1
|
Wall EK, Virakorn EA, Baker KD, Cohen EM, Richardson R. Preclinical behavioral and pharmacological treatments for enhancing fear extinction in adolescence. Neurosci Biobehav Rev 2025; 172:106090. [PMID: 40049540 DOI: 10.1016/j.neubiorev.2025.106090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Adolescence is a window of vulnerability for the development of anxiety disorders but also a window of opportunity for treatments to minimize the long-term impact of such disorders. Current first-line treatments, primarily exposure-based cognitive-behavioral therapy (CBT), have limited long-term efficacy in adolescents. The urgent need for more effective interventions is underscored by the frequent reports of extinction impairments in adolescents as well as the rising anxiety rates in youth, particularly post-COVID-19. Preclinical research on the extinction of learned fear in adolescents may contribute to developing better treatment approaches to anxiety in this age group. Unfortunately, this is still a largely under-explored area. However, both pharmacological and behavioral augmentation strategies can be used to enhance extinction learning and consolidation. Here we describe work exploring such adjuncts, focusing on pre-clinical work with rodents. Much of the research to date shows striking developmental differences in response to various pharmacological treatments, with only a few shown to be effective in adolescents. Further, recent experience of stress reduces the efficacy of these treatments in adolescence. This review highlights the necessity for tailored strategies, especially when it comes to pharmacological adjuncts, that address developmental differences in drug responses as well as the impact of stressful experiences on treatment efficacy.
Collapse
Affiliation(s)
- Emily K Wall
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia
| | | | - Kathryn D Baker
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia; Department of Psychology, Counselling and Therapy, La Trobe University, Melbourne, VIC 3086, Australia
| | - E Myfanwy Cohen
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Rick Richardson
- School of Psychology, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
2
|
Norwood MF, Marsh CH, Pretty D, Hollins I, Shirota C, Chen B, Gustafsson L, Kendall E, Jones S, Zeeman H. The environment as an important component of neurorehabilitation: introducing the BEEhive - brain and enriched environment (BEE) lab (hive). Disabil Rehabil 2025:1-11. [PMID: 39937038 DOI: 10.1080/09638288.2025.2461266] [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: 08/15/2024] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Contemporary healthcare design often overlooks the environment as a resource for supporting patient well-being and rehabilitation, particularly in neurotrauma care. The prioritisation of safety and efficiency has created stressful spaces that negatively impact patient needs. This paper explores whether environmental enrichment can enhance rehabilitation outcomes for individuals recovering from neurotrauma. It also introduces the BEEhive laboratory, a multidisciplinary initiative integrating environmental enrichment principles into healthcare. METHODOLOGY This paper reviews literature on the role of environmental enrichment in neurotrauma rehabilitation, synthesising empirical evidence on its benefits, and highlighting its potential to improve various aspects of neurorehabilitation. The findings are applied to the BEEhive laboratory's objectives. RESULTS Environmental enrichment is shown to stimulate neurogenesis, increase rehabilitation engagement, reduce disruptive behaviours and depressive symptoms, facilitate social relationships, improve cognitive functioning, reduce stress, and alleviate boredom. Despite these benefits, its application in neurotrauma rehabilitation remains underexplored. The BEEhive laboratory aims to address this gap through multidisciplinary collaboration, implementing strategies to enhance patient outcomes. CONCLUSION To optimise rehabilitation outcomes, healthcare environments must holistically support well-being. Environmentally focused, sustainable interventions in neurotrauma care, exemplified by the BEEhive initiative, are crucial for bridging the gap between research and practice, fostering innovative approaches to neurotrauma rehabilitation.
Collapse
Affiliation(s)
| | - Chelsea H Marsh
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
- School of Applied Psychology, Griffith University, Gold Coast, Australia
| | - Danielle Pretty
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
- School of Health Sciences and Social Work, Griffith University, Queensland, Australia
| | - Izak Hollins
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
| | - Camila Shirota
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
| | - Ben Chen
- Clinical Director, Allied Health and Rehabilitation, Emergency and Specialty Services, Gold Coast Health, Southport, Australia
| | | | - Elizabeth Kendall
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
- Inclusive Futures: Reimagining Disability, Griffith University, Southport, Australia
| | - Susan Jones
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
- Neurosciences Rehabilitation Unit, Gold Coast University Hospital, Gold Coast, Australia
| | - Heidi Zeeman
- The Hopkins Centre, Griffith University, Meadowbrook, Australia
| |
Collapse
|
3
|
Sullens DG, Gilley K, Moraglia LE, Dison S, Hoffman JT, Wiffler MB, Barnes RC, Ginty AT, Sekeres MJ. Sex in aging matters: exercise and chronic stress differentially impact females and males across the lifespan. Front Aging Neurosci 2025; 16:1508801. [PMID: 39881679 PMCID: PMC11774976 DOI: 10.3389/fnagi.2024.1508801] [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: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Assessing sex as a biological variable is critical to determining the influence of environmental and lifestyle risks and protective factors mediating behavior and neuroplasticity across the lifespan. We investigated sex differences in affective behavior, memory, and hippocampal neurogenesis following short- or long-term exposure to exercise or chronic mild stress in young and aged mice. Male and female mice were assigned control, running, or chronic stress rearing conditions for 1 month (young) or for 15 months (aged), then underwent a behavioral test battery to assess activity, affective behavior, and memory. Stress exposure into late-adulthood increased hyperactivity in both sexes, and enhanced anxiety-like and depressive-like behavior in aged female, but not male, mice. One month of stress or running had no differential effects on behavior in young males and females. Running increased survival of BrdU-labelled hippocampal cells in both young and aged mice, and enhanced spatial memory in aged mice. These findings highlight the importance of considering sex when determining how aging is differently impacted by modifiable lifestyle factors across the lifespan.
Collapse
Affiliation(s)
- D. Gregory Sullens
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Kayla Gilley
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, United States
| | - Luke E. Moraglia
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Psychology, The University of Texas at Dallas, Richardson, TX, United States
| | - Sarah Dison
- Department of Biology, Baylor University, Waco, TX, United States
| | - Jessica T. Hoffman
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Madison B. Wiffler
- Department of Biology, Baylor University, Waco, TX, United States
- Department of Neurobiology, University of Utah, Salt Lake City, UT, United States
| | - Robert C. Barnes
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Annie T. Ginty
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Melanie J. Sekeres
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
4
|
Han Z, Zhang L, Ma M, Keshavarzi M. Effects of MicroRNAs and Long Non-coding RNAs on Beneficial Action of Exercise on Cognition in Degenerative Diseases: A Review. Mol Neurobiol 2025; 62:485-500. [PMID: 38869810 DOI: 10.1007/s12035-024-04292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Recent research has exposed a growing body of proof underscoring the importance of microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) in maintaining the physical composition of neurons and influencing cognitive functioning in both standard and atypical circumstances. Extensive research has been conducted on the possible application of miRNAs and lncRNAs as biomarkers for various diseases, with a particular focus on brain disorders, as they possess remarkable durability in cell-free surroundings and can endure repeated freezing and thawing processes. It is intriguing to note that miRNAs and lncRNAs have the ability to function through paracrine mechanisms, thereby playing a role in communication between different organs. Recent research has proposed that the improvement of cognitive abilities through physical exercise in mentally healthy individuals is a valuable method for uncovering potential connections between miRNAs, or microRNAs, and lncRNAs, and human cognitive function. The process of cross-correlating data from disease models and patients with existing data will be crucial in identifying essential miRNAs and lncRNAs, which can potentially act as biomarkers or drug targets in the treatment of cognitive disorders. By combining this method with additional research in animal models, we can determine the function of these molecules and their potential impact on therapy. This article discusses the latest research about the primary miRNAs, lncRNAs, and their exosomes that are affected by physical activity in terms of human cognitive function.
Collapse
Affiliation(s)
- Zhen Han
- Department of Physical Education, Zhejiang International Studies University, Hangzhou, 310023, Zhejiang, China
| | - Lei Zhang
- Institute of Physical Education and Sports, Capital University Of Physical Education And Sports, Beijing, 100191, China.
| | - Minhang Ma
- Department of Physical Education, Zhejiang International Studies University, Hangzhou, 310023, Zhejiang, China
| | - Maryam Keshavarzi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Methi A, Islam MR, Kaurani L, Sakib MS, Krüger DM, Pena T, Burkhardt S, Liebetanz D, Fischer A. A Single-Cell Transcriptomic Analysis of the Mouse Hippocampus After Voluntary Exercise. Mol Neurobiol 2024; 61:5628-5645. [PMID: 38217668 PMCID: PMC11249425 DOI: 10.1007/s12035-023-03869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/29/2023] [Indexed: 01/15/2024]
Abstract
Exercise has been recognized as a beneficial factor for cognitive health, particularly in relation to the hippocampus, a vital brain region responsible for learning and memory. Previous research has demonstrated that exercise-mediated improvement of learning and memory in humans and rodents correlates with increased adult neurogenesis and processes related to enhanced synaptic plasticity. Nevertheless, the underlying molecular mechanisms are not fully understood. With the aim to further elucidate these mechanisms, we provide a comprehensive dataset of the mouse hippocampal transcriptome at the single-cell level after 4 weeks of voluntary wheel-running. Our analysis provides a number of interesting observations. For example, the results suggest that exercise affects adult neurogenesis by accelerating the maturation of a subpopulation of Prdm16-expressing neurons. Moreover, we uncover the existence of an intricate crosstalk among multiple vital signaling pathways such as NF-κB, Wnt/β-catenin, Notch, and retinoic acid (RA) pathways altered upon exercise in a specific cluster of excitatory neurons within the Cornu Ammonis (CA) region of the hippocampus. In conclusion, our study provides an important resource dataset and sheds further light on the molecular changes induced by exercise in the hippocampus. These findings have implications for developing targeted interventions aimed at optimizing cognitive health and preventing age-related cognitive decline.
Collapse
Affiliation(s)
- Aditi Methi
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - M Sadman Sakib
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Dennis M Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Tonatiuh Pena
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Susanne Burkhardt
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - David Liebetanz
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany.
- Department for Psychiatry and Psychotherapy, University Medical Center of Göttingen, Georg-August University, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
- DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany.
| |
Collapse
|
6
|
Mohd Sahini SN, Mohd Nor Hazalin NA, Srikumar BN, Jayasingh Chellammal HS, Surindar Singh GK. Environmental enrichment improves cognitive function, learning, memory and anxiety-related behaviours in rodent models of dementia: Implications for future study. Neurobiol Learn Mem 2024; 208:107880. [PMID: 38103676 DOI: 10.1016/j.nlm.2023.107880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Environmental enrichment (EE) is a process of brain stimulation by modifying the surroundings, for example, by changing the sensory, social, or physical conditions. Rodents have been used in such experimental strategies through exposure to diverse physical, social, and exploration conditions. The present study conducted an extensive analysis of the existing literature surrounding the impact of EE on dementia rodent models. The review emphasised the two principal aspects that are very closely related to dementia: cognitive function (learning and memory) as well as psychological factors (anxiety-related behaviours such as phobias and unrealistic worries). Also highlighted were the mechanisms involved in the rodent models of dementia showing EE effects. Two search engines, PubMed and Science Direct, were used for data collection using the following keywords: environmental enrichment, dementia, rodent model, cognitive performance, and anxiety-related behaviour. Fifty-five articles were chosen depending on the criteria for inclusion and exclusion. The rodent models with dementia demonstrated improved learning and memory in the form of hampered inflammatory responses, enhanced neuronal plasticity, and sustained neuronal activity. EE housing also prevented memory impairment through the prevention of amyloid beta (Aβ) seeding formation, an early stage of Aβ plaque formation. The rodents subjected to EE were observed to present increased exploratory activity and exert less anxiety-related behaviour, compared to those in standard housing. However, some studies have proposed that EE intervention through exercise would be too mild to counteract the anxiety-related behaviour and risk assessment behaviour deficits in the Alzheimer's disease rodent model. Future studies should be conducted on old-aged rodents and the duration of EE exposure that would elicit the greatest benefits since the existing studies have been conducted on a range of ages and EE durations. In summary, EE had a considerable effect on dementia rodent models, with the most evident being improved cognitive function.
Collapse
Affiliation(s)
- Siti Norhafizah Mohd Sahini
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Outpatient Pharmacy Department, Hospital Raja Permaisuri Bainun, 30450 Ipoh, Perak, Malaysia
| | - Nurul Aqmar Mohd Nor Hazalin
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Integrative Pharmacogenomics Institute (iPROMiSE), Level 7, FF3, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia
| | - Bettadapura N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru - 560029, India
| | - Hanish Singh Jayasingh Chellammal
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Selangor Branch, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia; Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia.
| |
Collapse
|
7
|
Jin M, Wei Z, Ramalingam N, Xiao M, Xu A, Yu X, Song Q, Liu W, Zhao J, Zhang D, Selkoe DJ, Li S. Activation of β 2-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28:4877-4888. [PMID: 37365243 DOI: 10.1038/s41380-023-02145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of β2-adrenergic receptor (β2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid β-protein (oAβ). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aβ-species. We found that pharmacological activation of β2-AR, but not β1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAβ-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in β2-AR knockout mice, suggesting that activating β2-AR prevents oAβ-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of β-ARs each decreased HDAC2, whereas Aβ oligomers increased HDAC2 levels in the hippocampus. Further, oAβ-induced inflammatory effects and neurite degeneration were prevented by either β2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of β2-AR is a novel potential therapeutic strategy to mitigate oAβ-mediated features of AD.
Collapse
Affiliation(s)
- Ming Jin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Meng Xiao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China
| | - Anqi Xu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Xiaohan Yu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Qingyang Song
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zhao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Henan Key Laboratory of Neurorestoratology, Xinxiang, Henan, 453100, China
| | - Dainan Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Haley M, Bertrand J, Anderson VT, Fuad M, Frenguelli BG, Corrêa SAL, Wall MJ. Arc expression regulates long-term potentiation magnitude and metaplasticity in area CA1 of the hippocampus in ArcKR mice. Eur J Neurosci 2023; 58:4166-4180. [PMID: 37821126 DOI: 10.1111/ejn.16172] [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: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
Expression of the immediate early gene Arc/Arg3.1 (Arc), a key mediator of synaptic plasticity, is enhanced by neural activity and then reduced by proteasome-dependent degradation. We have previously shown that the disruption of Arc degradation, in an Arc knock-in mouse (ArcKR), where the predominant Arc ubiquitination sites were mutated, reduced the threshold to induce, and also enhanced, the strength of Group I metabotropic glutamate receptor-mediated long-term depression (DHPG-LTD). Here, we have investigated if ArcKR expression changes long-term potentiation (LTP) in CA1 area of the hippocampus. As previously reported, there was no change in basal synaptic transmission at Schaffer collateral/commissural-CA1 (SC-CA1) synapses in ArcKR versus wild-type (WT) mice. There was, however, a significant increase in the amplitude of synaptically induced (with low frequency paired-pulse stimulation) LTD in ArcKR mice. Theta burst stimulation (TBS)-evoked LTP at SC-CA1 synapses was significantly reduced in ArcKR versus WT mice (after 2 h). Group 1 mGluR priming of LTP was abolished in ArcKR mice, which could also potentially contribute to a depression of LTP. Although high frequency stimulation (HFS)-induced LTP was not significantly different in ArcKR compared with WT mice (after 1 h), there was a phenotype in environmentally enriched mice, with the ratio of LTP to short-term potentiation (STP) significantly reduced in ArcKR mice. These findings support the hypothesis that Arc ubiquitination supports the induction and expression of LTP, likely via limiting Arc-dependent removal of AMPA receptors at synapses.
Collapse
Affiliation(s)
- Maisy Haley
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Jeanri Bertrand
- School of Life Sciences, University of Warwick, Coventry, UK
| | | | - Mukattar Fuad
- School of Life Sciences, University of Warwick, Coventry, UK
| | | | - Sonia A L Corrêa
- Faculty of Science and Engineering, Department of Life Sciences, John Dalton Building, Room E210, Manchester Metropolitan University, Manchester, UK
| | - Mark J Wall
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
9
|
Kobiec T, Mardaraz C, Toro-Urrego N, Kölliker-Frers R, Capani F, Otero-Losada M. Neuroprotection in metabolic syndrome by environmental enrichment. A lifespan perspective. Front Neurosci 2023; 17:1214468. [PMID: 37638319 PMCID: PMC10447983 DOI: 10.3389/fnins.2023.1214468] [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: 04/29/2023] [Accepted: 07/17/2023] [Indexed: 08/29/2023] Open
Abstract
Metabolic syndrome (MetS) is defined by the concurrence of different metabolic conditions: obesity, hypertension, dyslipidemia, and hyperglycemia. Its incidence has been increasingly rising over the past decades and has become a global health problem. MetS has deleterious consequences on the central nervous system (CNS) and neurological development. MetS can last several years or be lifelong, affecting the CNS in different ways and treatments can help manage condition, though there is no known cure. The early childhood years are extremely important in neurodevelopment, which extends beyond, encompassing a lifetime. Neuroplastic changes take place all life through - childhood, adolescence, adulthood, and old age - are highly sensitive to environmental input. Environmental factors have an important role in the etiopathogenesis and treatment of MetS, so environmental enrichment (EE) stands as a promising non-invasive therapeutic approach. While the EE paradigm has been designed for animal housing, its principles can be and actually are applied in cognitive, sensory, social, and physical stimulation programs for humans. Here, we briefly review the central milestones in neurodevelopment at each life stage, along with the research studies carried out on how MetS affects neurodevelopment at each life stage and the contributions that EE models can provide to improve health over the lifespan.
Collapse
Affiliation(s)
- Tamara Kobiec
- Facultad de Psicología, Centro de Investigaciones en Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Claudia Mardaraz
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Nicolás Toro-Urrego
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Rodolfo Kölliker-Frers
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
10
|
Alarcón TA, Presti-Silva SM, Simões APT, Ribeiro FM, Pires RGW. Molecular mechanisms underlying the neuroprotection of environmental enrichment in Parkinson's disease. Neural Regen Res 2023; 18:1450-1456. [PMID: 36571341 PMCID: PMC10075132 DOI: 10.4103/1673-5374.360264] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease is the most common movement disorder, affecting about 1% of the population over the age of 60 years. Parkinson's disease is characterized clinically by resting tremor, bradykinesia, rigidity and postural instability, as a result of the progressive loss of nigrostriatal dopaminergic neurons. In addition to this neuronal cell loss, Parkinson's disease is characterized by the accumulation of intracellular protein aggregates, Lewy bodies and Lewy neurites, composed primarily of the protein α-synuclein. Although it was first described almost 200 years ago, there are no disease-modifying drugs to treat patients with Parkinson's disease. In addition to conventional therapies, non-pharmacological treatment strategies are under investigation in patients and animal models of neurodegenerative disorders. Among such strategies, environmental enrichment, comprising physical exercise, cognitive stimulus, and social interactions, has been assessed in preclinical models of Parkinson's disease. Environmental enrichment can cause structural and functional changes in the brain and promote neurogenesis and dendritic growth by modifying gene expression, enhancing the expression of neurotrophic factors and modulating neurotransmission. In this review article, we focus on the current knowledge about the molecular mechanisms underlying environmental enrichment neuroprotection in Parkinson's disease, highlighting its influence on the dopaminergic, cholinergic, glutamatergic and GABAergic systems, as well as the involvement of neurotrophic factors. We describe experimental pre-clinical data showing how environmental enrichment can act as a modulator in a neurochemical and behavioral context in different animal models of Parkinson's disease, highlighting the potential of environmental enrichment as an additional strategy in the management and prevention of this complex disease.
Collapse
Affiliation(s)
- Tamara Andrea Alarcón
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Sarah Martins Presti-Silva
- Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria; Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Ana Paula Toniato Simões
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| | - Fabiola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute o Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, Brazil
| | - Rita Gomes Wanderley Pires
- Department of Physiological Sciences; Laboratory of Molecular and Behavioral Neurobiology, Health Science Center, Universidade Federal do Espirito Santo, Vitoria, Brazil
| |
Collapse
|
11
|
Sałaciak K, Koszałka A, Lustyk K, Żmudzka E, Jagielska A, Pytka K. Memory impairments in rodent depression models: A link with depression theories. Prog Neuropsychopharmacol Biol Psychiatry 2023; 125:110774. [PMID: 37088171 DOI: 10.1016/j.pnpbp.2023.110774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
More than 80% of depressed patients struggle with learning new tasks, remembering positive events, or concentrating on a single topic. These neurocognitive deficits accompanying depression may be linked to functional and structural changes in the prefrontal cortex and hippocampus. However, their mechanisms are not yet completely understood. We conducted a narrative review of articles regarding animal studies to assess the state of knowledge. First, we argue the contribution of changes in neurotransmitters and hormone levels in the pathomechanism of cognitive dysfunction in animal depression models. Then, we used numerous neuroinflammation studies to explore its possible implication in cognitive decline. Encouragingly, we also observed a positive correlation between increased oxidative stress and a depressive-like state with concomitant memory deficits. Finally, we discuss the undeniable role of neurotrophin deficits in developing cognitive decline in animal models of depression. This review reveals the complexity of depression-related memory impairments and highlights the potential clinical importance of gathered findings for developing more reliable animal models and designing novel antidepressants with procognitive properties.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Aleksandra Koszałka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Klaudia Lustyk
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Elżbieta Żmudzka
- Department of Social Pharmacy, Faculty of Pharmacy, Jagiellonian University Medical College Medyczna, 9 Street, Kraków 30-688, Poland
| | - Angelika Jagielska
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Krakow 30-688, Poland.
| |
Collapse
|
12
|
Ismail TR, Yap CG, Naidu R, Pamidi N. Environmental Enrichment and Metformin Improve Metabolic Functions, Hippocampal Neuron Survival, and Hippocampal-Dependent Memory in High-Fat/High-Sucrose Diet-Induced Type 2 Diabetic Rats. BIOLOGY 2023; 12:biology12030480. [PMID: 36979171 PMCID: PMC10045208 DOI: 10.3390/biology12030480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Background: The Western-style diet-induced type 2 diabetes mellitus (T2D) may eventually trigger neurodegeneration and memory impairment. Thus, it is essential to identify effective therapeutic strategies to overcome T2D complications. This study aimed to investigate the effects of environmental enrichment (EE) and metformin interventions on metabolic dysfunctions, hippocampal neuronal death, and hippocampal-dependent memory impairments in high-fat/high-sucrose (HFS) diet-induced T2D rats. Methods: Thirty-two male rats (200-250 g) were divided into four groups: C group (standard diet + conventional cage); D group (HFS diet + conventional cage); DE group (HFS diet + EE cage/6hr daily); and DM group (HFS diet + metformin + conventional cage). Body weight was measured every week. T-maze tasks, anthropometric, biochemical, histological, and morphometric parameters were measured. The expression changes of hippocampal genes were also analyzed. Results: The anthropometric and biochemical parameters were improved in DE and DM groups compared with the D group. DE and DM groups had significantly higher T-maze percentages than the D group. These groups also had better histological and morphometric parameters than the D group. The interventions of EE and metformin enhanced the expression of hippocampal genes related to neurogenesis and synaptic plasticity (BDNF/TrkB binding, PI3K-Akt, Ras-MAPK, PLCγ-Ca2+, and LTP). Conclusion: Environmental enrichment (EE) and metformin improved metabolic functions, hippocampal neuron survival, and hippocampal-dependent memory in HFS diet-induced T2D rats. The underlying mechanisms of these interventions involved the expression of genes that regulate neurogenesis and synaptic plasticity.
Collapse
Affiliation(s)
- Teh Rasyidah Ismail
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia
- Clinical Laboratory Science Section, Institute of Medical Science Technology, Universiti Kuala Lumpur, Kajang, Kuala Lumpur 43000, Selangor Darul Ehsan, Malaysia
| | - Christina Gertrude Yap
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia
| | - Narendra Pamidi
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
13
|
Ren Y, Savadlou A, Park S, Siska P, Epp JR, Sargin D. The impact of loneliness and social isolation on the development of cognitive decline and Alzheimer's Disease. Front Neuroendocrinol 2023; 69:101061. [PMID: 36758770 DOI: 10.1016/j.yfrne.2023.101061] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Alzheimer's Disease (AD) is the leading cause of dementia, observed at a higher incidence in women compared with men. Treatments aimed at improving pathology in AD remain ineffective to stop disease progression. This makes the detection of the early intervention strategies to reduce future disease risk extremely important. Isolation and loneliness have been identified among the major risk factors for AD. The increasing prevalence of both loneliness and AD emphasizes the urgent need to understand this association to inform treatment. Here we present a comprehensive review of both clinical and preclinical studies that investigated loneliness and social isolation as risk factors for AD. We discuss that understanding the mechanisms of how loneliness exacerbates cognitive impairment and AD with a focus on sex differences will shed the light for the underlying mechanisms regarding loneliness as a risk factor for AD and to develop effective prevention or treatment strategies.
Collapse
Affiliation(s)
- Yi Ren
- Department of Cell Biology and Anatomy, University of Calgary, Canada; Cumming School of Medicine, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada
| | - Aisouda Savadlou
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Soobin Park
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Paul Siska
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, University of Calgary, Canada; Cumming School of Medicine, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada
| | - Derya Sargin
- Department of Psychology, University of Calgary, Canada; Department of Physiology & Pharmacology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada.
| |
Collapse
|
14
|
Lipinski M, Niñerola S, Fuentes-Ramos M, Valor LM, Del Blanco B, López-Atalaya JP, Barco A. CBP Is Required for Establishing Adaptive Gene Programs in the Adult Mouse Brain. J Neurosci 2022; 42:7984-8001. [PMID: 36109165 PMCID: PMC9617619 DOI: 10.1523/jneurosci.0970-22.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Environmental factors and life experiences impinge on brain circuits triggering adaptive changes. Epigenetic regulators contribute to this neuroadaptation by enhancing or suppressing specific gene programs. The paralogous transcriptional coactivators and lysine acetyltransferases CREB binding protein (CBP) and p300 are involved in brain plasticity and stimulus-dependent transcription, but their specific roles in neuroadaptation are not fully understood. Here we investigated the impact of eliminating either CBP or p300 in excitatory neurons of the adult forebrain of mice from both sexes using inducible and cell type-restricted knock-out strains. The elimination of CBP, but not p300, reduced the expression and chromatin acetylation of plasticity genes, dampened activity-driven transcription, and caused memory deficits. The defects became more prominent in elderly mice and in paradigms that involved enduring changes in transcription, such as kindling and environmental enrichment, in which CBP loss interfered with the establishment of activity-induced transcriptional and epigenetic changes in response to stimulus or experience. These findings further strengthen the link between CBP deficiency in excitatory neurons and etiopathology in the nervous system.SIGNIFICANCE STATEMENT How environmental conditions and life experiences impinge on mature brain circuits to elicit adaptive responses that favor the survival of the organism remains an outstanding question in neurosciences. Epigenetic regulators are thought to contribute to neuroadaptation by initiating or enhancing adaptive gene programs. In this article, we examined the role of CREB binding protein (CBP) and p300, two paralogous transcriptional coactivators and histone acetyltransferases involved in cognitive processes and intellectual disability, in neuroadaptation in adult hippocampal circuits. Our experiments demonstrate that CBP, but not its paralog p300, plays a highly specific role in the epigenetic regulation of neuronal plasticity gene programs in response to stimulus and provide unprecedented insight into the molecular mechanisms underlying neuroadaptation.
Collapse
Affiliation(s)
- Michal Lipinski
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Sergio Niñerola
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Miguel Fuentes-Ramos
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Luis M Valor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Beatriz Del Blanco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Jose P López-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| | - Angel Barco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Campus de Sant Joan, 03550 Alicante, Spain
| |
Collapse
|
15
|
Zhu G, Guo M, Zhao J, Zhang H, Wang G, Chen W. Bifidobacterium breve intervention combined with environmental enrichment alleviates cognitive impairment by regulating the gut microbiota and microbial metabolites in Alzheimer's disease mice. Front Immunol 2022; 13:1013664. [PMID: 36203603 PMCID: PMC9530393 DOI: 10.3389/fimmu.2022.1013664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by behavioral and cognitive impairments and its increasing prevalence imposes a healthcare burden on society. To date, most intervention studies have only focused on a single AD-related factor and have yielded modest cognitive improvements. Here, we show that environmental enrichment (EE) training combined with Bifidobacterium breve CCFM1025 intervention significantly alleviated amyloid-β (Aβ)-induced cognitive impairment and inhibited neuroinflammation in mice. Moreover, we found that EE combined with B. breve CCFM1025 treatment restored AD-associated gut microbiota dysbiosis and reversed microbial metabolites changes. By integrating behavioral and neurological data with metabolomic profiles, we corroborated the microbiota-metabolite-brain interactions, with acetate and tryptophan metabolism as potential drivers. Taken together, our results provide a promising multidomain intervention strategy to prevent cognitive decline and delay the progression of AD through a combination of dietary microbiome-based approaches and lifestyle interventions.
Collapse
Affiliation(s)
- Guangsu Zhu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Min Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
- National Engineering Center of Functional Food, Jiangnan University, Wuxi, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Center of Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
16
|
Améndola L, Weary D, Zobel G. Effects of personality on assessments of anxiety and cognition. Neurosci Biobehav Rev 2022; 141:104827. [PMID: 35970418 DOI: 10.1016/j.neubiorev.2022.104827] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/10/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
Abstract
Individual variation in responses to commonly used tests of anxiety and spatial memory is often reported. While this variation is frequently considered to be 'noise', evidence suggests that it is, at least partially, related to consistent individual differences in behavioral responses (i.e., personality). The same tests used to assess anxiety are often used to profile personality traits, but personality differences are rarely considered when testing treatment differences in anxiety. Focusing on the rat literature, we describe fundamental principles involved in anxiety and spatial memory tests and we discuss how personality differences and housing conditions can influence behavioral responses in these tests. We propose that an opportunity exists to increase stress resiliency in environmentally sensitive individuals by providing environmental enrichment. We conclude by discussing different approaches to incorporating personality measures into the design and analysis of future studies; given the potential that variation masks research outcomes, we suggest that a strategy which considers the individual and its housing can contribute to improving research reproducibility.
Collapse
Affiliation(s)
- Lucia Améndola
- Animal Welfare Program, University of British Columbia, Canada.
| | - Daniel Weary
- Animal Welfare Program, University of British Columbia, Canada.
| | - Gosia Zobel
- Animal Behaviour and Welfare Team, AgResearch Ltd., Ruakura Research Centre, 10 Bisley Road, Private Bag 3123, Hamilton 3214, New Zealand.
| |
Collapse
|
17
|
Han Y, Yuan M, Guo YS, Shen XY, Gao ZK, Bi X. The role of enriched environment in neural development and repair. Front Cell Neurosci 2022; 16:890666. [PMID: 35936498 PMCID: PMC9350910 DOI: 10.3389/fncel.2022.890666] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/29/2022] [Indexed: 12/01/2022] Open
Abstract
In addition to genetic information, environmental factors play an important role in the structure and function of nervous system and the occurrence and development of some nervous system diseases. Enriched environment (EE) can not only promote normal neural development through enhancing neuroplasticity but also play a nerve repair role in restoring functional activities during CNS injury by morphological and cellular and molecular adaptations in the brain. Different stages of development after birth respond to the environment to varying degrees. Therefore, we systematically review the pro-developmental and anti-stress value of EE during pregnancy, pre-weaning, and “adolescence” and analyze the difference in the effects of EE and its sub-components, especially with physical exercise. In our exploration of potential mechanisms that promote neurodevelopment, we have found that not all sub-components exert maximum value throughout the developmental phase, such as animals that do not respond to physical activity before weaning, and that EE is not superior to its sub-components in all respects. EE affects the developing and adult brain, resulting in some neuroplastic changes in the microscopic and macroscopic anatomy, finally contributing to enhanced learning and memory capacity. These positive promoting influences are particularly prominent regarding neural repair after neurobiological disorders. Taking cerebral ischemia as an example, we analyzed the molecular mediators of EE promoting repair from various dimensions. We found that EE does not always lead to positive effects on nerve repair, such as infarct size. In view of the classic issues such as standardization and relativity of EE have been thoroughly discussed, we finally focus on analyzing the essentiality of the time window of EE action and clinical translation in order to devote to the future research direction of EE and rapid and reasonable clinical application.
Collapse
Affiliation(s)
- Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Mei Yuan
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yi-Sha Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Department of Graduate School, Shanghai University of Medicine and Health Sciences Affiliated Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Xia Bi
| |
Collapse
|
18
|
Dandi E, Spandou E, Tata DA. Investigating the role of environmental enrichment initiated in adolescence against the detrimental effects of chronic unpredictable stress in adulthood: Sex-specific differences in behavioral and neuroendocrinological findings. Behav Processes 2022; 200:104707. [PMID: 35842198 DOI: 10.1016/j.beproc.2022.104707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/10/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022]
Abstract
Environmental Enrichment (EE) improves cognitive function and enhances brain plasticity, while chronic stress increases emotionality, impairs learning and memory, and has adverse effects on brain anatomy and biochemistry. We explored the beneficial role of environmental enrichment initiated in adolescence against the negative outcomes of Chronic Unpredictable Stress (CUS) during adulthood on emotional behavior, cognitive function, as well as somatic and neuroendocrine markers in both sexes. Adolescent Wistar rats housed either in enriched or standard housing conditions for 10 weeks. On postnatal day 66, a subgroup from each housing condition was daily exposed to a 4-week stress protocol. Following stress, adult rats underwent behavioral testing to evaluate anxiety, exploration/locomotor activity, depressive-like behavior and spatial learning/memory. Upon completion of behavioral testing, animals were exposed to a 10-m stressful event to test the neuroendocrine response to acute stress. CUS decreased body weight gain and increased adrenal weight. Some stress-induced behavioral adverse effects were sex-specific since learning impairments were limited to males while depressive-like behavior to females. EE housing protected against CUS-related behavioral deficits and body weight loss. Exposure to CUS affected the neuroendocrine response of males to acute stress as revealed by the increased corticosterone levels. Our findings highlight the significant role of EE in adolescence as a protective factor against the negative effects of stress and underline the importance of inclusion of both sexes in animal studies.
Collapse
Affiliation(s)
- Evgenia Dandi
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Spandou
- Laboratory of Experimental Physiology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despina A Tata
- Laboratory of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
19
|
Higarza SG, Arboleya S, Arias JL, Gueimonde M, Arias N. Akkermansia muciniphila and environmental enrichment reverse cognitive impairment associated with high-fat high-cholesterol consumption in rats. Gut Microbes 2022; 13:1-20. [PMID: 33678110 PMCID: PMC7946069 DOI: 10.1080/19490976.2021.1880240] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is one of the most prevalent diseases globally. A high-fat, high-cholesterol (HFHC) diet leads to an early NASH model. It has been suggested that gut microbiota mediates the effects of diet through the microbiota-gut-brain axis, modifying the host's brain metabolism and disrupting cognition. Here, we target NASH-induced cognitive damage by testing the impact of environmental enrichment (EE) and the administration of either Lacticaseibacillus rhamnosus GG (LGG) or Akkermansia muciniphila CIP107961 (AKK). EE and AKK, but not LGG, reverse the HFHC-induced cognitive dysfunction, including impaired spatial working memory and novel object recognition; however, whereas AKK restores brain metabolism, EE results in an overall decrease. Moreover, AKK and LGG did not induce major rearrangements in the intestinal microbiota, with only slight changes in bacterial composition and diversity, whereas EE led to an increase in Firmicutes and Verrucomicrobia members. Our findings illustrate the interplay between gut microbiota, the host's brain energy metabolism, and cognition. In addition, the findings suggest intervention strategies, such as the administration of AKK, for the management of the cognitive dysfunction related to NASH.
Collapse
Affiliation(s)
- Sara G. Higarza
- Laboratory of Neuroscience, Department of Psychology. University of Oviedo, Oviedo, Asturias, Spain,Instituto De Neurociencias Del Principado De Asturias (INEUROPA), Asturias, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto De Productos Lácteos De Asturias (IPLA-CSIC), Villaviciosa, Asturias, Spain
| | - Jorge L. Arias
- Laboratory of Neuroscience, Department of Psychology. University of Oviedo, Oviedo, Asturias, Spain,Instituto De Neurociencias Del Principado De Asturias (INEUROPA), Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto De Productos Lácteos De Asturias (IPLA-CSIC), Villaviciosa, Asturias, Spain,Miguel Gueimonde Department of Microbiology and Biochemistry of Dairy Products, Instituto De Productos Lácteos De Asturias (IPLA-CSIC), Villaviciosa, Asturias 33300, Spain
| | - Natalia Arias
- Instituto De Neurociencias Del Principado De Asturias (INEUROPA), Asturias, Spain,UK Dementia Research Institute, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK,CONTACT Natalia Arias Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, LondonSE5 8AF, United Kingdom
| |
Collapse
|
20
|
Goldberg M, Islam MR, Kerimoglu C, Lancelin C, Gisa V, Burkhardt S, Krüger DM, Marquardt T, Malchow B, Schmitt A, Falkai P, Sananbenesi F, Fischer A. Exercise as a model to identify microRNAs linked to human cognition: a role for microRNA-409 and microRNA-501. Transl Psychiatry 2021; 11:514. [PMID: 34625536 PMCID: PMC8501071 DOI: 10.1038/s41398-021-01627-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/20/2021] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs have been linked to synaptic plasticity and memory function and are emerging as potential biomarkers and therapeutic targets for cognitive diseases. Most of these data stem from the analysis of model systems or postmortem tissue from patients which mainly represents an advanced stage of pathology. Due to the in-accessibility of human brain tissue upon experimental manipulation, it is still challenging to identify microRNAs relevant to human cognition, which is however a key step for future translational studies. Here, we employ exercise as an experimental model for memory enhancement in healthy humans with the aim to identify microRNAs linked to memory function. By analyzing the circulating smallRNAome we find a cluster of 18 microRNAs that are highly correlated to cognition. MicroRNA-409-5p and microRNA-501-3p were the most significantly regulated candidates. Functional analysis revealed that the two microRNAs are important for neuronal integrity, synaptic plasticity, and morphology. In conclusion, we provide a novel approach to identify microRNAs linked to human memory function.
Collapse
Affiliation(s)
- Maria Goldberg
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany
| | - Md Rezaul Islam
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany.
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany
| | - Camille Lancelin
- Developmental Neurobiology Laboratory, European Neuroscience Institute, Grisebachstrasse 5, 37077, Goettingen, Germany
| | - Verena Gisa
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany
| | - Susanne Burkhardt
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany
| | - Dennis M Krüger
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany
| | - Till Marquardt
- Interfaculty Chair for Neurobiological Research, RWTH Aachen University: Medical Faculty, Clinic for Neurology & Faculty for Mathematics, Computer and Natural Sciences, Institute for Biology 2, Worringer Weg 3, 52074, Aachen, Germany
| | - Berend Malchow
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University Munich, Nußbaumstr. 7, 80336, München, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Von-Siebold-Str. 5, 37075, Göttingen, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University Munich, Nußbaumstr. 7, 80336, München, Germany
- Laboratory of Neuroscience (LIM27), Institute of Psychiatry, University of Sao Paulo, 05403-010, São Paulo, Brazil
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-University Munich, Nußbaumstr. 7, 80336, München, Germany
| | - Farahnaz Sananbenesi
- German Center for Neurodegenerative Diseases, Research Group for Genome Dynamics in Brain Diseases, Von Siebold Str. 3A, 37075, Göttingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, Von Siebold Str 3A, 37075, Goettingen, Germany.
- Department of Psychiatry and Psychotherapy, University Medical Center, Von-Siebold-Str. 5, 37075, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
21
|
Ceanga M, Dahab M, Witte OW, Keiner S. Adult Neurogenesis and Stroke: A Tale of Two Neurogenic Niches. Front Neurosci 2021; 15:700297. [PMID: 34447293 PMCID: PMC8382802 DOI: 10.3389/fnins.2021.700297] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/30/2021] [Indexed: 01/17/2023] Open
Abstract
In the aftermath of an acute stroke, numerous signaling cascades that reshape the brain both in the perilesional zone as well as in more distal regions are activated. Despite continuous improvement in the acute treatment of stroke and the sustained research efforts into the pathophysiology of stroke, we critically lag in our integrated understanding of the delayed and chronic responses to ischemic injury. As such, the beneficial or maladaptive effect of some stroke-induced cellular responses is unclear, restricting the advancement of therapeutic strategies to target long-term complications. A prominent delayed effect of stroke is the robust increase in adult neurogenesis, which raises hopes for a regenerative strategy to counter neurological deficits in stroke survivors. In the adult brain, two regions are known to generate new neurons from endogenous stem cells: the subventricular zone (SVZ) and the dentate subgranular zone (SGZ) of the hippocampus. While both niches respond with an increase in neurogenesis post-stroke, there are significant regional differences in the ensuing stages of survival, migration, and maturation, which may differently influence functional outcome. External interventions such as rehabilitative training add a further layer of complexity by independently modulating the process of adult neurogenesis. In this review we summarize the current knowledge regarding the effects of ischemic stroke on neurogenesis in the SVZ and in the SGZ, and the influence of exogenous stimuli such as motor activity or enriched environment (EE). In addition, we discuss the contribution of SVZ or SGZ post-stroke neurogenesis to sensory, motor and cognitive recovery.
Collapse
Affiliation(s)
- Mihai Ceanga
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Mahmoud Dahab
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W. Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Silke Keiner
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
22
|
Lambert CT, Guillette LM. The impact of environmental and social factors on learning abilities: a meta-analysis. Biol Rev Camb Philos Soc 2021; 96:2871-2889. [PMID: 34342125 DOI: 10.1111/brv.12783] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/20/2022]
Abstract
Since the 1950s, researchers have examined how differences in the social and asocial environment affect learning in rats, mice, and, more recently, a variety of other species. Despite this large body of research, little has been done to synthesize these findings and to examine if social and asocial environmental factors have consistent effects on cognitive abilities, and if so, what aspects of these factors have greater or lesser impact. Here, we conducted a systematic review and meta-analysis examining how different external environmental features, including the social environment, impact learning (both speed of acquisition and performance). Using 531 mean-differences from 176 published articles across 27 species (with studies on rats and mice being most prominent) we conducted phylogenetically corrected mixed-effects models that reveal: (i) an average absolute effect size |d| = 0.55 and directional effect size d = 0.34; (ii) interventions manipulating the asocial environment result in larger effects than social interventions alone; and (iii) the length of the intervention is a significant predictor of effect size, with longer interventions resulting in larger effects. Additionally, much of the variation in effect size remained unexplained, possibly suggesting that species differ widely in how they are affected by environmental interventions due to varying ecological and evolutionary histories. Overall our results suggest that social and asocial environmental factors do significantly affect learning, but these effects are highly variable and perhaps not always as predicted. Most notably, the type (social or asocial) and length of interventions are important in determining the strength of the effect.
Collapse
Affiliation(s)
- Connor T Lambert
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2R3, Canada
| | - Lauren M Guillette
- Department of Psychology, University of Alberta, P217 Biological Sciences Building, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
23
|
Xiao R, Ali S, Caligiuri MA, Cao L. Enhancing Effects of Environmental Enrichment on the Functions of Natural Killer Cells in Mice. Front Immunol 2021; 12:695859. [PMID: 34394087 PMCID: PMC8355812 DOI: 10.3389/fimmu.2021.695859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
The environment of an organism can convey a powerful influence over its biology. Environmental enrichment (EE), as a eustress model, has been used extensively in neuroscience to study neurogenesis and brain plasticity. EE has also been used as an intervention for the treatment and prevention of neurological and psychiatric disorders with limited clinical application. By contrast, the effects of EE on the immune system are relatively less investigated. Recently, accumulating evidence has demonstrated that EE can robustly impact immune function. In this review, we summarize the major components of EE, the impact of EE on natural killer (NK) cells, EE's immunoprotective roles in cancer, and the underlying mechanisms of EE-induced NK cell regulation. Moreover, we discuss opportunities for translational application based on insights from animal research of EE-induced NK cell regulation.
Collapse
Affiliation(s)
- Run Xiao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, United States
| | - Michael A. Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA, United States
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, United States
| |
Collapse
|
24
|
Zocher S, Overall RW, Lesche M, Dahl A, Kempermann G. Environmental enrichment preserves a young DNA methylation landscape in the aged mouse hippocampus. Nat Commun 2021; 12:3892. [PMID: 34162876 PMCID: PMC8222384 DOI: 10.1038/s41467-021-23993-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
The decline of brain function during aging is associated with epigenetic changes, including DNA methylation. Lifestyle interventions can improve brain function during aging, but their influence on age-related epigenetic changes is unknown. Using genome-wide DNA methylation sequencing, we here show that experiencing a stimulus-rich environment counteracts age-related DNA methylation changes in the hippocampal dentate gyrus of mice. Specifically, environmental enrichment prevented the aging-induced CpG hypomethylation at target sites of the methyl-CpG-binding protein Mecp2, which is critical to neuronal function. The genes at which environmental enrichment counteracted aging effects have described roles in neuronal plasticity, neuronal cell communication and adult hippocampal neurogenesis and are dysregulated with age-related cognitive decline in the human brain. Our results highlight the stimulating effects of environmental enrichment on hippocampal plasticity at the level of DNA methylation and give molecular insights into the specific aspects of brain aging that can be counteracted by lifestyle interventions.
Collapse
Affiliation(s)
- Sara Zocher
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Rupert W Overall
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Mathias Lesche
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- DRESDEN-concept Genome Center c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
- DRESDEN-concept Genome Center c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
25
|
Heidarianpour A, Mohammadi F, Keshvari M, Mirazi N. Ameliorative effects of endurance training and Matricaria chamomilla flowers hydroethanolic extract on cognitive deficit in type 2 diabetes rats. Biomed Pharmacother 2021; 135:111230. [PMID: 33434853 DOI: 10.1016/j.biopha.2021.111230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/18/2020] [Accepted: 12/31/2020] [Indexed: 12/26/2022] Open
Abstract
Diabetes mellitus is mainly associated with degeneration of the central nervous system, which eventually leads to cognitive deficit. Although some studies suggest that exercise can improve the cognitive decline associated with diabetes, the potential effects of endurance training (ET) accompanied by Matricaria chamomilla (M.ch) flowers extract on cognitive impairment in type 2 diabetes has been poorly understood. Forty male Wistar rats were randomized into 5 equal groups of 8: healthy-sedentary (H-sed), diabetes-sedentary (D-sed), diabetes-endurance training (D-ET), diabetes-Matricaria chamomilla. (D-M.ch), and diabetes-endurance training-Matricaria chamomilla. (D-ET-M.ch). Nicotinamide (110 mg/kg, i.p.) and Streptozotocin (65 mg/kg, i.p.) were utilized to initiate type 2 diabetes. Then, ET (5 days/week) and M.ch (200 mg/kg body weight/daily) were administered for 12 weeks. After 12 weeks of the experiment, cognitive functions were assessed using the Morris Water Maze (MWM) test and a passive avoidance paradigm using a shuttle box device. Subsequently, using crystal violet staining, neuron necrosis was examined in the CA3 area of the hippocampus. Diabetic rats showed cognitive impairment following an increase in the number of necrotic cells in region CA3 of the hippocampal tissue. Also, diabetes increased serum levels of lipid peroxidation and decreased total antioxidant capacity in serum and hippocampal tissue. ET + M.ch treatment prevented the necrosis of neurons in the hippocampal tissue. Following positive changes in hippocampal tissue and serum antioxidant enzyme levels, an improvement was observed in the cognitive impairment of the diabetic rats receiving ET + M.ch. Therefore the results showed that treatment with ET + M.ch could ameliorate memory and inactive avoidance in diabetic rats. Hence, the use of ET + M.ch interventions is proposed as a new therapeutic perspective on the death of hippocampal neurons and cognitive deficit caused by diabetes.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/metabolism
- CA3 Region, Hippocampal/pathology
- Cognition/drug effects
- Cognitive Dysfunction/metabolism
- Cognitive Dysfunction/pathology
- Cognitive Dysfunction/prevention & control
- Cognitive Dysfunction/psychology
- Combined Modality Therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/psychology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/psychology
- Diabetes Mellitus, Type 2/therapy
- Endurance Training
- Flowers
- Lipid Peroxidation
- Male
- Matricaria/chemistry
- Morris Water Maze Test/drug effects
- Necrosis
- Oxidative Stress/drug effects
- Physical Conditioning, Animal
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Wistar
- Rats
Collapse
Affiliation(s)
- Ali Heidarianpour
- Department of Exercise Physiology, Faculty of Sport Science, Bu-Ali Sina University, Hamedan, Iran.
| | - Fereshteh Mohammadi
- Department of Exercise Physiology, Faculty of Sport Science, Bu-Ali Sina University, Hamedan, Iran
| | - Maryam Keshvari
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khorramabad, Iran
| | - Naser Mirazi
- Department of Biology, Faculty of Basic Sciences, Bu- Ali Sina University, Hamedan, Iran
| |
Collapse
|
26
|
Goli P, Yazdi M, Poursafa P, Kelishadi R. Intergenerational influence of paternal physical activity on the offspring's brain: A systematic review and meta-analysis. Int J Dev Neurosci 2021; 81:10-25. [PMID: 33252826 DOI: 10.1002/jdn.10081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND It is well established that parents can influence their offspring's neurodevelopment. It is shown that paternal environment and lifestyle is beneficial for the progeny's fitness and might affect their metabolic mechanisms; however, the effects of paternal exercise on brain in the offspring have not been explored in detail. OBJECTIVE This study aims to review the impact of paternal physical exercise on memory and learning, neuroplasticity, as well as DNA methylation levels in the offspring's hippocampus. STUDY DESIGN In this systematic review and meta-analysis, electronic literature search was conducted in databases including PubMed, Scopus, and Web of Science. Eligible studies were those with an experimental design, including an exercise intervention arm, with assessment of any type of memory function, learning ability, or any type of brain plasticity as the outcome measures. Standardized mean difference (SMD) and 95% confidence intervals (CI) were computed as effect size. RESULTS The systematic review revealed the important role of environmental enrichment in the behavioral development of next generation. Also, offspring of exercised fathers displayed higher levels of memory ability, and lower level of brain-derived neurotrophic factor. A significant effect of paternal exercise on the hippocampal volume was also reported in the few available studies. CONCLUSION These results suggest an intergenerational effect of paternal physical activity on cognitive benefit, which may be associated with hippocampal epigenetic programming in offspring. However, the biological mechanisms of this modulation remain to be determined.
Collapse
Affiliation(s)
- Parvin Goli
- Pediatrics Department, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Yazdi
- Pediatrics Department, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parnian Poursafa
- Cellular and Molecular Biology Department, Faculty of Science, University of Isfahan, Isfahan, Iran
| | - Roya Kelishadi
- Pediatrics Department, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
27
|
Fulopova B, Stuart KE, Bennett W, Bindoff A, King AE, Vickers JC, Canty AJ. Regional differences in beta amyloid plaque deposition and variable response to midlife environmental enrichment in the cortex of APP/PS1 mice. J Comp Neurol 2020; 529:1849-1862. [PMID: 33104234 DOI: 10.1002/cne.25060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/21/2023]
Abstract
Environmentally enriched housing conditions can increase performance on cognitive tasks in APP/PS1 mice; however, the potential effects of environmental enrichment (EE) on disease modification in terms of pathological change are inconclusive. We hypothesized that previous contrasting findings may be attributable to regional differences in susceptibility to amyloid beta (Aβ) plaque deposition in cortical regions that are functionally associated with EE. We characterized fibrillar plaque deposition in 6, 12, and 18-22 months old APP/PS1 mice in the prefrontal (PFC), somatosensory (SS2), and primary motor cortex (M1). We found a significant increase in plaque load between 6 and 12 months in all regions. In animals over 12 months, only the PFC region continued to significantly accumulate plaques. Additionally, 12 months old animals subjected to 6 months of EE showed improved spatial navigation and had significantly fewer plaques in M1 and SS2, but not in the PFC. These findings suggest that the PFC region is selectively susceptible to Aβ deposition and less responsive to the attenuating effects of EE. In contrast, M1 and SS2 regions plateau with respect to Aβ deposition by 12 months of age and are susceptible to amyloid pathology modification by midlife EE.
Collapse
Affiliation(s)
- Barbora Fulopova
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley E Stuart
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - William Bennett
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Alison J Canty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
28
|
Silva BA, Miglietta EA, Ferrari CC. Training the brain: could it improve multiple sclerosis treatment? Rev Neurosci 2020; 31:779-792. [PMID: 32712593 DOI: 10.1515/revneuro-2020-0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis (MS) is a neurological disease characterized by neuroinflammation, demyelination and axonal degeneration along with loss of function in the central nervous system. For many years, research in MS has focused on the efficacy of pharmacological treatments. However, during the last years, many publications have been dedicated to the study of the efficacy of non-pharmacological strategies, such as physical exercise and cognitive training. Beneficial effects of the combination of both strategies on cognitive function have been described in both ageing adults and patients with neurodegenerative diseases, such as MS. The analysis of combining both physical and cognitive stimulation can be summarized by the environmental enrichment (EE) experiments, which are more suitable for animal models. EE refers to housing conditions consisting of exercise and cognitive and social stimulation. In this review, we will summarize the available studies that describe the influence of EE in both MS patients and MS animal models.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET, Potosí 4240, Buenos Aires, C1181ACH, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| | - Esteban Alberto Miglietta
- Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET, Potosí 4240, Buenos Aires, C1181ACH, Argentina.,Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA-CONICET, Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| |
Collapse
|
29
|
Pritchett-Corning KR. Environmental Complexity and Research Outcomes. ILAR J 2020; 60:239-251. [PMID: 32559304 DOI: 10.1093/ilar/ilaa007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 11/14/2022] Open
Abstract
Environmental complexity is an experimental paradigm as well as a potential part of animals' everyday housing experiences. In experimental uses, researchers add complexity to stimulate brain development, delay degenerative brain changes, elicit more naturalistic behaviors, and test learning and memory. Complexity can exacerbate or mitigate behavioral problems, give animals a sense of control, and allow for expression of highly driven, species-typical behaviors that can improve animal welfare. Complex environments should be designed thoughtfully with the animal's natural behaviors in mind, reported faithfully in the literature, and evaluated carefully for unexpected effects.
Collapse
Affiliation(s)
- Kathleen R Pritchett-Corning
- Office of Animal Resources, Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts.,Department of Comparative Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
30
|
Cordier JM, Aguggia JP, Danelon V, Mir FR, Rivarola MA, Mascó D. Postweaning Enriched Environment Enhances Cognitive Function and Brain-Derived Neurotrophic Factor Signaling in the Hippocampus in Maternally Separated Rats. Neuroscience 2020; 453:138-147. [PMID: 33039520 DOI: 10.1016/j.neuroscience.2020.09.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Adverse environments during early life may lead to different neurophysiological and behavioral consequences, including depression and learning and memory deficits that persist into adulthood. Previously, we demonstrated that exposure to an enriched environment during adolescence mitigates the cognitive impairment observed after maternal separation in a task-specific manner. However, underlying neural mechanisms are still not fully understood. The current study examines the effects of neonatal maternal separation (MS) and postweaning environmental enrichment (EE) on spatial learning and memory performance in a short version of the Barnes Maze, active and passive behaviors in the forced swim test, and on TrkB/BDNF receptor expression in the hippocampus. Our results revealed that MS impaired acquisition learning and that enriched rats performed better than non-enriched rats in acquisition trials, regardless of early conditions. During the probe, enriched-housed rats demonstrated better performance than those reared in standard conditions. No significant differences between groups were found in the forced swim test. Both MS and EE increase full-length TrkB expression, and the combination of MS and EE treatment caused the highest levels of this protein expression. Similarly, truncated TrkB expression was higher in the MS/EE group. Animal facility rearing (AFR) non-enriched groups present the lowest activation of phosphorylated Erk, a canonical downstream kinase of TrkB signaling. Taken together, our results demonstrate the importance of enriched environment as an intervention to ameliorate the effects of maternal separation on spatial learning and memory. TrkB/BDNF signaling could mediate neuroplastic changes related to learning and memory during exposure to enriched environment.
Collapse
Affiliation(s)
- Javier Maximiliano Cordier
- Instituto de Diversidad y Ecología Animal (IDEA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba -Córdoba, Argentina
| | - Julieta Paola Aguggia
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Enrique Barros esq. Enfermera Gordillo. Ciudad Universitaria, CP: 5016, Córdoba, Argentina
| | - Víctor Danelon
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, CP: 5016, Córdoba, Argentina
| | - Franco Rafael Mir
- Cátedra de Fisiología Animal, Departamento de Ciencias Exactas Físicas y Naturales, Universidad Nacional de La Rioja, Av. Luis M. de la Fuente S/N, Ciudad Universitaria de la Ciencia y de la Técnica, F5300 La Rioja, Argentina; Cátedra de Fisiología Animal, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 299 X5000JJC- Córdoba, Argentina
| | - María Angélica Rivarola
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Enrique Barros esq. Enfermera Gordillo. Ciudad Universitaria, CP: 5016, Córdoba, Argentina; Cátedra de Fisiología Animal, Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 299 X5000JJC- Córdoba, Argentina.
| | - Daniel Mascó
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de Córdoba, Av. Vélez Sarsfield 1611, CP: 5016, Córdoba, Argentina
| |
Collapse
|
31
|
Marrocco J, Einhorn NR, McEwen BS. Environmental epigenetics of sex differences in the brain. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:209-220. [PMID: 33008526 DOI: 10.1016/b978-0-444-64123-6.00015-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Experiences throughout the life course lead to unique phenotypes even among those with the same genotype. Genotype sets the substrate on which physiologic processes, which communicate with the brain, mediate the effects of life experiences via epigenetics. Epigenetics modify the expression of genes in the brain and body in response to circulating hormones and other mediators, which are activated to facilitate survival responses through a process called allostasis. Epigenetic signatures can even be inherited, resulting in transgenerational effects. This chapter addresses epigenetics in the context of sex differences, discussing the intersection between genetics and gonadal hormones and their effect in the brain at discrete developmental periods.
Collapse
Affiliation(s)
- Jordan Marrocco
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States.
| | - Nathan R Einhorn
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, Rockefeller University, New York, NY, United States
| |
Collapse
|
32
|
Stevenson W, Hase Y, Wilson E, Hollins A, Hase M, Ennaceur A, Craggs L, Ihara M, Horsburgh K, Kalaria RN. Long-term effects of experimental carotid stenosis on hippocampal infarct pathology, neurons and glia and amelioration by environmental enrichment. Brain Res Bull 2020; 163:72-83. [PMID: 32707262 DOI: 10.1016/j.brainresbull.2020.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/03/2020] [Accepted: 07/15/2020] [Indexed: 01/17/2023]
Abstract
Hippocampal atrophy and pathology are common in ageing-related disorders and associated with cognitive impairment and dementia. We explored whether environmental enrichment (EE) ameliorated the pathological sequelae in the hippocampus subsequent to chronic cerebral hypoperfusion induced by bilateral common carotid artery stenosis (BCAS). Seventy-four male C57BL/6 J mice underwent BCAS or sham surgery. One-week after surgery, mice were exposed to three different degrees of EE; either standard housing conditions (std), limited 3 -h exposure to EE per day (3 h) or full-time exposure to EE (full) for 3 months. Four months after surgery, the hippocampus was examined for the extent of vascular brain injury and neuronal and glial changes. Results showed that long-term BCAS induced strokes, most often in CA1 subfield, reduced 40-50 % CA1 neurons (P < 0.01) and increased microglia/macrophage in CA1-CA3 subfields (P < 0.02). Remarkably, both 3 h and full-time EE regimes attenuated hippocampal neuronal death and repressed recurrent strokes with complete prevention of larger infarcts in mice on full-time EE (P < 0.01). Full-time EE also reduced astrocytic clasmatodendrosis and microglial/macrophage activation in all CA subfields. Our results suggest that exposure to EE differentially reduces long-term hypoperfusive hippocampal damage. The implementation of even limited EE may be beneficial for patients diagnosed with vascular cognitive impairment.
Collapse
Affiliation(s)
- William Stevenson
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Yoshiki Hase
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Elle Wilson
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Annabel Hollins
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mai Hase
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abdel Ennaceur
- Department of Pharmacy, Sunderland Pharmacy School, The University of Sunderland, Sunderland, UK
| | - Lucy Craggs
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Centre, Osaka, Japan
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Raj N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Heimer-McGinn VR, Wise TB, Hemmer BM, Dayaw JNT, Templer VL. Social housing enhances acquisition of task set independently of environmental enrichment: A longitudinal study in the Barnes maze. Learn Behav 2020; 48:322-334. [PMID: 32040697 PMCID: PMC7415481 DOI: 10.3758/s13420-020-00418-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human studies suggest that healthy social relationships benefit cognition, yet little is known about the underlying neural mechanisms of this protective effect. In rodents, studies on acute isolation and environmental enrichment (EE) confirm the importance of social exposure. Despite the widely recognized importance of sociality, however, rodent models have yet to explore the independent contributions of social housing divorced of other forms of enrichment. This study dissociates the effects of social and physical enrichment on spatial learning and memory from adulthood to old age. Rats were placed in either single or group housing, provided with ample enrichment, and tested at three time points on several phases/versions of the Barnes maze (BM) (standard, retention probes, variable location, and reversal). We found that sustained social housing enhanced cognitive flexibility, as evidenced by superior acquisition of task set (standard BM), adaptability to a new task set (variable BM), and improved reversal learning (reversal BM). Long-term retention (BM retention probes) of spatial memory was unaffected by housing conditions. Recent studies from our lab, including this report, are the first to show that social housing confers cognitive benefits beyond those of physical enrichment. Importantly, our experimental design is ideal for exploring the neural underpinnings of this socially induced cognitive protection. Understanding how sociality influences cognition will be invaluable to translational models of aging, neuropsychiatric disease, and neurological injury.
Collapse
Affiliation(s)
- Victoria R Heimer-McGinn
- Department of Psychology, Providence College, 1 Cunningham Square, Providence, RI, 02918, USA
- Department of Psychology, Roger Williams University, 1 Old Ferry Road, Bristol, RI, 02809, USA
| | - Taylor B Wise
- Department of Psychology, Providence College, 1 Cunningham Square, Providence, RI, 02918, USA
| | - Brittany M Hemmer
- Department of Psychology, Providence College, 1 Cunningham Square, Providence, RI, 02918, USA
| | - Judith N T Dayaw
- Department of Psychology, Providence College, 1 Cunningham Square, Providence, RI, 02918, USA
| | - Victoria L Templer
- Department of Psychology, Providence College, 1 Cunningham Square, Providence, RI, 02918, USA.
| |
Collapse
|
34
|
Early exposure to environmental enrichment protects male rats against neuropathic pain development after nerve injury. Exp Neurol 2020; 332:113390. [PMID: 32598929 DOI: 10.1016/j.expneurol.2020.113390] [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] [Received: 04/22/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 12/30/2022]
Abstract
Because environmental elements modify chronic pain development and endogenous mechanisms of pain control are still a great therapeutic source, we investigated the effects of an early exposure to environmental enrichment (EE) in a translational model of neuropathic pain. Young male rats born and bred in an enriched environment, which did not count on running wheel, underwent chronic constriction injury (CCI) of sciatic nerve. EE abolished neuropathic pain behavior 14 days after CCI. Opioid receptors' antagonism reversed EE-analgesic effect. β-endorphin and met-enkephalin serum levels were increased only in EE-CCI group. Blockade of glucocorticoid receptors did not alter EE-analgesic effect, although corticosterone circulating levels were increased in EE animals. In the spinal cord, EE controlled CCI-induced serotonin increase. In DRG, EE blunted the expression of ATF-3 after CCI. Surprisingly, EE-CCI group showed a remarkable preservation of sciatic nerve fibers compared to NE-CCI group. This work demonstrated global effects induced by an EE protocol that explain, in part, the protective role of EE upon chronic noxious stimulation, reinforcing the importance of endogenous mechanisms in the prevention of chronic pain development.
Collapse
|
35
|
Silva BA, Ferrari CC. Environmental enrichment as a promising strategy for aiding multiple sclerosis treatment. Neural Regen Res 2020; 15:1660-1661. [PMID: 32209769 PMCID: PMC7437581 DOI: 10.4103/1673-5374.276334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Translacional e Ingeniería Biomédica del Hospital italiano (IMTIB-CONICET); Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA- CONICET, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Translacional e Ingeniería Biomédica del Hospital italiano (IMTIB-CONICET); Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA- CONICET, Buenos Aires, Argentina
| |
Collapse
|
36
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
37
|
Wei Z, Meng X, El Fatimy R, Sun B, Mai D, Zhang J, Arora R, Zeng A, Xu P, Qu S, Krichevsky AM, Selkoe DJ, Li S. Environmental enrichment prevents Aβ oligomer-induced synaptic dysfunction through mirna-132 and hdac3 signaling pathways. Neurobiol Dis 2020; 134:104617. [PMID: 31669733 PMCID: PMC7243177 DOI: 10.1016/j.nbd.2019.104617] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/04/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022] Open
Abstract
As the most common cause of progressive cognitive decline in humans, Alzheimer's disease (AD) has been intensively studied, but the mechanisms underlying its profound synaptic dysfunction remain unclear. Here we confirm that exposing wild-type mice to an enriched environment (EE) facilitates signaling in the hippocampus that promotes long-term potentiation (LTP). Exposing the hippocampus of mice kept in standard housing to soluble Aβ oligomers impairs LTP, but EE can fully prevent this. Mechanistically, the key molecular features of the EE benefit are an upregulation of miRNA-132 and an inhibition of histone deacetylase (HDAC) signaling. Specifically, soluble Aβ oligomers decreased miR-132 expression and increased HDAC3 levels in cultured primary neurons. Further, we provide evidence that HDAC3 is a direct target of miR-132. Overexpressing miR-132 or injecting an HDAC3 inhibitor into mice in standard housing mimics the benefits of EE in enhancing hippocampal LTP and preventing hippocampal impairment by Aβ oligomers in vivo. We conclude that EE enhances hippocampal synaptic plasticity by upregulating miRNA-132 and reducing HDAC3 signaling in a way that counteracts the synaptotoxicity of human Aβ oligomers. Our findings provide a rationale for prolonged exposure to cognitive novelty and/or epigenetic modulation to lessen the progressive effects of Aβ accumulation during human brain aging.
Collapse
Affiliation(s)
- Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America; Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingjun Meng
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan 528300, Guangdong, China
| | - Rachid El Fatimy
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Bowen Sun
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Dongmei Mai
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan 528300, Guangdong, China
| | - Junfang Zhang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America; Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, HMS Initiative for RNA Medicine, Zhejiang, China
| | - Ramil Arora
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Ailiang Zeng
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shaogang Qu
- Central Laboratory and Department of Neurology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan 528300, Guangdong, China
| | - Anna M Krichevsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
38
|
Silva BA, Leal MC, Farías MI, Erhardt B, Galeano P, Pitossi FJ, Ferrari CC. Environmental enrichment improves cognitive symptoms and pathological features in a focal model of cortical damage of multiple sclerosis. Brain Res 2020; 1727:146520. [PMID: 31669283 DOI: 10.1016/j.brainres.2019.146520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/27/2019] [Accepted: 10/20/2019] [Indexed: 10/25/2022]
Abstract
Multiple Sclerosis (MS) is a neuroinflammatory disease affecting white and grey matter, it is characterized by demyelination, axonal degeneration along with loss of motor, sensitive and cognitive functions. MS is a heterogeneous disease that displays different clinical courses: relapsing/remitting MS (RRMS), and MS progressive forms: primary progressive (PPMS) and secondary progressive (SPMS). Cortical damage in the progressive MS forms has considerable clinical relevance due to its association with cognitive impairment and disability progression in patients. One treatment is available for the progressive forms of the disease, but none are specific for cognitive deficits. We developed an animal model that reflects most of the characteristics of the cortical damage, such as cortical neuroinflammation, demyelination, neurodegeneration and meningeal inflammation, which was associated with cognitive impairment. Cognitive rehabilitation, exercise and social support have begun to be evaluated in patients and animal models of neurodegenerative diseases. Environmental enrichment (EE) provides exercise as well as cognitive and social stimulation. EE has been demonstrated to exert positive effects on cognitive domains, such as learning and memory, and improving anxiety-like symptoms. We proposed to study the effect of EE on peripherally stimulated cortical lesion induced by the long term expression of interleukin IL-1β (IL-1β) in adult rats. Here, we demonstrated that EE: 1) reduces the peripheral inflammatory response to the stimulus, 2) ameliorates cognitive deficits and anxiety-like symptoms, 3) modulates neurodegeneration, demyelination and glial activation, 4) regulates neuroinflammation by reducing the expression of pro-inflammatory cytokines and enhancing the expression of anti-inflammatory ones. Our findings correlate with the fact that EE housing could be considered an effective non- pharmacological therapeutic agent that can synergistically aid in the rehabilitation of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Translational Medicine and Biomedical Engineering of the Italian Hospital (IMTIB, CONICET), Potosí 4240, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - María Celeste Leal
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - María Isabel Farías
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Brenda Erhardt
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Pablo Galeano
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Fernando Juan Pitossi
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Institute of Translational Medicine and Biomedical Engineering of the Italian Hospital (IMTIB, CONICET), Potosí 4240, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Patricias Argentinas 435, Buenos Aires, Argentina.
| |
Collapse
|
39
|
Khoo SYS, Correia V, Uhrig A. Nesting material enrichment reduces severity of overgrooming-related self-injury in individually housed rats. Lab Anim 2020; 54:546-558. [PMID: 31924130 DOI: 10.1177/0023677219894356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Individual or singly-housing laboratory rats is common in many animal facilities, but has an adverse impact on the welfare of this social species. It has previously been shown that a small proportion of individually housed mice (∼5%) engage in pathological overgrooming behaviour, but this has not been assessed in rats. We performed an observational study to determine the prevalence of overgrooming-related self-injury and whether providing nesting material enrichment throughout an animal's life would affect the prevalence or severity of overgrooming-related self-injury. Due to protocol differences between projects in our behavioural neuroscience lab, unenriched rats received a nylabone and a shelter (n = 167), while baseline-enriched rats received a nylabone, shelter and shredded paper nesting material throughout experiments (n = 238). Unenriched rats received nesting material enrichment after the onset of overgrooming-related self-injury. Over 18 months, rats were monitored by their experimenters on a daily basis (5-7 days/week over 2-3 months/project) and any cases of overgrooming-related self-injury were recorded. Replicating the findings of previous studies in mice, we observed 20 cases of overgrooming-related self-injury (∼5%) with no difference in prevalence between rats on the basis of supplier, cage position, experimental procedure (behavioural only or involving surgical procedures), reinforcer (ethanol or sugar) or level of baseline-enrichment. While there was no difference in onset severity between rats that were unenriched at baseline and baseline-enriched rats, baseline-enriched rats had lower self-injury severity scores at one-, two- and four-week follow-ups. These results suggest that nesting material enrichment provided throughout an animal's life may reduce overgrooming-related self-injury.
Collapse
Affiliation(s)
- Shaun Y-S Khoo
- Center for Studies in Behavioral Neurobiology, Department of Psychology, 5618Concordia University, Montreal, Canada
| | - Vanessa Correia
- Center for Studies in Behavioral Neurobiology, Department of Psychology, 5618Concordia University, Montreal, Canada
| | - Alexandra Uhrig
- Center for Studies in Behavioral Neurobiology, Department of Psychology, 5618Concordia University, Montreal, Canada
| |
Collapse
|
40
|
Rabadán R, Ramos-Campos M, Redolat R, Mesa-Gresa P. Physical activity and environmental enrichment: Behavioural effects of exposure to different housing conditions in mice. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2019-035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
41
|
Sampedro-Piquero P, Ladrón de Guevara-Miranda D, Pavón FJ, Serrano A, Suárez J, Rodríguez de Fonseca F, Santín LJ, Castilla-Ortega E. Neuroplastic and cognitive impairment in substance use disorders: a therapeutic potential of cognitive stimulation. Neurosci Biobehav Rev 2019; 106:23-48. [DOI: 10.1016/j.neubiorev.2018.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 10/15/2018] [Accepted: 11/23/2018] [Indexed: 01/08/2023]
|
42
|
Djawas FA, Redjeki S, Kodariah R, Kartinah NT. Aerobic exercise combined with environmental enrichment improves spatial memory and enhances neuroligin 1 expression: an animal study. MEDICAL JOURNAL OF INDONESIA 2019. [DOI: 10.13181/mji.v28i3.2109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND Numerous studies have revealed that aerobic exercise (AE) or environmental enrichment (EE) exert positive effects on the molecular, cellular, and structural changes responsible for functional plasticity. Therefore, this study was aimed to investigate whether the combination of AE and EE yields a greater effect on spatial memory and increases the expression of the adhesion molecule neuroligin 1 compared with either of the therapies alone. METHODS Twenty 6-month-old male Wistar rats were randomly divided into four groups: (1) the control group (C), (2) the AE group, (3) the EE group, and (4) the AE-EE group. Escape latency in a Water-E-maze (WEM) task was used as a parameter to assess spatial memory function. Neuroligin 1 protein expression was examined via the immunohistochemistry (IHC) technique followed by IHC optical density (OD) score analysis of the CA1 hippocampal region. RESULTS Data acquired from the WEM task show that escape latency progressively decreased over time in all groups. However, animals in the AE-EE group required less time to complete the task compared with those in the control, AE, and EE groups. IHC OD scores revealed that the combination of AE and EE caused the highest expression of neuroligin 1 in the CA1 hippocampal region among the therapies studied. Statistical analysis indicated significant differences in OD score between the AE-EE and control, AE, and EE groups (p ≤ 0.001). CONCLUSIONS The combination of AE and EE rats increased neuroligin 1 expression and improved the spatial memory of male Wistar rats compared with application of either therapy alone.
Collapse
|
43
|
Benito E, Kerimoglu C, Ramachandran B, Pena-Centeno T, Jain G, Stilling RM, Islam MR, Capece V, Zhou Q, Edbauer D, Dean C, Fischer A. RNA-Dependent Intergenerational Inheritance of Enhanced Synaptic Plasticity after Environmental Enrichment. Cell Rep 2019; 23:546-554. [PMID: 29642011 PMCID: PMC5912949 DOI: 10.1016/j.celrep.2018.03.059] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/05/2018] [Accepted: 03/14/2018] [Indexed: 01/05/2023] Open
Abstract
Physical exercise in combination with cognitive training is known to enhance synaptic plasticity, learning, and memory and lower the risk for various complex diseases including Alzheimer's disease. Here, we show that exposure of adult male mice to an environmental enrichment paradigm leads to enhancement of synaptic plasticity and cognition also in the next generation. We show that this effect is mediated through sperm RNA and especially miRs 212/132. In conclusion, our study reports intergenerational inheritance of an acquired cognitive benefit and points to specific miRs as candidates mechanistically involved in this type of transmission.
Collapse
Affiliation(s)
- Eva Benito
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Cemil Kerimoglu
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Binu Ramachandran
- Trans-synaptic Signaling Group, European Neuroscience Institute, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - Tonatiuh Pena-Centeno
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany; Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany; Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Roman Manuel Stilling
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Md Rezaul Islam
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Vincenzo Capece
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany; Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), Munich, Feodor Lynen Strasse 17, 81377 Munich, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Feodor Lynen Strasse 17, 81377 Munich, Germany
| | - Camin Dean
- Trans-synaptic Signaling Group, European Neuroscience Institute, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - André Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, von Siebold Strasse 3A, 37075 Göttingen, Germany; Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Von Siebold Strasse 5, 37075 Göttingen, Germany.
| |
Collapse
|
44
|
Haider S, Nawaz A, Batool Z, Tabassum S, Perveen T. Alleviation of diazepam-induced conditioned place preference and its withdrawal-associated neurobehavioral deficits following pre-exposure to enriched environment in rats. Physiol Behav 2019; 208:112564. [PMID: 31145918 DOI: 10.1016/j.physbeh.2019.112564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 05/24/2019] [Accepted: 05/26/2019] [Indexed: 12/18/2022]
Abstract
Diazepam is one of the widely prescribed sedative drugs for the treatment of anxiety and sleep disorders. However, its continuous use can induce addiction, tolerance, and withdrawal symptoms and, therefore, the pharmacological use of diazepam is restricted. Exposure to enriched environment can reduce the addiction to stimulants including amphetamine, cocaine, and nicotine. However, the protective effect of enriched environment against preference of sedative drugs is not yet investigated. This study, therefore, determined the effects of enriched environment to prevent diazepam-preference using conditioned place preference (CPP) paradigm. Adult rats were reared in social (n = 12) or physically (n = 12) enriched environment for four weeks. Each group was then sub-divided into two groups and were administered either saline (Control; n = 6) or diazepam (1 mg/kg; n = 6) on alternate days for thirteen days. During the administration of diazepam, the CPP was conducted to monitor drug preference on 5th, 9th and 13th day of experiment. It was observed that the diazepam administration significantly (p < .01) induced preference in rats. Neurobehavioral deficits including hypolocomotor activity, depression-like behavior, impaired learning and memory functions were also observed after 24 h of drug abstinence. Exposure to enriched environment significantly reduced diazepam-preference and other neurobehavioral deficits. This study provides preliminary evidence to highlight the importance of enriched environment in the attenuation of diazepam-preference.
Collapse
Affiliation(s)
- Saida Haider
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan.
| | - Amber Nawaz
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan; Department of Biomedical Engineering, Sir Syed University of Engineering and Technology, Karachi 75300, Pakistan
| | - Zehra Batool
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Saiqa Tabassum
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan; Department of Biosciences, Shaheed Zulfiqar Ali Bhutto Institute of Science and Technology, Karachi, Pakistan
| | - Tahira Perveen
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
45
|
Key periods of cognitive decline in a nonhuman primate model of cognitive aging, the common marmoset (Callithrix jacchus). Neurobiol Aging 2019; 74:1-14. [DOI: 10.1016/j.neurobiolaging.2018.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/18/2022]
|
46
|
Martinez Hernandez A, Urbanke H, Gillman AL, Lee J, Ryazanov S, Agbemenyah HY, Benito E, Jain G, Kaurani L, Grigorian G, Leonov A, Rezaei-Ghaleh N, Wilken P, Arce FT, Wagner J, Fuhrmann M, Caruana M, Camilleri A, Vassallo N, Zweckstetter M, Benz R, Giese A, Schneider A, Korte M, Lal R, Griesinger C, Eichele G, Fischer A. The diphenylpyrazole compound anle138b blocks Aβ channels and rescues disease phenotypes in a mouse model for amyloid pathology. EMBO Mol Med 2019; 10:32-47. [PMID: 29208638 PMCID: PMC5760857 DOI: 10.15252/emmm.201707825] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease is a devastating neurodegenerative disease eventually leading to dementia. An effective treatment does not yet exist. Here we show that oral application of the compound anle138b restores hippocampal synaptic and transcriptional plasticity as well as spatial memory in a mouse model for Alzheimer's disease, when given orally before or after the onset of pathology. At the mechanistic level, we provide evidence that anle138b blocks the activity of conducting Aβ pores without changing the membrane embedded Aβ-oligomer structure. In conclusion, our data suggest that anle138b is a novel and promising compound to treat AD-related pathology that should be investigated further.
Collapse
Affiliation(s)
- Ana Martinez Hernandez
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,Department for Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Hendrik Urbanke
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Alan L Gillman
- Department of Bioengineering, Materials Science and Engineering, Department of Mechanical and Aerospace Engineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joon Lee
- Department of Bioengineering, Materials Science and Engineering, Department of Mechanical and Aerospace Engineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sergey Ryazanov
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Hope Y Agbemenyah
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Eva Benito
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Lalit Kaurani
- DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Gayane Grigorian
- Department of Cellular Neurobiology, Technical University Braunschweig, Braunschweig, Germany
| | - Andrei Leonov
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Nasrollah Rezaei-Ghaleh
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Department of Translational Structural Biology of Dementia, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Petra Wilken
- DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.,Group for Translational Research in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Fernando Teran Arce
- Department of Bioengineering, Materials Science and Engineering, Department of Mechanical and Aerospace Engineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jens Wagner
- Group for Neuroimmunology and Imaging, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Martin Fuhrmann
- Group for Neuroimmunology and Imaging, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Mario Caruana
- Department of Physiology and Biochemistry, Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Angelique Camilleri
- Department of Physiology and Biochemistry, Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Markus Zweckstetter
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany.,Department of Translational Structural Biology of Dementia, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.,Department of Neurology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Roland Benz
- Life Sciences and Chemistry, Jacobs University of Bremen, Bremen, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anja Schneider
- DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany.,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.,Group for Translational Research in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen, Göttingen, Germany
| | - Martin Korte
- Department of Cellular Neurobiology, Technical University Braunschweig, Braunschweig, Germany .,Helmholtz Center for Infections Research, Braunschweig, Germany
| | - Ratnesh Lal
- Department of Bioengineering, Materials Science and Engineering, Department of Mechanical and Aerospace Engineering and Institute of Engineering in Medicine, University of California San Diego, La Jolla, CA, USA
| | - Christian Griesinger
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany .,DFG Research Center Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Gregor Eichele
- Department for Genes and Behavior, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany .,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
47
|
Kokras N, Sotiropoulos I, Besinis D, Tzouveka EL, Almeida OFX, Sousa N, Dalla C. Neuroplasticity-related correlates of environmental enrichment combined with physical activity differ between the sexes. Eur Neuropsychopharmacol 2019; 29:1-15. [PMID: 30497839 DOI: 10.1016/j.euroneuro.2018.11.1107] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 11/01/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
Abstract
Environmental enrichment (EE), comprising positive physical (exercise) and cognitive stimuli, influences neuronal structure and usually improves brain function. The promise of EE as a preventative strategy against neuropsychiatric disease is especially high during early postnatal development when the brain is still amenable to reorganization. Despite the fact that male and female brains differ in terms of connectivity and function that may reflect early life experiences, knowledge of the neural substrates and mechanisms by which such changes arise remains limited. This study compared the impact of EE combined with physical activity on neuroplasticity and its functional consequences in adult male and female rats; EE was provided during the first 3 months of life and our analysis focused on the hippocampus, an area implicated in cognitive behavior as well as the neuroendocrine response to stress. Both male and female rats reared in EE displayed better object recognition memory than their control counterparts. Interestingly, sex differences were revealed in the effects of EE on time spent exploring the objects during this test. Independently of sex, EE increased hippocampal turnover rates of dopamine and serotonin and reduced expression of 5-HT1A receptors; in addition, EE upregulated expression of synaptophysin, a presynaptic protein, in the hippocampus. As compared to their respective controls, EE-exposed males exhibited parallel increases in phosphorylated Tau and the GluN2B receptor, whereas females responded to EE with reduced hippocampal levels of glutamate and GluN2B. Together, these observations provide further evidence on the differential effects of EE on markers of hippocampal neuroplasticity in males and females.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece; First Department of Psychiatry, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - I Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal; Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - D Besinis
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | - E L Tzouveka
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece
| | | | - N Sousa
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal; ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - C Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens 11527, Greece.
| |
Collapse
|
48
|
Training Microglia to Resist Alzheimer's Disease. J Neurosci 2018; 37:477-479. [PMID: 28100733 DOI: 10.1523/jneurosci.3345-16.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022] Open
|
49
|
Hase Y, Craggs L, Hase M, Stevenson W, Slade J, Chen A, Liang D, Ennaceur A, Oakley A, Ihara M, Horsburgh K, Kalaria RN. The effects of environmental enrichment on white matter pathology in a mouse model of chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2018; 38:151-165. [PMID: 28273725 PMCID: PMC5757440 DOI: 10.1177/0271678x17694904] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
White matter (WM) disintegration is common in the older population and is associated with vascular cognitive impairment (VCI). This study explored the effects of environmental enrichment (EE) on pathological sequelae in a mouse model of chronic cerebral hypoperfusion induced by bilateral common carotid artery stenosis (BCAS). Male C57BL/6 J mice underwent BCAS or sham surgery. One-week after surgery, mice were exposed to three different degrees of EE; either standard housing conditions (std), limited 3 h exposure to EE per day (3 h) or full-time exposure to EE (full) for 12 weeks. At 13 weeks after surgery, cognitive testing was performed using a three-dimensional 9-arm radial maze. At 16 weeks after surgery, nesting ability was assessed in each mouse immediately before euthanasia. Brains retrieved after perfusion fixation were examined for WM pathology. BCAS caused WM changes, as demonstrated by corpus callosum atrophy and greater WM disintegrity. BCAS also caused impaired nesting ability and cognitive function. These pathological changes and working memory deficits were attenuated, more so by limited rather than full-time exposure to EE regime. Our results suggest that limited exposure to EE delays the onset of WM degeneration. Therefore, the implementation of even limited EE may be beneficial for patients diagnosed with VCI.
Collapse
Affiliation(s)
- Yoshiki Hase
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Lucinda Craggs
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Mai Hase
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - William Stevenson
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Janet Slade
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Aiqing Chen
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Di Liang
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Abdel Ennaceur
- 2 Department of Pharmacy, Sunderland Pharmacy School, University of Sunderland, Sunderland, UK
| | - Arthur Oakley
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- 3 Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Centre, Osaka, Japan
| | - Karen Horsburgh
- 4 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Raj N Kalaria
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
50
|
Cao M, Hu PP, Zhang YL, Yan YX, Shields CB, Zhang YP, Hu G, Xiao M. Enriched physical environment reverses spatial cognitive impairment of socially isolated APPswe/PS1dE9 transgenic mice before amyloidosis onset. CNS Neurosci Ther 2017; 24:202-211. [PMID: 29274291 DOI: 10.1111/cns.12790] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/03/2017] [Accepted: 12/04/2017] [Indexed: 12/31/2022] Open
Abstract
AIMS Social isolation increases the onset of Alzheimer's disease (AD). Environmental enrichment, a complicated social and physical construct, plays beneficial effects on brain plasticity and function. This study was designed to determine whether physical enrichment can reduce the deleterious consequences of social isolation on the onset of AD. METHODS One-month-old APPswe/PS1dE9 transgenic AD model mice were singly housed in the enriched physical environment for 8 weeks and then received behavioral tests, neuropathological analyses, and Western blot of the hippocampus. RESULTS The enriched physical environment reversed spatial cognitive decline of socially isolated APPswe/PS1dE9 mice. The functional reversal was associated with decreases in cellular apoptosis, synaptic protein loss, inflammation, and glial activation in the hippocampus, without changes in amyloid β neuropathology. CONCLUSION These results suggest that the enriched physical environment may serve as a nonpharmacological intervention for delaying the onset of AD accompanied with social isolation.
Collapse
Affiliation(s)
- Min Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Key Laboratory for Aging & Disease, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, China
| | - Pan-Pan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yan-Li Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yi-Xin Yan
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | | | | | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Key Laboratory for Aging & Disease, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Gerontology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|