1
|
Pittaras EC, Artal JM, Ajibola G, Allocca G, Bennett M, Camargo A, Carpio A, Gessner N, Hinton M, Pizzitola R, Tan N, Zhang E, Zhong A, Heller HC. Short-term γ-aminobutyric acid antagonist treatment improves long-term sleep quality, memory, and decision-making in a Down syndrome mouse model. Sleep 2025; 48:zsae300. [PMID: 39719304 DOI: 10.1093/sleep/zsae300] [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: 06/05/2024] [Revised: 11/08/2024] [Indexed: 12/26/2024] Open
Abstract
Down syndrome (DS) is a common genetic condition affecting people worldwide. It involves cognitive disabilities for which there are no drug therapies. The Ts65Dn mouse model of DS shows cognitive impairment due to a reduction in neuron number and connectivity as well as excessive neuronal activity, as γ-aminobutyric acid (GABA) antagonist treatment restores memory in these mice. Our study showed the effects of GABA antagonist treatment on sleep and decision-making in Ts65Dn mice. We administered a daily, low oral dose of pentylenetetrazol (PTZ) in milk to Ts65Dn mice for 17 days. Decision-making was tested with and without PTZ treatment. Short and long-term memories were tested before, immediately after, and 1 month following PTZ treatment. Electro-encephalography was also recorded at these three time points to study the effect of the treatment on sleep. We showed that PTZ treatment improved long-term recognition, but not short term memory and led to more Ts65Dn mice showing safer decision-making behavior. PTZ treatment showed a moderate and only global beneficial effect on sleep by decreasing the global amount of wake and increasing non-rapid eye movement sleep in the Ts65Dn mice, which may explain the observed cognitive improvements. These results bring new knowledge on the role of GABA in sleep, memory consolidation, and decision-making abilities in DS.
Collapse
Affiliation(s)
- Elsa C Pittaras
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jonathan M Artal
- Department of Biology, Stanford University, Stanford, CA, USA
- Stanford Law School, Stanford University, Stanford, CA, USA
| | - Grace Ajibola
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Giancarlo Allocca
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- School of Biomedical Sciences, University of Melbourne, Parkville, Victoria, Australia
- Somnivore Pty. Ltd., Bacchus Marsh, Victoria, Australia
| | - Mia Bennett
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Human Biology, School of Humanities and Sciences, Stanford University, Stanford, CA, USA
| | | | - Angelica Carpio
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Myles Hinton
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Rebecca Pizzitola
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Symbolic Systems, School of Humanities and Sciences, Stanford University, Stanford, CA, USA
| | - Natalie Tan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Evelyn Zhang
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alan Zhong
- Department of Biology, Stanford University, Stanford, CA, USA
- BASIS Independent Silicon Valley, San Jose, CA, USA
| | - Horace C Heller
- Department of Biology, Stanford University, Stanford, CA, USA
| |
Collapse
|
2
|
Islam MR, Jackson B, Naomi MT, Schatz B, Tan N, Murdock M, Park DS, Amorim DR, Jiang X, Pineda SS, Adaikkan C, Fernandez Avalos V, Geigenmuller U, Firenze RM, Kellis M, Boyden ES, Tsai LH. Multisensory gamma stimulation enhances adult neurogenesis and improves cognitive function in male mice with Down Syndrome. PLoS One 2025; 20:e0317428. [PMID: 40273201 PMCID: PMC12021272 DOI: 10.1371/journal.pone.0317428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/27/2024] [Indexed: 04/26/2025] Open
Affiliation(s)
- Md Rezaul Islam
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Brennan Jackson
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Maeesha Tasnim Naomi
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Brooke Schatz
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Noah Tan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Mitchell Murdock
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Dong Shin Park
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Daniela Rodrigues Amorim
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Xueqiao Jiang
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, Massachusetts, United States of America
| | - S. Sebastian Pineda
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Chinnakkaruppan Adaikkan
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Vanesa Fernandez Avalos
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ute Geigenmuller
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Rosalind Mott Firenze
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Edward S. Boyden
- Departments of Biological Engineering and Brain and Cognitive Sciences, McGovern Institute, Cambridge, Massachusetts, United States of America
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
3
|
Singhal G, Baune BT. A bibliometric analysis of studies on environmental enrichment spanning 1967-2024: patterns and trends over the years. Front Behav Neurosci 2024; 18:1501377. [PMID: 39697184 PMCID: PMC11652173 DOI: 10.3389/fnbeh.2024.1501377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
Environmental Enrichment (EE) has received considerable attention for its potential to enhance cognitive and neurobiological outcomes in animal models. This bibliometric analysis offers a comprehensive evaluation of the EE research spanning from 1967 to 2024, utilizing data extracted from Scopus and analyzed through R and VOSviewer. The volume of publications, citation patterns, and collaborations were systematically reviewed, highlighting important contributions and emerging trends within the field of animal research. Core concepts of EE research are mapped, revealing key themes such as neuroplasticity, cognitive function, and behavioral outcomes. A significant increase in EE research is demonstrated, particularly after the year 2000, reflecting growing scientific and public interest in EE paradigms. This analysis provides insights into the global contributions and collaborative networks that have shaped EE studies over time. The role of EE in advancing the understanding of neurobiological, neurodevelopmental, and neurodegenerative processes is underscored. Influential contributors, leading countries, and high-impact journals in the field of EE are identified, offering a valuable resource for researchers seeking to understand or extend the current knowledge base. The strategic selection of keywords and rigorous data curation methods ensure that the findings accurately reflect the most impactful aspects of EE research in animals. This study serves as an essential reference for future explorations and applications of EE across disciplines. By providing a clear and structured overview of the field, this paper aims to serve as a foundation for ongoing and future research initiatives, encouraging more robust investigations and applications of EE to enhance cognitive and neurological health globally.
Collapse
Affiliation(s)
- Gaurav Singhal
- Division of Otolaryngology - Head & Neck Surgery, Department of Surgery, University of Wisconsin, Madison, WI, United States
| | - Bernhard T. Baune
- Department of Mental Health, University of Münster, Münster, Germany
| |
Collapse
|
4
|
Faralli A, Fucà E, Lazzaro G, Menghini D, Vicari S, Costanzo F. Transcranial Direct Current Stimulation in neurogenetic syndromes: new treatment perspectives for Down syndrome? Front Cell Neurosci 2024; 18:1328963. [PMID: 38456063 PMCID: PMC10917937 DOI: 10.3389/fncel.2024.1328963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/25/2024] [Indexed: 03/09/2024] Open
Abstract
This perspective review aims to explore the potential neurobiological mechanisms involved in the application of transcranial Direct Current Stimulation (tDCS) for Down syndrome (DS), the leading cause of genetically-based intellectual disability. The neural mechanisms underlying tDCS interventions in genetic disorders, typically characterized by cognitive deficits, are grounded in the concept of brain plasticity. We initially present the neurobiological and functional effects elicited by tDCS applications in enhancing neuroplasticity and in regulating the excitatory/inhibitory balance, both associated with cognitive improvement in the general population. The review begins with evidence on tDCS applications in five neurogenetic disorders, including Rett, Prader-Willi, Phelan-McDermid, and Neurofibromatosis 1 syndromes, as well as DS. Available evidence supports tDCS as a potential intervention tool and underscores the importance of advancing neurobiological research into the mechanisms of tDCS action in these conditions. We then discuss the potential of tDCS as a promising non-invasive strategy to mitigate deficits in plasticity and promote fine-tuning of the excitatory/inhibitory balance in DS, exploring implications for cognitive treatment perspectives in this population.
Collapse
Affiliation(s)
- Alessio Faralli
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Elisa Fucà
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Giulia Lazzaro
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Deny Menghini
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
- Life Sciences and Public Health Department, Catholic University of Sacred Heart, Rome, Italy
| | - Floriana Costanzo
- Child and Adolescent Neuropsychiatry Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| |
Collapse
|
5
|
Manubens-Gil L, Pons-Espinal M, Gener T, Ballesteros-Yañez I, de Lagrán MM, Dierssen M. Deficits in neuronal architecture but not over-inhibition are main determinants of reduced neuronal network activity in a mouse model of overexpression of Dyrk1A. Cereb Cortex 2024; 34:bhad431. [PMID: 37997361 PMCID: PMC10793573 DOI: 10.1093/cercor/bhad431] [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: 07/04/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/25/2023] Open
Abstract
In this study, we investigated the impact of Dual specificity tyrosine-phosphorylation-regulated kinase 1A (Dyrk1A) overexpression, a gene associated with Down syndrome, on hippocampal neuronal deficits in mice. Our findings revealed that mice overexpressing Dyrk1A (TgDyrk1A; TG) exhibited impaired hippocampal recognition memory, disrupted excitation-inhibition balance, and deficits in long-term potentiation (LTP). Specifically, we observed layer-specific deficits in dendritic arborization of TG CA1 pyramidal neurons in the stratum radiatum. Through computational modeling, we determined that these alterations resulted in reduced storage capacity and compromised integration of inputs, with decreased high γ oscillations. Contrary to prevailing assumptions, our model suggests that deficits in neuronal architecture, rather than over-inhibition, primarily contribute to the reduced network. We explored the potential of environmental enrichment (EE) as a therapeutic intervention and found that it normalized the excitation-inhibition balance, restored LTP, and improved short-term recognition memory. Interestingly, we observed transient significant dendritic remodeling, leading to recovered high γ. However, these effects were not sustained after EE discontinuation. Based on our findings, we conclude that Dyrk1A overexpression-induced layer-specific neuromorphological disturbances impair the encoding of place and temporal context. These findings contribute to our understanding of the underlying mechanisms of Dyrk1A-related hippocampal deficits and highlight the challenges associated with long-term therapeutic interventions for cognitive impairments.
Collapse
Affiliation(s)
- Linus Manubens-Gil
- Institute for Brain Science and Intelligent Technology, Southeast University (SEU), Biomedical engineering, Sipailou street No. 2, Xuanwu district, 210096, Nanjing, China
- School of Biological Science and Medical Engineering, Southeast University (SEU), Sipailou street No. 2, Xuanwu district, 210096, Nanjing, China
| | - Meritxell Pons-Espinal
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Avinguda de la Granvia de l'Hospitalet, 199, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Institute of Biomedicine (IBUB) of the University of Barcelona (UB), Avda. Diagonal, 643 Edifici Prevosti, planta -108028, Barcelona, Spain
| | - Thomas Gener
- Advanced Electronic Materials and Devices Group (AEMD), Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, UAB Campus, Bellaterra Barcelona 08193, Spain
| | - Inmaculada Ballesteros-Yañez
- Inorganic and Organic Chemistry and Biochemistry, Faculty of Medicine, University of Castilla- La Mancha, Camino de Moledores, 13071, Ciudad Real, Spain
| | - María Martínez de Lagrán
- Cellular and Systems Neurobiology, Systems and Synthetic Biology Program, Center for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Mara Dierssen
- Cellular and Systems Neurobiology, Systems and Synthetic Biology Program, Center for Genomic Regulation, Dr. Aiguader 88, 08003 Barcelona, Spain
- Center for Biomedical Research in the Network of Rare Diseases (CIBERER), v. Monforte de Lemos, 3-5. Pabellón 11. Planta 0 28029, Madrid, Spain
- Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003 Barcelona, Spain
| |
Collapse
|
6
|
Hawley LE, Stringer M, Deal AJ, Folz A, Goodlett CR, Roper RJ. Sex-specific developmental alterations in DYRK1A expression in the brain of a Down syndrome mouse model. Neurobiol Dis 2024; 190:106359. [PMID: 37992782 PMCID: PMC10843801 DOI: 10.1016/j.nbd.2023.106359] [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: 09/19/2023] [Revised: 11/02/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
Aberrant neurodevelopment in Down syndrome (DS)-caused by triplication of human chromosome 21-is commonly attributed to gene dosage imbalance, linking overexpression of trisomic genes with disrupted developmental processes, with DYRK1A particularly implicated. We hypothesized that regional brain DYRK1A protein overexpression in trisomic mice varies over development in sex-specific patterns that may be distinct from Dyrk1a transcription, and reduction of Dyrk1a copy number from 3 to 2 in otherwise trisomic mice reduces DYRK1A, independent of other trisomic genes. DYRK1A overexpression varied with age, sex, and brain region, with peak overexpression on postnatal day (P) 6 in both sexes. Sex-dependent differences were also evident from P15-P24. Reducing Dyrk1a copy number confirmed that these differences depended on Dyrk1a gene dosage and not other trisomic genes. Trisomic Dyrk1a mRNA and protein expression were not highly correlated. Sex-specific patterns of DYRK1A overexpression during trisomic neurodevelopment may provide mechanistic targets for therapeutic intervention in DS.
Collapse
Affiliation(s)
- Laura E Hawley
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Megan Stringer
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford Street, LD124, Indianapolis, IN, 46202, USA
| | - Abigail J Deal
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Andrew Folz
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA
| | - Charles R Goodlett
- Department of Psychology, Indiana University - Purdue University Indianapolis, 402 N. Blackford Street, LD124, Indianapolis, IN, 46202, USA
| | - Randall J Roper
- Department of Biology, Indiana University - Purdue University Indianapolis, 723 W. Michigan Street, SL306, Indianapolis, IN, 46202, USA.
| |
Collapse
|
7
|
Schaffner SL, Wassouf Z, Hentrich T, Nuesch-Germano M, Kobor MS, Schulze-Hentrich JM. Distinct impacts of alpha-synuclein overexpression on the hippocampal epigenome of mice in standard and enriched environments. Neurobiol Dis 2023; 186:106274. [PMID: 37648037 DOI: 10.1016/j.nbd.2023.106274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/18/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023] Open
Abstract
Elevated alpha-synuclein (SNCA) gene expression is associated with transcriptional deregulation and increased risk of Parkinson's disease, which may be partially ameliorated by environmental enrichment. At the molecular level, there is emerging evidence that excess alpha-synuclein protein (aSyn) impacts the epigenome through direct and/or indirect mechanisms. However, the extents to which the effects of both aSyn and the environment converge at the epigenome and whether epigenetic alterations underpin the preventive effects of environmental factors on transcription remain to be elucidated. Here, we profiled five DNA and histone modifications in the hippocampus of wild-type and transgenic mice overexpressing human SNCA. Mice of each genotype were housed under either standard conditions or in an enriched environment (EE) for 12 months. SNCA overexpression induced hippocampal CpG hydroxymethylation and histone H3K27 acetylation changes that associated with genotype more than environment. Excess aSyn was also associated with genotype- and environment-dependent changes in non-CpG (CpH) DNA methylation and H3K4 methylation. These H3K4 methylation changes included loci where the EE ameliorated the impacts of the transgene as well as loci resistant to the effects of environmental enrichment in transgenic mice. In addition, select H3K4 monomethylation alterations were associated with changes in mRNA expression. Our results suggested an environment-dependent impact of excess aSyn on some functionally relevant parts of the epigenome, and will ultimately enhance our understanding of the molecular etiology of Parkinson's disease and other synucleinopathies.
Collapse
Affiliation(s)
- Samantha L Schaffner
- Edwin S. H. Leong Centre for Healthy Aging, Faculty of Medicine, 117-2194 Health Sciences Mall, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada; Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, V5Z 4H4 Vancouver, BC, Canada.
| | - Zinah Wassouf
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany.
| | - Thomas Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany
| | | | - Michael S Kobor
- Edwin S. H. Leong Centre for Healthy Aging, Faculty of Medicine, 117-2194 Health Sciences Mall, University of British Columbia, V6T 1Z3 Vancouver, BC, Canada; Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, University of British Columbia, V5Z 4H4 Vancouver, BC, Canada.
| | - Julia M Schulze-Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
8
|
Manubens-Gil L, Pons-Espinal M, Gener T, Ballesteros-Yañez I, de Lagrán MM, Dierssen M. Deficits in neuronal architecture but not over-inhibition are main determinants of reduced neuronal network activity in a mouse model of overexpression of Dyrk1A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531874. [PMID: 36945607 PMCID: PMC10028951 DOI: 10.1101/2023.03.09.531874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Abnormal dendritic arbors, dendritic spine "dysgenesis" and excitation inhibition imbalance are main traits assumed to underlie impaired cognition and behavioral adaptation in intellectual disability. However, how these modifications actually contribute to functional properties of neuronal networks, such as signal integration or storage capacity is unknown. Here, we used a mouse model overexpressing Dyrk1A (Dual-specificity tyrosine [Y]-regulated kinase), one of the most relevant Down syndrome (DS) candidate genes, to gather quantitative data regarding hippocampal neuronal deficits produced by the overexpression of Dyrk1A in mice (TgDyrk1A; TG). TG mice showed impaired hippocampal recognition memory, altered excitation-inhibition balance and deficits in hippocampal CA1 LTP. We also detected for the first time that deficits in dendritic arborization in TG CA1 pyramidal neurons are layer-specific, with a reduction in the width of the stratum radiatum, the postsynaptic target site of CA3 excitatory neurons, but not in the stratum lacunosum-moleculare, which receives temporo-ammonic projections. To interrogate about the functional impact of layer-specific TG dendritic deficits we developed tailored computational multicompartmental models. Computational modelling revealed that neuronal microarchitecture alterations in TG mice lead to deficits in storage capacity, altered the integration of inputs from entorhinal cortex and hippocampal CA3 region onto CA1 pyramidal cells, important for coding place and temporal context and on connectivity and activity dynamics, with impaired the ability to reach high γ oscillations. Contrary to what is assumed in the field, the reduced network activity in TG is mainly contributed by the deficits in neuronal architecture and to a lesser extent by over-inhibition. Finally, given that therapies aimed at improving cognition have also been tested for their capability to recover dendritic spine deficits and excitation-inhibition imbalance, we also tested the short- and long-term changes produced by exposure to environmental enrichment (EE). Exposure to EE normalized the excitation inhibition imbalance and LTP, and had beneficial effects on short-term recognition memory. Importantly, it produced massive but transient dendritic remodeling of hippocampal CA1, that led to recovery of high γ oscillations, the main readout of synchronization of CA1 neurons, in our simulations. However, those effects where not stable and were lost after EE discontinuation. We conclude that layer-specific neuromorphological disturbances produced by Dyrk1A overexpression impair coding place and temporal context. Our results also suggest that treatments targeting structural plasticity, such as EE, even though hold promise towards improved treatment of intellectual disabilities, only produce temporary recovery, due to transient dendritic remodeling.
Collapse
Affiliation(s)
- Linus Manubens-Gil
- SEU-Allen Joint Center, Institute for Brain and Intelligence, Southeast University (SEU), China
| | - Meritxell Pons-Espinal
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Barcelona, Spain
- Institute of Biomedicine (IBUB) of the University of Barcelona (UB), Barcelona, Spain
| | - Thomas Gener
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), Campus UAB, Bellaterra, Barcelona, Spain
| | - Inmaculada Ballesteros-Yañez
- Department of Inorganic and Organic Chemistry and Biochemistry, Faculty of Medicine, University of Castilla-La Mancha, Ciudad Real, Spain (UCLM), CRIB, Spain
| | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), BIST, Spain
- Center for Biomedical Research in the Network of Rare Diseases (CIBERER), Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), BIST, Spain
- Center for Biomedical Research in the Network of Rare Diseases (CIBERER), Spain
| |
Collapse
|
9
|
Scott-McKean JJ, Jones R, Johnson MW, Mier J, Basten IA, Stasko MR, Costa ACS. Emergence of Treadmill Running Ability and Quantitative Assessment of Gait Dynamics in Young Ts65Dn Mice: A Mouse Model for Down Syndrome. Brain Sci 2023; 13:brainsci13050743. [PMID: 37239215 DOI: 10.3390/brainsci13050743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/18/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Down syndrome (DS), which results from the complete or partial trisomy of chromosome 21 (trisomy-21), is the most common genetically defined cause of intellectual disability. Trisomy-21 also produces, or is associated with, many neurodevelopmental phenotypes and neurological comorbidities, including delays and deficits in fine and gross motor development. The Ts65Dn mouse is the most studied animal model for DS and displays the largest known subset of DS-like phenotypes. To date, however, only a small number of developmental phenotypes have been quantitatively defined in these animals. Here, we used a commercially available high-speed, video-based system to record and analyze the gait of Ts65Dn and euploid control mice. Longitudinal treadmill recordings were performed from p17 to p35. One of the main findings was the detection of genotype- and sex-dependent developmental delays in the emergence of consistent, progressive-intensity gait in Ts65Dn mice when compared to control mice. Gait dynamic analysis showed wider normalized front and hind stances in Ts65Dn mice compared to control mice, which may reflect deficits in dynamic postural balance. Ts65Dn mice also displayed statistically significant differences in the variability in several normalized gait measures, which were indicative of deficits in precise motor control in generating gait.
Collapse
Affiliation(s)
- Jonah J Scott-McKean
- Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| | - Ryan Jones
- College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43606-3390, USA
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| | - Mark W Johnson
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| | - Joyce Mier
- Physical Therapy Program, University of Wisconsin, Madison, WI 53706-1532, USA
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| | - Ines A Basten
- Psychiatric Hospital Asster, 3800 Sint-Truiden, Belgium
| | - Melissa R Stasko
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| | - Alberto C S Costa
- Department of Macromolecular Science and Engineering, Case Western Reserve University, Cleveland, OH 44106-6090, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106-6090, USA
| |
Collapse
|
10
|
Bibollet-Bahena O, Tissier S, Ho-Tran S, Rojewski A, Casanova C. Enriched environment exposure during development positively impacts the structure and function of the visual cortex in mice. Sci Rep 2023; 13:7020. [PMID: 37120630 PMCID: PMC10148800 DOI: 10.1038/s41598-023-33951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023] Open
Abstract
Optimal conditions of development have been of interest for decades, since genetics alone cannot fully explain how an individual matures. In the present study, we used optical brain imaging to investigate whether a relatively simple enrichment can positively influence the development of the visual cortex of mice. The enrichment paradigm was composed of larger cages housing multiple mice that contained several toys, hiding places, nesting material and a spinning wheel that were moved or replaced at regular intervals. We compared C57BL/6N adult mice (> P60) that had been raised either in an enriched environment (EE; n = 16) or a standard (ST; n = 12) environment from 1 week before birth to adulthood, encompassing all cortical developmental stages. Here, we report significant beneficial changes on the structure and function of the visual cortex following environmental enrichment throughout the lifespan. More specifically, retinotopic mapping through intrinsic signal optical imaging revealed that the size of the primary visual cortex was greater in mice reared in an EE compared to controls. In addition, the visual field coverage of EE mice was wider. Finally, the organization of the cortical representation of the visual field (as determined by cortical magnification) versus its eccentricity also differed between the two groups. We did not observe any significant differences between females and males within each group. Taken together, these data demonstrate specific benefits of an EE throughout development on the visual cortex, which suggests adaptation to their environmental realities.
Collapse
Affiliation(s)
- O Bibollet-Bahena
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada.
| | - S Tissier
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - S Ho-Tran
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - A Rojewski
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| | - C Casanova
- Laboratoire des Neurosciences de la Vision, School of Optometry, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
11
|
Kapusta J, Siewierska D, Kruczek M, Pochron E, Olejniczak P. Species specific differences in short-term behavioral reaction of voles to cage elements removal. Appl Anim Behav Sci 2023. [DOI: 10.1016/j.applanim.2023.105899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
12
|
Duchon A, del Mar Muñiz Moreno M, Chevalier C, Nalesso V, Andre P, Fructuoso-Castellar M, Mondino M, Po C, Noblet V, Birling MC, Potier MC, Herault Y. Ts66Yah, a mouse model of Down syndrome with improved construct and face validity. Dis Model Mech 2022; 15:282398. [PMID: 36374158 PMCID: PMC9789398 DOI: 10.1242/dmm.049721] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Down syndrome (DS) is caused by trisomy of human chromosome 21 (Hsa21). The understanding of genotype-phenotype relationships, the identification of driver genes and various proofs of concept for therapeutics have benefited from mouse models. The premier model, named Ts(1716)65Dn/J (Ts65Dn), displayed phenotypes related to human DS features. It carries an additional minichromosome with the Mir155 to Zbtb21 region of mouse chromosome 16, homologous to Hsa21, encompassing around 90 genes, fused to the centromeric part of mouse chromosome 17 from Pisd-ps2/Scaf8 to Pde10a, containing 46 genes not related to Hsa21. Here, we report the investigation of a new model, Ts66Yah, generated by CRISPR/Cas9 without the genomic region unrelated to Hsa21 on the minichromosome. As expected, Ts66Yah replicated DS cognitive features. However, certain phenotypes related to increased activity, spatial learning and molecular signatures were changed, suggesting genetic interactions between the Mir155-Zbtb21 and Scaf8-Pde10a intervals. Thus, Ts66Yah mice have stronger construct and face validity than Ts65Dn mice for mimicking consequences of DS genetic overdosage. Furthermore, this study is the first to demonstrate genetic interactions between triplicated regions homologous to Hsa21 and others unrelated to Hsa21. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Arnaud Duchon
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Maria del Mar Muñiz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Philippe Andre
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris (ICS), 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Marta Fructuoso-Castellar
- Paris Brain Institute ICM, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Centre National de la Recherche Scientifique, UMR 7225, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Mary Mondino
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Chrystelle Po
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Vincent Noblet
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Marie-Christine Birling
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris (ICS), 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France
| | - Marie-Claude Potier
- Paris Brain Institute ICM, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Institut National de la Santé et de la Recherche Médicale, U1127, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Centre National de la Recherche Scientifique, UMR 7225, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France,Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France,Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN-Institut Clinique de la Souris (ICS), 1 rue Laurent Fries, 67404 Illkirch-Graffenstaden, France,Author for correspondence ()
| |
Collapse
|
13
|
Fulton SL, Wenderski W, Lepack AE, Eagle AL, Fanutza T, Bastle RM, Ramakrishnan A, Hays EC, Neal A, Bendl J, Farrelly LA, Al-Kachak A, Lyu Y, Cetin B, Chan JC, Tran TN, Neve RL, Roper RJ, Brennand KJ, Roussos P, Schimenti JC, Friedman AK, Shen L, Blitzer RD, Robison AJ, Crabtree GR, Maze I. Rescue of deficits by Brwd1 copy number restoration in the Ts65Dn mouse model of Down syndrome. Nat Commun 2022; 13:6384. [PMID: 36289231 PMCID: PMC9606253 DOI: 10.1038/s41467-022-34200-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/17/2022] [Indexed: 12/25/2022] Open
Abstract
With an incidence of ~1 in 800 births, Down syndrome (DS) is the most common chromosomal condition linked to intellectual disability worldwide. While the genetic basis of DS has been identified as a triplication of chromosome 21 (HSA21), the genes encoded from HSA21 that directly contribute to cognitive deficits remain incompletely understood. Here, we found that the HSA21-encoded chromatin effector, BRWD1, was upregulated in neurons derived from iPS cells from an individual with Down syndrome and brain of trisomic mice. We showed that selective copy number restoration of Brwd1 in trisomic animals rescued deficits in hippocampal LTP, cognition and gene expression. We demonstrated that Brwd1 tightly binds the BAF chromatin remodeling complex, and that increased Brwd1 expression promotes BAF genomic mistargeting. Importantly, Brwd1 renormalization rescued aberrant BAF localization, along with associated changes in chromatin accessibility and gene expression. These findings establish BRWD1 as a key epigenomic mediator of normal neurodevelopment and an important contributor to DS-related phenotypes.
Collapse
Affiliation(s)
- Sasha L Fulton
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Wendy Wenderski
- Department of Pathology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Genetics, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Ashley E Lepack
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Tomas Fanutza
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ryan M Bastle
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emma C Hays
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arianna Neal
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jaroslav Bendl
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Disease Neuroepigenomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lorna A Farrelly
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Amni Al-Kachak
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yang Lyu
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bulent Cetin
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jennifer C Chan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tina N Tran
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Rachael L Neve
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Randall J Roper
- Department of Biology, Indiana University-Purdue University, Indianapolis, IN, 46202, USA
| | - Kristen J Brennand
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Departments of Psychiatry and Genetics, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, 065109, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Disease Neuroepigenomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- J.J. Peters Veterans Affairs Hospital, Bronx, NY, 10468, USA
| | - John C Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, 14853, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Allyson K Friedman
- Department of Biological Sciences, City University of New York-Hunter College, New York, NY, 10065, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Gerald R Crabtree
- Department of Pathology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Genetics, Stanford Medical School, Palo Alto, CA, 94305, USA
- Department of Developmental Biology, Stanford Medical School, Palo Alto, CA, 94305, USA
- Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
14
|
Alemany-González M, Vilademunt M, Gener T, Puig MV. Postnatal environmental enrichment enhances memory through distinct neural mechanisms in healthy and trisomic female mice. Neurobiol Dis 2022; 173:105841. [PMID: 35988873 DOI: 10.1016/j.nbd.2022.105841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022] Open
Abstract
Stimulating lifestyles have powerful effects on cognitive abilities, especially when they are experienced early in life. Cognitive therapies are widely used to improve cognitive impairment due to intellectual disability, aging, and neurodegeneration, however the underlying neural mechanisms are poorly understood. We investigated the neural correlates of memory amelioration produced by postnatal environmental enrichment (EE) in diploid mice and the Ts65Dn mouse model of Down syndrome (trisomy 21). We recorded neural activities in brain structures key for memory processing, the hippocampus and the prefrontal cortex, during rest, sleep and memory performance in mice reared in non-enriched or enriched environments. Enriched wild-type animals exhibited enhanced neural synchrony in the hippocampus across different brain states (increased gamma oscillations, theta-gamma coupling, sleep ripples). Trisomic females showed increased theta and gamma rhythms in the hippocampus and prefrontal cortex across different brain states along with enlarged ripples and disrupted circuit gamma signals that were associated with memory deficits. These pathological activities were attenuated in their trisomic EE-reared peers. Our results suggest distinct neural mechanisms for the generation and rescue of healthy and pathological brain synchrony, respectively, by EE and put forward hippocampal-prefrontal hypersynchrony and miscommunication as major targets underlying the beneficial effects of EE in intellectual disability.
Collapse
Affiliation(s)
- Maria Alemany-González
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Marta Vilademunt
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Thomas Gener
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Catalan Institute of Nanoscience and Nanotechnology (ICN2), the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - M Victoria Puig
- Integrative Pharmacology and Systems Neuroscience, Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Catalan Institute of Nanoscience and Nanotechnology (ICN2), the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193 Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
15
|
Andrews EJ, Martini AC, Head E. Exploring the role of sex differences in Alzheimer's disease pathogenesis in Down syndrome. Front Neurosci 2022; 16:954999. [PMID: 36033603 PMCID: PMC9411995 DOI: 10.3389/fnins.2022.954999] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/25/2022] [Indexed: 11/14/2022] Open
Abstract
Women are disproportionately affected by Alzheimer's disease (AD), yet little is known about sex-specific effects on the development of AD in the Down syndrome (DS) population. DS is caused by a full or partial triplication of chromosome 21, which harbors the amyloid precursor protein (APP) gene, among others. The majority of people with DS in their early- to mid-40s will accumulate sufficient amyloid-beta (Aβ) in their brains along with neurofibrillary tangles (NFT) for a neuropathological diagnosis of AD, and the triplication of the APP gene is regarded as the main cause. Studies addressing sex differences with age and impact on dementia in people with DS are inconsistent. However, women with DS experience earlier age of onset of menopause, marked by a drop in estrogen, than women without DS. This review focuses on key sex differences observed with age and AD in people with DS and a discussion of possible underlying mechanisms that could be driving or protecting from AD development in DS. Understanding how biological sex influences the brain will lead to development of dedicated therapeutics and interventions to improve the quality of life for people with DS and AD.
Collapse
Affiliation(s)
- Elizabeth J. Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
16
|
Hawley LE, Prochaska F, Stringer M, Goodlett CR, Roper RJ. Sexually dimorphic DYRK1A overexpression on postnatal day 15 in the Ts65Dn mouse model of Down syndrome: Effects of pharmacological targeting on behavioral phenotypes. Pharmacol Biochem Behav 2022; 217:173404. [DOI: 10.1016/j.pbb.2022.173404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
17
|
Lalonde R, Strazielle C. The Hole-Board Test in Mutant Mice. Behav Genet 2022; 52:158-169. [PMID: 35482162 DOI: 10.1007/s10519-022-10102-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/01/2022] [Indexed: 11/02/2022]
Abstract
First described by Boissier and Simon in (Ther Recreat J 17:1225-1232, 1962), the hole-board has become a recognized test of anxiety and spatial memory. Benzodiazepines acting at the GABAA-BZD site increase hole-pokes in rats and mice, indicating a loss in behavioral inhibition concordant with the behavior of mutant mice deficient in the GABA transporter. Hole-poking also depends on arousal mechanisms dependent on dopaminergic transmission, as indicated by drug and null mutant studies. In addition, the behavior is modified in natural and null mutants affecting the cerebellum as well as null mutants affecting neuropeptides, growth factors, cell adhesion, and inflammation. Further research is required to determine convergences between genetic and pharmacological effects.
Collapse
Affiliation(s)
- Robert Lalonde
- Laboratory of Stress, Immunity, Pathogens (EA7300), Medical School, University of Lorraine, 54500, Vandœuvre-les-Nancy, France
| | - Catherine Strazielle
- Laboratory of Stress, Immunity, Pathogens (EA7300), Medical School, University of Lorraine, 54500, Vandœuvre-les-Nancy, France. .,CHRU Nancy, Vandœuvre-les-Nancy, France.
| |
Collapse
|
18
|
Di Franco N, Drutel G, Roullot-Lacarrière V, Julio-Kalajzic F, Lalanne V, Grel A, Leste-Lasserre T, Matias I, Cannich A, Gonzales D, Simon V, Cota D, Marsicano G, Piazza PV, Vallée M, Revest JM. Differential expression of the neuronal CB1 cannabinoid receptor in the hippocampus of male Ts65Dn Down syndrome mouse model. Mol Cell Neurosci 2022; 119:103705. [PMID: 35158060 DOI: 10.1016/j.mcn.2022.103705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/27/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022] Open
Abstract
Down syndrome (DS) or Trisomy 21 is the most common genetic cause of mental retardation with severe learning and memory deficits. DS is due to the complete or partial triplication of human chromosome 21 (HSA21) triggering gene overexpression and protein synthesis alterations responsible for a plethora of mental and physical phenotypes. Among the diverse brain target systems that affect hippocampal-dependent learning and memory deficit impairments in DS, the upregulation of the endocannabinoid system (ECS), and notably the overexpression of the cannabinoid type-1 receptor (CB1), seems to play a major role. Combining various protein and gene expression targeted approaches using western blot, qRT-PCR and FISH techniques, we investigated the expression pattern of ECS components in the hippocampus (HPC) of male Ts65Dn mice. Among all the molecules that constitute the ECS, we found that the expression of the CB1 is altered in the HPC of Ts65Dn mice. CB1 distribution is differentially segregated between the dorsal and ventral part of the HPC and within the different cell populations that compose the HPC. CB1 expression is upregulated in GABAergic neurons of Ts65Dn mice whereas it is downregulated in glutamatergic neurons. These results highlight a complex regulation of the CB1 encoding gene (Cnr1) in Ts65Dn mice that could open new therapeutic solutions for this syndrome.
Collapse
Affiliation(s)
- Nadia Di Franco
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Guillaume Drutel
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | | | | | - Valérie Lalanne
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Agnès Grel
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | | | - Isabelle Matias
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Astrid Cannich
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Delphine Gonzales
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Vincent Simon
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Daniela Cota
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Giovanni Marsicano
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | | | - Monique Vallée
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Jean-Michel Revest
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000 Bordeaux, France.
| |
Collapse
|
19
|
Cosgrove JA, Kelly LK, Kiffmeyer EA, Kloth AD. Sex-dependent influence of postweaning environmental enrichment in Angelman syndrome model mice. Brain Behav 2022; 12:e2468. [PMID: 34985196 PMCID: PMC8865162 DOI: 10.1002/brb3.2468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/09/2021] [Accepted: 12/12/2021] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Angelman syndrome (AS) is a rare neurodevelopmental disorder caused by mutation or loss of UBE3A and marked by intellectual disability, ataxia, autism-like symptoms, and other atypical behaviors. One route to treatment may lie in the role that environment plays early in postnatal life. Environmental enrichment (EE) is one manipulation that has shown therapeutic potential in preclinical models of many brain disorders, including neurodevelopmental disorders. Here, we examined whether postweaning EE can rescue behavioral phenotypes in Ube3a maternal deletion mice (AS mice), and whether any improvements are sex-dependent. METHODS Male and female mice (C57BL/6J Ube3atm1Alb mice and wild-type (WT) littermates; ≥10 mice/group) were randomly assigned to standard housing (SH) or EE at weaning. EE had a larger footprint, a running wheel, and a variety of toys that promoted foraging, burrowing, and climbing. Following 6 weeks of EE, animals were submitted to a battery of tests that reliably elicit behavioral deficits in AS mice, including rotarod, open field, marble burying, and forced swim; weights were also monitored. RESULTS In male AS-EE mice, we found complete restoration of motor coordination, marble burying, and forced swim behavior to the level of WT-SH mice. We also observed a complete normalization of exploratory distance traveled in the open field, but we found no rescue of vertical behavior or center time. AS-EE mice also had weights comparable to WT-SH mice. Intriguingly, in the female AS-EE mice, we found a failure of EE to rescue the same behavioral deficits relative to female WT-SH mice. CONCLUSIONS Environmental enrichment is an effective route to correcting the most penetrant phenotypes in male AS mice but not female AS mice. This finding has important implications for the translatability of early behavioral intervention for AS patients, most importantly the potential dependency of treatment response on sex.
Collapse
Affiliation(s)
- Jameson A. Cosgrove
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Lauren K. Kelly
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Elizabeth A. Kiffmeyer
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| | - Alexander D. Kloth
- Department of BiologyAugustana University2001 S. Summit AvenueSioux FallsSouth DakotaUSA
| |
Collapse
|
20
|
Sukegawa M, Yoshihara T, Hou S, Asano M, Hannan AJ, Wang DO. Long‐lasting Housing Environment Manipulation and Acute Loss of Environmental Enrichment Impact BALB/c Mice Behavior in Multiple Functional Domains. Eur J Neurosci 2022; 55:1118-1140. [PMID: 35060219 PMCID: PMC9306724 DOI: 10.1111/ejn.15602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/28/2021] [Accepted: 01/06/2022] [Indexed: 12/01/2022]
Abstract
Understanding environmental influences on individuals' behaviour is challenging. Here we have investigated the housing impact of 9 weeks of enriched environment (EE) and social isolation (SI) and the impact of abrupt deprivation of EE (enrichment removal: ER) on BALB/c mice. Compared with the widely used C57BL/6 strain in research, BALB/c synthesises serotonin less efficiently due to a genetic variation and thus may potentially represent human populations at higher risk of stress‐related disorders. We assessed the effects of EE and SI by conducting a behavioural test battery and the effects of acute ER by monitoring homecage activities and social behaviour. We found that EE and SI impact BALB/c's physiological states and behavioural performances from lower to higher cognitive processes: increased body weight, increased rectal temperature, altered performance in motor and sensory tasks, the activity level in a novel environment and altered performance in tests of anxiety‐like behaviour, stress‐coping strategies and learning and memory. Furthermore, acute ER triggered stress/frustration‐like behaviour in BALB/c, with increased aggression, increased social distancing and disrupted daily/nightly activities. Our results demonstrate that long‐lasting housing manipulation such as EE and SI, impact behaviour via multilayered processes over a wide range of functional domains, and unforeseen change to a negative environment, ER, is a major stressor that causes behavioural and psychological consequences through environment–gene interactions, a model of direct relevance to human health.
Collapse
Affiliation(s)
- Momoe Sukegawa
- Center for Biosystems Dynamics Research (BDR), RIKEN Japan
- Graduate School of Biostudies Kyoto University Japan
- Institute for Integrated Cell‐Material Sciences (iCeMS) Kyoto University Japan
| | - Toru Yoshihara
- Institute of Laboratory Animals, Graduate School of Medicine Kyoto University Japan
| | - Shengqun Hou
- Center for Biosystems Dynamics Research (BDR), RIKEN Japan
- Graduate School of Biostudies Kyoto University Japan
- Institute for Integrated Cell‐Material Sciences (iCeMS) Kyoto University Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine Kyoto University Japan
| | - Anthony J. Hannan
- Department of Anatomy and Neuroscience University of Melbourne Australia
- Melbourne Brain Centre Australia
| | - Dan Ohtan Wang
- Center for Biosystems Dynamics Research (BDR), RIKEN Japan
- Graduate School of Biostudies Kyoto University Japan
- Institute for Integrated Cell‐Material Sciences (iCeMS) Kyoto University Japan
| |
Collapse
|
21
|
Sierra C, De Toma I, Cascio LL, Vegas E, Dierssen M. Social Factors Influence Behavior in the Novel Object Recognition Task in a Mouse Model of Down Syndrome. Front Behav Neurosci 2021; 15:772734. [PMID: 34803627 PMCID: PMC8602686 DOI: 10.3389/fnbeh.2021.772734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/05/2021] [Indexed: 11/24/2022] Open
Abstract
The use of mouse models has revolutionized the field of Down syndrome (DS), increasing our knowledge about neuropathology and helping to propose new therapies for cognitive impairment. However, concerns about the reproducibility of results in mice and their translatability to humans have become a major issue, and controlling for moderators of behavior is essential. Social and environmental factors, the experience of the researcher, and the sex and strain of the animals can all have effects on behavior, and their impact on DS mouse models has not been explored. Here we analyzed the influence of a number of social and environmental factors, usually not taken into consideration, on the behavior of male and female wild-type and trisomic mice (the Ts65Dn model) in one of the most used tests for proving drug effects on memory, the novel object recognition (NOR) test. Using principal component analysis and correlation matrices, we show that the ratio of trisomic mice in the cage, the experience of the experimenter, and the timing of the test have a differential impact on male and female and on wild-type and trisomic behavior. We conclude that although the NOR test is quite robust and less susceptible to environmental influences than expected, to obtain useful results, the phenotype expression must be contrasted against the influences of social and environmental factors.
Collapse
Affiliation(s)
- Cesar Sierra
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Ilario De Toma
- Neurosciences Research Program, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Lorenzo Lo Cascio
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Esteban Vegas
- Department of Genetics, Microbiology and Statistics, Section of Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Mara Dierssen
- Center for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, Spain
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
- Biomedical Research Networking Center for Rare Diseases (CIBERER), Barcelona, Spain
| |
Collapse
|
22
|
Espeso-Gil S, Holik AZ, Bonnin S, Jhanwar S, Chandrasekaran S, Pique-Regi R, Albaigès-Ràfols J, Maher M, Permanyer J, Irimia M, Friedländer MR, Pons-Espinal M, Akbarian S, Dierssen M, Maass PG, Hor CN, Ossowski S. Environmental Enrichment Induces Epigenomic and Genome Organization Changes Relevant for Cognition. Front Mol Neurosci 2021; 14:664912. [PMID: 34025350 PMCID: PMC8131874 DOI: 10.3389/fnmol.2021.664912] [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: 02/06/2021] [Accepted: 04/09/2021] [Indexed: 01/11/2023] Open
Abstract
In early development, the environment triggers mnemonic epigenomic programs resulting in memory and learning experiences to confer cognitive phenotypes into adulthood. To uncover how environmental stimulation impacts the epigenome and genome organization, we used the paradigm of environmental enrichment (EE) in young mice constantly receiving novel stimulation. We profiled epigenome and chromatin architecture in whole cortex and sorted neurons by deep-sequencing techniques. Specifically, we studied chromatin accessibility, gene and protein regulation, and 3D genome conformation, combined with predicted enhancer and chromatin interactions. We identified increased chromatin accessibility, transcription factor binding including CTCF-mediated insulation, differential occupancy of H3K36me3 and H3K79me2, and changes in transcriptional programs required for neuronal development. EE stimuli led to local genome re-organization by inducing increased contacts between chromosomes 7 and 17 (inter-chromosomal). Our findings support the notion that EE-induced learning and memory processes are directly associated with the epigenome and genome organization.
Collapse
Affiliation(s)
- Sergio Espeso-Gil
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada
| | - Aliaksei Z. Holik
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sarah Bonnin
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Shalu Jhanwar
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sandhya Chandrasekaran
- MD/PhD Program in the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Júlia Albaigès-Ràfols
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Michael Maher
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jon Permanyer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain
| | - Marc R. Friedländer
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Meritxell Pons-Espinal
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Schahram Akbarian
- Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Philipp G. Maass
- Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Charlotte N. Hor
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Stephan Ossowski
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
23
|
Duchon A, Del Mar Muniz Moreno M, Martin Lorenzo S, Silva de Souza MP, Chevalier C, Nalesso V, Meziane H, Loureiro de Sousa P, Noblet V, Armspach JP, Brault V, Herault Y. Multi-influential genetic interactions alter behaviour and cognition through six main biological cascades in Down syndrome mouse models. Hum Mol Genet 2021; 30:771-788. [PMID: 33693642 PMCID: PMC8161522 DOI: 10.1093/hmg/ddab012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Down syndrome (DS) is the most common genetic form of intellectual disability caused by the presence of an additional copy of human chromosome 21 (Hsa21). To provide novel insights into genotype–phenotype correlations, we used standardized behavioural tests, magnetic resonance imaging and hippocampal gene expression to screen several DS mouse models for the mouse chromosome 16 region homologous to Hsa21. First, we unravelled several genetic interactions between different regions of chromosome 16 and how they contribute significantly to altering the outcome of the phenotypes in brain cognition, function and structure. Then, in-depth analysis of misregulated expressed genes involved in synaptic dysfunction highlighted six biological cascades centred around DYRK1A, GSK3β, NPY, SNARE, RHOA and NPAS4. Finally, we provide a novel vision of the existing altered gene–gene crosstalk and molecular mechanisms targeting specific hubs in DS models that should become central to better understanding of DS and improving the development of therapies.
Collapse
Affiliation(s)
- Arnaud Duchon
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Maria Del Mar Muniz Moreno
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Sandra Martin Lorenzo
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Marcia Priscilla Silva de Souza
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Claire Chevalier
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Valérie Nalesso
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | | | - Vincent Noblet
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Jean-Paul Armspach
- Université de Strasbourg, CNRS UMR 7357, ICube, FMTS, 67000 Strasbourg, France
| | - Veronique Brault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), department of translational medicine and neurogenetics 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France.,Université de Strasbourg, CNRS, INSERM, Institut Clinique de la Souris (ICS), CELPHEDIA, PHENOMIN, 1 rue Laurent Fries, 67404 Illkirch Graffenstaden, France
| |
Collapse
|
24
|
De Toma I, Ortega M, Catuara-Solarz S, Sierra C, Sabidó E, Dierssen M. Re-establishment of the epigenetic state and rescue of kinome deregulation in Ts65Dn mice upon treatment with green tea extract and environmental enrichment. Sci Rep 2020; 10:16023. [PMID: 32994493 PMCID: PMC7524756 DOI: 10.1038/s41598-020-72625-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Down syndrome (DS) is the main genetic cause of intellectual disability due to triplication of human chromosome 21 (HSA21). Although there is no treatment for intellectual disability, environmental enrichment (EE) and the administration of green tea extracts containing epigallocatechin-3-gallate (EGCG) improve cognition in mouse models and individuals with DS. Using proteome, and phosphoproteome analysis in the hippocampi of a DS mouse model (Ts65Dn), we investigated the possible mechanisms underlying the effects of green tea extracts, EE and their combination. Our results revealed disturbances in cognitive-related (synaptic proteins, neuronal projection, neuron development, microtubule), GTPase/kinase activity and chromatin proteins. Green tea extracts, EE, and their combination restored more than 70% of the phosphoprotein deregulation in Ts65Dn, and induced possible compensatory effects. Our downstream analyses indicate that re-establishment of a proper epigenetic state and rescue of the kinome deregulation may contribute to the cognitive rescue induced by green tea extracts.
Collapse
Affiliation(s)
- I De Toma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - M Ortega
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - S Catuara-Solarz
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - C Sierra
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - E Sabidó
- Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003, Barcelona, Spain.,Proteomics Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - M Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003, Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Dr. Aiguader 88, 08003, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.
| |
Collapse
|
25
|
Shaw PR, Klein JA, Aziz NM, Haydar TF. Longitudinal neuroanatomical and behavioral analyses show phenotypic drift and variability in the Ts65Dn mouse model of Down syndrome. Dis Model Mech 2020; 13:dmm046243. [PMID: 32817053 PMCID: PMC7522024 DOI: 10.1242/dmm.046243] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022] Open
Abstract
Mouse models of Down syndrome (DS) have been invaluable tools for advancing knowledge of the underlying mechanisms of intellectual disability in people with DS. The Ts(1716)65Dn (Ts65Dn) mouse is one of the most commonly used models as it recapitulates many of the phenotypes seen in individuals with DS, including neuroanatomical changes and impaired learning and memory. In this study, we use rigorous metrics to evaluate multiple cohorts of Ts65Dn ranging from 2014 to the present, including a stock of animals recovered from embryos frozen within ten generations after the colony was first created in 2010. Through quantification of prenatal and postnatal brain development and several behavioral tasks, our results provide a comprehensive comparison of Ts65Dn across time and show a significant amount of variability both across cohorts as well as within cohorts. The inconsistent phenotypes in Ts65Dn mice highlight specific cautions and caveats for use of this model. We outline important steps for ensuring responsible use of Ts65Dn in future research.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Patricia R Shaw
- Graduate Program in Neuroscience, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jenny A Klein
- Graduate Program in Neuroscience, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nadine M Aziz
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Tarik F Haydar
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| |
Collapse
|
26
|
Rueda N, Vidal V, García-Cerro S, Puente A, Campa V, Lantigua S, Narcís O, Bartesaghi R, Martínez-Cué C. Prenatal, but not Postnatal, Curcumin Administration Rescues Neuromorphological and Cognitive Alterations in Ts65Dn Down Syndrome Mice. J Nutr 2020; 150:2478-2489. [PMID: 32729926 DOI: 10.1093/jn/nxaa207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/27/2020] [Accepted: 06/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The cognitive dysfunction in Down syndrome (DS) is partially caused by deficient neurogenesis during fetal stages. Curcumin enhances neurogenesis and learning and memory. OBJECTIVES We aimed to test the ability of curcumin to rescue the neuromorphological and cognitive alterations of the Ts65Dn (TS) mouse model of DS when administered prenatally or during early postnatal stages, and to evaluate whether these effects were maintained several weeks after the treatment. METHODS To evaluate the effects of prenatal curcumin administration, 65 pregnant TS females were subcutaneously treated with curcumin (300 mg/kg) or vehicle from ED (Embryonic Day) 10 to PD (Postnatal Day) 2. All the analyses were performed on their TS and Control (CO) male and female progeny. At PD2, the changes in neurogenesis, cellularity, and brain weight were analyzed in 30 TS and CO pups. The long-term effects of prenatal curcumin were evaluated in another cohort of 44 TS and CO mice between PD30 and PD45. The neuromorphological effects of the early postnatal administration of curcumin were assessed on PD15 in 30 male and female TS and CO pups treated with curcumin (300 mg/kg) or vehicle from PD2 to PD15. The long-term neuromorphological and cognitive effects were assessed from PD60 to PD90 in 45 mice. Data was compared by ANOVAs. RESULTS Prenatal administration of curcumin increased the brain weight (+45%, P < 0.001), the density of BrdU (bromodeoxyuridine)- (+150%, P < 0.001) and DAPI (4',6-diamidino-2-phenylindole)- (+38%, P = 0.005) positive cells, and produced a long-term improvement of cognition in TS (+35%, P = 0.007) mice with respect to vehicle-treated mice. Postnatal administration of curcumin did not rescue any of the short- or long-term altered phenotypes of TS mice. CONCLUSION The beneficial effects of prenatal curcumin administration to TS mice suggest that it could be a therapeutic strategy to treat DS cognitive disabilities.
Collapse
Affiliation(s)
- Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Alba Puente
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Víctor Campa
- Institute of Molecular Biology and Biomedicine, Santander, Cantabria, Spain
| | - Sara Lantigua
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Oriol Narcís
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
27
|
Roper RJ, Goodlett CR, Martínez de Lagrán M, Dierssen M. Behavioral Phenotyping for Down Syndrome in Mice. ACTA ACUST UNITED AC 2020; 10:e79. [PMID: 32780566 DOI: 10.1002/cpmo.79] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability, characterized by alterations in different behavioral symptom domains: neurodevelopment, motor behavior, and cognition. As mouse models have the potential to generate data regarding the neurological basis for the specific behavioral profile of DS, and may indicate pharmacological treatments with the potential to affect their behavioral phenotype, it is important to be able to assess disease-relevant behavioral traits in animal models in order to provide biological plausibility to the potential findings. The field is at a juncture that requires assessments that may effectively translate the findings acquired in mouse models to humans with DS. In this article, behavioral tests are described that are relevant to the domains affected in DS. A neurodevelopmental behavioral screen, the balance beam test, and the Multivariate Concentric Square Field test to assess multiple behavioral phenotypes and locomotion are described, discussing the ways to merge these findings to more fully understand cognitive strengths and weaknesses in this population. New directions for approaches to cognitive assessment in mice and humans are discussed. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Preweaning neurodevelopmental battery Basic Protocol 2: Balance beam Basic Protocol 3: Multivariate concentric square field test (MCSF).
Collapse
Affiliation(s)
| | | | - María Martínez de Lagrán
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Phenotype microarrays reveal metabolic dysregulations of neurospheres derived from embryonic Ts1Cje mouse model of Down syndrome. PLoS One 2020; 15:e0236826. [PMID: 32730314 PMCID: PMC7392322 DOI: 10.1371/journal.pone.0236826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/14/2020] [Indexed: 11/20/2022] Open
Abstract
Down syndrome (DS), is the most common cause of intellectual disability, and is characterized by defective neurogenesis during perinatal development. To identify metabolic aberrations in early neurogenesis, we profiled neurospheres derived from the embryonic brain of Ts1Cje, a mouse model of Down syndrome. High-throughput phenotypic microarray revealed a significant decrease in utilisation of 17 out of 367 substrates and significantly higher utilisation of 6 substrates in the Ts1Cje neurospheres compared to controls. Specifically, Ts1Cje neurospheres were less efficient in the utilisation of glucose-6-phosphate suggesting a dysregulation in the energy-producing pathway. T Cje neurospheres were significantly smaller in diameter than the controls. Subsequent preliminary study on supplementation with 6-phosphogluconic acid, an intermediate of glucose-6-phosphate metabolism, was able to rescue the Ts1Cje neurosphere size. This study confirmed the perturbed pentose phosphate pathway, contributing to defects observed in Ts1Cje neurospheres. We show for the first time that this comprehensive energetic assay platform facilitates the metabolic characterisation of Ts1Cje cells and confirmed their distinguishable metabolic profiles compared to the controls.
Collapse
|
29
|
Zheng JJ, Zou R, Huang S, Song TJ, Yu X. Enriched Environment Rearing from Birth Reduced Anxiety, Improved Learning and Memory, and Promoted Social Interactions in Adult Male Mice. Neuroscience 2020; 442:138-150. [PMID: 32652178 DOI: 10.1016/j.neuroscience.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022]
Abstract
Rearing rodents in an enriched environment (EE), with increased sensory stimulations and social interactions, is a well-established model for naturally increasing neural activity. It is well-known that EE-rearing of rodents from adolescence or during adulthood leads to extensive biochemical, morphological, electrophysiological and behavioral changes. Here, we examine the effects of EE-rearing from birth on adult behavior. Through a battery of assays, we found that mice EE-reared from birth had better acquisition and consolidation of memory, in both aversive-based fear conditioning and reward-based contextual association tasks. Moreover, EE-reared mice showed reduced anxiety in novel environments and enhanced social interactions. Together, these results demonstrated that EE-rearing from birth significantly improved motor ability, learning and memory and sociability, while reducing anxiety. A better understanding of how early environmental influences affect behavior is not only important for understanding neural circuit wiring, but also provides insight into developing more effective intervention programs for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jing-Jing Zheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rong Zou
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shajin Huang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tian-Jia Song
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| | - Xiang Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| |
Collapse
|
30
|
Shimizu C, Wakita Y, Tsuchiya Y, Nabeshima T. Influence of Housing Systems on Physical, Emotional, and Cognitive Functions with Aging in DBA/2CrSlc Mice. Animals (Basel) 2020; 10:ani10040746. [PMID: 32344780 PMCID: PMC7222825 DOI: 10.3390/ani10040746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Many scientists carefully monitor the experimental protocols, mouse strain , use of group-housing, and atmospheric enrichment in a housing-cage, but not commercially available housing-systems. The environmental conditions of mice as well as humans affects their emotional behaviors or physical activities. However, limited information is available regarding the influence of housing systems on experimental data. We used two types of housing system in the same laboratory. The difference in the structure of the two cages (chamber vs. individually ventilated cages: IVC) was whether the mouse could dangle or not. The dangling increases the amounts and quality of physical activities. Using the two-different housing systems, we investigated whether differences in physical, emotional, and cognitive functions can be observed in mice with aging. The IVC group demonstrated significantly less food intake, higher body weight, lower rectal core temperature, less muscle and balancing powers with aging, and fewer anxiety-like behaviors than the chamber group. Based on this experiment, the daily physical activities derived from housing systems significantly affected the results of body weight, body temperature, as well as their behaviors. Scientists should pay attention to the structure of housing systems and experimental parameters, particularly when changing the housing systems. Abstract Environmental conditions, including enrichment and stress, affect animal behaviors, but limited information is available regarding the differences in animal functions between the chamber (ventilated system) vs. IVC (individually ventilated cages) housing systems. Therefore, the effects of different housing systems were examined on physical, emotional, and cognitive functions and the intestinal flora with aging. DBA/2CrSlc mice were divided into chamber and IVC groups. Differences in the structure of the two cages considered whether the mouse could dangle or not. Physical, emotional, and cognitive functions were examined using the open field, black and white box, object recognition, horizontal bar, wire hanging, balancing, footprint, and locomotor tests. The IVC group demonstrated significantly less food intake, higher body weight (by approximately 5 g), lower rectal core temperature, less muscle and balancing powers with aging, and fewer anxiety-like behaviors than the chamber group. No differences were observed in the cognitive function and intestinal microbiota between the groups. The housing environment affected the rodent basal temperature and body weight as well as the physical and emotional functions. Scientists should be attentive to the type of cages used in the housing system for an experiment, especially when comparing the results with animals reared in different systems.
Collapse
Affiliation(s)
- Chikako Shimizu
- Frontier Laboratories for Value Creation, SAPPORO HOLDINGS LTD., 10 Okatome, Yaizu, Shizuoka 425-0013, Japan; (Y.W.); (Y.T.)
- Correspondence: ; Tel.: +81-54-629-7980
| | - Yoshihisa Wakita
- Frontier Laboratories for Value Creation, SAPPORO HOLDINGS LTD., 10 Okatome, Yaizu, Shizuoka 425-0013, Japan; (Y.W.); (Y.T.)
| | - Youichi Tsuchiya
- Frontier Laboratories for Value Creation, SAPPORO HOLDINGS LTD., 10 Okatome, Yaizu, Shizuoka 425-0013, Japan; (Y.W.); (Y.T.)
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University,1–98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan;
- NPO Japanese Drug Organization of Appropriate Use and Research, 3-1509 Omoteyama, Tenpaku-ku, Nagoya, Aichi 468-0069, Japan
| |
Collapse
|
31
|
Tsuji M, Ohshima M, Yamamoto Y, Saito S, Hattori Y, Tanaka E, Taguchi A, Ihara M, Ogawa Y. Cilostazol, a Phosphodiesterase 3 Inhibitor, Moderately Attenuates Behaviors Depending on Sex in the Ts65Dn Mouse Model of Down Syndrome. Front Aging Neurosci 2020; 12:106. [PMID: 32372946 PMCID: PMC7186592 DOI: 10.3389/fnagi.2020.00106] [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: 01/31/2020] [Accepted: 03/30/2020] [Indexed: 11/16/2022] Open
Abstract
People with Down syndrome, which is a trisomy of chromosome 21, exhibit intellectual disability from infancy and neuropathology similar to Alzheimer’s disease, such as amyloid plaques, from an early age. Recently, we showed that cilostazol, a selective inhibitor of phosphodiesterase (PDE) 3, promotes the clearance of amyloid β and rescues cognitive deficits in a mouse model of Alzheimer’s disease. The objective of the present study was to examine whether cilostazol improves behaviors in the most widely used animal model of Down syndrome, i.e., Ts65Dn mice. Mice were supplemented with cilostazol from the fetal period until young adulthood. Supplementation significantly ameliorated novel-object recognition in Ts65Dn females and partially ameliorated sensorimotor function as determined by the rotarod test in Ts65Dn females and hyperactive locomotion in Ts65Dn males. Cilostazol supplementation significantly shortened swimming distance in Ts65Dn males in the Morris water maze test, suggesting that the drug improved cognitive function, although it did not shorten swimming duration, which was due to decreased swimming speed. Thus, this study suggests that early supplementation with cilostazol partially rescues behavioral abnormalities seen in Down syndrome and indicates that the effects are sex-dependent.
Collapse
Affiliation(s)
- Masahiro Tsuji
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Makiko Ohshima
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Saito
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yorito Hattori
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Emi Tanaka
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yuko Ogawa
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
32
|
Mertens S, Gass P, Palme R, Hiebl B, Chourbaji S. Effect of a partial cage dividing enrichment on aggression-associated parameters in group-housed male C57BL/6NCrl mice. Appl Anim Behav Sci 2020. [DOI: 10.1016/j.applanim.2020.104939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Martínez Cué C, Dierssen M. Plasticity as a therapeutic target for improving cognition and behavior in Down syndrome. PROGRESS IN BRAIN RESEARCH 2020; 251:269-302. [DOI: 10.1016/bs.pbr.2019.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Martínez de Lagrán M. Mapping behavioral landscapes in Down syndrome animal models. PROGRESS IN BRAIN RESEARCH 2020; 251:145-179. [DOI: 10.1016/bs.pbr.2020.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
35
|
De Toma I, Ortega M, Aloy P, Sabidó E, Dierssen M. DYRK1A Overexpression Alters Cognition and Neural-Related Proteomic Pathways in the Hippocampus That Are Rescued by Green Tea Extract and/or Environmental Enrichment. Front Mol Neurosci 2019; 12:272. [PMID: 31803016 PMCID: PMC6873902 DOI: 10.3389/fnmol.2019.00272] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21, is the most common genetic cause of intellectual disability. We recently discovered that green tea extracts containing epigallocatechin-3-gallate (EGCG) improve cognition in mice transgenic for Dyrk1a (TgDyrk1A) and in a trisomic DS mouse model (Ts65Dn). Interestingly, paired with cognitive stimulation, green tea has beneficial pro-cognitive effects in DS individuals. Dual Specificity Tyrosine-Phosphorylation-Regulated Kinase 1A (DYRK1A) is a major candidate to explain the cognitive phenotypes of DS, and inhibiting its activity is a promising pro-cognitive therapy. DYRK1A kinase activity can be normalized in the hippocampus of transgenic DYRK1A mice administering green tea extracts, but also submitting the animals to environmental enrichment (EE). However, many other mechanisms could also explain the pro-cognitive effects of green tea extracts and EE. To underpin the overall alterations arising upon DYRK1A overexpression and the molecular processes underneath the pro-cognitive effects, we used quantitative proteomics. We investigated the hippocampal (phospho)proteome in basal conditions and after treatment with a green tea extract containing EGCG and/or EE in TgDyrk1A and control mice. We found that Dyrk1A overexpression alters protein and phosphoprotein levels of key postsynaptic and plasticity-related pathways and that these alterations were rescued upon the cognitive enhancer treatments.
Collapse
Affiliation(s)
- Ilario De Toma
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Mireia Ortega
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Eduard Sabidó
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Proteomic Unit, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| |
Collapse
|
36
|
From Basic Visual Science to Neurodevelopmental Disorders: The Voyage of Environmental Enrichment-Like Stimulation. Neural Plast 2019; 2019:5653180. [PMID: 31198418 PMCID: PMC6526521 DOI: 10.1155/2019/5653180] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/06/2019] [Accepted: 04/16/2019] [Indexed: 12/31/2022] Open
Abstract
Genes and environmental stimuli cooperate in the regulation of brain development and formation of the adult neuronal architecture. Genetic alterations or exposure to perturbing environmental conditions, therefore, can lead to altered neural processes associated with neurodevelopmental disorders and brain disabilities. In this context, environmental enrichment emerged as a promising and noninvasive experimental treatment for favoring recovery of cognitive and sensory functions in different neurodevelopmental disorders. The aim of this review is to depict, mainly through the much explicative examples of amblyopia, Down syndrome, and Rett syndrome, the increasing interest in the potentialities and applications of enriched environment-like protocols in the field of neurodevelopmental disorders and the understanding of the molecular mechanisms underlying the beneficial effects of these protocols, which might lead to development of pharmacological interventions.
Collapse
|
37
|
Illouz T, Madar R, Biragyn A, Okun E. Restoring microglial and astroglial homeostasis using DNA immunization in a Down Syndrome mouse model. Brain Behav Immun 2019; 75:163-180. [PMID: 30389461 PMCID: PMC6358279 DOI: 10.1016/j.bbi.2018.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/22/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022] Open
Abstract
Down Syndrome (DS), the most common cause of genetic intellectual disability, is characterized by over-expression of the APP and DYRK1A genes, located on the triplicated chromosome 21. This chromosomal abnormality leads to a cognitive decline mediated by Amyloid-β (Aβ) overproduction and tau hyper-phosphorylation as early as the age of 40. In this study, we used the Ts65Dn mouse model of DS to evaluate the beneficial effect of a DNA vaccination against the Aβ1-11 fragment, in ameliorating Aβ-related neuropathology and rescue of cognitive and behavioral abilities. Anti-Aβ1-11 vaccination induced antibody production and facilitated clearance of soluble oligomers and small extracellular inclusions of Aβ from the hippocampus and cortex of Ts65Dn mice. This was correlated with reduced neurodegeneration and restoration of the homeostatic phenotype of microglial and astroglial cells. Vaccinated Ts65Dn mice performed better in spatial-learning tasks, exhibited reduced motor hyperactivity typical for this strain, and restored short-term memory abilities. Our findings support the hypothesis that DS individuals may benefit from active immunotherapy against Aβ from a young age by slowing the progression of dementia.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
38
|
Moretto E, Murru L, Martano G, Sassone J, Passafaro M. Glutamatergic synapses in neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:328-342. [PMID: 28935587 DOI: 10.1016/j.pnpbp.2017.09.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are a group of diseases whose symptoms arise during childhood or adolescence and that impact several higher cognitive functions such as learning, sociability and mood. Accruing evidence suggests that a shared pathogenic mechanism underlying these diseases is the dysfunction of glutamatergic synapses. We summarize present knowledge on autism spectrum disorders (ASD), intellectual disability (ID), Down syndrome (DS), Rett syndrome (RS) and attention-deficit hyperactivity disorder (ADHD), highlighting the involvement of glutamatergic synapses and receptors in these disorders. The most commonly shared defects involve α-amino-3-hydroxy-5-methyl- 4-isoxazole propionic acid receptors (AMPARs), N-methyl-d-aspartate receptors (NMDARs) and metabotropic glutamate receptors (mGluRs), whose functions are strongly linked to synaptic plasticity, affecting both cell-autonomous features as well as circuit formation. Moreover, the major scaffolding proteins and, thus, the general structure of the synapse are often deregulated in neurodevelopmental disorders, which is not surprising considering their crucial role in the regulation of glutamate receptor positioning and functioning. This convergence of defects supports the definition of neurodevelopmental disorders as a continuum of pathological manifestations, suggesting that glutamatergic synapses could be a therapeutic target to ameliorate patient symptomatology.
Collapse
Affiliation(s)
- Edoardo Moretto
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Luca Murru
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Giuseppe Martano
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy
| | - Jenny Sassone
- San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Maria Passafaro
- CNR, Institute of Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy.
| |
Collapse
|
39
|
Bayne K. Environmental enrichment and mouse models: Current perspectives. Animal Model Exp Med 2018; 1:82-90. [PMID: 30891552 PMCID: PMC6388067 DOI: 10.1002/ame2.12015] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/19/2018] [Indexed: 11/28/2022] Open
Abstract
The provision of environmental enrichment to numerous species of laboratory animals is generally considered routine husbandry. However, mouse enrichment has proven to be very complex due to the often contradictory outcomes (animal health and welfare, variability in scientific data, etc.) associated with strain, age of the animal when enrichment is provided, gender of the animal, scientific use of the animal, and other housing attributes. While this has led to some suggesting that mice should not be provided enrichment, more recently opinion is trending toward acknowledging that enrichment actually normalizes the animal and data obtained from a mouse living in a barren environment are likely not to be representative or even reliable. This article offers an overview of the types of impact enrichment can have on various strains of mice and demonstrates that enrichment not only has a role in mouse husbandry, but also can lead to new areas of scientific enquiry in a number of different fields.
Collapse
|
40
|
Sampedro-Piquero P, Begega A. Environmental Enrichment as a Positive Behavioral Intervention Across the Lifespan. Curr Neuropharmacol 2018; 15:459-470. [PMID: 27012955 PMCID: PMC5543669 DOI: 10.2174/1570159x14666160325115909] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/30/2015] [Accepted: 03/16/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In recent decades, the interest in behavioral interventions has been growing due to the higher prevalence of age-related cognitive impairments. Hence, behavioral interventions, such as cognitive stimulation and physical activity, and along with these, our lifestyle (education level, work position, frequency of cognitive and social activities) have shown important benefits during the cognitive impairment, dementia and even recovery after brain injury. This is due to the fact that this type of intervention and activities promote the formation of a cognitive and brain reserve that allows tolerating brain damage during a long period of time without the appearance of cognitive symptoms. With regard to this, animal models have proved very useful in providing information about the brain mechanisms involved in the development of these cognitive and brain reserves and how they interact with each other. METHODS We summarize several studies showing the positive effects of Environmental Enrichment (EE), understood as a housing condition in which animals benefit from the sensory, physical, cognitive and social stimulation provided, on brain and cognitive functions usually impaired during aging. RESULTS Most of studies have shown that EE is a successful protocol to improve cognitive functions and reduce anxiety-related behaviors across the lifespan, as well as in animal models of neurodegenerative diseases. CONCLUSION Therefore, EE is a laboratory condition in which some aspects of an active lifestyle are reproduced.
Collapse
Affiliation(s)
- P Sampedro-Piquero
- Department of Biological and Health Psychology, Autonomous University of Madrid, Cantoblanco 28049, Madrid, Spain
| | - A Begega
- Neuroscience Laboratory, Psychology Department, University of Oviedo, Plaza Feijoo s/n 33003 Oviedo, INEUROPA, Spain
| |
Collapse
|
41
|
Xing R, Zhang Y, Xu H, Luo X, Chang RCC, Liu J, Yang X. Spatial memory impairment by TRPC1 depletion is ameliorated by environmental enrichment. Oncotarget 2017; 7:27855-73. [PMID: 27034165 PMCID: PMC5053693 DOI: 10.18632/oncotarget.8428] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/08/2016] [Indexed: 01/22/2023] Open
Abstract
Canonical transient receptor potential (TRPC) channels are widely expressed throughout the nervous system whereas their functions remain largely unclear. Here we investigated the effects of TRPC1 deletion on spatial memory ability of mice and the potential intervention by environmental enrichment (EE). Significant spatial memory impairment assessed by conditional fearing test, Y maze test and step-down test in TRPC1 knockout mice was revealed. The behavioral abnormality were attenuated by the treatment of EE. Proteomic analysis by two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) coupled with a matrix-assisted laser desorption/ionisation-time of flight (MALDI-TOF) and tandem mass spectrometry (MS) revealed that TRPC1 deletion caused differential expression of a total of 10 proteins (8 up-regulated and 2 down-regulated) in hippocampus. EE treatment resulted in differential expression of a total of 22 proteins (2 up-regulated and 20 down-regulated) in hippocampus of TRPC1 knockout mice. Among these differentially expressed proteins, the expression of α-internexin and glia maturation factor β (GMF-β), two proteins shown to impair memory, were significantly down-regulated in hippocampus of TRPC1 knockout mice by EE treatment. Taken together, these data suggested that TRPC1 regulated directly or indirectly the expression of multiple proteins, which may be crucial for the maintenance of memory ability, and that EE treatment modulated spatial memory impairment caused by TRPC1 depletion and the mechanisms may involve the modulation of EE on the expression of those dys-regulated proteins such as α-internexin and GMF-β in hippocampus.
Collapse
Affiliation(s)
- Renzhong Xing
- College of Pharmacy, Jinan University, Guangdong, China.,Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Yanling Zhang
- College of Pharmacy, Jinan University, Guangdong, China.,Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Hua Xu
- College of Pharmacy, Jinan University, Guangdong, China
| | - Xiaobin Luo
- AND Biotech, Shenzhen, China.,Guang Zhou Kai-Tuo Biotech, Guangzhou, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Medical Key Laboratory of Guangdong Province, Medical Key Laboratory of Health Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
42
|
McLaughlin KA, Sheridan MA, Nelson CA. Neglect as a Violation of Species-Expectant Experience: Neurodevelopmental Consequences. Biol Psychiatry 2017; 82:462-471. [PMID: 28392082 PMCID: PMC5572554 DOI: 10.1016/j.biopsych.2017.02.1096] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/03/2017] [Accepted: 02/20/2017] [Indexed: 12/25/2022]
Abstract
The human brain requires a wide variety of experiences and environmental inputs in order to develop normally. Children who are neglected by caregivers or raised in institutional environments are deprived of numerous types of species-expectant environmental experiences. In this review, we articulate a model of how the absence of cognitive stimulation and sensory, motor, linguistic, and social experiences common among children raised in deprived early environments constrains early forms of learning, producing long-term deficits in complex cognitive function and associative learning. Building on evidence from animal models, we propose that deprivation accelerates the neurodevelopmental process of synaptic pruning and limits myelination, resulting in age-specific reductions in cortical thickness and white matter integrity among children raised in deprived early environments. We review evidence linking early experiences of psychosocial deprivation to reductions in cognitive ability, associative and implicit learning, language skills, and executive functions as well as atypical patterns of cortical and white matter development-domains that should be profoundly influenced by deprivation through the learning and neural mechanisms we propose. These patterns of atypical development are difficult to explain with existing models that emphasize stress pathways and accelerated limbic system development. A learning account of how deprived early environments influence cognitive and neural development provides a complementary perspective to stress models and highlights novel pathways through which deprivation might confer risk for internalizing and externalizing psychopathology. We end by reviewing evidence for plasticity in cognitive and neural development among children raised in deprived environments following interventions that improve caregiving quality.
Collapse
Affiliation(s)
- Katie A McLaughlin
- Department of Psychology, University of Washington, Seattle, Washington.
| | - Margaret A Sheridan
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Charles A Nelson
- Division of Developmental Medicine and Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston; Harvard Graduate School of Education, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
43
|
Smith-Hicks CL, Cai P, Savonenko AV, Reeves RH, Worley PF. Increased Sparsity of Hippocampal CA1 Neuronal Ensembles in a Mouse Model of Down Syndrome Assayed by Arc Expression. Front Neural Circuits 2017; 11:6. [PMID: 28217086 PMCID: PMC5289947 DOI: 10.3389/fncir.2017.00006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 01/19/2017] [Indexed: 02/05/2023] Open
Abstract
Down syndrome (DS) is the leading chromosomal cause of intellectual disability, yet the neural substrates of learning and memory deficits remain poorly understood. Here, we interrogate neural networks linked to learning and memory in a well-characterized model of DS, the Ts65Dn mouse. We report that Ts65Dn mice exhibit exploratory behavior that is not different from littermate wild-type (WT) controls yet behavioral activation of Arc mRNA transcription in pyramidal neurons of the CA1 region of the hippocampus is altered in Ts65Dn mice. In WT mice, a 5 min period of exploration of a novel environment resulted in Arc mRNA transcription in 39% of CA1 neurons. By contrast, the same period of exploration resulted in only ~20% of CA1 neurons transcribing Arc mRNA in Ts65Dn mice indicating increased sparsity of the behaviorally induced ensemble. Like WT mice the CA1 pyramidal neurons of Ts65Dn mice reactivated Arc transcription during a second exposure to the same environment 20 min after the first experience, but the size of the reactivated ensemble was only ~60% of that in WT mice. After repeated daily exposures there was a further decline in the size of the reactivated ensemble in Ts65Dn and a disruption of reactivation. Together these data demonstrate reduction in the size of the behaviorally induced network that expresses Arc in Ts65Dn mice and disruption of the long-term stability of the ensemble. We propose that these deficits in network formation and stability contribute to cognitive symptoms in DS.
Collapse
Affiliation(s)
- Constance L Smith-Hicks
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA; Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Peiling Cai
- The State Key Laboratory of Biotherapy, West-China Hospital, Sichuan University Chengdu, China
| | - Alena V Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Roger H Reeves
- Department of Physiology and Institute of Genetic Medicine, Johns Hopkins University, School of Medicine Baltimore, MD, USA
| | - Paul F Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA; Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
44
|
Combined Treatment With Environmental Enrichment and (-)-Epigallocatechin-3-Gallate Ameliorates Learning Deficits and Hippocampal Alterations in a Mouse Model of Down Syndrome. eNeuro 2016; 3:eN-NWR-0103-16. [PMID: 27844057 PMCID: PMC5099603 DOI: 10.1523/eneuro.0103-16.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/26/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022] Open
Abstract
Intellectual disability in Down syndrome (DS) is accompanied by altered neuro-architecture, deficient synaptic plasticity, and excitation-inhibition imbalance in critical brain regions for learning and memory. Recently, we have demonstrated beneficial effects of a combined treatment with green tea extract containing (-)-epigallocatechin-3-gallate (EGCG) and cognitive stimulation in young adult DS individuals. Although we could reproduce the cognitive-enhancing effects in mouse models, the underlying mechanisms of these beneficial effects are unknown. Here, we explored the effects of a combined therapy with environmental enrichment (EE) and EGCG in the Ts65Dn mouse model of DS at young age. Our results show that combined EE-EGCG treatment improved corticohippocampal-dependent learning and memory. Cognitive improvements were accompanied by a rescue of cornu ammonis 1 (CA1) dendritic spine density and a normalization of the proportion of excitatory and inhibitory synaptic markers in CA1 and dentate gyrus.
Collapse
|
45
|
Romano E, Cosentino L, Laviola G, De Filippis B. Genes and sex hormones interaction in neurodevelopmental disorders. Neurosci Biobehav Rev 2016; 67:9-24. [DOI: 10.1016/j.neubiorev.2016.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
|
46
|
Sansevero G, Begenisic T, Mainardi M, Sale A. Experience-dependent reduction of soluble β-amyloid oligomers and rescue of cognitive abilities in middle-age Ts65Dn mice, a model of Down syndrome. Exp Neurol 2016; 283:49-56. [PMID: 27288239 DOI: 10.1016/j.expneurol.2016.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/30/2016] [Accepted: 06/06/2016] [Indexed: 11/17/2022]
Abstract
Down syndrome (DS) is the most diffused genetic cause of intellectual disability and, after the age of forty, is invariantly associated with Alzheimer's disease (AD). In the last years, the prolongation of life expectancy in people with DS renders the need for intervention paradigms aimed at improving mental disability and counteracting AD pathology particularly urgent. At present, however, there are no effective therapeutic strategies for DS and concomitant AD in mid-life people. The most intensively studied mouse model of DS is the Ts65Dn line, which summarizes the main hallmarks of the DS phenotype, included severe learning and memory deficits and age-dependent AD-like pathology. Here we report for the first time that middle-age Ts65Dn mice display a marked increase in soluble Aβ oligomer levels in their hippocampus. Moreover, we found that long-term exposure to environmental enrichment (EE), a widely used paradigm that increases sensory-motor stimulation, reduces Aβ oligomers and rescues spatial memory abilities in trisomic mice. Our findings underscore the potential of EE procedures as a non-invasive paradigm for counteracting brain aging processes in DS subjects.
Collapse
Affiliation(s)
- Gabriele Sansevero
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy; NEUROFARBA, University of Florence, Florence, Italy
| | - Tatjana Begenisic
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Marco Mainardi
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Alessandro Sale
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy.
| |
Collapse
|
47
|
Widespread cerebellar transcriptome changes in Ts65Dn Down syndrome mouse model after lifelong running. Behav Brain Res 2016; 296:35-46. [DOI: 10.1016/j.bbr.2015.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/28/2015] [Accepted: 08/17/2015] [Indexed: 12/22/2022]
|
48
|
Catuara-Solarz S, Espinosa-Carrasco J, Erb I, Langohr K, Notredame C, Gonzalez JR, Dierssen M. Principal Component Analysis of the Effects of Environmental Enrichment and (-)-epigallocatechin-3-gallate on Age-Associated Learning Deficits in a Mouse Model of Down Syndrome. Front Behav Neurosci 2015; 9:330. [PMID: 26696850 PMCID: PMC4675859 DOI: 10.3389/fnbeh.2015.00330] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/16/2015] [Indexed: 11/13/2022] Open
Abstract
Down syndrome (DS) individuals present increased risk for Alzheimer's disease (AD) neuropathology and AD-type dementia. Here, we investigated the use of green tea extracts containing (-)-epigallocatechin-3-gallate (EGCG), as co-adjuvant to enhance the effects of environmental enrichment (EE) in Ts65Dn mice, a segmental trisomy model of DS that partially mimics DS/AD pathology, at the age of initiation of cognitive decline. Classical repeated measures ANOVA showed that combined EE-EGCG treatment was more efficient than EE or EGCG alone to improve specific spatial learning related variables. Using principal component analysis (PCA) we found that several spatial learning parameters contributed similarly to a first PC and explained a large proportion of the variance among groups, thus representing a composite learning measure. This PC1 revealed that EGCG or EE alone had no significant effect. However, combined EE-EGCG significantly ameliorated learning alterations of middle age Ts65Dn mice. Interestingly, PCA revealed an increased variability along learning sessions with good and poor learners in Ts65Dn, and this stratification did not disappear upon treatments. Our results suggest that combining EE and EGCG represents a viable therapeutic approach for amelioration of age-related cognitive decline in DS, although its efficacy may vary across individuals.
Collapse
Affiliation(s)
- Silvina Catuara-Solarz
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology Barcelona, Spain ; Centre for Genomic Regulation, Universitat Pompeu Fabra Barcelona, Spain
| | - Jose Espinosa-Carrasco
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology Barcelona, Spain ; Centre for Genomic Regulation, Universitat Pompeu Fabra Barcelona, Spain ; Bioinformatics and Genomics Program, Comparative Bioinformatics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology Barcelona, Spain
| | - Ionas Erb
- Centre for Genomic Regulation, Universitat Pompeu Fabra Barcelona, Spain ; Bioinformatics and Genomics Program, Comparative Bioinformatics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology Barcelona, Spain
| | - Klaus Langohr
- Neurosciences Research Program, Human Pharmacology and Clinical Neurosciences Research Group, IMIM (Hospital del Mar Medical Research Institute) Barcelona, Spain ; Department of Statistics and Operations Research, Universitat Politècnica de Catalunya/BARCELONATECH Barcelona, Spain
| | - Cedric Notredame
- Centre for Genomic Regulation, Universitat Pompeu Fabra Barcelona, Spain ; Bioinformatics and Genomics Program, Comparative Bioinformatics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology Barcelona, Spain
| | - Juan R Gonzalez
- Bioinformatics and Genomics Program, Comparative Bioinformatics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology Barcelona, Spain ; Centre for Research in Environmental Epidemiology Barcelona, Spain ; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública Barcelona, Spain
| | - Mara Dierssen
- Systems Biology Program, Cellular and Systems Neurobiology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology Barcelona, Spain ; Centre for Genomic Regulation, Universitat Pompeu Fabra Barcelona, Spain ; Centro de Investigación Biomédica en Red de Enfermedades Raras Barcelona, Spain
| |
Collapse
|
49
|
Ahmadalipour A, Sadeghzadeh J, Vafaei AA, Bandegi AR, Mohammadkhani R, Rashidy-Pour A. Effects of environmental enrichment on behavioral deficits and alterations in hippocampal BDNF induced by prenatal exposure to morphine in juvenile rats. Neuroscience 2015; 305:372-383. [PMID: 26272536 DOI: 10.1016/j.neuroscience.2015.08.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/06/2015] [Accepted: 08/06/2015] [Indexed: 01/25/2023]
Abstract
Prenatal morphine exposure throughout pregnancy can induce a series of neurobehavioral and neurochemical disturbances by affecting central nervous system development. This study was designed to investigate the effects of an enriched environment on behavioral deficits and changes in hippocampal brain-derived neurotrophic factor (BDNF) levels induced by prenatal morphine in rats. On pregnancy days 11-18, female Wistar rats were randomly injected twice daily with saline or morphine. Offspring were weaned on postnatal day (PND) 21. They were subjected to a standard rearing environment or an enriched environment on PNDs 22-50. On PNDs 51-57, the behavioral responses including anxiety and depression-like behaviors, and passive avoidance memory as well as hippocampal BDNF levels were investigated. The light/dark (L/D) box and elevated plus maze (EPM) were used for the study of anxiety, forced swimming test (FST) was used to assess depression-like behavior and passive avoidance task was used to evaluate learning and memory. Prenatal morphine exposure caused a reduction in time spent in the EPM open arms and a reduction in time spent in the lit side of the L/D box. It also decreased step-through latency and increased time spent in the dark side of passive avoidance task. Prenatal morphine exposure also reduced immobility time and increased swimming time in FST. Postnatal rearing in an enriched environment counteracted with behavioral deficits in the EPM and passive avoidance task, but not in the L/D box. This suggests that exposure to an enriched environment during adolescence period alters anxiety profile in a task-specific manner. Prenatal morphine exposure reduced hippocampal BDNF levels, but enriched environment significantly increased BDNF levels in both saline- and morphine-exposed groups. Our results demonstrate that exposure to an enriched environment alleviates behavioral deficits induced by prenatal morphine exposure and up-regulates the decreased levels of BDNF. BDNF may contribute to the beneficial effects of an enriched environment on prenatal morphine-exposed to rats.
Collapse
Affiliation(s)
- A Ahmadalipour
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, 15131-38111 Semnan, Iran; Student's Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - J Sadeghzadeh
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, 15131-38111 Semnan, Iran; Student's Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - A A Vafaei
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, 15131-38111 Semnan, Iran
| | - A R Bandegi
- Laboratory of Endocrine Research, Research Center of Physiology and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - R Mohammadkhani
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, 15131-38111 Semnan, Iran
| | - A Rashidy-Pour
- Laboratory of Learning and Memory, Research Center and Department of Physiology, School of Medicine, Semnan University of Medical Sciences, 15131-38111 Semnan, Iran.
| |
Collapse
|
50
|
Begenisic T, Sansevero G, Baroncelli L, Cioni G, Sale A. Early environmental therapy rescues brain development in a mouse model of Down syndrome. Neurobiol Dis 2015; 82:409-419. [PMID: 26244989 DOI: 10.1016/j.nbd.2015.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/26/2015] [Accepted: 07/31/2015] [Indexed: 01/15/2023] Open
Abstract
Down syndrome (DS), the most common genetic disorder associated with intellectual disabilities, is an untreatable condition characterized by a number of developmental defects and permanent deficits in the adulthood. Ts65Dn mice, the major animal model for DS, display severe cognitive and synaptic plasticity defects closely resembling the human phenotype. Here, we employed a multidisciplinary approach to investigate, for the first time in developing Ts65Dn mice, the effects elicited by early environmental enrichment (EE) on brain maturation and function. We report that exposure to EE resulted in a robust increase in maternal care levels displayed by Ts65Dn mothers and led to a normalization of declarative memory abilities and hippocampal plasticity in trisomic offspring. The positive effects of EE on Ts65Dn phenotype were not limited to the cognitive domain, but also included a rescue of visual system maturation. The beneficial EE effects were accompanied by increased BDNF and correction of over-expression of the GABA vesicular transporter vGAT. These findings highlight the beneficial impact of early environmental stimuli and their potential for application in the treatment of major functional deficits in children with DS.
Collapse
Affiliation(s)
| | | | | | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Stella Maris, University of Pisa, Calambrone, I-56100 Pisa, Italy
| | | |
Collapse
|