Collapse
|
22
|
Xiong M, Jones OD, Peppercorn K, Ohline SM, Tate WP, Abraham WC. Secreted amyloid precursor protein-alpha can restore novel object location memory and hippocampal LTP in aged rats. Neurobiol Learn Mem 2017; 138:291-299. [DOI: 10.1016/j.nlm.2016.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/27/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
|
23
|
Shaban H, O’Connor R, Ovsepian SV, Dinan TG, Cryan JF, Schellekens H. Electrophysiological approaches to unravel the neurobiological basis of appetite and satiety: use of the multielectrode array as a screening strategy. Drug Discov Today 2017; 22:31-42. [DOI: 10.1016/j.drudis.2016.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/03/2016] [Accepted: 09/06/2016] [Indexed: 01/10/2023]
|
24
|
Pinho J, Vale R, Batalha VL, Costenla AR, Dias R, Rombo D, Sebastião AM, de Mendonça A, Diógenes MJ. Enhanced LTP in aged rats: Detrimental or compensatory? Neuropharmacology 2016; 114:12-19. [PMID: 27889488 DOI: 10.1016/j.neuropharm.2016.11.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022]
Abstract
Age-dependent memory deterioration has been well documented and yet an increase in rat hippocampal LTP upon aging has been reported. This poses the question of whether the enhanced LTP is a cause or an attempt to compensate the memory deficits described in aged rats. Hippocampal slices from young, adult and aged Wistar rats were pre-incubated, with an NMDA receptor (NMDAR) antagonist, memantine (1 μM, 4 h), and hippocampal LTP was evaluated. The results show that memantine significantly decreases the larger LTP magnitude recorded in hippocampal slices from aged rats without compromising LTP recorded in slices from young and adult animals. To unveil the impact of in vivo administration of memantine, different doses (1, 5 and 10 mg/kg/day) or saline vehicle solution were intraperitoneally administered, for 15-20 days, to both young and aged animals. Memantine did not significantly affect neither the place learning of young animals, evaluated by Morris Water Maze, nor LTP recorded from hippocampal slices from the same group of animals. However, memantine (5 and 10 mg/kg/day) significantly decreased the large LTP recorded in hippocampal slices from aged animals. Moreover, aged animals treated with memantine (10 mg/kg/day) showed a significantly compromised place learning when compared to aged control animals. Overall, these results suggest that the larger LTP observed in aged animals is a compensatory phenomenon, rather than pathological. The finding that age-dependent blockade of LTP by a NMDAR antagonist leads to learning deficits, implies that the increased LTP observed upon aging may be playing an important role in the learning process.
Collapse
Affiliation(s)
- Júlia Pinho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Ruben Vale
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Vânia L Batalha
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Ana Rita Costenla
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Raquel Dias
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Diogo Rombo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Alexandre de Mendonça
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, Portugal.
| |
Collapse
|
25
|
Yu XW, Oh MM, Disterhoft JF. CREB, cellular excitability, and cognition: Implications for aging. Behav Brain Res 2016; 322:206-211. [PMID: 27478142 DOI: 10.1016/j.bbr.2016.07.042] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/11/2016] [Accepted: 07/27/2016] [Indexed: 10/21/2022]
Abstract
Humans and laboratory animals display cognitive deficits as they age. However, there are currently no effective therapies available to treat these deficits, as the underlying mechanisms are poorly understood. Studies using pharmacological compounds have found a link between cognitive performance and the intrinsic cellular excitability of CA1 hippocampal neurons. Therefore, it is of great interest to identify molecular regulators that may be influencing both cognition and neuronal excitability, which could be changed with age. One possible regulator is the transcription factor cAMP response element binding-protein (CREB). In young adult animals, manipulation of CREB activity has resulted in modulation of both cognitive performance on behavioral tasks, and neuronal excitability. While evidence is sparse, studies also point to a dysfunction in CREB signaling with aging. We propose that CREB may be a viable therapeutic target for the treatment of age-related cognitive deficits, along with potential experiments to test this hypothesis.
Collapse
Affiliation(s)
- Xiao-Wen Yu
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA.
| | - M Matthew Oh
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA.
| | - John F Disterhoft
- Department of Physiology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
26
|
Stein LR, O'Dell KA, Funatsu M, Zorumski CF, Izumi Y. Short-term environmental enrichment enhances synaptic plasticity in hippocampal slices from aged rats. Neuroscience 2016; 329:294-305. [PMID: 27208617 DOI: 10.1016/j.neuroscience.2016.05.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 12/26/2022]
Abstract
Age-associated changes in cognition are mirrored by impairments in cellular models of memory and learning, such as long-term potentiation (LTP) and long-term depression (LTD). In young rodents, environmental enrichment (EE) can enhance memory, alter LTP and LTD, as well as reverse cognitive deficits induced by aging. Whether short-term EE can benefit cognition and synaptic plasticity in aged rodents is unclear. Here, we tested if short-term EE could overcome age-associated impairments in induction of LTP and LTD. LTP and LTD could not be induced in the CA1 region of hippocampal slices in control, aged rats using standard stimuli that are highly effective in young rats. However, exposure of aged littermates to EE for three weeks enabled successful induction of LTP and LTD. EE-facilitated LTP was dependent upon N-methyl-d-aspartate receptors (NMDARs). These alterations in synaptic plasticity occurred with elevated levels of phosphorylated cAMP response element-binding protein and vascular endothelial growth factor, but in the absence of changes in several other synaptic and cellular markers. Importantly, our study suggests that even a relatively short period of EE is sufficient to alter synaptic plasticity and molecular markers linked to cognitive function in aged animals.
Collapse
Affiliation(s)
- Liana R Stein
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Kazuko A O'Dell
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Michiyo Funatsu
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Center for Brain Research in Mood Disorders, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Center for Brain Research in Mood Disorders, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
27
|
Tsai SF, Chen PC, Calkins MJ, Wu SY, Kuo YM. Exercise Counteracts Aging-Related Memory Impairment: A Potential Role for the Astrocytic Metabolic Shuttle. Front Aging Neurosci 2016; 8:57. [PMID: 27047373 PMCID: PMC4801859 DOI: 10.3389/fnagi.2016.00057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/08/2016] [Indexed: 01/19/2023] Open
Abstract
Age-related cognitive impairment has become one of the most common health threats in many countries. The biological substrate of cognition is the interconnection of neurons to form complex information processing networks. Experience-based alterations in the activities of these information processing networks lead to neuroadaptation, which is physically represented at the cellular level as synaptic plasticity. Although synaptic plasticity is known to be affected by aging, the underlying molecular mechanisms are not well described. Astrocytes, a glial cell type that is infrequently investigated in cognitive science, have emerged as energy suppliers which are necessary for meeting the abundant energy demand resulting from glutamatergic synaptic activity. Moreover, the concerted action of an astrocyte-neuron metabolic shuttle is essential for cognitive function; whereas, energetic incoordination between astrocytes and neurons may contribute to cognitive impairment. Whether altered function of the astrocyte-neuron metabolic shuttle links aging to reduced synaptic plasticity is unexplored. However, accumulated evidence documents significant beneficial effects of long-term, regular exercise on cognition and synaptic plasticity. Furthermore, exercise increases the effectiveness of astrocyte-neuron metabolic shuttle by upregulation of astrocytic lactate transporter levels. This review summarizes previous findings related to the neuronal activity-dependent astrocyte-neuron metabolic shuttle. Moreover, we discuss how aging and exercise may shape the astrocyte-neuron metabolic shuttle in cognition-associated brain areas.
Collapse
Affiliation(s)
- Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Marcus J Calkins
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Shih-Ying Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| |
Collapse
|
28
|
Patel R, Sesti F. Oxidation of ion channels in the aging nervous system. Brain Res 2016; 1639:174-85. [PMID: 26947620 DOI: 10.1016/j.brainres.2016.02.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 12/19/2022]
Abstract
Ion channels are integral membrane proteins that allow passive diffusion of ions across membranes. In neurons and in other excitable cells, the harmonious coordination between the numerous types of ion channels shape and propagate electrical signals. Increased accumulation of reactive oxidative species (ROS), and subsequent oxidation of proteins, including ion channels, is a hallmark feature of aging and may contribute to cell failure as a result. In this review we discuss the effects of ROS on three major types of ion channels of the central nervous system, namely the potassium (K(+)), calcium (Ca(2+)) and sodium (Na(+)) channels. We examine two general mechanisms through which ROS affect ion channels: via direct oxidation of specific residues and via indirect interference of pathways that regulate the channels. The overall status of the present studies indicates that the interaction of ion channels with ROS is multimodal and pervasive in the central nervous system and likely constitutes a general mechanism of aging susceptibility.
Collapse
Affiliation(s)
- Rahul Patel
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, 683 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, 683 Hoes Lane West, Piscataway, NJ 08854, USA.
| |
Collapse
|
29
|
Fernández-Fernández D, Dorner-Ciossek C, Kroker KS, Rosenbrock H. Age-related synaptic dysfunction in Tg2576 mice starts as a failure in early long-term potentiation which develops into a full abolishment of late long-term potentiation. J Neurosci Res 2015; 94:266-81. [PMID: 26629777 DOI: 10.1002/jnr.23701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 11/12/2022]
Abstract
Tg2576 mice are widely used to study amyloid-dependent synaptic dysfunction related to Alzheimer's disease. However, conflicting data have been reported for these mice with regard to basal transmission as well as the in vitro correlate of memory, long-term potentiation (LTP). Some studies show clear impairments, whereas others report no deficiency. The present study uses hippocampal slices from 3-, 10-, and 15-month-old wild-type (WT) and Tg2576 mice to evaluate synaptic function in each group, including experiments to investigate basal synaptic transmission, short- and long-term plasticity by inducing paired-pulse facilitation, and both early and late LTP. We show that synaptic function remains intact in hippocampal slices from Tg2576 mice at 3 months of age. However, both early and late LTP decline progressively during aging in these mice. This deterioration of synaptic plasticity starts affecting early LTP, ultimately leading to the abolishment of both forms of LTP in 15-month-old animals. In comparison, WT littermates display normal synaptic parameters during aging. Additional pharmacological investigation into the involvement of NMDA receptors and L-type voltage-gated calcium channels in LTP suggests a distinct mechanism of induction among age groups, demonstrating that both early and late LTP are differentially affected by these channels in Tg2576 mice during aging.
Collapse
Affiliation(s)
- Diego Fernández-Fernández
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Cornelia Dorner-Ciossek
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Katja S Kroker
- Deptartment of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Holger Rosenbrock
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| |
Collapse
|
30
|
Abstract
Senescence-accelerated mouse (SAM) is an experimental model animal showing a short lifespan and rapid advancement of senescence. Especially, SAM prone 8 (SAMP8) shows age-related impairment of learning and memory, and thus, it is a good model for age-related cognitive function. However, the synaptic characteristics related to cognitive function of SAMP8 have been poorly understood. In this study, we quantitatively evaluated the synaptic transmission and synaptic plasticity using hippocampal slices obtained from SAMP8 with electrophysiological methods to elucidate the synaptic features of SAMP8. We used the field recordings to measure some synaptic parameters. The slope of field excitatory postsynaptic potentials decreased with age in both SAMP8 and SAM resistant 1 (SAMR1), the control strain of SAMP8. The paired-pulse ratio (PPR), a representative of short-term synaptic plasticity, also decreased in both strains with age. On the other hand, although both SAMR1 and SAMP8 exhibited age-dependent decrease in long-term potentiation (LTP), a representative of long-term synaptic plasticity, the decrease in LTP in SAMP8 started at 6 months of age, while in SAMR1, it was observed at 14 months but not at 6 months of age. The PPRs after high-frequency stimulation for LTP induction were smaller than those before the stimulation. These results indicate that synaptic plasticity in SAMP8 deteriorates at an earlier age compared to SAMR1, and are consistent with behavioral tests showing early impairment of learning and memory of SAMP8. Our study is the first report on quantitative analysis of synaptic function at SAMP8 hippocampus and corroborates the behavioral studies showing cognitive dysfunction with age; therefore, it will be helpful for future studies on aging.
Collapse
|
31
|
Azpurua J, Eaton BA. Neuronal epigenetics and the aging synapse. Front Cell Neurosci 2015; 9:208. [PMID: 26074775 PMCID: PMC4444820 DOI: 10.3389/fncel.2015.00208] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022] Open
Abstract
Two of the most salient phenotypes of aging are cognitive decline and loss of motor function, both of which are controlled by the nervous system. Cognition and muscle contraction require that neuronal synapses develop and maintain proper structure and function. We review the literature on how normal physiological aging disrupts central and peripheral synapse function including the degradation of structure and/or control of neurotransmission. Here we also attempt to connect the work done on the epigenetics of aging to the growing literature of how epigenetic mechanisms control synapse structure and function. Lastly, we address possible roles of epigenetic mechanisms to explain why the basal rates of age-related dysfunction vary so widely across individuals.
Collapse
Affiliation(s)
- Jorge Azpurua
- Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Benjamin A Eaton
- Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| |
Collapse
|
32
|
Vitamin D prevents cognitive decline and enhances hippocampal synaptic function in aging rats. Proc Natl Acad Sci U S A 2014; 111:E4359-66. [PMID: 25267625 DOI: 10.1073/pnas.1404477111] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vitamin D is an important calcium-regulating hormone with diverse functions in numerous tissues, including the brain. Increasing evidence suggests that vitamin D may play a role in maintaining cognitive function and that vitamin D deficiency may accelerate age-related cognitive decline. Using aging rodents, we attempted to model the range of human serum vitamin D levels, from deficient to sufficient, to test whether vitamin D could preserve or improve cognitive function with aging. For 5-6 mo, middle-aged F344 rats were fed diets containing low, medium (typical amount), or high (100, 1,000, or 10,000 international units/kg diet, respectively) vitamin D3, and hippocampal-dependent learning and memory were then tested in the Morris water maze. Rats on high vitamin D achieved the highest blood levels (in the sufficient range) and significantly outperformed low and medium groups on maze reversal, a particularly challenging task that detects more subtle changes in memory. In addition to calcium-related processes, hippocampal gene expression microarrays identified pathways pertaining to synaptic transmission, cell communication, and G protein function as being up-regulated with high vitamin D. Basal synaptic transmission also was enhanced, corroborating observed effects on gene expression and learning and memory. Our studies demonstrate a causal relationship between vitamin D status and cognitive function, and they suggest that vitamin D-mediated changes in hippocampal gene expression may improve the likelihood of successful brain aging.
Collapse
|
33
|
Abstract
Homeostatic plasticity functions within the nervous system to maintain normal neural functions, such as neurotransmission, within predefined optimal ranges. The defined output of these neuronal processes is referred to as the set point, which is the value that the homeostatic system defends against fluctuations. Currently, it is unknown how stable homeostatic set points are within the nervous system. In the present study we used the CM9 neuromuscular junctions (NMJs) in the adult Drosophila to investigate the stability of the set point of synaptic homeostasis across the lifespan of the fly. At the fly NMJ, it is believed that the depolarization of the muscle by neurotransmitter during an action potential, represented by the EPSP, is a homeostatic set point that is precisely maintained via changes in synaptic vesicle release. We find that the amplitude of the EPSP abruptly increases during middle age and that this enhanced EPSP is maintained into late life, consistent with an age-dependent change to the homeostatic set point of the synapse during middle age. In support of this, comparison of the homeostatic response at the young versus the old synapse shows that the magnitude of the homeostatic response at the older synapse is significantly larger than the response at the young NMJ, appropriate for a synapse at which the set point has been increased. Our data demonstrate that the amplitude of the EPSP at the Drosophila NMJ increases during aging and that the homeostatic signaling system adjusts its response to accommodate the new set point.
Collapse
|
34
|
Buechel HM, Popovic J, Staggs K, Anderson KL, Thibault O, Blalock EM. Aged rats are hypo-responsive to acute restraint: implications for psychosocial stress in aging. Front Aging Neurosci 2014; 6:13. [PMID: 24575039 PMCID: PMC3921565 DOI: 10.3389/fnagi.2014.00013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/20/2014] [Indexed: 11/13/2022] Open
Abstract
Cognitive processes associated with prefrontal cortex and hippocampus decline with age and are vulnerable to disruption by stress. The stress/stress hormone/allostatic load hypotheses of brain aging posit that brain aging, at least in part, is the manifestation of life-long stress exposure. In addition, as humans age, there is a profound increase in the incidence of new onset stressors, many of which are psychosocial (e.g., loss of job, death of spouse, social isolation), and aged humans are well-understood to be more vulnerable to the negative consequences of such new-onset chronic psychosocial stress events. However, the mechanistic underpinnings of this age-related shift in chronic psychosocial stress response, or the initial acute phase of that chronic response, have been less well-studied. Here, we separated young (3 month) and aged (21 month) male F344 rats into control and acute restraint (an animal model of psychosocial stress) groups (n = 9–12/group). We then assessed hippocampus-associated behavioral, electrophysiological, and transcriptional outcomes, as well as blood glucocorticoid and sleep architecture changes. Aged rats showed characteristic water maze, deep sleep, transcriptome, and synaptic sensitivity changes compared to young. Young and aged rats showed similar levels of distress during the 3 h restraint, as well as highly significant increases in blood glucocorticoid levels 21 h after restraint. However, young, but not aged, animals responded to stress exposure with water maze deficits, loss of deep sleep and hyperthermia. These results demonstrate that aged subjects are hypo-responsive to new-onset acute psychosocial stress, which may have negative consequences for long-term stress adaptation and suggest that age itself may act as a stressor occluding the influence of new onset stressors.
Collapse
Affiliation(s)
- Heather M Buechel
- Blalock Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Jelena Popovic
- Blalock Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Kendra Staggs
- Blalock Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Katie L Anderson
- Thibault Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Olivier Thibault
- Thibault Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Eric M Blalock
- Blalock Laboratory, Department of Molecular and Biomedical Pharmacology, College of Medicine, University of Kentucky Lexington, KY, USA
| |
Collapse
|
35
|
Akhmedov K, Rizzo V, Kadakkuzha BM, Carter CJ, Magoski NS, Capo TR, Puthanveettil SV. Decreased response to acetylcholine during aging of aplysia neuron R15. PLoS One 2013; 8:e84793. [PMID: 24386417 PMCID: PMC3874043 DOI: 10.1371/journal.pone.0084793] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/18/2013] [Indexed: 12/03/2022] Open
Abstract
How aging affects the communication between neurons is poorly understood. To address this question, we have studied the electrophysiological properties of identified neuron R15 of the marine mollusk Aplysia californica. R15 is a bursting neuron in the abdominal ganglia of the central nervous system and is implicated in reproduction, water balance, and heart function. Exposure to acetylcholine (ACh) causes an increase in R15 burst firing. Whole-cell recordings of R15 in the intact ganglia dissected from mature and old Aplysia showed specific changes in burst firing and properties of action potentials induced by ACh. We found that while there were no significant changes in resting membrane potential and latency in response to ACh, the burst number and burst duration is altered during aging. The action potential waveform analysis showed that unlike mature neurons, the duration of depolarization and the repolarization amplitude and duration did not change in old neurons in response to ACh. Furthermore, single neuron quantitative analysis of acetylcholine receptors (AChRs) suggested alteration of expression of specific AChRs in R15 neurons during aging. These results suggest a defect in cholinergic transmission during aging of the R15 neuron.
Collapse
Affiliation(s)
- Komolitdin Akhmedov
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Valerio Rizzo
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Beena M. Kadakkuzha
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Christopher J. Carter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Neil S. Magoski
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Thomas R. Capo
- Division of Marine Biology and Fisheries, University of Miami Rosenstiel School of Marine and Atmospheric Science, Miami, Florida, United States of America
| | - Sathyanarayanan V. Puthanveettil
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
36
|
Artola A. Diabetes mellitus- and ageing-induced changes in the capacity for long-term depression and long-term potentiation inductions: Toward a unified mechanism. Eur J Pharmacol 2013; 719:161-169. [DOI: 10.1016/j.ejphar.2013.04.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/21/2013] [Accepted: 04/03/2013] [Indexed: 12/01/2022]
|
37
|
Griffith WH, Dubois DW, Fincher A, Peebles KA, Bizon JL, Murchison D. Characterization of age-related changes in synaptic transmission onto F344 rat basal forebrain cholinergic neurons using a reduced synaptic preparation. J Neurophysiol 2013; 111:273-86. [PMID: 24133226 DOI: 10.1152/jn.00129.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Basal forebrain (BF) cholinergic neurons participate in a number of cognitive processes that become impaired during aging. We previously found that age-related enhancement of Ca(2+) buffering in rat cholinergic BF neurons was associated with impaired performance in the water maze spatial learning task (Murchison D, McDermott AN, Lasarge CL, Peebles KA, Bizon JL, and Griffith WH. J Neurophysiol 102: 2194-2207, 2009). One way that altered Ca(2+) buffering could contribute to cognitive impairment involves synaptic function. In this report we show that synaptic transmission in the BF is altered with age and cognitive status. We have examined the properties of spontaneous postsynaptic currents (sPSCs) in cholinergic BF neurons that have been mechanically dissociated without enzymes from behaviorally characterized F344 rats. These isolated neurons retain functional presynaptic terminals on their somata and proximal dendrites. Using whole cell patch-clamp recording, we show that sPSCs and miniature PSCs are predominately GABAergic (bicuculline sensitive) and in all ways closely resemble PSCs recorded in a BF in vitro slice preparation. Adult (4-7 mo) and aged (22-24 mo) male rats were cognitively assessed using the water maze. Neuronal phenotype was identified post hoc using single-cell RT-PCR. The frequency of sPSCs was reduced during aging, and this was most pronounced in cognitively impaired subjects. This is the same population that demonstrated increased intracellular Ca(2+) buffering. We also show that increasing Ca(2+) buffering in the synaptic terminals of young BF neurons can mimic the reduced frequency of sPSCs observed in aged BF neurons.
Collapse
Affiliation(s)
- William H Griffith
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, Texas; and
| | | | | | | | | | | |
Collapse
|
38
|
Vallence AM, Goldsworthy MR. Can noninvasive brain stimulation enhance function in the ageing brain? J Neurophysiol 2013; 111:1-3. [PMID: 24004526 DOI: 10.1152/jn.00088.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Advancing age is associated with cognitive and motor performance deficits and a reduced capacity for plasticity. Zimerman and colleagues (Zimerman M, Nitsch M, Giraux P, Gerloff C, Cohen LG, Hummel FC. Ann Neurol 73: 10-15, 2013) have recently shown that noninvasive brain stimulation can enhance behavioral improvements following training on a motor sequence task in older adults. The work is of high clinical importance given the rapidly growing ageing population and the accompanying costs to health systems globally.
Collapse
Affiliation(s)
- Ann-Maree Vallence
- The Robinson Institute, School of Pediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | | |
Collapse
|
39
|
Marrone DF, Satvat E, Shaner MJ, Worley PF, Barnes CA. Attenuated long-term Arc expression in the aged fascia dentata. Neurobiol Aging 2012; 33:979-90. [PMID: 20850902 PMCID: PMC3010431 DOI: 10.1016/j.neurobiolaging.2010.07.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 07/19/2010] [Accepted: 07/30/2010] [Indexed: 12/26/2022]
Abstract
One prominent component of aging is a defect in memory stabilization. To understand how the formation of enduring memories is altered in the aged brain, long-term markers of the biological events that may mediate memory consolidation were used to examine the activity dynamics of hippocampal circuits over extended intervals. The immediate early gene Arc, which is implicated in both durable memory and synaptic plasticity, is expressed in the fascia dentata (FD) for long periods following behavioral experience. To test the hypothesis that aging alters long-term Arc transcription in the FD, a region critical for spatial memory and impaired with progressive age, young and aged rats explored a novel environment twice, separated by an 8-hour interval, and FD Arc transcription was assessed. Relative to young rats, (a) fewer granule cells in the aged FD transcribe arc 8 hours after spatial exploration, and (b) this decrease is correlated with impaired spatial memory. These findings are consistent with behavioral evidence of age-related decline in hippocampal-dependent memory processing long after an event is to be remembered, and reaffirm the integral role of the FD in the neural circuits supporting durable memory.
Collapse
Affiliation(s)
- Diano F. Marrone
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA, 85724
- Arizona Research Laboratories, Division of Neural Systems, Memory, and Aging, University of Arizona, Tucson, AZ, USA, 85724
- Dept. of Psychology, Wilfrid Laurier University, Waterloo, ON, Canada, N2L 3C5
| | - Elham Satvat
- Dept. of Psychology, Wilfrid Laurier University, Waterloo, ON, Canada, N2L 3C5
| | - Michael J. Shaner
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA, 85724
- Arizona Research Laboratories, Division of Neural Systems, Memory, and Aging, University of Arizona, Tucson, AZ, USA, 85724
| | - Paul F. Worley
- Depts. of Neuroscience and Neurology, Johns Hopkins University, Baltimore, MD, USA, 21218
| | - Carol A. Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA, 85724
- Arizona Research Laboratories, Division of Neural Systems, Memory, and Aging, University of Arizona, Tucson, AZ, USA, 85724
| |
Collapse
|
40
|
Foster TC. Dissecting the age-related decline on spatial learning and memory tasks in rodent models: N-methyl-D-aspartate receptors and voltage-dependent Ca2+ channels in senescent synaptic plasticity. Prog Neurobiol 2012; 96:283-303. [PMID: 22307057 DOI: 10.1016/j.pneurobio.2012.01.007] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/09/2012] [Accepted: 01/19/2012] [Indexed: 10/14/2022]
Abstract
In humans, heterogeneity in the decline of hippocampal-dependent episodic memory is observed during aging. Rodents have been employed as models of age-related cognitive decline and the spatial water maze has been used to show variability in the emergence and extent of impaired hippocampal-dependent memory. Impairment in the consolidation of intermediate-term memory for rapidly acquired and flexible spatial information emerges early, in middle-age. As aging proceeds, deficits may broaden to include impaired incremental learning of a spatial reference memory. The extent and time course of impairment has been be linked to senescence of calcium (Ca²⁺) regulation and Ca²⁺-dependent synaptic plasticity mechanisms in region CA1. Specifically, aging is associated with altered function of N-methyl-D-aspartate receptors (NMDARs), voltage-dependent Ca²⁺ channels (VDCCs), and ryanodine receptors (RyRs) linked to intracellular Ca²⁺ stores (ICS). In young animals, NMDAR activation induces long-term potentiation of synaptic transmission (NMDAR-LTP), which is thought to mediate the rapid consolidation of intermediate-term memory. Oxidative stress, starting in middle-age, reduces NMDAR function. In addition, VDCCs and ICS can actively inhibit NMDAR-dependent LTP and oxidative stress enhances the role of VDCC and RyR-ICS in regulating synaptic plasticity. Blockade of L-type VDCCs promotes NMDAR-LTP and memory in older animals. Interestingly, pharmacological or genetic manipulations to reduce hippocampal NMDAR function readily impair memory consolidation or rapid learning, generally leaving incremental learning intact. Finally, evidence is mounting to indicate a role for VDCC-dependent synaptic plasticity in associative learning and the consolidation of remote memories. Thus, VDCC-dependent synaptic plasticity and extrahippocampal systems may contribute to incremental learning deficits observed with advanced aging.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, Evelyn F. and William L. McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL 32610-0244, USA. ,
| |
Collapse
|
41
|
Tremblay MÈ, Zettel ML, Ison JR, Allen PD, Majewska AK. Effects of aging and sensory loss on glial cells in mouse visual and auditory cortices. Glia 2012; 60:541-58. [PMID: 22223464 DOI: 10.1002/glia.22287] [Citation(s) in RCA: 243] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 12/05/2011] [Indexed: 12/11/2022]
Abstract
Normal aging is often accompanied by a progressive loss of receptor sensitivity in hearing and vision, whose consequences on cellular function in cortical sensory areas have remained largely unknown. By examining the primary auditory (A1) and visual (V1) cortices in two inbred strains of mice undergoing either age-related loss of audition (C57BL/6J) or vision (CBA/CaJ), we were able to describe cellular and subcellular changes that were associated with normal aging (occurring in A1 and V1 of both strains) or specifically with age-related sensory loss (only in A1 of C57BL/6J or V1 of CBA/CaJ), using immunocytochemical electron microscopy and light microscopy. While the changes were subtle in neurons, glial cells and especially microglia were transformed in aged animals. Microglia became more numerous and irregularly distributed, displayed more variable cell body and process morphologies, occupied smaller territories, and accumulated phagocytic inclusions that often displayed ultrastructural features of synaptic elements. Additionally, evidence of myelination defects were observed, and aged oligodendrocytes became more numerous and were more often encountered in contiguous pairs. Most of these effects were profoundly exacerbated by age-related sensory loss. Together, our results suggest that the age-related alteration of glial cells in sensory cortical areas can be accelerated by activity-driven central mechanisms that result from an age-related loss of peripheral sensitivity. In light of our observations, these age-related changes in sensory function should be considered when investigating cellular, cortical, and behavioral functions throughout the lifespan in these commonly used C57BL/6J and CBA/CaJ mouse models.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Department of Neurobiology and Anatomy and Center for Visual Science, University of Rochester, Rochester, New York 14642, USA
| | | | | | | | | |
Collapse
|
42
|
McClelland S, Korosi A, Cope J, Ivy A, Baram TZ. Emerging roles of epigenetic mechanisms in the enduring effects of early-life stress and experience on learning and memory. Neurobiol Learn Mem 2011; 96:79-88. [PMID: 21338703 PMCID: PMC3111759 DOI: 10.1016/j.nlm.2011.02.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 02/04/2011] [Accepted: 02/10/2011] [Indexed: 12/25/2022]
Abstract
Epigenetic mechanisms are involved in programming gene expression throughout development. In addition, they are key contributors to the processes by which early-life experience fine-tunes the expression levels of key neuronal genes, governing learning and memory throughout life. Here we describe the long-lasting, bi-directional effects of early-life experience on learning and memory. We discuss how enriched postnatal experience enduringly augments spatial learning, and how chronic early-life stress results in persistent and progressive deficits in the structure and function of hippocampal neurons. The existing and emerging roles of epigenetic mechanisms in these fundamental neuroplasticity phenomena are illustrated.
Collapse
|
43
|
Isaias IU, Volkmann J, Kupsch A, Burgunder JM, Ostrem JL, Alterman RL, Mehdorn HM, Schönecker T, Krauss JK, Starr P, Reese R, Kühn AA, Schüpbach WMM, Tagliati M. Factors predicting protracted improvement after pallidal DBS for primary dystonia: the role of age and disease duration. J Neurol 2011; 258:1469-76. [DOI: 10.1007/s00415-011-5961-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 02/10/2011] [Accepted: 02/11/2011] [Indexed: 11/30/2022]
|
44
|
|
45
|
Schimanski LA, Barnes CA. Neural Protein Synthesis during Aging: Effects on Plasticity and Memory. Front Aging Neurosci 2010; 2:26. [PMID: 20802800 PMCID: PMC2928699 DOI: 10.3389/fnagi.2010.00026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 06/15/2010] [Indexed: 12/13/2022] Open
Abstract
During aging, many experience a decline in cognitive function that includes memory loss. The encoding of long-term memories depends on new protein synthesis, and this is also reduced during aging. Thus, it is possible that changes in the regulation of protein synthesis contribute to the memory impairments observed in older animals. Several lines of evidence support this hypothesis. For instance, protein synthesis is required for a longer period following learning to establish long-term memory in aged rodents. Also, under some conditions, synaptic activity or pharmacological activation can induce de novo protein synthesis and lasting changes in synaptic transmission in aged, but not young, rodents; the opposite results can be observed in other conditions. These changes in plasticity likely play a role in manifesting the altered place field properties observed in awake and behaving aged rats. The collective evidence suggests a link between memory loss and the regulation of protein synthesis in senescence. In fact, pharmaceuticals that target the signaling pathways required for induction of protein synthesis have improved memory, synaptic plasticity, and place cell properties in aged animals. We suggest that a better understanding of the mechanisms that lead to different protein expression patterns in the neural circuits that change as a function of age will enable the development of more effective therapeutic treatments for memory loss.
Collapse
Affiliation(s)
- Lesley A. Schimanski
- Evelyn F. McKnight Brain Institute and Division of Neural Systems, Memory and Aging, Arizona Research Laboratories, University of ArizonaTucson, AZ, USA
| | - Carol A. Barnes
- Evelyn F. McKnight Brain Institute and Division of Neural Systems, Memory and Aging, Arizona Research Laboratories, University of ArizonaTucson, AZ, USA
- Department of Psychology, University of ArizonaTucson, AZ, USA
- Department of Neurology, University of ArizonaTucson, AZ, USA
| |
Collapse
|
46
|
Penner MR, Roth TL, Barnes CA, Sweatt JD. An epigenetic hypothesis of aging-related cognitive dysfunction. Front Aging Neurosci 2010; 2:9. [PMID: 20552047 PMCID: PMC2874394 DOI: 10.3389/fnagi.2010.00009] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 02/24/2010] [Indexed: 12/19/2022] Open
Abstract
This brief review will focus on a new hypothesis for the role of epigenetic mechanisms in aging-related disruptions of synaptic plasticity and memory. Epigenetics refers to a set of potentially self-perpetuating, covalent modifications of DNA and post-translational modifications of nuclear proteins that produce lasting alterations in chromatin structure. These mechanisms, in turn, result in alterations in specific patterns of gene expression. Aging-related memory decline is manifest prominently in declarative/episodic memory and working memory, memory modalities anatomically based largely in the hippocampus and prefrontal cortex, respectively. The neurobiological underpinnings of age-related memory deficits include aberrant changes in gene transcription that ultimately affect the ability of the aged brain to be "plastic". The molecular mechanisms underlying these changes in gene transcription are not currently known, but recent work points toward a potential novel mechanism, dysregulation of epigenetic mechanisms. This has led us to hypothesize that dysregulation of epigenetic control mechanisms and aberrant epigenetic "marks" drive aging-related cognitive dysfunction. Here we focus on this theme, reviewing current knowledge concerning epigenetic molecular mechanisms, as well as recent results suggesting disruption of plasticity and memory formation during aging. Finally, several open questions will be discussed that we believe will fuel experimental discovery.
Collapse
Affiliation(s)
- Marsha R. Penner
- Arizona Research Laboratories Division of Neural Systems, Memory and Aging and Evelyn F. McKnight Brain Institute, University of ArizonaTucson, AZ, USA
| | - Tania L. Roth
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at BirminghamBirmingham, AL, USA
| | - Carol A. Barnes
- Arizona Research Laboratories Division of Neural Systems, Memory and Aging and Evelyn F. McKnight Brain Institute, University of ArizonaTucson, AZ, USA
- Departments of Psychology and Neurology, University of ArizonaTucson, AZ, USA
| | - J. David Sweatt
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at BirminghamBirmingham, AL, USA
| |
Collapse
|
47
|
Burke SN, Barnes CA. Senescent synapses and hippocampal circuit dynamics. Trends Neurosci 2010; 33:153-61. [PMID: 20071039 DOI: 10.1016/j.tins.2009.12.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 11/30/2009] [Accepted: 12/15/2009] [Indexed: 01/11/2023]
Abstract
Excitatory synaptic transmission is altered during aging in hippocampal granule cells, and in CA3 and CA1 pyramidal cells. These functional changes contribute to age-associated impairments in experimentally-induced plasticity in each of these primary hippocampal subregions. In CA1, plasticity evoked by stimulation shares common mechanisms with the synaptic modification observed following natural behavior. Aging results in deficits in both artificially- and behaviorally-induced plasticity, and this could in part reflect age-related changes in Ca2+ homeostasis. Other observations, however, suggest that increased intracellular Ca2+ levels are beneficial under some circumstances. This review focuses on age-associated changes in synaptic function, how these alterations might contribute to cognitive decline, and the extent to which altered hippocampal circuit properties are detrimental or reflect compensatory processes.
Collapse
Affiliation(s)
- Sara N Burke
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85724, USA
| | | |
Collapse
|
48
|
Kollen M, Stéphan A, Faivre-Bauman A, Loudes C, Sinet PM, Alliot J, Billard J, Epelbaum J, Dutar P, Jouvenceau A. Preserved memory capacities in aged Lou/C/Jall rats. Neurobiol Aging 2010; 31:129-42. [DOI: 10.1016/j.neurobiolaging.2008.03.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Revised: 03/10/2008] [Accepted: 03/14/2008] [Indexed: 10/22/2022]
|
49
|
A novel anti-inflammatory role of NCAM-derived mimetic peptide, FGL. Neurobiol Aging 2010; 31:118-28. [DOI: 10.1016/j.neurobiolaging.2008.03.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 03/03/2008] [Accepted: 03/20/2008] [Indexed: 11/22/2022]
|
50
|
Gant JC, Thibault O. Action potential throughput in aged rat hippocampal neurons: regulation by selective forms of hyperpolarization. Neurobiol Aging 2009; 30:2053-64. [PMID: 18367293 PMCID: PMC2776637 DOI: 10.1016/j.neurobiolaging.2008.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 02/07/2008] [Accepted: 02/14/2008] [Indexed: 01/23/2023]
Abstract
At hippocampal synapses, repetitive synaptic stimulation (RSS) in the theta frequency range (3-12Hz) is associated with robust EPSP frequency facilitation (FF) and consequently, enhanced action potential (spike) generation and throughput. A complex, synaptically induced hyperpolarization (SIHP) is also triggered by synaptic activation, and a Ca(2+)-dependent afterhyperpolarization (AHP) is triggered above spike threshold. With aging, the AHP is increased and impairs intracellular spike generation, at least in accommodation protocols. However, little is known about how these aging changes interact to affect spike generation at physiological frequencies of RSS, or if the SIHP also is modified in aging. Here we performed the first tests of the net impact of these excitatory and inhibitory aging changes on spike generation during RSS. We report that during RSS at spike threshold (1) spike throughput is well sustained at theta frequencies in young and aged neurons; (2) an interposed AHP dampens spike generation, particularly in aged neurons and at higher frequencies; (3) compared to the AHP, the SIHP does not exert an equivalent inhibitory effect on spike throughput; and (4) in contrast to the AHP, the SIHP is reduced with aging. Together, these results are consistent with a model in which the source of the hyperpolarization is important in determining hippocampal spike throughput within the theta frequency range.
Collapse
Affiliation(s)
- John C. Gant
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center (UKMC), MS320, Lexington, KY, 40503
| | - Olivier Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky Medical Center (UKMC), MS320, Lexington, KY, 40503
| |
Collapse
|