1
|
Biose IJ, Chastain WH, Solch-Ottaiano RJ, Grayson VS, Wang H, Banerjee S, Bix GJ. The Effects of Physical Activity on Experimental Models of Vascular Dementia: A Systematic Review and Meta-Analysis. Ann Neurosci 2024; 31:204-224. [PMID: 39156626 PMCID: PMC11325693 DOI: 10.1177/09727531231192759] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/16/2023] [Indexed: 08/20/2024] Open
Abstract
Background Physical activity is associated with improved brain health and cognition in humans. However, the validity, range, and quality of evidence for the beneficial outcomes linked to exercise in experimental models of vascular dementia (VaD) have not been evaluated. We performed a systematic review and meta-analysis of studies that assessed the effect of exercise intervention on models of VaD to provide an unbiased and comprehensive determination of the cognitive function and brain morphology benefits of exercise. Summary A systematic search in three databases as well as study design characteristics and experimental data extraction were completed in December 2021. We investigated the effects of exercise on cognitive function and brain-morphology outcomes in VaD models. Twenty-five studies were included for systematic review, while 21 studies were included in the meta-analysis. These studies included seven models of VaD in rats (60%, 15 studies), mice (36%, 9 studies), and pigs (4%, 1 study). None of the included studies used aged animals, and the majority of studies (80%) used only male animals. Key Message Exercise improves cognition but increased neuro-inflammation in VaD models Exercise improved cognitive function as well as some markers of brain morphology in models of VaD. However, exercise increased anxiety and neuro-inflammatory signals in VaD models. Further, we observed increased reporting anomalies such as a lack of blinding to group treatment or data analysis and randomization of animals to groups. Our report could help in the appropriate design of experimental studies seeking to investigate the effects of exercise as a non-pharmacological intervention on VaD models with a high translational impact.
Collapse
Affiliation(s)
- Ifechukwude J. Biose
- Department of Pharmacology and Experimental Therapeutics, Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Rebecca J. Solch-Ottaiano
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Hanyun Wang
- Department of Pharmacology and Experimental Therapeutics, Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Gregory J. Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| |
Collapse
|
2
|
Fan B, Lin J, Luo Q, Liao W, Hao C. Enriched Environment Inhibits Neurotoxic Reactive Astrocytes via JAK2-STAT3 to Promote Glutamatergic Synaptogenesis and Cognitive Improvement in Chronic Cerebral Hypoperfusion Rats. Neurotox Res 2024; 42:22. [PMID: 38564082 DOI: 10.1007/s12640-024-00704-4] [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] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) is a primary contributor to cognitive decline in the elderly. Enriched environment (EE) is proved to improve cognitive function. However, mechanisms involved remain unclear. The purpose of the study was exploring the mechanisms of EE in alleviating cognitive deficit in rats with CCH. To create a rat model of CCH, 2-vessel occlusion (2-VO) surgery was performed. All rats lived in standard or enriched environments for 4 weeks. Cognitive function was assessed using the novel object recognition test and Morris water maze test. The protein levels of glutamatergic synapses, neurotoxic reactive astrocytes, reactive microglia, and JAK2-STAT3 signaling pathway were measured using Western blot. The mRNA levels of synaptic regulatory factors, C1q, TNF-α, and IL-1α were identified using quantitative PCR. Immunofluorescence was used to detect glutamatergic synapses, neurotoxic reactive astrocytes, and reactive microglia, as well as the expression of p-STAT3 in astrocytes in the hippocampus. The results demonstrated that the EE mitigated cognitive impairment in rats with CCH and enhanced glutamatergic synaptogenesis. EE also inhibited the activation of neurotoxic reactive astrocytes. Moreover, EE downregulated microglial activation, levels of C1q, TNF-α and IL-1α and phosphorylation of JAK2 and STAT3. Our results suggest that inhibition of neurotoxic reactive astrocytes may be one of the mechanisms by which EE promotes glutamatergic synaptogenesis and improves cognitive function in rats with CCH. The downregulation of reactive microglia and JAK2-STAT3 signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Bin Fan
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junbin Lin
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qihang Luo
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weijing Liao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Chizi Hao
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
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
|
4
|
Biskupiak Z, Ha VV, Rohaj A, Bulaj G. Digital Therapeutics for Improving Effectiveness of Pharmaceutical Drugs and Biological Products: Preclinical and Clinical Studies Supporting Development of Drug + Digital Combination Therapies for Chronic Diseases. J Clin Med 2024; 13:403. [PMID: 38256537 PMCID: PMC10816409 DOI: 10.3390/jcm13020403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Limitations of pharmaceutical drugs and biologics for chronic diseases (e.g., medication non-adherence, adverse effects, toxicity, or inadequate efficacy) can be mitigated by mobile medical apps, known as digital therapeutics (DTx). Authorization of adjunct DTx by the US Food and Drug Administration and draft guidelines on "prescription drug use-related software" illustrate opportunities to create drug + digital combination therapies, ultimately leading towards drug-device combination products (DTx has a status of medical devices). Digital interventions (mobile, web-based, virtual reality, and video game applications) demonstrate clinically meaningful benefits for people living with Alzheimer's disease, dementia, rheumatoid arthritis, cancer, chronic pain, epilepsy, depression, and anxiety. In the respective animal disease models, preclinical studies on environmental enrichment and other non-pharmacological modalities (physical activity, social interactions, learning, and music) as surrogates for DTx "active ingredients" also show improved outcomes. In this narrative review, we discuss how drug + digital combination therapies can impact translational research, drug discovery and development, generic drug repurposing, and gene therapies. Market-driven incentives to create drug-device combination products are illustrated by Humira® (adalimumab) facing a "patent-cliff" competition with cheaper and more effective biosimilars seamlessly integrated with DTx. In conclusion, pharma and biotech companies, patients, and healthcare professionals will benefit from accelerating integration of digital interventions with pharmacotherapies.
Collapse
Affiliation(s)
- Zack Biskupiak
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| | - Victor Vinh Ha
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| | - Aarushi Rohaj
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
- The Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT 84113, USA
| | - Grzegorz Bulaj
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
5
|
Paton SEJ, Solano JL, Coulombe-Rozon F, Lebel M, Menard C. Barrier-environment interactions along the gut-brain axis and their influence on cognition and behaviour throughout the lifespan. J Psychiatry Neurosci 2023; 48:E190-E208. [PMID: 37253482 PMCID: PMC10234620 DOI: 10.1503/jpn.220218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/01/2023] [Accepted: 03/19/2023] [Indexed: 06/01/2023] Open
Abstract
Environment is known to substantially alter mental state and behaviour across the lifespan. Biological barriers such as the blood-brain barrier (BBB) and gut barrier (GB) are major hubs for communication of environmental information. Alterations in the structural, social and motor environment at different stages of life can influence function of the BBB and GB and their integrity to exert behavioural consequences. Importantly, each of these environmental components is associated with a distinct immune profile, glucocorticoid response and gut microbiome composition, creating unique effects on the BBB and GB. These barrier-environment interactions are sensitive to change throughout life, and positive or negative alterations at critical stages of development can exert long-lasting cognitive and behavioural consequences. Furthermore, because loss of barrier integrity is implicated in pathogenesis of mental disorders, the pathways of environmental influence represent important areas for understanding these diseases. Positive environments can be protective against stress- and age-related damage, raising the possibility of novel pharmacological targets. This review summarizes known mechanisms of environmental influence - such as social interactions, structural complexity and physical exercise - on barrier composition, morphology and development, and considers the outcomes and implications of these interactions in the context of psychiatric disorders.
Collapse
Affiliation(s)
- Sam E J Paton
- From the Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Centre, Université Laval, Québec, Que. (Paton, Solano, Coulombe-Rozon, Lebel, Menard)
| | - José L Solano
- From the Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Centre, Université Laval, Québec, Que. (Paton, Solano, Coulombe-Rozon, Lebel, Menard)
| | - François Coulombe-Rozon
- From the Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Centre, Université Laval, Québec, Que. (Paton, Solano, Coulombe-Rozon, Lebel, Menard)
| | - Manon Lebel
- From the Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Centre, Université Laval, Québec, Que. (Paton, Solano, Coulombe-Rozon, Lebel, Menard)
| | - Caroline Menard
- From the Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Centre, Université Laval, Québec, Que. (Paton, Solano, Coulombe-Rozon, Lebel, Menard)
| |
Collapse
|
6
|
The Molecular Effects of Environmental Enrichment on Alzheimer's Disease. Mol Neurobiol 2022; 59:7095-7118. [PMID: 36083518 PMCID: PMC9616781 DOI: 10.1007/s12035-022-03016-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022]
Abstract
Environmental enrichment (EE) is an environmental paradigm encompassing sensory, cognitive, and physical stimulation at a heightened level. Previous studies have reported the beneficial effects of EE in the brain, particularly in the hippocampus. EE improves cognitive function as well as ameliorates depressive and anxiety-like behaviors, making it a potentially effective neuroprotective strategy against neurodegenerative diseases such as Alzheimer's disease (AD). Here, we summarize the current evidence for EE as a neuroprotective strategy as well as the potential molecular pathways that can explain the effects of EE from a biochemical perspective using animal models. The effectiveness of EE in enhancing brain activity against neurodegeneration is explored with a view to differences present in early and late life EE exposure, with its potential application in human being discussed. We discuss EE as one of the non pharmacological approaches in preventing or delaying the onset of AD for future research.
Collapse
|
7
|
Zhao K, Zeng L, Cai Z, Liu M, Sun T, Li Z, Liu R. RNA sequencing-based identification of the regulatory mechanism of microRNAs, transcription factors, and corresponding target genes involved in vascular dementia. Front Neurosci 2022; 16:917489. [PMID: 36203804 PMCID: PMC9531238 DOI: 10.3389/fnins.2022.917489] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia with uncertain mechanisms and no effective treatments. microRNAs (miRNAs) and transcription factors (TFs) are considered regulatory factors of genes involved in many diseases. Therefore, this work investigated the aberrantly expressed miRNAs, TFs, corresponding target genes, and their co-regulatory networks in the cortex of rats with bilateral common carotid artery occlusion (2VO) to uncover the potential mechanism and biomarkers of VaD. Differentially expressed genes (DEGs), miRNAs (DEMs), and TFs (DETFs) were identified using RNA sequencing, and their interaction networks were constructed using Cytoscape. The results showed that rats with 2VO had declined cognitive abilities and neuronal loss in the cortex than sham rats. DEGs, DEMs, and DETFs were discriminated between rats with 2VO and sham rats in the cortex, as shown by the 13 aberrantly expressed miRNAs, 805 mRNAs, and 63 TFs. The miRNA-TF-target gene network was constructed, showing 523 nodes and 7237 edges. Five miRNAs (miR-5132-5p, miR-764-3p, miR-223-3p, miR-145-5p, and miR-122-5p), ten TFs (Mxi1, Nfatc4, Rxrg, Zfp523, Foxj2, Nkx6-1, Klf4, Klf5, Csrnp1, and Prdm6), and seven target genes (Serpine1, Nedd4l, Pxn, Col1a1, Plec, Trip12, and Tpm1) were chosen as the significant nodes to construct feed-forward loops (FFLs). Gene Ontology and pathway enrichment analysis revealed that these miRNA and TF-associated genes are mostly involved in the PI3K/Akt, neuroactive ligand–receptor interaction, calcium signaling, and Wnt signaling pathways, along with central locations around the cell membrane. They exert functions such as growth factor binding, integrin binding, and extracellular matrix structural constituent, with representative biological processes like vasculature development, cell–substrate adhesion, cellular response to growth factor stimulus, and synaptic transmission. Furthermore, the expression of three miRNAs (miR-145-5p, miR-122-5p, and miR-5132-5p), six TFs (Csrnp1, Klf4, Nfatc4, Rxrg, Foxj2, and Klf5), and five mRNAs (Serpine1, Plec, Nedd4l, Trip12, and Tpm1) were significantly changed in rats with VaD, in line with the outcome of RNA sequencing. In the potential FFL, miR-145-5p directly bound Csrnp1 and decreased its mRNA expression. These results might help the understanding of the underlying regulatory mechanisms of miRNA-TF-genes, providing potential therapeutic targets in VaD.
Collapse
|
8
|
Effects of Housing and Management Systems on the Growth, Immunity, Antioxidation, and Related Physiological and Biochemical Indicators of Donkeys in Cold Weather. Animals (Basel) 2022; 12:ani12182405. [PMID: 36139265 PMCID: PMC9494980 DOI: 10.3390/ani12182405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
The study was designed with a 2 × 2 factorial experiment to evaluate the effects of growth performance, immune function, antioxidant status, blood biochemical indexes, and hormone levels of donkeys in different housing and management systems in cold weather. Twenty-four male donkeys with similar body weight and age were randomly allocated into four treatment groups that were as follows: a cold-water-drinking group without a windproof facility, a lukewarm-water-drinking group without windproof facilities, a cold-water-drinking group with a windproof facility, and a lukewarm-water-drinking group with a windproof facility. The experiment lasted for 42 days. The results showed that windproof facilities increased average daily gain (ADG) and decreased average daily feed intake (ADFI) and feed-to-gain ratio (F:G) at all time periods (p < 0.01) of the experiment. Windproof facilities increased the digestibility of dry matter (DM), crude fat (CF), crude protein (CP), ash, calcium (Ca), and phosphate (P) on day 21 (p < 0.01), and increased the digestibility of DM, CF, ash, and P on day 42 (p < 0.01). The respiration rate and the skin temperature of the abdomen and legs increased (p < 0.05) and rectal temperature tended to increase (p = 0.083) by adopting windproof facilities at 07:00; the windproof facilities tended to increase the skin temperature of the ears and abdomen (p = 0.081, p = 0.091) at 14:00. For the blood parameters, with windproof facilities, the concentrations of total protein (TP), blood urea nitrogen (BUN), triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C) increased (p < 0.05) and glucose (GLU) concentration decreased (p < 0.05) at 07:00 on day 21; the concentrations of TG and cholesterol (CHO) increased and the concentrations of TP, BUN, and GLU decreased at 07:00 on day 42 (p < 0.05). The concentrations of adrenocorticotropic hormone (ACTH), cortisol (COR), triiodothyronine (T3), and thyroxine (T4) decreased (p < 0.05) at 07:00 on day 21, and T4 concentration decreased (p < 0.05) at 07:00 on day 42. The concentrations of interleukin-4 (IL-4), immunoglobulin A (IgA), immunoglobulin G (IgG), and immunoglobulin M (IgM) increased (p < 0.01) and the concentrations of interleukin-1β (IL-1β) and tumor necrosis factor-alpha (TNF-α) decreased (p < 0.01) on days 21 and 42. The activities of total superoxide dismutase (T-SOD) and glutathione peroxidase (GPx) increased (p < 0.05), and malondialdehyde (MDA) concentration decreased (p < 0.01) on day 21; the activities of T-SOD and catalase (CAT) increased (p < 0.05), and MDA concentration decreased (p < 0.01) on day 42. However, under the conditions of this experiment, water temperature did not affect the above indexes on days 21 and 42. These results indicated that adopting windproof facilities in a cold climate can mitigate the effects of atrocious weather on the production performance of donkeys.
Collapse
|
9
|
Sun M, Wu L, Chen G, Mo X, Shi C. Hemodynamic changes and neuronal damage detected by 9.4 T MRI in rats with chronic cerebral ischemia and cognitive impairment. Brain Behav 2022; 12:e2642. [PMID: 35687797 PMCID: PMC9304847 DOI: 10.1002/brb3.2642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The bilateral common carotid artery occlusion (BCCAO) rat model is an ideal animal model for simulating the pathology of chronic brain hypoperfusion in humans. However, dynamic changes in neuronal activity, cellular edema, and neuronal structural integrity in vivo after BCCAO have rarely been reported. The purpose of this study is to use a 9.4 T MRI to explore the pathophysiological mechanisms of vascular dementia. MATERIALS AND METHODS Twelve Sprague-Dawley (SD) rats were randomly divided into two groups: the sham group and the model group (n = 6 for each group). Rats were subjected to MRI using T2*WI, diffusion tensor imaging (DTI), and DWI sequences by MRI at the following six time points: presurgery and 6 h, 3 days, 7 days, 21 days, and 28 days postsurgery. Then, the T2*, fractional anisotropy (FA), and average apparent diffusion coefficient (ADC) values were measured in the bilateral cortices and hippocampi. After MRI scanning, all rats in both groups were subjected to the Y-maze test, novel object recognition test, and open-field test to assess their learning, memory, cognition, and locomotor activity. RESULTS The T2*, FA, and ADC values in the cerebral cortex and hippocampus decreased sharply at 6 h after BCCAO in the model group compared with those of the sham group. By Day 28, the T2* and ADC values gradually increased to close to those in the sham group, but the FA values changed little, and the rats in the model group had worse learning, memory, and cognition and less locomotor activity than the rats in the sham group. CONCLUSIONS The BCCAO is an ideal rat model for studying the pathophysiological mechanisms of vascular dementia.
Collapse
Affiliation(s)
- Minghua Sun
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, No 613 Huangpu Dadao West, Guangzhou, People's Republic of China.,Department of Radiology, The Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, People's Republic of China.,The Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, Guangzhou, Guangdong Province, People's Republic of China
| | - Liangmiao Wu
- Institute of New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Moderation and innovative Drug Development of Chinese Ministry of Education, Jinan University College of Pharmacy, Guangzhou, People's Republic of China.,Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guangying Chen
- Institute of New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Moderation and innovative Drug Development of Chinese Ministry of Education, Jinan University College of Pharmacy, Guangzhou, People's Republic of China
| | - Xukai Mo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, No 613 Huangpu Dadao West, Guangzhou, People's Republic of China
| | - Changzheng Shi
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, No 613 Huangpu Dadao West, Guangzhou, People's Republic of China.,The Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
10
|
Enriched Environment Effects on Myelination of the Central Nervous System: Role of Glial Cells. Neural Plast 2022; 2022:5766993. [PMID: 35465398 PMCID: PMC9023233 DOI: 10.1155/2022/5766993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/20/2022] [Accepted: 03/09/2022] [Indexed: 12/24/2022] Open
Abstract
Myelination is regulated by various glial cells in the central nervous system (CNS), including oligodendrocytes (OLs), microglia, and astrocytes. Myelination of the CNS requires the generation of functionally mature OLs from OPCs. OLs are the myelin-forming cells in the CNS. Microglia play both beneficial and detrimental roles during myelin damage and repair. Astrocyte is responsible for myelin formation and regeneration by direct interaction with oligodendrocyte lineage cells. These glial cells are influenced by experience-dependent activities such as environmental enrichment (EE). To date, there are few studies that have investigated the association between EE and glial cells. EE with a complex combination of sensorimotor, cognitive, and social stimulation has a significant effect on cognitive impairment and brain plasticity. Hence, one mechanism through EE improving cognitive function may rely on the mutual effect of EE and glial cells. The purpose of this paper is to review recent research into the efficacy of EE for myelination and glial cells at cellular and molecular levels and offers critical insights for future research directions of EE and the treatment of EE in cognitive impairment disease.
Collapse
|
11
|
Pamidi N, Yap CG, Nayak N. Environmental enrichment preserves hippocampal neurons in diabetes and stressed rats. Histol Histopathol 2022; 37:385-395. [PMID: 34978332 DOI: 10.14670/hh-18-418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
This study evaluated the effect of Environmental Enrichment (EE) on neuron morphology in the CA1, CA3 and dentate hilus (DH) regions of the hippocampus by quantitating the total dendritic arborizations. EE is a potential intervention for stress and diabetes. It is capable of mitigating diabetes and stress-induced cognitive and memory deficit. Diabetes and stress were induced in male Wistar rats (4-5 weeks). Diabetic and stressed rats were exposed to EE on Day 2 post STZ injection and subsequently once daily for 30 days. All animals were sacrificed on Day 30. The hippocampus was dissected and processed for Golgi staining to quantitate dendritic arborizations at the CA1, CA2 and DH regions. Diabetes (D) and Diabetes+stress (D+S) groups had significantly fewer apical and basal dendritic branching points (ADBP, BDBP) at CA1 (p<0.01), CA3 (p<0.001) and DH (p<0.001) relative to control group (NC). Diabetes and stressed rats exposed to EE: [D+EE and D+S+EE groups] exhibited significantly denser ADBP and BDBP at all regions relative to D (p<0.001) and (D+S+EE) (p<0.001) groups respectively. EE significantly preserved neuronal arborizations in hippocampus of diabetic and stressed rats, suggesting a potential entity of diabetes and stress management.
Collapse
Affiliation(s)
- N Pamidi
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Kuala Lumpur, Malaysia
| | - C G Yap
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Kuala Lumpur, Malaysia.
| | - N Nayak
- Department of Anatomy, Melaka Manipal Medical College, Manipal University, Manipal, Karnataka, India
| |
Collapse
|
12
|
Zhang X, Shi X, Wang J, Xu Z, He J. Enriched environment remedies cognitive dysfunctions and synaptic plasticity through NMDAR-Ca 2+-Activin A circuit in chronic cerebral hypoperfusion rats. Aging (Albany NY) 2021; 13:20748-20761. [PMID: 34462377 PMCID: PMC8436900 DOI: 10.18632/aging.203462] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022]
Abstract
Chronic cerebral ischemia (CCI) is one of the critical factors in the occurrence and development of vascular cognitive impairment (VCI). Apoptosis of nerve cells and changes in synaptic activity after CCI are the key factors to induce VCI. Synaptic stimulation up-regulates intraneuronal Ca2+ level through N-methyl-D-aspartic acid receptor (NMDAR) via induction of the activity-regulated inhibitor of death (AID) expression to produce active-dependent neuroprotection. Moreover, the regulation of synaptic plasticity could improve cognition and learning ability. Activin A (ActA), an exocrine protein of AID, can promote NMDAR phosphorylation and participate in the regulation of synaptic plasticity. We previously found that exogenous ActA can improve the cognitive function of rats with chronic cerebral ischemia and enhance the oxygenated glucose deprivation of intracellular Ca2+ level. In addition to NMDAR, the Wnt pathway is critical in the positive regulation of LTP through activation or inhibition. It plays an essential role in synaptic transmission and activity-dependent synaptic plasticity. The enriched environment can increase ActA expression during CCI injury. We speculated that the NMDAR-Ca2+-ActA signal pathway has a loop-acting mode, and the environmental enrichment could improve chronic cerebral ischemia cognitive impairment via NMDAR-Ca2+-ActA, Wnt/β-catenin pathway is involved in this process. For the hypothesis verification, this study intends to establish chronic cerebral hypoperfusion (CCH) rat model, explore the improvement effect of enriched environment on VCI, detect the changes in plasticity of synaptic morphology and investigate the regulatory mechanism NMDAR-Ca2+-ActA-Wnt/β-catenin signaling loop, providing a therapeutic method for the treatment of CCH.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Zhuang Y, Wang X. Humanin-S14G Ameliorates Vascular Dementia Through Regulating miR-134. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Humanin-S14G is a synthetic derivative of Humanin with neuroprotective effects. miR-134 involves in the regulation of the nervous system. However, whether Humanin-S14G ameliorates VD through miR-134 remains poorly understood. Healthy male rats were assigned into sham group; VD group
and Humanin-S14G group followed by analysis of learning ability by the Morris water maze test, expression of miR-134, Bcl-2 and Bax by Real time PCR, BDNF protein level by Western blot, IL-6 secretion by ELISA as well as pathological changes of hippocampal nerve region by HE staining. In VD
model group, the learning and cognitive ability of the rats was significantly decreased and miR- 134 and IL-6 was significantly upregulated along with downregulated Bcl-2 and BDNF and upregulated Bax expression compared to sham group (P <0.05). Humanin-S14G significantly improved
the learning and cognitive ability of VD model rats, decreased miR-134 and IL-6 level, increased Bcl-2 and BDNF expression, as well as inhibited Bax expression (P <0.05) and nerve damage was significantly improved. Humanin-S14G regulates miR-134 expression in the brain tissue of
VD rats, promote the expression of BDNF, regulate cell apoptosis, inhibit inflammation, improve the learning function of vascular dementia, and delay the occurrence and development of vascular dementia.
Collapse
Affiliation(s)
- Yuan Zhuang
- Linyi Social Welfare Institute, Linyi, Shandong, 276000, China
| | - Xutang Wang
- Neurosurgery of Junan People’s Hospital, Junan Shandong, 276600, China
| |
Collapse
|
14
|
Lin H, Jin T, Chen L, Dai Y, Jia W, He X, Yang M, Li J, Liang S, Wu J, Huang J, Chen L, Liu W, Tao J. Longitudinal tracing of neurochemical metabolic disorders in working memory neural circuit and optogenetics modulation in rats with vascular cognitive impairment. Brain Res Bull 2021; 170:174-186. [PMID: 33600886 DOI: 10.1016/j.brainresbull.2021.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 11/30/2022]
Abstract
Chronic cerebral ischemia leads to vascular cognitive impairment (VCI) that exacerbates along with ischemia time and eventually develops into dementia. Recent advances in molecular neuroimaging contribute to understand its pathological characteristics. We previously traced the anisotropic diffusion of water molecules suggests that chronic cerebral ischemia leads to irreversible progressive damage to white matter integrity. However, the abnormalities of gray matter activity following chronic cerebral ischemia remains not entirely understood. In this study, in vivo hydrogen proton magnetic resonance spectroscopy (1H-MRS) was applied to longitudinally track the neurochemical metabolic disorder of gray matter associated with working memory, and optogenetics modulation of neurochemical metabolism was performed for targeted treatment of VCI. The results showed that the concentration of N-acetylaspartate (NAA) in the right hippocampus, left hippocampus, right medial prefrontal cortex (mPFC) and mediodorsal thalamus was decreased as early as 7 days after chronic cerebral ischemia, subsequently gamma-aminobutyric acid (GABA) declined whereas myo-inositol (mI) and glutamate (Glu) increased at 14 days, as well as choline (Cho) lost at 28 days, concurrently the change of Glu and GABA in the mPFC and hippocampus was ischemia time-dependent manner within 1 month. Behaviorally, working memory and object recognition memory were impaired at 14 days, 28 days that significantly correlated with neurochemical metabolic disorders. Interestingly, using optogenetics modulation of PV neurons in the mPFC, the metabolic abnormalities of NAA and GABA in working memory neural circuit could be repaired after chronic cerebral ischemia, together with behavior improvements. These findings suggested that as early as 1∼4 weeks after chronic cerebral ischemia, the metabolism of NAA, Glu, mI and Cho was synchronously impaired in neural circuit of hippocampus-mediodorsal thalamus-mPFC, and the loss of GABA delayed in the hippocampus, and optogenetics modulation of parvalbumin (PV) neurons in the mPFC can improve the neurochemical metabolism of working memory neural circuit and enhance working memory.
Collapse
Affiliation(s)
- Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Tingting Jin
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Lewen Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Weiwei Jia
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Xiaojun He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Minguang Yang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Jianhong Li
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Shengxiang Liang
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Jinsong Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Lidian Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China
| | - Weilin Liu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China.
| | - Jing Tao
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology & Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China; Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, PR China.
| |
Collapse
|
15
|
Xu GJ, Zhang Q, Li SY, Zhu YT, Yu KW, Wang CJ, Xie HY, Wu Y. Environmental enrichment combined with fasudil treatment inhibits neuronal death in the hippocampal CA1 region and ameliorates memory deficits. Neural Regen Res 2021; 16:1460-1466. [PMID: 33433459 PMCID: PMC8323697 DOI: 10.4103/1673-5374.303034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Currently, no specific treatment exists to promote recovery from cognitive impairment after a stroke. Dysfunction of the actin cytoskeleton correlates well with poststroke cognitive declines, and its reorganization requires proper regulation of Rho-associated kinase (ROCK) proteins. Fasudil downregulates ROCK activation and protects neurons against cytoskeleton collapse in the acute phase after stroke. An enriched environment can reduce poststroke cognitive impairment. However, the efficacy of environmental enrichment combined with fasudil treatment remains poorly understood. A photothrombotic stroke model was established in 6-week-old male C57BL/6 mice. Twenty-four hours after modeling, these animals were intraperitoneally administered fasudil (10 mg/kg) once daily for 14 successive days and/or provided with environmental enrichment for 21 successive days. After exposure to environmental enrichment combined with fasudil treatment, the number of neurons in the hippocampal CA1 region increased significantly, the expression and proportion of p-cofilin in the hippocampus decreased, and the distribution of F-actin in the hippocampal CA1 region increased significantly. Furthermore, the performance of mouse stroke models in the tail suspension test and step-through passive avoidance test improved significantly. These findings suggest that environmental enrichment combined with fasudil treatment can ameliorate memory dysfunction through inhibition of the hippocampal ROCK/cofilin pathway, alteration of the dynamic distribution of F-actin, and inhibition of neuronal death in the hippocampal CA1 region. The efficacy of environmental enrichment combined with fasudil treatment was superior to that of fasudil treatment alone. This study was approved by the Animal Ethics Committee of Fudan University of China (approval No. 2019-Huashan Hospital JS-139) on February 20, 2019.
Collapse
Affiliation(s)
- Gao-Jing Xu
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qun Zhang
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Si-Yue Li
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi-Tong Zhu
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke-Wei Yu
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuan-Jie Wang
- Department of Rehabilitation Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Hong-Yu Xie
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yi Wu
- Department of rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Yun SJ, Kang MG, Yang D, Choi Y, Kim H, Oh BM, Seo HG. Cognitive Training Using Fully Immersive, Enriched Environment Virtual Reality for Patients With Mild Cognitive Impairment and Mild Dementia: Feasibility and Usability Study. JMIR Serious Games 2020; 8:e18127. [PMID: 33052115 PMCID: PMC7593866 DOI: 10.2196/18127] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/12/2020] [Accepted: 09/16/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Cognitive training using virtual reality (VR) may result in motivational and playful training for patients with mild cognitive impairment and mild dementia. Fully immersive VR sets patients free from external interference and thus encourages patients with cognitive impairment to maintain selective attention. The enriched environment, which refers to a rich and stimulating environment, has a positive effect on cognitive function and mood. OBJECTIVE The aim of this study was to investigate the feasibility and usability of cognitive training using fully immersive VR programs in enriched environments with physiatrists, occupational therapists (OTs), and patients with mild cognitive impairment and mild dementia. METHODS The VR interface system consisted of a commercialized head-mounted display and a custom-made hand motion tracking module. We developed the virtual harvest and cook programs in enriched environments representing rural scenery. Physiatrists, OTs, and patients with mild cognitive impairment and mild dementia received 30 minutes of VR training to evaluate the feasibility and usability of the test for cognitive training. At the end of the test, the usability and feasibility were assessed by a self-report questionnaire based on a 7-point Likert-type scale. Response time and finger tapping were measured in patients before and after the test. RESULTS Participants included 10 physiatrists, 6 OTs, and 11 patients with mild cognitive impairment and mild dementia. The mean scores for overall satisfaction with the program were 5.75 (SD 1.00) for rehabilitation specialists and 5.64 (SD 1.43) for patients. The response time of the dominant hand in patients decreased after the single session of cognitive training using VR, but this was not statistically significant (P=.25). There was no significant change in finger tapping in either the right or left hand (P=.48 and P=.42, respectively). None of the participants reported headaches, dizziness, or any other motion sickness after the test. CONCLUSIONS A fully immersive VR cognitive training program may be feasible and usable in patients with mild cognitive impairment and mild dementia based on the positive satisfaction and willingness to use the program reported by physiatrists, OTs, and patients. Although not statistically significant, decreased response time without a change in finger tapping rate may reflect a temporary increase in attention after the test. Additional clinical trials are needed to investigate the effect on cognitive function, mood, and physical outcomes.
Collapse
Affiliation(s)
- Seo Jung Yun
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Min-Gu Kang
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dongseok Yang
- Department of Computer Engineering, Dankook University, Gyeonggi, Republic of Korea
| | - Younggeun Choi
- Department of Computer Engineering, Dankook University, Gyeonggi, Republic of Korea
| | - Heejae Kim
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
17
|
Keymoradzadeh A, Hedayati Ch M, Abedinzade M, Gazor R, Rostampour M, Taleghani BK. Enriched environment effect on lipopolysaccharide-induced spatial learning, memory impairment and hippocampal inflammatory cytokine levels in male rats. Behav Brain Res 2020; 394:112814. [PMID: 32707137 DOI: 10.1016/j.bbr.2020.112814] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/02/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022]
Abstract
Neuro-inflammation is responsible for cognitive impairments and neurodegenerative diseases such as Alzheimer's disease. In this study, we aimed to investigate the enriched environment (EE) effect on learning and memory impairment as well as on pro-inflammatory cytokines changes induced by lipopolysaccharide (LPS). LPS injection (1 mg/kg/i.p, days 1, 3, 5, and 7) was used to develop the animal model of neuro-inflammation. Twenty-eight male Wistar rats were used in the experiment and randomly divided into 4 groups: 1) sham (S), 2) sham + enriched environment (SE), 3) LPS (L), and 4) LPS + EE (LE). Two different housing conditions, including standard environment (SE) and enriched environment, were used. The Morris Water Maze (MWM) test was used to examine animals learning and memory. IL-1β, IL-10, and TNF-α levels were measured in the brain using ELISA. We found that LPS significantly impaired learning and memory (p < 0.05) in the MWM task, but EE could significantly improve learning and memory impairment (p < 0.05). IL-1 and IL-10 levels dramatically increased in the LPS group (P < 0.05), whereas EE could decrease and increase IL-1β and IL-10 values in the LPS + EE group (P < 0.05), respectively. TNF-α levels were traced but had not detectable values in the hippocampus. Thus, we can conclude that EE has healing effects on LPS induced neuro-inflammation and can improve learning and memory deficit; however, further studies are needed to support the findings of our study.
Collapse
Affiliation(s)
- Arman Keymoradzadeh
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mojtaba Hedayati Ch
- Department of Microbiology, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmood Abedinzade
- Medical Biotechnology Research Center, School of Nursing, Midwifery and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Rohollah Gazor
- Department of Anatomical Sciences, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Rostampour
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Behrooz Khakpour Taleghani
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
18
|
Yang Y, Zhao L, Li N, Dai C, Yin N, Chu Z, Duan X, Niu X, Yan P, Lv P. Estrogen Exerts Neuroprotective Effects in Vascular Dementia Rats by Suppressing Autophagy and Activating the Wnt/β-Catenin Signaling Pathway. Neurochem Res 2020; 45:2100-2112. [PMID: 32719979 DOI: 10.1007/s11064-020-03072-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/23/2020] [Accepted: 06/12/2020] [Indexed: 01/24/2023]
Abstract
Vascular dementia (VD) is a clinical syndrome of acquired cognitive dysfunction caused by various cerebrovascular factors. Estrogen is a steroid hormone involved in promoting neuronal survival and in regulating many signaling pathways. However, the mechanism by which it confers neuroprotective effects in VD remains unclear. Here, we aimed to investigate the effect of estrogen on neuronal injury and cognitive impairment in VD rats. Adult female rats were randomly divided into four groups (sham, model, estrogen early and estrogen later treatment) and received sham surgery or bilateral ovariectomy and permanent occlusion of bilateral common carotid arteries (BCCAO). The early treatment group received daily intraperitoneal injections of 17β-estradiol (100 µg/kg/day) for 8 weeks starting the day after BCCAO. The later treatment group was administered the same starting 1 week after BCCAO. Learning and memory functions were assessed using the Morris water maze. Morphological changes within the hippocampal CA1 region were observed by hematoxylin/eosin staining and electron microscopy. Expression of proteins associated with autophagy and signaling were detected by immunohistochemical staining and Western blot. We found that estrogen significantly alleviated cognitive damage and neuronal injury and reduced the expression of Beclin1 and LC3B, indicating a suppression of autophagy. Moreover, estrogen enhanced expression of β-catenin and Cyclin D1, while reducing glycogen synthase kinase 3β, suggesting activation of Wnt/β-catenin signaling. These results indicate that estrogen ameliorates learning and memory deficiencies in VD rats, and that this neuroprotective effect may be explained by the suppression of autophagy and activation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yanyan Yang
- Department of Neurology, Hebei Medical University, Shijiazhuang, 050017, China.,Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Lei Zhao
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Na Li
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Congwei Dai
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Nan Yin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Zhaoping Chu
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Xiaoyan Duan
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Xiaoli Niu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Ping Yan
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, 050051, China
| | - Peiyuan Lv
- Department of Neurology, Hebei General Hospital, Shijiazhuang, 050051, China.
| |
Collapse
|
19
|
Schoentgen B, Gagliardi G, Défontaines B. Environmental and Cognitive Enrichment in Childhood as Protective Factors in the Adult and Aging Brain. Front Psychol 2020; 11:1814. [PMID: 32793081 PMCID: PMC7385286 DOI: 10.3389/fpsyg.2020.01814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
Some recent studies have highlighted a link between a favorable childhood environment and the strengthening of neuronal resilience against the changes that occur in natural aging neurodegenerative disease. Many works have assessed the factors – both internal and external – that can contribute to delay the phenotype of an ongoing neurodegenerative brain pathology. At the crossroads of genetic, environmental and lifestyle factors, these relationships are unified by the concept of cognitive reserve (CR). This review focuses on the protective effects of maintaining this CR through the cognitive aging process, and emphasizes the most essential time in life for the development and strengthening of this CR. The in-depth study of this research shows that early stimulation with regard to social and sensory interactions, contributes to the proper development of cognitive, affective and psychosocial capacities. Childhood thus appears to be the most active phase in the development of CR, and as such we hypothesize that this constitutes the first essential period of primary prevention of pathological aging and loss of cognitive capacities. If this hypothesis is correct, early stimulation of the environment would therefore be considered as a true primary prevention and a public health issue. The earlier identification of neurodevelopmental disorders, which can affect personal and professional development across the lifespan, could therefore have longer-term impacts and provide better protection against aging.
Collapse
Affiliation(s)
- Bertrand Schoentgen
- Réseau Aloïs Pôle Enfant (Pediatric Aloïs Network), Paris, France.,Réseau Aloïs (Aloïs Network), Paris, France
| | - Geoffroy Gagliardi
- Réseau Aloïs (Aloïs Network), Paris, France.,UPMC Univ Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle (ICM) - Hôpital Pitié-Salpêtrière, Sorbonne Universités, Paris, France
| | - Bénédicte Défontaines
- Réseau Aloïs Pôle Enfant (Pediatric Aloïs Network), Paris, France.,Réseau Aloïs (Aloïs Network), Paris, France
| |
Collapse
|
20
|
Hsu WL, Ma YL, Liu YC, Tai DJC, Lee EHY. Restoring Wnt6 signaling ameliorates behavioral deficits in MeCP2 T158A mouse model of Rett syndrome. Sci Rep 2020; 10:1074. [PMID: 31974426 PMCID: PMC6978308 DOI: 10.1038/s41598-020-57745-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/09/2019] [Indexed: 01/23/2023] Open
Abstract
The methyl-CpG-binding protein 2 gene, MECP2, is an X chromosome-linked gene encoding the MeCP2 protein, and mutations of MECP2 cause Rett syndrome (RTT). Previous study has shown that re-expression of SUMO-modified MeCP2 in Mecp2-null neurons rescues synaptic and behavioral deficits in Mecp2 conditional knockout mice, whereas about 12-fold decrease in Wnt6 mRNA level was found in MeCP2K412R sumo-mutant mice. Here, we examined the role of Wnt6 in MeCP2 T158A mouse model of RTT. Results show that lentiviral delivery of Wnt6 to the amygdala ameliorates locomotor impairment and social behavioral deficits in these animals. MeCP2 T158A mice show decreased level of GSK-3β phosphorylation and increased level of β-catenin phosphorylation. They also show reduced level of MeCP2 SUMOylation. These alterations were also restored by lenti-Wnt6 transduction. Further, both BDNF and IGF-1 expressions are decreased in MeCP2 T158A mice. Overexpression of Wnt6 increases Bdnf and Igf-1 promoter activity in HEK293T cells in a dose-dependent manner. Lenti-Wnt6 transduction to the amygdala similarly increases the mRNA level and protein expression of BDNF and IGF-1 in MeCP2 T158A mice. Moreover, environmental enrichment (EE) similarly ameliorates the locomotor and social behavioral deficits in MeCP2 T158A mice. One of the mechanisms underlying EE is mediated through enhanced MeCP2 SUMOylation and increased Wnt6 expression in these animals by EE.
Collapse
Affiliation(s)
- Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yen-Chen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Derek J C Tai
- Molecular Neurogenetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eminy H Y Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
21
|
Alquezar C, Felix JB, McCandlish E, Buckley BT, Caparros-Lefebvre D, Karch CM, Golbe LI, Kao AW. Heavy metals contaminating the environment of a progressive supranuclear palsy cluster induce tau accumulation and cell death in cultured neurons. Sci Rep 2020; 10:569. [PMID: 31953414 PMCID: PMC6969162 DOI: 10.1038/s41598-019-56930-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022] Open
Abstract
Progressive supranuclear palsy (PSP) is a neurodegenerative disorder characterized by the presence of intracellular aggregates of tau protein and neuronal loss leading to cognitive and motor impairment. Occurrence is mostly sporadic, but rare family clusters have been described. Although the etiopathology of PSP is unknown, mutations in the MAPT/tau gene and exposure to environmental toxins can increase the risk of PSP. Here, we used cell models to investigate the potential neurotoxic effects of heavy metals enriched in a highly industrialized region in France with a cluster of sporadic PSP cases. We found that iPSC-derived iNeurons from a MAPT mutation carrier tend to be more sensitive to cell death induced by chromium (Cr) and nickel (Ni) exposure than an isogenic control line. We hypothesize that genetic variations may predispose to neurodegeneration induced by those heavy metals. Furthermore, using an SH-SY5Y neuroblastoma cell line, we showed that both heavy metals induce cell death by an apoptotic mechanism. Interestingly, Cr and Ni treatments increased total and phosphorylated tau levels in both cell types, implicating Cr and Ni exposure in tau pathology. Overall, this study suggests that chromium and nickel could contribute to the pathophysiology of tauopathies such as PSP by promoting tau accumulation and neuronal cell death.
Collapse
Affiliation(s)
- Carolina Alquezar
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA
| | - Jessica B Felix
- Graduate Program, Department of Molecular and Cellular Biology Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | - Brian T Buckley
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, 170, Frelinghuysen Road Piscataway NJ, 08854, New Brunswick, NJ, United States
| | | | - Celeste M Karch
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, 63110, USA
| | - Lawrence I Golbe
- Division of Movement Disorders. Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Aimee W Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, 94158, USA.
| |
Collapse
|
22
|
Protection of blood-brain barrier as a potential mechanism for enriched environments to improve cognitive impairment caused by chronic cerebral hypoperfusion. Behav Brain Res 2019; 379:112385. [PMID: 31778736 DOI: 10.1016/j.bbr.2019.112385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/27/2019] [Accepted: 11/24/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a common pathophysiological basis for Alzheimer's Disease and vascular dementia in the early stages. It has been confirmed that blood-brain barrier (BBB) destruction is a key factor in CCH-related cognitive impairment. Here we explored the effects of an enriched environment (EE) intervention on CCH-induced BBB destruction and cognitive impairment, and the underlying mechanism. METHODS Rats in the EE group were exposed to an EE, while the standard environment (SE) group was maintained in a standard cage with bedding but no other objects. On day 14, CCH was induced via permanent bilateral common carotid artery occlusion (2VO). Next, Evans blue (EB) leakage in the hippocampus was measured by chemical colorimetry to dynamically evaluate BBB permeability. On day 28, the BBB ultrastructure was observed using transmission electron microscopy. The expression levels of BBB integrity-related proteins, matrix metalloproteinases-2/-9 (MMP-2/-9), and the classical Wnt/β-catenin signaling pathway-related proteins were detected using western-blotting techniques. On day 43, cognitive function was assessed using the Morris water maze. RESULTS After 2VO, CCH rats exposed to the SE developed obvious cognitive impairment and BBB destruction. BBB damage was manifested through increased EB leakage, ultrastructural destruction, degradation of BBB integrity-related proteins, and up-regulation of MMP-2/-9. These changes were significantly alleviated after the EE intervention. In addition, EEs activated the Wnt/β-catenin signaling pathway in the hippocampus of rats. CONCLUSIONS These results suggest that protection of the BBB may be a novel mechanism by which EEs ameliorate CCH-induced cognitive impairment, and this effect may be related to the activation of the Wnt/β-catenin pathway.
Collapse
|
23
|
Xu J, Qi Q, Lv P, Dong Y, Jiang X, Liu Z. Oxiracetam ameliorates cognitive deficits in vascular dementia rats by regulating the expression of neuronal apoptosis/autophagy-related genes associated with the activation of the Akt/mTOR signaling pathway. ACTA ACUST UNITED AC 2019; 52:e8371. [PMID: 31721903 PMCID: PMC6853072 DOI: 10.1590/1414-431x20198371] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/04/2019] [Indexed: 11/25/2022]
Abstract
Oxiracetam (ORC) is a commonly used nootropic drug for improving cognition and memory impairments. The therapeutic effect and underlying mechanism of ORC in vascular dementia (VaD) treatment remain unknown. In this study, 3-month-old male Sprague-Dawley rats with permanent bilateral common carotid artery occlusion-induced VaD were treated orally with low (100 mg/kg) or high (200 mg/kg) dose ORC once a day for 4 weeks. The results of the Morris water maze test and Nissl staining showed that ORC treatment significantly alleviated learning and memory deficits and neuronal damage in rats with VaD. Mechanistically, the protein levels of a panel of genes associated with neuronal apoptosis (Bcl-2, Bax) and autophagy (microtubule-associated protein 1 chain 3, Beclin1, p62) were significantly altered by ORC treatment compared with VaD, suggesting a protective role of ORC against VaD-induced neuronal apoptosis and autophagy. Moreover, the Akt/mTOR pathway, which is known to be the upstream signaling governing apoptosis and autophagy, was found to be activated in ORC-treated rats, suggesting an involvement of Akt/mTOR activation in ORC-rendered protection in VaD rats. Taken together, this study demonstrated that ORC may alleviate learning and memory impairments and neuronal damage in VaD rats by altering the expression of apoptosis/autophagy-related genes and activation of the Akt/mTOR signaling pathway in neurons.
Collapse
Affiliation(s)
- Jing Xu
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| | - Qianqian Qi
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| | - Peiyuan Lv
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| | - Xin Jiang
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| | - Zhijuan Liu
- Department of Neurology, Hebei General Hospital, Shijiazhuang City, Hebei Province, China
| |
Collapse
|
24
|
Gelfo F. Does Experience Enhance Cognitive Flexibility? An Overview of the Evidence Provided by the Environmental Enrichment Studies. Front Behav Neurosci 2019; 13:150. [PMID: 31338030 PMCID: PMC6629767 DOI: 10.3389/fnbeh.2019.00150] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/21/2019] [Indexed: 12/16/2022] Open
Abstract
Neuroplasticity accounts for the ability of the brain to change in both structure and function in consequence of life experiences. An enhanced stimulation provided by the environment is able to create a form of brain, neural, and cognitive reserve, which allows an individual to cope better with the environmental demands, also in case of neural damage leading to cognitive decline. With its complex manipulation of several stimuli, the animal experimental paradigm of environmental enrichment (EE) appears particularly effective in modulating the ability to successfully respond to the ever-changing characteristics of the environment. According to this point, it could be very relevant to analyze the specific effects of EE on cognitive flexibility (CF). CF could be defined as the ability to effectively change behavior in response to the environmental condition changing. This review article is specifically aimed to summarize and focus on the available evidence in relation to the effects of EE on CF. To this aim, findings obtained in behavioral tasks specifically structured to investigate animal CF, such as reversal learning and attentional set-shifting tests (tasks based on the request of responding to a rewarding rule that changes, within one or multiple perceptual dimensions), are reviewed. Data provided on the structural and biochemical correlates of these findings are also enumerated. Studies realized in healthy animals and also in pathological models are considered. On the whole, the summarized evidence clearly supports the specific beneficial effects of EE on CF. However, further studies on this key topic are strictly required to gain a comprehensive and detailed framework on the mechanisms by which an enhanced stimulation could improve CF.
Collapse
Affiliation(s)
- Francesca Gelfo
- Department of Human Sciences, Guglielmo Marconi University, Rome, Italy.,Department of Clinical and Behavioural Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
25
|
Llorens-Martín M. Exercising New Neurons to Vanquish Alzheimer Disease. Brain Plast 2018; 4:111-126. [PMID: 30564550 PMCID: PMC6296267 DOI: 10.3233/bpl-180065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer disease (AD) is the most common type of dementia in individuals over 65 years of age. The neuropathological hallmarks of the condition are Tau neurofibrillary tangles and Amyloid-β senile plaques. Moreover, certain susceptible regions of the brain experience a generalized lack of neural plasticity and marked synaptic alterations during the progression of this as yet incurable disease. One of these regions, the hippocampus, is characterized by the continuous addition of new neurons throughout life. This phenomenon, named adult hippocampal neurogenesis (AHN), provides a potentially endless source of new synaptic elements that increase the complexity and plasticity of the hippocampal circuitry. Numerous lines of evidence show that physical activity and environmental enrichment (EE) are among the most potent positive regulators of AHN. Given that neural plasticity is markedly decreased in many neurodegenerative diseases, the therapeutic potential of making certain lifestyle changes, such as increasing physical activity, is being recognised in several non-pharmacologic strategies seeking to slow down or prevent the progression of these diseases. This review article summarizes current evidence supporting the putative therapeutic potential of EE and physical exercise to increase AHN and hippocampal plasticity both under physiological and pathological circumstances, with a special emphasis on neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- María Llorens-Martín
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa”, CBMSO, CSIC-UAM, Madrid, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases CIBERNED, Madrid, Spain
- Department of Molecular Biology, Faculty of Sciences, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
26
|
Grech AM, Ratnayake U, Hannan AJ, van den Buuse M, Hill RA. Sex-Dependent Effects of Environmental Enrichment on Spatial Memory and Brain-Derived Neurotrophic Factor (BDNF) Signaling in a Developmental "Two-Hit" Mouse Model Combining BDNF Haploinsufficiency and Chronic Glucocorticoid Stimulation. Front Behav Neurosci 2018; 12:227. [PMID: 30356704 PMCID: PMC6189322 DOI: 10.3389/fnbeh.2018.00227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/11/2018] [Indexed: 01/02/2023] Open
Abstract
Neurodevelopmental disorders are thought to be caused by a combination of adverse genetic and environmental insults. The "two-hit" hypothesis suggests that an early first "hit" primes the developing brain to be vulnerable to a second "hit" during adolescence which triggers behavioral dysfunction. We have previously modeled this scenario in mice and found that the combined effect of a genetic hapolinsuffuciency in the brain-derived neurotrophic factor (BDNF) gene (1st hit) and chronic corticosterone (CORT) treatment during adolescence (2nd hit), caused spatial memory impairments in adulthood. Environmental enrichment (EE) protocols are designed to stimulate experience-dependent plasticity and have shown therapeutic actions. This study investigated whether EE can reverse these spatial memory impairments. Wild-type (WT) and BDNF heterozygous (HET) mice were treated with corticosterone (CORT) in their drinking water (50 mg/L) from weeks 6 to 8 and exposed to EE from 7 to 9 weeks. Enriched housing included open top cages with additional toys, tunnels, housing, and platforms. Y-maze novel preference testing, to assess short-term spatial memory, was performed at 10 weeks of age. At week 16 dorsal hippocampus tissue was obtained for Western blot analysis of expression levels of BDNF, the BDNF receptor TrkB, and NMDA receptor subunits, GluNR1, 2A and 2B. As in our previous studies, spatial memory was impaired in our two-hit (BDNF HET + CORT) mice. Simultaneous EE prevented these impairments. However, EE appeared to worsen spatial memory performance in WT mice, particularly those exposed to CORT. While BDNF levels were lower in BDNF HET mice as expected, there were no further effects of CORT or EE in males but a close to significant female CORT × EE × genotype interaction which qualitatively corresponded with Y-maze performance. However, EE caused both sex- and genotype-specific effects on phosphorylated TrkB residues and GluNR expression within the dorsal hippocampus, with GluNR2B levels in males changing in parallel with spatial memory performance. In conclusion, beneficial effects of EE on spatial memory emerge only following two developmental disruptions. The mechanisms by which EE exerts its effects are likely via regulation of multiple activity-dependent pathways, including TrkB and NMDA receptor signaling.
Collapse
Affiliation(s)
- Adrienne M. Grech
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Udani Ratnayake
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J. Hannan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Rachel A. Hill
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|