1
|
Wu K, Gollo LL. Mapping and modeling age-related changes in intrinsic neural timescales. Commun Biol 2025; 8:167. [PMID: 39901043 PMCID: PMC11791184 DOI: 10.1038/s42003-025-07517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 02/05/2025] Open
Abstract
Intrinsic timescales of brain regions exhibit heterogeneity, escalating with hierarchical levels, and are crucial for the temporal integration of external stimuli. Aging, often associated with cognitive decline, involves progressive neuronal and synaptic loss, reshaping brain structure and dynamics. However, the impact of these structural changes on temporal coding in the aging brain remains unclear. We mapped intrinsic timescales and gray matter volume (GMV) using magnetic resonance imaging (MRI) in young and elderly adults. We found shorter intrinsic timescales across multiple large-scale functional networks in the elderly cohort, and a significant positive association between intrinsic timescales and GMV. Additionally, age-related decline in performance on visual discrimination tasks was linked to a reduction in intrinsic timescales in the cuneus. To explain these age-related shifts, we developed an age-dependent spiking neuron network model. In younger subjects, brain regions were near a critical branching regime, while regions in elderly subjects had fewer neurons and synapses, pushing the dynamics toward a subcritical regime. The model accurately reproduced the empirical results, showing longer intrinsic timescales in young adults due to critical slowing down. Our findings reveal how age-related structural brain changes may drive alterations in brain dynamics, offering testable predictions and informing possible interventions targeting cognitive decline.
Collapse
Affiliation(s)
- Kaichao Wu
- Brain Networks and Modelling Laboratory and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | - Leonardo L Gollo
- Brain Networks and Modelling Laboratory and The Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia.
- Instituto de Física Interdisciplinary Sistemas Complejos, IFISC (UIB-CSIC), Campus Universitat de les Illes Balears, Palma de Mallorca, Spain.
| |
Collapse
|
2
|
Fei L, Liang Y, Kintscher U, Sigrist SJ. Coupling of mitochondrial state with active zone plasticity in early brain aging. Redox Biol 2025; 79:103454. [PMID: 39642596 PMCID: PMC11666929 DOI: 10.1016/j.redox.2024.103454] [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: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Neurodegenerative diseases typically emerge after an extended prodromal period, underscoring the critical importance of initiating interventions during the early stages of brain aging to enhance later resilience. Changes in presynaptic active zone proteins ("PreScale") are considered a dynamic, resilience-enhancing form of plasticity in the process of early, still reversible aging of the Drosophila brain. Aging, however, triggers significant changes not only of synapses but also mitochondria. While the two organelles are spaced in close proximity, likely reflecting a direct functional coupling in regard to ATP and Ca2+ homeostasis, the exact modes of coupling in the aging process remain to understood. We here show that genetic manipulations of mitochondrial functional status, which alters brain oxidative phosphorylation, ATP levels, or the production of reactive oxygen species (ROS), can bidirectionally regulate PreScale during early Drosophila brain aging. Conversely, genetic mimicry of PreScale resulted in decreased oxidative phosphorylation and ATP production, potentially due to reduced mitochondrial calcium (Ca2+) import. Our findings indicate the existence of a positive feedback loop where mitochondrial functional state and PreScale are reciprocally coupled to optimize protection during the early stages of brain aging.
Collapse
Affiliation(s)
- Lu Fei
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany
| | - Yongtian Liang
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, 10117, Berlin, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Center for Cardiovascular Research, Charité Universitätmedizin Berlin, 10115, Berlin, Germany; German Centre for Cardiovascular Research (DZHK), partner site Berlin, 10117, Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, 14195, Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
3
|
Ngcobo NN. Influence of Ageing on the Pharmacodynamics and Pharmacokinetics of Chronically Administered Medicines in Geriatric Patients: A Review. Clin Pharmacokinet 2025:10.1007/s40262-024-01466-0. [PMID: 39798015 DOI: 10.1007/s40262-024-01466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2024] [Indexed: 01/13/2025]
Abstract
As people age, the efficiency of various regulatory processes that ensure proper communication between cells and organs tends to decline. This deterioration can lead to difficulties in maintaining homeostasis during physiological stress. This includes but is not limited to cognitive impairments, functional difficulties, and issues related to caregivers which contribute significantly to medication errors and non-adherence. These factors can lead to higher morbidity, extended hospital stays, reduced quality of life, and even mortality. The decrease in homeostatic capacity varies among individuals, contributing to the greater variability observed in geriatric populations. Significant pharmacokinetic and pharmacodynamic alterations accompany ageing. Pharmacokinetic changes include decreased renal and hepatic clearance and an increased volume of distribution for lipid-soluble drugs, which prolong their elimination half-life. Pharmacodynamic changes typically involve increased sensitivity to various drug classes, such as anticoagulants, antidiabetic and psychotropic medications. This review examines the primary age-related physiological changes in geriatrics and their impact on the pharmacokinetics and pharmacodynamics of medications.
Collapse
Affiliation(s)
- Nokwanda N Ngcobo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
4
|
Alsegiani AS, Shah ZA. Age-dependent sex differences in cofilin1 pathway (LIMK1/SSH1) and its association with AD biomarkers after chronic systemic inflammation in mice. Neurobiol Aging 2024; 144:43-55. [PMID: 39265451 DOI: 10.1016/j.neurobiolaging.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/15/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
Chronic systemic inflammation (CSI) results in neuroinflammation and neurodegeneration. Cofilin1 is a stress protein that activates microglia and induces neuroinflammation, but its role in CSI at different aging stages remains unidentified. Therefore, the study aims to identify cofilin1 and its upstream regulators LIMK1 and SSH1 after CSI in young-, middle-, and advanced-aged mice. CSI was induced by injecting the male and female mice with a sub-lethal dose of Lipopolysaccharide weekly for six weeks. The results showed that normal male mice did not show cofilin pathway dysregulation, but a significant dysregulation was observed in CSI advanced-aged mice. In females, cofilin1 dysregulation was observed in healthy and CSI advanced-aged mice, while significant cofilin1 dysregulation was observed in middle-aged mice during CSI. Furthermore, cofilin1 pathway dysregulations correlated with Alzheimer's disease (AD) biomarkers in the brain and saliva, astrocyte activation, synaptic degeneration, neurobehavioral impairments, gut-microbiota abnormalities, and circulatory inflammation. These results provide new insights into cofilin1 sex and age-dependent mechanistic differences that might help identify targets for modulating neuroinflammation and early onset of AD.
Collapse
Affiliation(s)
- Amsha S Alsegiani
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
5
|
Dziewa M, Złotek M, Herbet M, Piątkowska-Chmiel I. Molecular and Cellular Foundations of Aging of the Brain: Anti-aging Strategies in Alzheimer's Disease. Cell Mol Neurobiol 2024; 44:80. [PMID: 39607636 PMCID: PMC11604688 DOI: 10.1007/s10571-024-01514-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Alzheimer's disease (AD) is a condition characterized by the gradual degeneration of the nervous system that poses significant challenges to cognitive function and overall mental health. Given the increasing global life expectancy, there is an urgent need for effective strategies to prevent and manage Alzheimer's disease, with a particular focus on anti-aging interventions. Recent scientific advancements have unveiled several promising strategies for combating Alzheimer's disease (AD), ranging from lifestyle interventions to cutting-edge pharmacological treatments and therapies targeting the underlying biological processes of aging and AD. Regular physical exercise, cognitive engagement, a balanced diet, and social interaction serve as key pillars in maintaining brain health. At the same time, therapies target key pathological mechanisms of AD, such as amyloid-beta accumulation, tau abnormalities, neuroinflammation, mitochondrial dysfunction, and synaptic loss, offering potential breakthroughs in treatment. Moreover, cutting-edge innovations such as gene therapy, stem cell transplantation, and novel drug delivery systems are emerging as potential game-changers in the fight against AD. This review critically evaluates the latest research on anti-aging interventions and their potential in preventing and treating Alzheimer's disease (AD) by exploring the connections between aging mechanisms and AD pathogenesis. It provides a comprehensive analysis of both well-established and emerging strategies, while also identifying key gaps in current knowledge to guide future research efforts.
Collapse
Affiliation(s)
- Magdalena Dziewa
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090, Lublin, Poland
| | - Magdalena Złotek
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090, Lublin, Poland
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090, Lublin, Poland
| | - Iwona Piątkowska-Chmiel
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090, Lublin, Poland.
| |
Collapse
|
6
|
Goldberg D, Wadhwani AR, Dehghani N, Sreepada LP, Fu H, De Jager PL, Bennett DA, Wolk DA, Lee EB, Farrell K, Crary JF, Zhou W, McMillan CT. Epigenetic signatures of regional tau pathology and cognition in the aging and pathological brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.07.24316933. [PMID: 39606399 PMCID: PMC11601699 DOI: 10.1101/2024.11.07.24316933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Primary age-related tauopathy (PART) and Alzheimer's disease (AD) share hippocampal phospho-tau (p-tau) pathology but differ in p-tau extent and amyloid presence. As a result, PART uniquely enables investigation of amyloid-independent p-tau mechanisms during brain aging. We conducted the first epigenome-wide association (EWAS) study of PART, which yielded 13 new and robust p-tau/methylation associations. We then jointly analyzed PART and AD epigenomes to develop "TauAge", novel epigenetic clocks that predict p-tau severity in region-specific, age-, and amyloid-independent manners. Integrative transcriptomic analyses revealed that genes involved in synaptic transmission are related to hippocampal p-tau severity in both PART and AD, while neuroinflammatory genes are related to frontal cortex p-tau severity in AD only. Further, a machine learning classifier based on PART-vs-AD epigenetic differences discriminates neuropathological diagnoses and stratifies indeterminate cases into subgroups with disparity in cognitive impairment. Together, these findings demonstrate the brain epigenome's substantial role in linking tau pathology to cognitive outcomes in aging and AD.
Collapse
|
7
|
Andraka JM, Sharma N, Marchalant Y. New Insights on the Effects of Krill Oil Supplementation, a High-Fat Diet, and Aging on Hippocampal-Dependent Memory, Neuroinflammation, Synaptic Density, and Neurogenesis. Int J Mol Sci 2024; 25:11554. [PMID: 39519107 PMCID: PMC11545834 DOI: 10.3390/ijms252111554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Krill oil (KO) has been described as having the potential to ameliorate the detrimental consequences of a high-fat diet (HFD) on the aging brain, though the magnitude and mechanism of this benefit is unclear. We thus hypothesized that dietary KO supplementation could counteract the effects of cognitive aging and an HFD on spatial learning, neuroinflammation, neurogenesis, and synaptic density in the cortex and hippocampus of aged rats. Sixteen-month-old Sprague Dawley rats were fed for 12 weeks while being divided into four groups: control (CON); control with KO supplementation (CONKO); high-fat diet (HF); and high-fat diet with KO supplementation (HFKO). We measured food consumption, body mass, spatial memory (Morris water maze), microglia, neurogenesis, cytokine concentrations, and synaptic markers (post-synaptic density-95 and synaptophysin). Predictably, an HFD did induce significant differences in body weights, with the high-fat groups gaining more weight than the low-fat controls. However, KO supplementation did not produce significant changes in the other quantified parameters. Our results demonstrate that the dietary KO dose provided in the current study does not benefit hippocampal or cortical functions in an aging model. Our results provide a benchmark for future dosing protocols that may eventually prove to be beneficial.
Collapse
Affiliation(s)
- John M. Andraka
- Department of Physical Therapy, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
| | - Naveen Sharma
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
- School of Health Sciences, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Yannick Marchalant
- Neuroscience Program, Central Michigan University, Mt. Pleasant, MI 48859, USA; (N.S.); (Y.M.)
- Psychology Department, Central Michigan University, Mt. Pleasant, MI 48859, USA
| |
Collapse
|
8
|
Chujan S, Cholpraipimolrat W, Satayavivad J. Integrated Transcriptomics and Network Analysis Identified Altered Neural Mechanisms in Frontal Aging Brain-Associated Alzheimer's Disease. Biochem Genet 2024; 62:2382-2398. [PMID: 37934339 DOI: 10.1007/s10528-023-10549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. The late stage of AD typically develops after 60 years of age and AD pathogenesis can be detected predominately in the frontal lobe, which is responsible for memory. Multiple alterations in cellular mechanisms have been associated with AD, but there is no clear information on AD pathogenesis during brain aging. This study aimed to explore the differentially expressed genes (DEGs) in the frontal lobe of aging brains and to identify shared crucial mechanisms in the aging brain linked to AD pathogenesis. Three datasets were downloaded from the Gene Expression Omnibus (GEO). Biological function analysis was performed by DAVID and KEGG databases. An AD patient's cohort (GSE150696) was collected for verification of the enriched pathway. The results demonstrated that multiple neurochemical synapsis and regulation of the cytoskeleton are linked to AD pathogenesis during aging. Taken together, this study contributes to our further understanding of neural alterations during aging in AD that could be used to develop therapeutics for early intervention to prevent or slow progression.
Collapse
Affiliation(s)
- Suthipong Chujan
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, 10400, Thailand
| | | | - Jutamaad Satayavivad
- Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, 10210, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, 10400, Thailand.
| |
Collapse
|
9
|
Burmistrov DE, Gudkov SV, Franceschi C, Vedunova MV. Sex as a Determinant of Age-Related Changes in the Brain. Int J Mol Sci 2024; 25:7122. [PMID: 39000227 PMCID: PMC11241365 DOI: 10.3390/ijms25137122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.
Collapse
Affiliation(s)
- Dmitriy E. Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
10
|
Tang J, Huang H, Muirhead RCJ, Zhou Y, Li J, DeFelice J, Kopanitsa MV, Serneels L, Davey K, Tilley BS, Gentleman S, Matthews PM. Associations of amyloid-β oligomers and plaques with neuropathology in the App NL-G-F mouse. Brain Commun 2024; 6:fcae218. [PMID: 39035420 PMCID: PMC11258573 DOI: 10.1093/braincomms/fcae218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 03/22/2024] [Accepted: 06/23/2024] [Indexed: 07/23/2024] Open
Abstract
Amyloid-β pathology and neurofibrillary tangles lead to glial activation and neurodegeneration in Alzheimer's disease. In this study, we investigated the relationships between the levels of amyloid-β oligomers, amyloid-β plaques, glial activation and markers related to neurodegeneration in the App NL-G-F triple mutation mouse line and in a knock-in line homozygous for the common human amyloid precursor protein (App hu mouse). The relationships between neuropathological features were characterized with immunohistochemistry and imaging mass cytometry. Markers assessing human amyloid-β proteins, microglial and astrocytic activation and neuronal and synaptic densities were used in mice between 2.5 and 12 months of age. We found that amyloid-β oligomers were abundant in the brains of App hu mice in the absence of classical amyloid-β plaques. These brains showed morphological changes consistent with astrocyte activation but no evidence of microglial activation or synaptic or neuronal pathology. In contrast, both high levels of amyloid-β oligomers and numerous plaques accumulated in App NL-G-F mice in association with substantial astrocytic and microglial activation. The increase in amyloid-β oligomers over time was more strongly correlated with astrocytic than with microglia activation. Spatial analyses suggested that activated microglia were more closely associated with amyloid-β oligomers than with amyloid-β plaques in App NL-G-F mice, which also showed age-dependent decreases in neuronal and synaptic density markers. A comparative study of the two models highlighted the dependence of glial and neuronal pathology on the nature and aggregation state of the amyloid-β peptide. Astrocyte activation and neuronal pathology appeared to be more strongly associated with amyloid-β oligomers than with amyloid-β plaques, although amyloid-β plaques were associated with microglia activation.
Collapse
Affiliation(s)
- Jiabin Tang
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Department of Anesthesiology, Weill Cornell Medicine, Cornell University, New York, NY 11106, USA
| | - Helen Huang
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Robert C J Muirhead
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Randall Centre for Cell and Molecular Biophysics, Kings College London, London SE5 9RX, UK
| | - Yue Zhou
- Department of Mechanical Engineering, Roberts Engineering Building, University College London, London WC1E 7JE, UK
| | - Junheng Li
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
| | - John DeFelice
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Lutgarde Serneels
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), 9052 Gent, Belgium
| | - Karen Davey
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- UK Dementia Research Institute, Kings College London, Denmark Hill Campus, London SE5 9RX, UK
| | - Bension S Tilley
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Steve Gentleman
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Paul M Matthews
- UK Dementia Research Institute, Uren Building, Imperial College London, White City Campus, London W12 0BZ, UK
- Department of Brain Sciences, Burlington Danes Building, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| |
Collapse
|
11
|
Banerjee S, Vernon S, Ruchti E, Limoni G, Jiao W, Asadzadeh J, Van Campenhoudt M, McCabe BD. Trio preserves motor synapses and prolongs motor ability during aging. Cell Rep 2024; 43:114256. [PMID: 38795343 DOI: 10.1016/j.celrep.2024.114256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 01/24/2024] [Accepted: 05/05/2024] [Indexed: 05/27/2024] Open
Abstract
The decline of motor ability is a hallmark feature of aging and is accompanied by degeneration of motor synaptic terminals. Consistent with this, Drosophila motor synapses undergo characteristic age-dependent structural fragmentation co-incident with diminishing motor ability. Here, we show that motor synapse levels of Trio, an evolutionarily conserved guanine nucleotide exchange factor (GEF), decline with age. We demonstrate that increasing Trio expression in adult Drosophila can abrogate age-dependent synaptic structural fragmentation, postpone the decline of motor ability, and maintain the capacity of motor synapses to sustain high-intensity neurotransmitter release. This preservative activity is conserved in transgenic human Trio, requires Trio Rac GEF function, and can also ameliorate synapse degeneration induced by depletion of miniature neurotransmission. Our results support a paradigm where the structural dissolution of motor synapses precedes and promotes motor behavioral diminishment and where intervening in this process can postpone the decline of motor function during aging.
Collapse
Affiliation(s)
- Soumya Banerjee
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Samuel Vernon
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Marine Van Campenhoudt
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland.
| |
Collapse
|
12
|
He J, Hou T, Wang Q, Wang Q, Jiang Y, Chen L, Xu J, Qi Y, Jia D, Gu Y, Gao L, Yu Y, Wang L, Kang L, Si J, Wang L, Chen S. L-arginine metabolism ameliorates age-related cognitive impairment by Amuc_1100-mediated gut homeostasis maintaining. Aging Cell 2024; 23:e14081. [PMID: 38236004 PMCID: PMC11019123 DOI: 10.1111/acel.14081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
Aging-induced cognitive impairment is associated with a loss of metabolic homeostasis and plasticity. An emerging idea is that targeting key metabolites is sufficient to impact the function of other organisms. Therefore, more metabolism-targeted therapeutic intervention is needed to improve cognitive impairment. We first conducted untargeted metabolomic analyses and 16S rRNA to identify the aging-associated metabolic adaption and intestinal microbiome change. Untargeted metabolomic analyses of plasma revealed L-arginine metabolic homeostasis was altered during the aging process. Impaired L-arginine metabolic homeostasis was associated with low abundance of intestinal Akkermansia muciniphila (AKK) colonization in mice. Long-term supplementation of AKK outer membranes protein-Amuc_1100, rescued the L-arginine level and restored cognitive impairment in aging mice. Mechanically, Amuc_1100 acted directly as a source of L-arginine and enriched the L-arginine-producing bacteria. In aged brain, Amuc_1100 promoted the superoxide dismutase to alleviated oxidation stress, and increased nitric oxide, derivatives of L-arginine, to improve synaptic plasticity. Meanwhile, L-arginine repaired lipopolysaccharide-induced intestinal barrier damage and promoted growth of colon organoid. Our findings indicated that aging-related cognitive impairment was closely associated with the disorders of L-arginine metabolism. AKK-derived Amuc_1100, as a potential postbiotic, targeting the L-arginine metabolism, might provide a promising therapeutic strategy to maintain the intestinal homeostasis and cognitive function in aging.
Collapse
Affiliation(s)
- Jiamin He
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Tongyao Hou
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Qiwen Wang
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Qingyi Wang
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Yao Jiang
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Luyi Chen
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of General PracticeSir Run Run Shaw Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Jilei Xu
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Yadong Qi
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Dingjiacheng Jia
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Yanrou Gu
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of Gastroenterology, Wenzhou People's HospitalWenzhou Medical UniversityWenzhouChina
| | - Lidan Gao
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Third Affiliated Hospital of Shanghai University, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's HospitalWenzhouChina
| | - Yingcong Yu
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of Gastroenterology, Wenzhou People's HospitalWenzhou Medical UniversityWenzhouChina
| | - Lan Wang
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Lijun Kang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain‐Machine Integration, School of Brain Science and Brain MedicineZhejiang UniversityHangzhouChina
| | - Jianmin Si
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| | - Liangjing Wang
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
- Department of GastroenterologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Shujie Chen
- Department of GastroenterologySir Run Run Shaw Hospital, Zhejiang UniversityHangzhouChina
- Institution of GastroenterologyZhejiang UniversityHangzhouChina
- Prevention and Treatment Research Center for Senescent DiseaseZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
13
|
Suman A, Mahapatra A, Gupta P, Ray SS, Singh RK. Polystyrene microplastics induced disturbances in neuronal arborization and dendritic spine density in mice prefrontal cortex. CHEMOSPHERE 2024; 351:141165. [PMID: 38224746 DOI: 10.1016/j.chemosphere.2024.141165] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
An increasing use of plastics in daily life leads to the accumulation of microplastics (MPs) in the environment, posing a serious threat to the ecosystem, including humans. It has been reported that MPs cause neurotoxicity, but the deleterious effect of polystyrene (PS) MPs on neuronal cytoarchitectural morphology in the prefrontal cortex (PFC) region of mice brain remains to be established. In the present study, Swiss albino male mice were orally exposed to 0.1, 1, and 10 ppm PS-MPs for 28 days. After exposure, we found a significant accumulation of PS-MPs with a decreased number of Nissl bodies in the PFC region of the entire treated group compared to the control. Morphometric analysis in the PFC neurons using Golgi-Cox staining accompanied by Sholl analysis showed a significant reduction in basal dendritic length, dendritic intersections, nodes, and number of intersections at seventh branch order in PFC neurons of 1 ppm treated PS-MPs. In neurons of 0.1 ppm treated mice, we found only decrease in the number of intersections at the seventh branch order. While 10 ppm treated neurons decreased in basal dendritic length, dendritic intersections, followed by the number of intersections at the third and seventh branch order were observed. As well, spine density on the apical secondary branches along with mRNA level of BDNF was significantly reduced in all the PS-MPs treated PFC neurons, mainly at 1 ppm versus control. These results suggest that PS-MPs exposure affects overall basal neuronal arborization, with the highest levels at 1 and 10 ppm, followed by 0.1 ppm treated neurons, which may be related to the down-regulation of BDNF expression in PFC.
Collapse
Affiliation(s)
- Anjali Suman
- Molecular Endocrinology and Toxicology Laboratory (METLab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Archisman Mahapatra
- Molecular Endocrinology and Toxicology Laboratory (METLab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Priya Gupta
- Molecular Endocrinology and Toxicology Laboratory (METLab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shubhendu Shekhar Ray
- Molecular Endocrinology and Toxicology Laboratory (METLab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Rahul Kumar Singh
- Molecular Endocrinology and Toxicology Laboratory (METLab), Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
14
|
Martin SL, Uribe C, Strafella AP. PET imaging of synaptic density in Parkinsonian disorders. J Neurosci Res 2024; 102:e25253. [PMID: 37814917 DOI: 10.1002/jnr.25253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023]
Abstract
Synaptic dysfunction and altered synaptic pruning are present in people with Parkinsonian disorders. Dopamine loss and alpha-synuclein accumulation, two hallmarks of Parkinson's disease (PD) pathology, contribute to synaptic dysfunction and reduced synaptic density in PD. Atypical Parkinsonian disorders are likely to have unique spatiotemporal patterns of synaptic density, differentiating them from PD. Therefore, quantification of synaptic density has the potential to support diagnoses, monitor disease progression, and treatment efficacy. Novel radiotracers for positron emission tomography which target the presynaptic vesicle protein SV2A have been developed to quantify presynaptic density. The radiotracers have successfully investigated synaptic density in preclinical models of PD and people with Parkinsonian disorders. Therefore, this review will summarize the preclinical and clinical utilization of SV2A radiotracers in people with Parkinsonian disorders. We will evaluate how SV2A abundance is associated with other imaging modalities and the considerations for interpreting SV2A in Parkinsonian pathology.
Collapse
Affiliation(s)
- Sarah L Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Carme Uribe
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Unitat de Psicologia Medica, Departament de Medicina, Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
| | - Antonio P Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital & Krembil Brain Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Bozic I. Neuro-oscillations in memory consolidation and forgotten parts of the brain: Commentary on Weiner et al., 2023. Eur J Neurosci 2024; 59:481-482. [PMID: 37313790 DOI: 10.1111/ejn.16056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 06/15/2023]
Affiliation(s)
- Ivan Bozic
- Universitäre Psychiatrische Dienste Bern, Universität Bern, Bern, Switzerland
| |
Collapse
|
16
|
Wulaer B, Holtz MA, Nagai J. Homeostasis to Allostasis: Prefrontal Astrocyte Roles in Cognitive Flexibility and Stress Biology. ADVANCES IN NEUROBIOLOGY 2024; 39:137-163. [PMID: 39190074 DOI: 10.1007/978-3-031-64839-7_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
In the intricate landscape of neurophysiology, astrocytes have been traditionally cast as homeostatic cells; however, their mechanistic involvement in allostasis-particularly how they modulate the adaptive response to stress and its accumulative impact that disrupts cognitive functions and precipitates psychiatric disorders-is now starting to be unraveled. Here, we address the gap by positing astrocytes as crucial allostatic players whose molecular adaptations underlie cognitive flexibility in stress-related neuropsychiatric conditions. We review how astrocytes, responding to stress mediators such as glucocorticoid and epinephrine/norepinephrine, undergo morphological and functional transformations that parallel the maladaptive changes. Our synthesis of recent findings reveals that these glial changes, especially in the metabolically demanding prefrontal cortex, may underlie some of the neuropsychiatric mechanisms characterized by the disruption of energy metabolism and astrocytic networks, compromised glutamate clearance, and diminished synaptic support. We argue that astrocytes extend beyond their homeostatic role, actively participating in the brain's allostatic response, especially by modulating energy substrates critical for cognitive functions.
Collapse
Affiliation(s)
- Bolati Wulaer
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Mika A Holtz
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Jun Nagai
- Laboratory for Glia-Neuron Circuit Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan.
| |
Collapse
|
17
|
Chaves-Coira I, García-Magro N, Zegarra-Valdivia J, Torres-Alemán I, Núñez Á. Cognitive Deficits in Aging Related to Changes in Basal Forebrain Neuronal Activity. Cells 2023; 12:1477. [PMID: 37296598 PMCID: PMC10252596 DOI: 10.3390/cells12111477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Aging is a physiological process accompanied by a decline in cognitive performance. The cholinergic neurons of the basal forebrain provide projections to the cortex that are directly engaged in many cognitive processes in mammals. In addition, basal forebrain neurons contribute to the generation of different rhythms in the EEG along the sleep/wakefulness cycle. The aim of this review is to provide an overview of recent advances grouped around the changes in basal forebrain activity during healthy aging. Elucidating the underlying mechanisms of brain function and their decline is especially relevant in today's society as an increasingly aged population faces higher risks of developing neurodegenerative diseases such as Alzheimer's disease. The profound age-related cognitive deficits and neurodegenerative diseases associated with basal forebrain dysfunction highlight the importance of investigating the aging of this brain region.
Collapse
Affiliation(s)
- Irene Chaves-Coira
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| | - Nuria García-Magro
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain;
| | - Jonathan Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (J.Z.-V.); (I.T.-A.)
- Facultad de Ciencias de la Salud, Universidad Señor de Sipán, Chiclayo 02001, Peru
| | - Ignacio Torres-Alemán
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain; (J.Z.-V.); (I.T.-A.)
- Ikerbasque Science Foundation, 48009 Bilbao, Spain
| | - Ángel Núñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
| |
Collapse
|
18
|
Kesidou E, Theotokis P, Damianidou O, Boziki M, Konstantinidou N, Taloumtzis C, Sintila SA, Grigoriadis P, Evangelopoulos ME, Bakirtzis C, Simeonidou C. CNS Ageing in Health and Neurodegenerative Disorders. J Clin Med 2023; 12:2255. [PMID: 36983254 PMCID: PMC10054919 DOI: 10.3390/jcm12062255] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
The process of ageing is characteristic of multicellular organisms associated with late stages of the lifecycle and is manifested through a plethora of phenotypes. Its underlying mechanisms are correlated with age-dependent diseases, especially neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS) that are accompanied by social and financial difficulties for patients. Over time, people not only become more prone to neurodegeneration but they also lose the ability to trigger pivotal restorative mechanisms. In this review, we attempt to present the already known molecular and cellular hallmarks that characterize ageing in association with their impact on the central nervous system (CNS)'s structure and function intensifying possible preexisting pathogenetic conditions. A thorough and elucidative study of the underlying mechanisms of ageing will be able to contribute further to the development of new therapeutic interventions to effectively treat age-dependent manifestations of neurodegenerative diseases.
Collapse
Affiliation(s)
- Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
- Laboratory of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Olympia Damianidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Natalia Konstantinidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Charilaos Taloumtzis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Styliani-Aggeliki Sintila
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Panagiotis Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | | | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Greece (P.T.)
| | - Constantina Simeonidou
- Laboratory of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
19
|
Kawai H, Kishimoto M, Okahisa Y, Sakamoto S, Terada S, Takaki M. Initial Outcomes of the Safe and Sound Protocol on Patients with Adult Autism Spectrum Disorder: Exploratory Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4862. [PMID: 36981773 PMCID: PMC10049251 DOI: 10.3390/ijerph20064862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 06/18/2023]
Abstract
Sensory impairments are common features of autism spectrum disorder (ASD) and are associated with its social impairments. However, there is no established treatment for these impairments in adults with ASD. The Safe & Sound Protocol (SSP) is a listening program designed to improve social communication skills by reducing auditory hypersensitivity. We investigated the effectiveness of the SSP for adults with ASD. We administered the SSP to six participants with ASD aged 21-44 years old, and the effects were assessed using the Social Responsiveness Scale, Second Edition (SRS-2). Secondary outcomes were assessed using the Center for Epidemiological Studies Depression Scale (CES-D), State-Trait Anxiety Inventory (STAI), WHO Quality of Life 26 (WHOQOL-BREF), and Adolescent/Adult Sensory Profile (A/ASP). In this study, only the Social Awareness scale of the SRS-2 Family-Report showed a significant improvement after the intervention. In addition, it was significantly correlated with physical health of WHOQOL-BREF (r = -0.577, p = 0.012), state and trait anxiety of STAI (r = 0.576, p = 0.012; r = 0.708, p = 0.00009, respectively), and CES-D (r = 0.465, p = 0.05). In conclusion, the SSP has a partial effect on social impairments in adults with ASD, specifically on the Social Awareness subscale of the SRS-2.
Collapse
Affiliation(s)
- Hiroki Kawai
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
| | - Makiko Kishimoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
- National Center for Child Health and Development, Tokyo 157-0074, Japan
| | - Yuko Okahisa
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
| | - Shinji Sakamoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
| | - Seishi Terada
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
| | - Manabu Takaki
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan; (H.K.)
| |
Collapse
|
20
|
Ho PWL, Li L, Liu HF, Choi ZYK, Chang EES, Pang SYY, Malki Y, Leung CT, Kung MHW, Ramsden DB, Ho SL. In vivo overexpression of synaptogyrin-3 promotes striatal synaptic dopamine uptake in LRRK2 R1441G mutant mouse model of Parkinson's disease. Brain Behav 2023; 13:e2886. [PMID: 36624932 PMCID: PMC9927849 DOI: 10.1002/brb3.2886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/17/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Leucine-rich repeat kinase 2 (LRRK2) mutation is a common genetic risk factor of Parkinson's disease (PD). Presynaptic dysfunction is an early pathogenic event associated with dopamine (DA) dysregulation in striatum of the brain. DA uptake activity of DA uptake transporter (DAT) affects synaptic plasticity and motor and non-motor behavior. Synaptogyrin-3 (SYNGR3) is part of the synaptogyrin family, especially abundant in brain. Previous in vitro studies demonstrated interaction between SYNGR3 and DAT. Reduced SYNGR3 expression was observed in human PD brains with unclear reasons. METHODS Here, we further explored whether inducing SYNGR3 expression can influence (i) cellular DA uptake using differentiated human SH-SY5Y neuronal cells, (ii) striatal synaptosomal DA uptake in a mutant LRRK2R1441G knockin mouse model of PD, and (iii) innate rodent behavior using the marble burying test. RESULTS Young LRRK2 mutant mice exhibited significantly lower SYNGR3 levels in striatum compared to age-matched wild-type (WT) controls, resembling level in aged WT mice. SYNGR3 is spatially co-localized with DAT at striatal presynaptic terminals, visualized by immuno-gold transmission electron microscopy and immunohistochemistry. Their protein-protein interaction was confirmed by co-immunoprecipitation. Transient overexpression of SYNGR3 in differentiated SH-SY5Y cells increased cellular DA uptake activity without affecting total DAT levels. Inducing SYNGR3 overexpression by adeno-associated virus-7 (AAV7) injection in vivo into striatum increased ex vivo synaptosomal DA uptake in LRRK2 mutant mice and improved their innate marble burying behavior. CONCLUSION Brain SYNGR3 expression may be an important determinant to striatal DA homeostasis and synaptic function. Our preliminary behavioral test showed improved innate behavior after SYNGR3 overexpression in LRRK2 mutant mice, advocating further studies to determine the influence of SYNGR3 in the pathophysiology of DA neurons in PD.
Collapse
Affiliation(s)
- Philip Wing-Lok Ho
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Lingfei Li
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Hui-Fang Liu
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Zoe Yuen-Kiu Choi
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Eunice Eun Seo Chang
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Shirley Yin-Yu Pang
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Yasine Malki
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Chi-Ting Leung
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - Michelle Hiu-Wai Kung
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| | - David Boyer Ramsden
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Shu-Leong Ho
- Division of Neurology, Department of Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, China
| |
Collapse
|
21
|
Zaccard CR, Gippo I, Song A, Geula C, Penzes P. Dendritic spinule-mediated structural synaptic plasticity: Implications for development, aging, and psychiatric disease. Front Mol Neurosci 2023; 16:1059730. [PMID: 36741924 PMCID: PMC9895827 DOI: 10.3389/fnmol.2023.1059730] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
Dendritic spines are highly dynamic and changes in their density, size, and shape underlie structural synaptic plasticity in cognition and memory. Fine membranous protrusions of spines, termed dendritic spinules, can contact neighboring neurons or glial cells and are positively regulated by neuronal activity. Spinules are thinner than filopodia, variable in length, and often emerge from large mushroom spines. Due to their nanoscale, spinules have frequently been overlooked in diffraction-limited microscopy datasets. Until recently, our knowledge of spinules has been interpreted largely from single snapshots in time captured by electron microscopy. We summarize herein the current knowledge about the molecular mechanisms of spinule formation. Additionally, we discuss possible spinule functions in structural synaptic plasticity in the context of development, adulthood, aging, and psychiatric disorders. The literature collectively implicates spinules as a mode of structural synaptic plasticity and suggests the existence of morphologically and functionally distinct spinule subsets. A recent time-lapse, enhanced resolution imaging study demonstrated that the majority of spinules are small, short-lived, and dynamic, potentially exploring their environment or mediating retrograde signaling and membrane remodeling via trans-endocytosis. A subset of activity-enhanced, elongated, long-lived spinules is associated with complex PSDs, and preferentially contacts adjacent axonal boutons not presynaptic to the spine head. Hence, long-lived spinules can form secondary synapses with the potential to alter synaptic connectivity. Published studies further suggest that decreased spinules are associated with impaired synaptic plasticity and intellectual disability, while increased spinules are linked to hyperexcitability and neurodegenerative diseases. In summary, the literature indicates that spinules mediate structural synaptic plasticity and perturbations in spinules can contribute to synaptic dysfunction and psychiatric disease. Additional studies would be beneficial to further delineate the molecular mechanisms of spinule formation and determine the exact role of spinules in development, adulthood, aging, and psychiatric disorders.
Collapse
Affiliation(s)
- Colleen R. Zaccard
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Isabel Gippo
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Amy Song
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, United States,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States,*Correspondence: Peter Penzes,
| |
Collapse
|
22
|
Dundee JM, Puigdellívol M, Butler R, Cockram TOJ, Brown GC. P2Y 6 receptor-dependent microglial phagocytosis of synapses mediates synaptic and memory loss in aging. Aging Cell 2022; 22:e13761. [PMID: 36565471 PMCID: PMC9924939 DOI: 10.1111/acel.13761] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
Aging causes loss of brain synapses and memory, and microglial phagocytosis of synapses may contribute to this loss. Stressed neurons can release the nucleotide UTP, which is rapidly converted into UDP, that in turn activates the P2Y6 receptor (P2Y6 R) on the surface of microglia, inducing microglial phagocytosis of neurons. However, whether the activation of P2Y6 R affects microglial phagocytosis of synapses is unknown. We show here that inactivation of P2Y6 R decreases microglial phagocytosis of isolated synapses (synaptosomes) and synaptic loss in neuronal-glial co-cultures. In vivo, wild-type mice aged from 4 to 17 months exhibited reduced synaptic density in cortical and hippocampal regions, which correlated with increased internalization of synaptic material within microglia. However, this aging-induced synaptic loss and internalization were absent in P2Y6 R knockout mice, and these mice also lacked any aging-induced memory loss. Thus, P2Y6 R appears to mediate aging-induced loss of synapses and memory by increasing microglial phagocytosis of synapses. Consequently, blocking P2Y6 R has the potential to prevent age-associated memory impairment.
Collapse
Affiliation(s)
- Jacob M. Dundee
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Mar Puigdellívol
- Department of BiochemistryUniversity of CambridgeCambridgeUK,Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
| | - Richard Butler
- The Wellcome Trust Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUK
| | | | - Guy C. Brown
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
23
|
Stoeger T, Grant RA, McQuattie-Pimentel AC, Anekalla KR, Liu SS, Tejedor-Navarro H, Singer BD, Abdala-Valencia H, Schwake M, Tetreault MP, Perlman H, Balch WE, Chandel NS, Ridge KM, Sznajder JI, Morimoto RI, Misharin AV, Budinger GRS, Nunes Amaral LA. Aging is associated with a systemic length-associated transcriptome imbalance. NATURE AGING 2022; 2:1191-1206. [PMID: 37118543 PMCID: PMC10154227 DOI: 10.1038/s43587-022-00317-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/21/2022] [Indexed: 12/14/2022]
Abstract
Aging is among the most important risk factors for morbidity and mortality. To contribute toward a molecular understanding of aging, we analyzed age-resolved transcriptomic data from multiple studies. Here, we show that transcript length alone explains most transcriptional changes observed with aging in mice and humans. We present three lines of evidence supporting the biological importance of the uncovered transcriptome imbalance. First, in vertebrates the length association primarily displays a lower relative abundance of long transcripts in aging. Second, eight antiaging interventions of the Interventions Testing Program of the National Institute on Aging can counter this length association. Third, we find that in humans and mice the genes with the longest transcripts enrich for genes reported to extend lifespan, whereas those with the shortest transcripts enrich for genes reported to shorten lifespan. Our study opens fundamental questions on aging and the organization of transcriptomes.
Collapse
Affiliation(s)
- Thomas Stoeger
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA.
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, USA.
- Center for Genetic Medicine, Northwestern University, Evanston, IL, USA.
| | - Rogan A Grant
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
| | | | - Kishore R Anekalla
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
| | - Sophia S Liu
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | | | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Evanston, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
| | - Michael Schwake
- Department of Neurology, Northwestern University, Evanston, IL, USA
- Faculty of Chemistry, University of Bielefeld, Bielefeld, Germany
| | - Marie-Pier Tetreault
- Division of Gastroenterology and Hepatology, Northwestern University, Evanston, IL, USA
| | - Harris Perlman
- Division of Rheumatology, Northwestern University, Evanston, IL, USA
| | | | - Navdeep S Chandel
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA
| | - Richard I Morimoto
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
- Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA.
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA.
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Lung Institute for Translational Science at Northwestern University (SQLIFTSNU), Evanston, IL, USA.
| | - Luis A Nunes Amaral
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA.
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, USA.
- Department of Physics and Astronomy, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
24
|
Ni MZ, Zhang YM, Li Y, Wu QT, Zhang ZZ, Chen J, Luo BL, Li XW, Chen GH. Environmental enrichment improves declined cognition induced by prenatal inflammatory exposure in aged CD-1 mice: Role of NGPF2 and PSD-95. Front Aging Neurosci 2022; 14:1021237. [PMID: 36479357 PMCID: PMC9720164 DOI: 10.3389/fnagi.2022.1021237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 12/08/2023] Open
Abstract
INTRODUCTION Research suggests that prenatal inflammatory exposure could accelerate age-related cognitive decline that may be resulted from neuroinflammation and synaptic dysfunction during aging. Environmental enrichment (EE) may mitigate the cognitive and synaptic deficits. Neurite growth-promoting factor 2 (NGPF2) and postsynaptic density protein 95 (PSD-95) play critical roles in neuroinflammation and synaptic function, respectively. METHODS We examined whether this adversity and EE exposure can cause alterations in Ngpf2 and Psd-95 expression. In this study, CD-1 mice received intraperitoneal injection of lipopolysaccharide (50 μg/kg) or normal saline from gestational days 15-17. After weaning, half of the male offspring under each treatment were exposed to EE. The Morris water maze was used to assess spatial learning and memory at 3 and 15 months of age, whereas quantitative real-time polymerase chain reaction and Western blotting were used to measure hippocampal mRNA and protein levels of NGPF2 and PSD-95, respectively. Meanwhile, serum levels of IL-6, IL-1β, and TNF-α were determined by enzyme-linked immunosorbent assay. RESULTS The results showed that aged mice exhibited poor spatial learning and memory ability, elevated NGPF2 mRNA and protein levels, and decreased PSD-95 mRNA and protein levels relative to their young counterparts during natural aging. Embryonic inflammatory exposure accelerated age-related changes in spatial cognition, and in Ngpf2 and Psd-95 expression. Additionally, the levels of Ngpf2 and Psd-95 products were significantly positively and negatively correlated with cognitive dysfunction, respectively, particularly in prenatal inflammation-exposed aged mice. Changes in serum levels of IL-6, IL-1β, and TNF-α reflective of systemic inflammation and their correlation with cognitive decline during accelerated aging were similar to those of hippocampal NGPF2. EE exposure could partially restore the accelerated decline in age-related cognitive function and in Psd-95 expression, especially in aged mice. DISCUSSION Overall, the aggravated cognitive disabilities in aged mice may be related to the alterations in Ngpf2 and Psd-95 expression and in systemic state of inflammation due to prenatal inflammatory exposure, and long-term EE exposure may ameliorate this cognitive impairment by upregulating Psd-95 expression.
Collapse
Affiliation(s)
- Ming-Zhu Ni
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yun Li
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Qi-Tao Wu
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Zhe-Zhe Zhang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Jing Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Bao-Ling Luo
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Xue-Wei Li
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
Anusha-Kiran Y, Mol P, Dey G, Bhat FA, Chatterjee O, Deolankar SC, Philip M, Prasad TSK, Srinivas Bharath MM, Mahadevan A. Regional heterogeneity in mitochondrial function underlies region specific vulnerability in human brain ageing: Implications for neurodegeneration. Free Radic Biol Med 2022; 193:34-57. [PMID: 36195160 DOI: 10.1016/j.freeradbiomed.2022.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/22/2022] [Indexed: 12/01/2022]
Abstract
Selective neuronal vulnerability (SNV) of specific neuroanatomical regions such as frontal cortex (FC) and hippocampus (HC) is characteristic of age-associated neurodegenerative diseases (NDDs), although its pathogenetic basis remains unresolved. We hypothesized that physiological differences in mitochondrial function in neuroanatomical regions could contribute to SNV. To investigate this, we evaluated mitochondrial function in human brains (age range:1-90 y) in FC, striatum (ST), HC, cerebellum (CB) and medulla oblongata (MD), using enzyme assays and quantitative proteomics. Striking differences were noted in resistant regions- MD and CB compared to the vulnerable regions- FC, HC and ST. At younger age (25 ± 5 y), higher activity of electron transport chain enzymes and upregulation of metabolic and antioxidant proteins were noted in MD compared to FC and HC, that was sustained with increasing age (≥65 y). In contrast, the expression of synaptic proteins was higher in FC, HC and ST (vs. MD). In line with this, quantitative phospho-proteomics revealed activation of upstream regulators (ERS, PPARα) of mitochondrial metabolism and inhibition of synaptic pathways in MD. Microtubule Associated Protein Tau (MAPT) showed overexpression in FC, HC and ST both in young and older age (vs. MD). MAPT hyperphosphorylation and the activation of its kinases were noted in FC and HC with age. Our study demonstrates that regional heterogeneity in mitochondrial and other cellular functions contribute to SNV and protect regions such as MD, while rendering FC and HC vulnerable to NDDs. The findings also support the "last in, first out" hypothesis of ageing, wherein regions such as FC, that are the most recent to develop phylogenetically and ontogenetically, are the first to be affected in ageing and NDDs.
Collapse
Affiliation(s)
- Yarlagadda Anusha-Kiran
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India; Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - Praseeda Mol
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Gourav Dey
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Firdous Ahmad Bhat
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Sayali Chandrashekhar Deolankar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Mariamma Philip
- Department of Biostatistics, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India.
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India.
| |
Collapse
|
26
|
Lin L, Petralia RS, Holtzclaw L, Wang YX, Abebe D, Hoffman DA. Alzheimer's disease/dementia-associated brain pathology in aging DPP6-KO mice. Neurobiol Dis 2022; 174:105887. [PMID: 36209950 PMCID: PMC9617781 DOI: 10.1016/j.nbd.2022.105887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
We have previously reported that the single transmembrane protein Dipeptidyl Peptidase Like 6 (DPP6) impacts neuronal and synaptic development. DPP6-KO mice are impaired in hippocampal-dependent learning and memory and exhibit smaller brain size. Recently, we have described novel structures in hippocampal area CA1 in aging mice, apparently derived from degenerating presynaptic terminals, that are significantly more prevalent in DPP6-KO mice compared to WT mice of the same age and that these structures were observed earlier in development in DPP6-KO mice. These novel structures appear as clusters of large puncta that colocalize NeuN, synaptophysin, and chromogranin A, and also partially label for MAP2, amyloid β, APP, α-synuclein, and phosphorylated tau, with synapsin-1 and VGluT1 labeling on their periphery. In this current study, using immunofluorescence and electron microscopy, we confirm that both APP and amyloid β are prevalent in these structures; and we show with immunofluorescence the presence of similar structures in humans with Alzheimer's disease. Here we also found evidence that aging DPP6-KO mutants show additional changes related to Alzheimer's disease. We used in vivo MRI to show reduced size of the DPP6-KO brain and hippocampus. Aging DPP6-KO hippocampi contained fewer total neurons and greater neuron death and had diagnostic biomarkers of Alzheimer's disease present including accumulation of amyloid β and APP and increase in expression of hyper-phosphorylated tau. The amyloid β and phosphorylated tau pathologies were associated with neuroinflammation characterized by increases in microglia and astrocytes. And levels of proinflammatory or anti-inflammatory cytokines increased in aging DPP6-KO mice. We finally show that aging DPP6-KO mice display circadian dysfunction, a common symptom of Alzheimer's disease. Together these results indicate that aging DPP6-KO mice show symptoms of enhanced neurodegeneration reminiscent of dementia associated with a novel structure resulting from synapse loss and neuronal death. This study continues our laboratory's work in discerning the function of DPP6 and here provides compelling evidence of a direct role of DPP6 in Alzheimer's disease.
Collapse
Affiliation(s)
- Lin Lin
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lynne Holtzclaw
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Abebe
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dax A Hoffman
- Molecular Neurophysiology and Biophysics Section, Program in Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Thomas J, Martinez-Reza MF, Thorwirth M, Zarb Y, Conzelmann KK, Hauck SM, Grade S, Götz M. Excessive local host-graft connectivity in aging and amyloid-loaded brain. SCIENCE ADVANCES 2022; 8:eabg9287. [PMID: 35687689 PMCID: PMC9187230 DOI: 10.1126/sciadv.abg9287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/25/2022] [Indexed: 05/24/2023]
Abstract
Transplantation is a clinically relevant approach for brain repair, but much remains to be understood about influences of the disease environment on transplant connectivity. To explore the effect of amyloid pathology in Alzheimer's disease (AD) and aging, we examined graft connectivity using monosynaptic rabies virus tracing in APP/PS1 mice and in 16- to 18-month-old wild-type (WT) mice. Transplanted neurons differentiated within 4 weeks and integrated well into the host visual cortex, receiving input from the appropriate brain regions for this area. Unexpectedly, we found a prominent several-fold increase in local inputs, in both amyloid-loaded and aged environments. State-of-the-art deep proteome analysis using mass spectrometry highlights complement system activation as a common denominator of environments promoting excessive local input connectivity. These data therefore reveal the key role of the host pathology in shaping the input connectome, calling for caution in extrapolating results from one pathological condition to another.
Collapse
Affiliation(s)
- Judith Thomas
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
| | - Maria Fernanda Martinez-Reza
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
| | - Manja Thorwirth
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Yvette Zarb
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer-Institute of Virology, Medical Faculty and Gene Center, Ludwig-Maximilians Universitaet Muenchen, D-81377 Muenchen, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Sofia Grade
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- SYNERGY, Excellence Cluster for Systems Neurology, Ludwig-Maximilians Universitaet Muenchen, D-82152 Planegg, Germany
| |
Collapse
|
28
|
Zhang J, Wu J, Lu D, To CH, Lam TC, Lin B. Retinal Proteomic Analysis in a Mouse Model of Endotoxin-Induced Uveitis Using Data-Independent Acquisition-Based Mass Spectrometry. Int J Mol Sci 2022; 23:ijms23126464. [PMID: 35742911 PMCID: PMC9223489 DOI: 10.3390/ijms23126464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/24/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Uveitis is a group of sight-threatening ocular inflammatory diseases, potentially leading to permanent vision loss in patients. However, it remains largely unknown how uveitis causes retinal malfunction and vision loss. Endotoxin-induced uveitis (EIU) in rodents is a good animal model to study uveitis and associated acute retinal inflammation. To understand the pathogenic mechanism of uveitis and screen potential targets for treatment, we analyzed the retinal proteomic profile of the EIU mouse model using a data-independent acquisition-based mass spectrometry (SWATH-MS). After systemic LPS administration, we observed activation of microglial cells accompanied with the elevation of pro-inflammatory mediators and visual function declines. In total, we observed 79 upregulated and 90 downregulated differentially expressed proteins (DEPs). Among the DEPs, we found that histone family members (histone H1, H2A, H2B) and blood proteins including haptoglobin (HP), hemopexin (HPX), and fibrinogen gamma chain (FGG) were dramatically increased in EIU groups relative to those in control groups. We identified phototransduction and synaptic vesicle cycle as the top two significant KEGG pathways. Moreover, canonical pathway analysis on DEPs using Ingenuity Pathway Analysis revealed top three most significant enriched pathways related to acute phase response signaling, synaptogenesis signaling, and eif2 signaling. We further confirmed upregulation of several DEPs associated with the acute phase response signaling including HP, HPX, and FGG in LPS-treated retinas by qPCR and Western blot. In summary, this study serves as the first report to detect retinal proteome changes in the EIU model. The study provides several potential candidates for exploring the mechanism and novel therapeutic targets for uveitis and other retinal inflammatory diseases.
Collapse
Affiliation(s)
- Jing Zhang
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
| | - Jiangmei Wu
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
| | - Daqian Lu
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
| | - Chi-Ho To
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
- Centre for Eye and Vision Research (CEVR), Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Thomas Chuen Lam
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
- Centre for Eye and Vision Research (CEVR), Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong SAR, China
- Correspondence: (T.C.L.); (B.L.)
| | - Bin Lin
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China; (J.Z.); (J.W.); (D.L.); (C.-H.T.)
- Centre for Eye and Vision Research (CEVR), Hong Kong SAR, China
- Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong SAR, China
- Correspondence: (T.C.L.); (B.L.)
| |
Collapse
|
29
|
Li X, Yang W, Shen Y, Liu F, Xiong X, Wu Q, Xiao Z, Yang X, Dang R, Manaenko A, Xie P, Li Q. Analysis of Age-Dependent Transcriptomic Changes in Response to Intracerebral Hemorrhage in Mice. Front Mol Neurosci 2022; 15:908683. [PMID: 35677585 PMCID: PMC9169040 DOI: 10.3389/fnmol.2022.908683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/18/2022] Open
Abstract
Age is a well-known risk factor that is independently associated with poor outcomes after intracerebral hemorrhage (ICH). However, the interrelationship between age and poor outcomes after ICH is not well defined. In this study, we aimed to investigate this relationship based on collagenase-induced ICH mice models. After being assessed neurological deficit 24 h after ICH, mice were euthanized and brain perihematomal tissues were used for RNA-sequencing (RNA-seq). And then the functions of differentially expressed genes (DEGs) identified by RNA-seq were analyzed using Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Ingenuity Pathway Analysis (IPA) and protein-protein interaction (PPI) analysis. In addition, we performed real-time quantitative polymerase chain reaction (RT-qPCR) for validation of candidate DEGs. In the behavioral tests, aged mice presented significantly worse neurological function than young mice and greater weight loss than aged sham controls 24 h after ICH. In DEGs analysis, ICH affected the expression of more genes in young mice (2,337 DEGs) compared with aged mice (2,005 DEGs). We found aged mice exhibited increased brain inflammatory responses compared with young animals and ICH induced significant activation of the interferon-β (IFN-β) and IFN signaling pathways exclusively in aged mice. Moreover, further analysis demonstrated that ICH resulted in the activation of cytosolic DNA-sensing pathway with the production of downstream molecule type I IFN, and the response to type I IFN was more significant in aged mice than in young mice. In agreement with the results of RNA-seq, RT-qPCR indicated that the expression of candidate genes of cyclic GMP-AMP synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), and IFN-β was significantly altered in aged mice after ICH. Taken together, our study indicated that compared to young animals, aged mice exhibit increased vulnerability to ICH and that the differences in transcriptional response patterns to ICH between young and aged mice. We believe that these findings will facilitate our understanding of ICH pathology and help to translate the results of preclinical studies into a clinical setting.
Collapse
Affiliation(s)
- Xinhui Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wensong Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiqing Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangyu Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Xiong
- Department of Neurology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Zhongsong Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xun Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruozhi Dang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anatol Manaenko
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Qi Li,
| | - Qi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Qi Li,
| |
Collapse
|
30
|
García-Magro N, Zegarra-Valdivia JA, Troyas-Martinez S, Torres-Aleman I, Nuñez A. Response Facilitation Induced by Insulin-Like Growth Factor-I in the Primary Somatosensory Cortex of Mice Was Reduced in Aging. Cells 2022; 11:cells11040717. [PMID: 35203366 PMCID: PMC8870291 DOI: 10.3390/cells11040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023] Open
Abstract
Aging is accompanied by a decline in cognition that can be due to a lower IGF-I level. We studied response facilitation induced in primary somatosensory (S1) cortical neurons by repetitive stimulation of whiskers in young and old mice. Layer 2/3 and 5/6 neurons were extracellularly recorded in young (≤ 6 months of age) and old (≥ 20 month of age) anesthetized mice. IGF-I injection in S1 cortex (10 nM; 0.2 μL) increased whisker responses in young and old animals. A stimulation train at 8 Hz induced a long-lasting response facilitation in only layer 2/3 neurons of young animals. However, all cortical neurons from young and old animals showed long-lasting response facilitation when IGF-I was applied in the S1 cortex. The reduction in response facilitation in old animals can be due to a reduction in the IGF-I receptors as was indicated by the immunohistochemistry study. Furthermore, a reduction in the performance of a whisker discrimination task was observed in old animals. In conclusion, our findings indicate that there is a reduction in the synaptic plasticity of S1 neurons during aging that can be recovered by IGF-I. Therefore, it opens the possibility of use IGF-I as a therapeutic tool to ameliorate the effects of heathy aging.
Collapse
Affiliation(s)
- Nuria García-Magro
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Facultad de Ciencias de la Salud, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Jonathan A. Zegarra-Valdivia
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Cajal Institute, Cibernet (CSIC), 28002 Madrid, Spain;
- Universidad Señor de Sipán, Chiclayo 02001, Peru
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Foundation for Science, 48009 Bilbao, Spain
| | - Sara Troyas-Martinez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
| | - Ignacio Torres-Aleman
- Cajal Institute, Cibernet (CSIC), 28002 Madrid, Spain;
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Foundation for Science, 48009 Bilbao, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (N.G.-M.); (J.A.Z.-V.); (S.T.-M.)
- Correspondence:
| |
Collapse
|
31
|
Hiroki CH, Sarden N, Hassanabad MF, Yipp BG. Innate Receptors Expression by Lung Nociceptors: Impact on COVID-19 and Aging. Front Immunol 2021; 12:785355. [PMID: 34975876 PMCID: PMC8716370 DOI: 10.3389/fimmu.2021.785355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022] Open
Abstract
The lungs are constantly exposed to non-sterile air which carries harmful threats, such as particles and pathogens. Nonetheless, this organ is equipped with fast and efficient mechanisms to eliminate these threats from the airways as well as prevent pathogen invasion. The respiratory tract is densely innervated by sensory neurons, also known as nociceptors, which are responsible for the detection of external stimuli and initiation of physiological and immunological responses. Furthermore, expression of functional innate receptors by nociceptors have been reported; however, the influence of these receptors to the lung function and local immune response is poorly described. The COVID-19 pandemic has shown the importance of coordinated and competent pulmonary immunity for the prevention of pathogen spread as well as prevention of excessive tissue injury. New findings suggest that lung nociceptors can be a target of SARS-CoV-2 infection; what remains unclear is whether innate receptor trigger sensory neuron activation during SARS-CoV-2 infection and what is the relevance for the outcomes. Moreover, elderly individuals often present with respiratory, neurological and immunological dysfunction. Whether aging in the context of sensory nerve function and innate receptors contributes to the disorders of these systems is currently unknown. Here we discuss the expression of innate receptors by nociceptors, particularly in the lungs, and the possible impact of their activation on pulmonary immunity. We then demonstrate recent evidence that suggests lung sensory neurons as reservoirs for SARS-CoV-2 and possible viral recognition via innate receptors. Lastly, we explore the mechanisms by which lung nociceptors might contribute to disturbance in respiratory and immunological responses during the aging process.
Collapse
Affiliation(s)
- Carlos H. Hiroki
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nicole Sarden
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mortaza F. Hassanabad
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G. Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
Seker FB, Fan Z, Gesierich B, Gaubert M, Sienel RI, Plesnila N. Neurovascular Reactivity in the Aging Mouse Brain Assessed by Laser Speckle Contrast Imaging and 2-Photon Microscopy: Quantification by an Investigator-Independent Analysis Tool. Front Neurol 2021; 12:745770. [PMID: 34858312 PMCID: PMC8631776 DOI: 10.3389/fneur.2021.745770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
The brain has a high energy demand but little to no energy stores. Therefore, proper brain function relies on the delivery of glucose and oxygen by the cerebral vasculature. The regulation of cerebral blood flow (CBF) occurs at the level of the cerebral capillaries and is driven by a fast and efficient crosstalk between neurons and vessels, a process termed neurovascular coupling (NVC). Experimentally NVC is mainly triggered by sensory stimulation and assessed by measuring either CBF by laser Doppler fluxmetry, laser speckle contrast imaging (LSCI), intrinsic optical imaging, BOLD fMRI, near infrared spectroscopy (NIRS) or functional ultrasound imaging (fUS). Since these techniques have relatively low spatial resolution, diameters of cerebral vessels are mainly assessed by 2-photon microscopy (2-PM). Results of studies on NVC rely on stable animal physiology, high-quality data acquisition, and unbiased data analysis, criteria, which are not easy to achieve. In the current study, we assessed NVC using two different imaging modalities, i.e., LSCI and 2-PM, and analyzed our data using an investigator-independent Matlab-based analysis tool, after manually defining the area of analysis in LSCI and vessels to measure in 2-PM. By investigating NVC in 6–8 weeks, 1-, and 2-year-old mice, we found that NVC was maximal in 1-year old mice and was significantly reduced in aged mice. These findings suggest that NVC is differently affected during the aging process. Most interestingly, specifically pial arterioles, seem to be distinctly affected by the aging. The main finding of our study is that the automated analysis tool works very efficiently in terms of time and accuracy. In fact, the tool reduces the analysis time of one animal from approximately 23 h to about 2 s while basically making no mistakes. In summary, we developed an experimental workflow, which allows us to reliably measure NVC with high spatial and temporal resolution in young and aged mice and to analyze these data in an investigator-independent manner.
Collapse
Affiliation(s)
- Fatma Burcu Seker
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Ziyu Fan
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Malo Gaubert
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Rebecca Isabella Sienel
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
33
|
Karabicici M, Azbazdar Y, Iscan E, Ozhan G. Misregulation of Wnt Signaling Pathways at the Plasma Membrane in Brain and Metabolic Diseases. MEMBRANES 2021; 11:844. [PMID: 34832073 PMCID: PMC8621778 DOI: 10.3390/membranes11110844] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/23/2021] [Accepted: 10/27/2021] [Indexed: 12/26/2022]
Abstract
Wnt signaling pathways constitute a group of signal transduction pathways that direct many physiological processes, such as development, growth, and differentiation. Dysregulation of these pathways is thus associated with many pathological processes, including neurodegenerative diseases, metabolic disorders, and cancer. At the same time, alterations are observed in plasma membrane compositions, lipid organizations, and ordered membrane domains in brain and metabolic diseases that are associated with Wnt signaling pathway activation. Here, we discuss the relationships between plasma membrane components-specifically ligands, (co) receptors, and extracellular or membrane-associated modulators-to activate Wnt pathways in several brain and metabolic diseases. Thus, the Wnt-receptor complex can be targeted based on the composition and organization of the plasma membrane, in order to develop effective targeted therapy drugs.
Collapse
Affiliation(s)
- Mustafa Karabicici
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir 35340, Turkey; (M.K.); (Y.A.); (E.I.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir 35340, Turkey
| | - Yagmur Azbazdar
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir 35340, Turkey; (M.K.); (Y.A.); (E.I.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir 35340, Turkey
| | - Evin Iscan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir 35340, Turkey; (M.K.); (Y.A.); (E.I.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir 35340, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center (IBG), Dokuz Eylul University Health Campus, Inciralti-Balcova, Izmir 35340, Turkey; (M.K.); (Y.A.); (E.I.)
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, Inciralti-Balcova, Izmir 35340, Turkey
| |
Collapse
|
34
|
Zegarra-Valdivia JA, Chaves-Coira I, Fernandez de Sevilla ME, Martinez-Rachadell L, Esparza J, Torres-Aleman I, Nuñez A. Reduced Insulin-Like Growth Factor-I Effects in the Basal Forebrain of Aging Mouse. Front Aging Neurosci 2021; 13:682388. [PMID: 34539376 PMCID: PMC8442768 DOI: 10.3389/fnagi.2021.682388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
It is known that aging is frequently accompanied by a decline in cognition. Furthermore, aging is associated with lower serum IGF-I levels that may contribute to this deterioration. We studied the effect of IGF-I in neurons of the horizontal diagonal band of Broca (HDB) of young (≤6 months old) and old (≥20-month-old) mice to determine if changes in the response of these neurons to IGF-I occur along with aging. Local injection of IGF-I in the HDB nucleus increased their neuronal activity and induced fast oscillatory activity in the electrocorticogram (ECoG). Furthermore, IGF-I facilitated tactile responses in the primary somatosensory cortex elicited by air-puffs delivered in the whiskers. These excitatory effects decreased in old mice. Immunohistochemistry showed that cholinergic HDB neurons express IGF-I receptors and that IGF-I injection increased the expression of c-fos in young, but not in old animals. IGF-I increased the activity of optogenetically-identified cholinergic neurons in young animals, suggesting that most of the IGF-I-induced excitatory effects were mediated by activation of these neurons. Effects of aging were partially ameliorated by chronic IGF-I treatment in old mice. The present findings suggest that reduced IGF-I activity in old animals participates in age-associated changes in cortical activity.
Collapse
Affiliation(s)
- Jonathan A Zegarra-Valdivia
- Cajal Institute (CSIC), Madrid, Spain.,CIBERNED, Madrid, Spain.,Universidad Nacional de San Agustín de Arequipa, Arequipa, Peru.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Irene Chaves-Coira
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | - Ignacio Torres-Aleman
- Cajal Institute (CSIC), Madrid, Spain.,CIBERNED, Madrid, Spain.,Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Angel Nuñez
- Department of Anatomy, Histology and Neurosciences, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Choudhury ME, Miyanishi K, Takeda H, Tanaka J. Microglia and the Aging Brain: Are Geriatric Microglia Linked to Poor Sleep Quality? Int J Mol Sci 2021; 22:ijms22157824. [PMID: 34360590 PMCID: PMC8345993 DOI: 10.3390/ijms22157824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Poor sleep quality and disrupted circadian behavior are a normal part of aging and include excessive daytime sleepiness, increased sleep fragmentation, and decreased total sleep time and sleep quality. Although the neuronal decline underlying the cellular mechanism of poor sleep has been extensively investigated, brain function is not fully dependent on neurons. A recent antemortem autographic study and postmortem RNA sequencing and immunohistochemical studies on aged human brain have investigated the relationship between sleep fragmentation and activation of the innate immune cells of the brain, microglia. In the process of aging, there are marked reductions in the number of brain microglial cells, and the depletion of microglial cells disrupts circadian rhythmicity of brain tissue. We also showed, in a previous study, that pharmacological suppression of microglial function induced sleep abnormalities. However, the mechanism underlying the contribution of microglial cells to sleep homeostasis is only beginning to be understood. This review revisits the impact of aging on the microglial population and activation, as well as microglial contribution to sleep maintenance and response to sleep loss. Most importantly, this review will answer questions such as whether there is any link between senescent microglia and age-related poor quality sleep and how this exacerbates neurodegenerative disease.
Collapse
Affiliation(s)
- Mohammed E. Choudhury
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan
- Correspondence: (M.E.C.); (J.T.)
| | - Kazuya Miyanishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan;
| | - Haruna Takeda
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Aoba, Sendai 980-8575, Miyagi, Japan;
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Shitsukawa, Toon 791-0295, Ehime, Japan
- Correspondence: (M.E.C.); (J.T.)
| |
Collapse
|
36
|
Banerjee S, Vernon S, Jiao W, Choi BJ, Ruchti E, Asadzadeh J, Burri O, Stowers RS, McCabe BD. Miniature neurotransmission is required to maintain Drosophila synaptic structures during ageing. Nat Commun 2021; 12:4399. [PMID: 34285221 PMCID: PMC8292383 DOI: 10.1038/s41467-021-24490-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 06/22/2021] [Indexed: 11/27/2022] Open
Abstract
The decline of neuronal synapses is an established feature of ageing accompanied by the diminishment of neuronal function, and in the motor system at least, a reduction of behavioural capacity. Here, we have investigated Drosophila motor neuron synaptic terminals during ageing. We observed cumulative fragmentation of presynaptic structures accompanied by diminishment of both evoked and miniature neurotransmission occurring in tandem with reduced motor ability. Through discrete manipulation of each neurotransmission modality, we find that miniature but not evoked neurotransmission is required to maintain presynaptic architecture and that increasing miniature events can both preserve synaptic structures and prolong motor ability during ageing. Our results establish that miniature neurotransmission, formerly viewed as an epiphenomenon, is necessary for the long-term stability of synaptic connections. Synaptic structures disintegrate and fragment as ageing progresses. Here the authors find that miniature neurotransmission is required to maintain adult motor synapse structures in Drosophila and that increasing miniature events can preserve motor ability during ageing.
Collapse
Affiliation(s)
- Soumya Banerjee
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Samuel Vernon
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Ben Jiwon Choi
- Department of Biology, New York University, New York, USA
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Olivier Burri
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - R Steven Stowers
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, USA
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.
| |
Collapse
|
37
|
Petralia RS, Yao PJ, Kapogiannis D, Wang YX. Invaginating Structures in Synapses - Perspective. Front Synaptic Neurosci 2021; 13:685052. [PMID: 34108873 PMCID: PMC8180840 DOI: 10.3389/fnsyn.2021.685052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/28/2021] [Indexed: 01/05/2023] Open
Abstract
Invaginating structures are common in the synapses of most animals. However, the details of these invaginating structures remain understudied in part because they are not well resolved in light microscopy and were often misidentified in early electron microscope (EM) studies. Utilizing experimental techniques along with the latest advances in microscopy, such as focused ion beam-scanning EM (FIB-SEM), evidence is gradually building to suggest that the synaptic invaginating structures contribute to synapse development, maintenance, and plasticity. These invaginating structures are most elaborate in synapses mediating rapid integration of signals, such as muscle contraction, mechanoreception, and vision. Here we argue that the synaptic invaginations should be considered in future studies seeking to understand their role in sensory integration and coordination, learning, and memory. We review the various types of invaginating structures in the synapses and discuss their potential functions. We also present several new examples of invaginating structures from a variety of animals including Drosophila and mice, mainly using FIB-SEM, with which we trace the form and arrangement of these structures.
Collapse
Affiliation(s)
- Ronald S. Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders/National Institutes of Health, Bethesda, MD, United States
| | - Pamela J. Yao
- Laboratory of Clinical Investigation, National Institute on Aging/National Institutes of Health, Bethesda, MD, United States
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging/National Institutes of Health, Bethesda, MD, United States
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders/National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
38
|
Burrinha T, Martinsson I, Gomes R, Terrasso AP, Gouras GK, Almeida CG. Up-regulation of APP endocytosis by neuronal aging drives amyloid dependent-synapse loss. J Cell Sci 2021; 134:240244. [PMID: 33910234 DOI: 10.1242/jcs.255752] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/03/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal aging increases the risk of late-onset Alzheimer's disease. During normal aging, synapses decline, and β-amyloid (Aβ) accumulates intraneuronally. However, little is known about the underlying cell biological mechanisms. We studied normal neuronal aging using normal aged brain and aged mouse primary neurons that accumulate lysosomal lipofuscin and show synapse loss. We identify the up-regulation of amyloid precursor protein (APP) endocytosis as a neuronal aging mechanism that potentiates APP processing and Aβ production in vitro and in vivo. The increased APP endocytosis may contribute to the observed early endosomes enlargement in the aged brain. Mechanistically, we show that clathrin-dependent APP endocytosis requires F-actin and that clathrin and endocytic F-actin increase with neuronal aging. Finally, Aβ production inhibition reverts synaptic decline in aged neurons while Aβ accumulation, promoted by endocytosis up-regulation in younger neurons, recapitulates aging-related synapse decline. Overall, we identify APP endocytosis up-regulation as a potential mechanism of neuronal aging and, thus, a novel target to prevent late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Tatiana Burrinha
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056 Lisboa,Portugal
| | - Isak Martinsson
- Experimental Dementia Research Unit, Lund University, 22184 Lund, Sweden
| | - Ricardo Gomes
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056 Lisboa,Portugal.,iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Ana Paula Terrasso
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056 Lisboa,Portugal.,iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Gunnar K Gouras
- Experimental Dementia Research Unit, Lund University, 22184 Lund, Sweden
| | - Cláudia Guimas Almeida
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056 Lisboa,Portugal
| |
Collapse
|
39
|
Björk V. Aging of the Suprachiasmatic Nucleus, CIRCLONSA Syndrome, Implications for Regenerative Medicine and Restoration of the Master Body Clock. Rejuvenation Res 2021; 24:274-282. [PMID: 33573456 DOI: 10.1089/rej.2020.2388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The suprachiasmatic nucleus (SCN) in the brain is the master regulator of the circadian clocks throughout the human body. With increasing age the circadian clock in humans and other mammals becomes increasingly disorganized leading to a large number of more or less well-categorized problems. While a lot of aging research has focused on the peripheral clocks in tissues across organisms, it remains a paramount task to quantify aging of the most important master clock, the human SCN. Furthermore, a pipeline needs to be developed with therapies to mitigate the systemic cellular circadian dysfunction in the elderly and ultimately repair and reverse aging of the SCN itself. A disease classification for the aging SCN, Circadian Clock Neuronal Senile Atrophy (CIRCLONSA syndrome), would improve research funding and goal-oriented biotechnological entrepreneurship.
Collapse
|
40
|
van Soest APM, van de Rest O, Witkamp RF, de Groot LCPGM. Positive effects of folic acid supplementation on cognitive aging are dependent on ω-3 fatty acid status: a post hoc analysis of the FACIT trial. Am J Clin Nutr 2021; 113:801-809. [PMID: 33564824 DOI: 10.1093/ajcn/nqaa373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although epidemiological studies suggest a protective role of B vitamins and omega-3 (ω-3) fatty acids in cognitive decline, findings from intervention studies are conflicting. Mechanistic studies suggest that the ω-3 (n-3) fatty acid status can modulate the effects of B vitamins on cognitive decline. OBJECTIVES We investigated the interaction between baseline ω-3 fatty acid statuses and folic acid treatment on cognitive decline in a placebo-controlled trial [FACIT (Folic Acid and Carotid Intima-media Thickness)]. METHODS This post hoc analysis included 791 older adults aged 50-70 y with plasma total homocysteine ≥13 µmol/L and ≤26 µmol/L and serum vitamin B12 ≥200 pmol/L. Participants received 800 µg folic acid or placebo daily for 3 y. Global cognitive functioning and domain-specific functioning (episodic memory, information processing speed, executive functioning) were assessed at baseline and after 3 y. The effect of the folic acid supplementation was analyzed according to tertiles of baseline ω-3 fatty acid concentrations using linear multiple regression. RESULTS The mean ± SD age of the study population was 60.2 ± 5.6 y, and the mean ± SD Mini-Mental State Examination score was 28.6 ± 1.5. The treatment effect of folic acid was significantly larger in participants in the low compared to high ω-3 fatty acid tertile for global cognition (difference in z-score: mean ± SE = 0.16 ± 0.059; P < 0.01). Regarding domain-specific functioning, similar results were observed for information processing speed (mean ± SE = 0.167 ± 0.068; P = 0.01). There were no overall interactions between folic acid treatment and ω-3 fatty acid tertiles for episodic memory (P = 0.14) and executive functioning (P = 0.21). CONCLUSIONS This post hoc analysis revealed that the efficacy of folic acid treatment on cognitive functioning is dependent on the ω-3 fatty acid status. Individuals with a lower ω-3 fatty acid status at baseline benefited from folic acid treatment, while individuals with a higher ω-3 fatty acid status did not. The results potentially explain the inconsistency in outcomes of B-vitamin supplementation trials and emphasize the importance of a personalized approach. This trial was registered at clinicaltrials.gov as NCT00110604.
Collapse
Affiliation(s)
- Annick P M van Soest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Ondine van de Rest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Renger F Witkamp
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| | - Lisette C P G M de Groot
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
41
|
Koch SC, Nelson A, Hartenstein V. Structural aspects of the aging invertebrate brain. Cell Tissue Res 2021; 383:931-947. [PMID: 33409654 PMCID: PMC7965346 DOI: 10.1007/s00441-020-03314-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/28/2020] [Indexed: 11/26/2022]
Abstract
Aging is characterized by a decline in neuronal function in all animal species investigated so far. Functional changes are accompanied by and may be in part caused by, structurally visible degenerative changes in neurons. In the mammalian brain, normal aging shows abnormalities in dendrites and axons, as well as ultrastructural changes in synapses, rather than global neuron loss. The analysis of the structural features of aging neurons, as well as their causal link to molecular mechanisms on the one hand, and the functional decline on the other hand is crucial in order to understand the aging process in the brain. Invertebrate model organisms like Drosophila and C. elegans offer the opportunity to apply a forward genetic approach to the analysis of aging. In the present review, we aim to summarize findings concerning abnormalities in morphology and ultrastructure in invertebrate brains during normal aging and compare them to what is known for the mammalian brain. It becomes clear that despite of their considerably shorter life span, invertebrates display several age-related changes very similar to the mammalian condition, including the retraction of dendritic and axonal branches at specific locations, changes in synaptic density and increased accumulation of presynaptic protein complexes. We anticipate that continued research efforts in invertebrate systems will significantly contribute to reveal (and possibly manipulate) the molecular/cellular pathways leading to neuronal aging in the mammalian brain.
Collapse
Affiliation(s)
- Sandra C Koch
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Annie Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles (UCLA), Los Angeles, California, USA.
| |
Collapse
|
42
|
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: 165] [Impact Index Per Article: 41.3] [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
|
43
|
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: 48] [Impact Index Per Article: 12.0] [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
|
44
|
Structural and Functional Synaptic Plasticity Induced by Convergent Synapse Loss in the Drosophila Neuromuscular Circuit. J Neurosci 2021; 41:1401-1417. [PMID: 33402422 DOI: 10.1523/jneurosci.1492-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/28/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022] Open
Abstract
Throughout the nervous system, the convergence of two or more presynaptic inputs on a target cell is commonly observed. The question we ask here is to what extent converging inputs influence each other's structural and functional synaptic plasticity. In complex circuits, isolating individual inputs is difficult because postsynaptic cells can receive thousands of inputs. An ideal model to address this question is the Drosophila larval neuromuscular junction (NMJ) where each postsynaptic muscle cell receives inputs from two glutamatergic types of motor neurons (MNs), known as 1b and 1s MNs. Notably, each muscle is unique and receives input from a different combination of 1b and 1s MNs; we surveyed multiple muscles for this reason. Here, we identified a cell-specific promoter that allows ablation of 1s MNs postinnervation and measured structural and functional responses of convergent 1b NMJs using microscopy and electrophysiology. For all muscles examined in both sexes, ablation of 1s MNs resulted in NMJ expansion and increased spontaneous neurotransmitter release at corresponding 1b NMJs. This demonstrates that 1b NMJs can compensate for the loss of convergent 1s MNs. However, only a subset of 1b NMJs showed compensatory evoked neurotransmission, suggesting target-specific plasticity. Silencing 1s MNs led to similar plasticity at 1b NMJs, suggesting that evoked neurotransmission from 1s MNs contributes to 1b synaptic plasticity. Finally, we genetically blocked 1s innervation in male larvae and robust 1b synaptic plasticity was eliminated, raising the possibility that 1s NMJ formation is required to set up a reference for subsequent synaptic perturbations.SIGNIFICANCE STATEMENT In complex neural circuits, multiple convergent inputs contribute to the activity of the target cell, but whether synaptic plasticity exists among these inputs has not been thoroughly explored. In this study, we examined synaptic plasticity in the structurally and functionally tractable Drosophila larval neuromuscular system. In this convergent circuit, each muscle is innervated by a unique pair of motor neurons. Removal of one neuron after innervation causes the adjacent neuron to increase neuromuscular junction outgrowth and functional output. However, this is not a general feature as each motor neuron differentially compensates. Further, robust compensation requires initial coinnervation by both neurons. Understanding how neurons respond to perturbations in adjacent neurons will provide insight into nervous system plasticity in both healthy and disease states.
Collapse
|
45
|
Habich A, Fehér KD, Antonenko D, Boraxbekk CJ, Flöel A, Nissen C, Siebner HR, Thielscher A, Klöppel S. Stimulating aged brains with transcranial direct current stimulation: Opportunities and challenges. Psychiatry Res Neuroimaging 2020; 306:111179. [PMID: 32972813 DOI: 10.1016/j.pscychresns.2020.111179] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/30/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Ageing involves significant neurophysiological changes that are both systematic while at the same time exhibiting divergent trajectories across individuals. These changes underlie cognitive impairments in elderly while also affecting the response of aged brains to interventions like transcranial direct current stimulation (tDCS). While the cognitive benefits of tDCS are more variable in elderly, older adults also respond differently to stimulation protocols compared to young adults. The age-related neurophysiological changes influencing the responsiveness to tDCS remain to be addressed in-depth. We review and discuss the premise that, in comparison to the better calibrated brain networks present in young adults, aged systems perform further away from a homoeostatic set-point. We argue that this age-related neurophysiological deviation from the homoeostatic optimum extends the leeway for tDCS to modulate the aged brain. This promotes the potency of immediate tDCS effects to induce directional plastic changes towards the homoeostatic equilibrium despite the impaired plasticity induction in elderly. We also consider how age-related neurophysiological changes pose specific challenges for tDCS that necessitate proper adaptations of stimulation protocols. Appreciating the distinctive properties of aged brains and the accompanying adjustment of stimulation parameters can increase the potency and reliability of tDCS as a treatment avenue in older adults.
Collapse
Affiliation(s)
- Annegret Habich
- University Hospital of Old Age Psychiatry and Psychotherpa, University of Bern, Bolligenstrasse 111, 3000 Bern, Switzerland; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany.
| | - Kristoffer D Fehér
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern, Switzerland
| | - Daria Antonenko
- Department of Neurology, University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Carl-Johan Boraxbekk
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Østvej, 2650 Hvidovre, Denmark; Department of Radiation Sciences, Umeå University, 90187 Umeå, Sweden; Institute of Sports Medicine Copenhagen (ISMC), Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Agnes Flöel
- Department of Neurology, University of Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany; German Center for Neurodegenerative Diseases, Ellernholzstraße 1-2, 17489 Greifswald, Germany
| | - Christoph Nissen
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bolligenstrasse 111, 3000 Bern, Switzerland; Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Freiburg, Hauptstraße 5, 79104 Freiburg, Germany
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Østvej, 2650 Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital Bispebjerg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Nørre Allé 20, 2200 Copenhagen, Denmark
| | - Axel Thielscher
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Hvidovre, Østvej, 2650 Hvidovre, Denmark; Department of Electrical Engineering, Technical University of Denmark, Ørsteds Pl. 348, 2800 Kgs. Lyngby, Denmark
| | - Stefan Klöppel
- University Hospital of Old Age Psychiatry and Psychotherpa, University of Bern, Bolligenstrasse 111, 3000 Bern, Switzerland
| |
Collapse
|
46
|
Ötzkan S, Muller WE, Gibson Wood W, Eckert GP. Effects of 7,8-Dihydroxyflavone on Lipid Isoprenoid and Rho Protein Levels in Brains of Aged C57BL/6 Mice. Neuromolecular Med 2020; 23:130-139. [PMID: 33377988 PMCID: PMC7929957 DOI: 10.1007/s12017-020-08640-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022]
Abstract
Synaptic impairment may be the main cause of cognitive dysfunction in brain aging that is probably due to a reduction in synaptic contact between the axonal buttons and dendritic spines. Rho proteins including the small GTPase Rac1 have become key regulators of neuronal morphogenesis that supports synaptic plasticity. Small Rho- and Ras-GTPases are post-translationally modified by the isoprenoids geranylgeranyl pyrophosphate (GGPP) and farnesyl pyrophosphate (FPP), respectively. For all GTPases, anchoring in the plasma membrane is essential for their activation by guanine nucleotide exchange factors (GEFs). Rac1-specific GEFs include the protein T lymphoma invasion and metastasis 1 (Tiam1). Tiam1 interacts with the TrkB receptor to mediate the brain-derived neurotrophic factor (BDNF)-induced activation of Rac1, resulting in cytoskeletal rearrangement and changes in cellular morphology. The flavonoid 7,8-dihydroxyflavone (7,8-DHF) acts as a highly affine-selective TrkB receptor agonist and causes the dimerization and autophosphorylation of the TrkB receptor and thus the activation of downstream signaling pathways. In the current study, we investigated the effects of 7,8-DHF on cerebral lipid isoprenoid and Rho protein levels in male C57BL/6 mice aged 3 and 23 months. Aged mice were daily treated with 100 mg/kg b.w. 7,8-DHF by oral gavage for 21 days. FPP, GGPP, and cholesterol levels were determined in brain tissue. In the same tissue, the protein content of Tiam1 and TrkB in was measured. The cellular localization of the small Rho-GTPase Rac1 and small Rab-GTPase Rab3A was studied in total brain homogenates and membrane preparations. We report the novel finding that 7,8-DHF restored levels of the Rho proteins Rac1 and Rab3A in membrane preparations isolated from brains of treated aged mice. The selective TrkB agonist 7,8-DHF did not affect BDNF and TrkB levels, but restored Tiam1 levels that were found to be reduced in brains of aged mice. FPP, GGPP, and cholesterol levels were significantly elevated in brains of aged mice but not changed by 7,8-DHF treatment. Hence, 7,8-DHF may be useful as pharmacological tool to treat age-related cognitive dysfunction although the underlying mechanisms need to be elucidated in detail.
Collapse
Affiliation(s)
- Sarah Ötzkan
- Department of Pharmacology, Biocenter Niederursel, University of Frankfurt, Goethe-University, Max-von-Laue-St. 9, 60438, Frankfurt, Germany
| | - Walter E Muller
- Department of Pharmacology, Biocenter Niederursel, University of Frankfurt, Goethe-University, Max-von-Laue-St. 9, 60438, Frankfurt, Germany
| | - W Gibson Wood
- Department of Pharmacology, Geriatric Research, Education and Clinical Center, University of Minnesota School of Medicine, VAMC, Minneapolis, MN, 55417, USA
| | - Gunter P Eckert
- Department of Pharmacology, Biocenter Niederursel, University of Frankfurt, Goethe-University, Max-von-Laue-St. 9, 60438, Frankfurt, Germany.
- Institute of Nutritional Sciences, Laboratory for Nutrition in Prevention and Therapy, Justus-Liebig-University of Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstr. 81, 35392, Giessen, Germany.
| |
Collapse
|
47
|
Aghaizu ND, Jin H, Whiting PJ. Dysregulated Wnt Signalling in the Alzheimer's Brain. Brain Sci 2020; 10:E902. [PMID: 33255414 PMCID: PMC7761504 DOI: 10.3390/brainsci10120902] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/16/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023] Open
Abstract
The Wnt signalling system is essential for both the developing and adult central nervous system. It regulates numerous cellular functions ranging from neurogenesis to blood brain barrier biology. Dysregulated Wnt signalling can thus have significant consequences for normal brain function, which is becoming increasingly clear in Alzheimer's disease (AD), an age-related neurodegenerative disorder that is the most prevalent form of dementia. AD exhibits a range of pathophysiological manifestations including aberrant amyloid precursor protein processing, tau pathology, synapse loss, neuroinflammation and blood brain barrier breakdown, which have been associated to a greater or lesser degree with abnormal Wnt signalling. Here we provide a comprehensive overview of the role of Wnt signalling in the CNS, and the research that implicates dysregulated Wnt signalling in the ageing brain and in AD pathogenesis. We also discuss the opportunities for therapeutic intervention in AD via modulation of the Wnt signalling pathway, and highlight some of the challenges and the gaps in our current understanding that need to be met to enable that goal.
Collapse
Affiliation(s)
- Nozie D. Aghaizu
- UK Dementia Research Institute at University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK;
| | - Hanqing Jin
- UK Dementia Research Institute at University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK;
| | - Paul J. Whiting
- UK Dementia Research Institute at University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK;
- ARUK Drug Discovery Institute (DDI), University College London, Cruciform Building, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
48
|
Age-Dependent Decline in Synaptic Mitochondrial Function Is Exacerbated in Vulnerable Brain Regions of Female 3xTg-AD Mice. Int J Mol Sci 2020; 21:ijms21228727. [PMID: 33227902 PMCID: PMC7699171 DOI: 10.3390/ijms21228727] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 02/04/2023] Open
Abstract
Synaptic aging has been associated with neuronal circuit dysfunction and cognitive decline. Reduced mitochondrial function may be an early event that compromises synaptic integrity and neurotransmission in vulnerable brain regions during physiological and pathological aging. Thus, we aimed to measure mitochondrial function in synapses from three brain regions at two different ages in the 3xTg-AD mouse model and in wild mice. We found that aging is the main factor associated with the decline in synaptic mitochondrial function, particularly in synapses isolated from the cerebellum. Accumulation of toxic compounds, such as tau and Aβ, that occurred in the 3xTg-AD mouse model seemed to participate in the worsening of this decline in the hippocampus. The changes in synaptic bioenergetics were also associated with increased activation of the mitochondrial fission protein Drp1. These results suggest the presence of altered mechanisms of synaptic mitochondrial dynamics and their quality control during aging and in the 3xTg-AD mouse model; they also point to bioenergetic restoration as a useful therapeutic strategy to preserve synaptic function during aging and at the early stages of Alzheimer's disease (AD).
Collapse
|
49
|
Lopez-Soley E, Solana E, Martínez-Heras E, Andorra M, Radua J, Prats-Uribe A, Montejo C, Sola-Valls N, Sepulveda M, Pulido-Valdeolivas I, Blanco Y, Martinez-Lapiscina EH, Saiz A, Llufriu S. Impact of Cognitive Reserve and Structural Connectivity on Cognitive Performance in Multiple Sclerosis. Front Neurol 2020; 11:581700. [PMID: 33193039 PMCID: PMC7662554 DOI: 10.3389/fneur.2020.581700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/30/2020] [Indexed: 01/07/2023] Open
Abstract
Background: Cognitive reserve (CR) could attenuate the impact of the brain burden on the cognition in people with multiple sclerosis (PwMS). Objective: To explore the relationship between CR and structural brain connectivity and investigate their role on cognition in PwMS cognitively impaired (PwMS-CI) and cognitively preserved (PwMS-CP). Methods: In this study, 181 PwMS (71% female; 42.9 ± 10.0 years) were evaluated using the Cognitive Reserve Questionnaire (CRQ), Brief Repeatable Battery of Neuropsychological tests, and MRI. Brain lesion and gray matter volumes were quantified, as was the structural network connectivity. Patients were classified as PwMS-CI (z scores = −1.5 SD in at least two tests) or PwMS-CP. Linear and multiple regression analyses were run to evaluate the association of CRQ and structural connectivity with cognition in each group. Hedges's effect size was used to compute the strength of associations. Results: We found a very low association between CRQ scores and connectivity metrics in PwMS-CP, while in PwMS-CI, this relation was low to moderate. The multiple regression model, adjusted for age, gender, mood, lesion volume, and graph metrics (local and global efficiency, and transitivity), indicated that the CRQ (β = 0.26, 95% CI: 0.17–0.35) was associated with cognition (adj R2 = 0.34) in PwMS-CP (55%). In PwMS-CI, CRQ (β = 0.18, 95% CI: 0.07–0.29), age, and network global efficiency were independently associated with cognition (adj R2 = 0.55). The age- and gender-adjusted association between CRQ score and global efficiency on having an impaired cognitive status was −0.338 (OR: 0.71, p = 0.036) and −0.531 (OR: 0.59, p = 0.002), respectively. Conclusions: CR seems to have a marginally significant effect on brain structural connectivity, observed in patients with more severe clinical impairment. It protects PwMS from cognitive decline regardless of their cognitive status, yet once cognitive impairment has set in, brain damage and aging are also influencing cognitive performance.
Collapse
Affiliation(s)
- Elisabet Lopez-Soley
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Elisabeth Solana
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Eloy Martínez-Heras
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Magi Andorra
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Joaquim Radua
- Imaging of Mood- and Anxiety-Related Disorders (IMARD) Group, Mental Health Research Networking Center (CIBERSAM), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Psychosis Studies, King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom.,Department of Clinical Neuroscience, Centre for Psychiatric Research and Education, Karolinska Institutet, Solna, Sweden
| | - Albert Prats-Uribe
- Centre for Statistics in Medicine, Botnar Research Centre, Nuffiel Department of Orthopeadics, rheumatology and musculoskeletal sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Carmen Montejo
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Nuria Sola-Valls
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Maria Sepulveda
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Irene Pulido-Valdeolivas
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Yolanda Blanco
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Elena H Martinez-Lapiscina
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Albert Saiz
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Sara Llufriu
- Laboratory of Advanced Imaging in Neuroimmunological Diseases, Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Hospital Clinic Barcelona, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
50
|
Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020; 12:247. [PMID: 32848716 PMCID: PMC7426063 DOI: 10.3389/fnagi.2020.00247] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
The adult central nervous system (CNS) contains resident stem cells within specific niches that maintain a self-renewal and proliferative capacity to generate new neurons, astrocytes, and oligodendrocytes throughout adulthood. Physiological aging is associated with a progressive loss of function and a decline in the self-renewal and regenerative capacities of CNS stem cells. Also, the biggest risk factor for neurodegenerative diseases is age, and current in vivo and in vitro models of neurodegenerative diseases rarely consider this. Therefore, combining both aging research and appropriate interrogation of animal disease models towards the understanding of the disease and age-related stem cell failure is imperative to the discovery of new therapies. This review article will highlight the main intrinsic and extrinsic regulators of neural stem cell (NSC) aging and discuss how these factors impact normal homeostatic functions within the adult brain. We will consider established in vivo animal and in vitro human disease model systems, and then discuss the current and future trajectories of novel senotherapeutics that target aging NSCs to ameliorate brain disease.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|