1
|
Chen H, Ouyang W, Cui X, Ma X, Hu S, Qing W, Tong J. miR-124 mediates the effects of gut microbial dysbiosis on brain function in chronic stressed mice. Behav Brain Res 2025; 476:115262. [PMID: 39306097 DOI: 10.1016/j.bbr.2024.115262] [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: 02/19/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024]
Abstract
The gut microbiota plays a key role in the brain function impairment caused by chronic stress, yet its exact mechanism remains unclear. Many studies have revealed the important role of miR-124 in the central nervous system. Meanwhile, previous studies have indicated that miR-124 may be regulated by chronic stress and gut microbiota. Here, we aimed to explore whether miR-124 serves as a mediator for the impacts of gut microbial dysbiosis on brain function in mice subjected to chronic stress. Repeated daily restraint stress for 4 weeks was used to induce chronic stress in mice. Chronic stress resulted in gut microbial dysbiosis, abnormal behaviors, and a decrease in hippocampal miR-124 levels. Treatment with different probiotic mixtures significantly alleviated the effects of chronic stress on hippocampal miR-124 levels and mouse behaviors. Suppression of hippocampal miR-124 expression reversed the beneficial effects of probiotics on cognitive function, neurogenesis, and related molecular markers in chronically stressed mice. Bioinformatics analysis and qPCR suggested that Ptpn11 might be a target gene for miR-124 in mediating the effects of gut microbial dysbiosis on brain function in these mice. These findings suggest that miR-124 is a pivotal regulator that mediates the detrimental effects of gut microbial dysbiosis on brain function and the subsequent cognitive impairment during chronic stress.
Collapse
Affiliation(s)
- Hui Chen
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Department of Laboratory Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoyu Cui
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xin Ma
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shanshan Hu
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wenxiang Qing
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Jianbin Tong
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha 410013, China; Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha 410013, China; Center for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha 410013, China; Brain Research Center, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
2
|
Shao J, Deng Q, Feng S, Wu C, Liu X, Yang L. Role of astrocytes in Alzheimer's disease pathogenesis and the impact of exercise-induced remodeling. Biochem Biophys Res Commun 2024; 732:150418. [PMID: 39032410 DOI: 10.1016/j.bbrc.2024.150418] [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: 04/26/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) is a prevalent and debilitating brain disorder that worsens progressively with age, characterized by cognitive decline and memory impairment. The accumulation of amyloid-beta (Aβ) leading to amyloid plaques and hyperphosphorylation of Tau, resulting in intracellular neurofibrillary tangles (NFTs), are primary pathological features of AD. Despite significant research investment and effort, therapies targeting Aβ and NFTs have proven limited in efficacy for treating or slowing AD progression. Consequently, there is a growing interest in non-invasive therapeutic strategies for AD prevention. Exercise, a low-cost and non-invasive intervention, has demonstrated promising neuroprotective potential in AD prevention. Astrocytes, among the most abundant glial cells in the brain, play essential roles in various physiological processes and are implicated in AD initiation and progression. Exercise delays pathological progression and mitigates cognitive dysfunction in AD by modulating astrocyte morphological and phenotypic changes and fostering crosstalk with other glial cells. This review aims to consolidate the current understanding of how exercise influences astrocyte dynamics in AD, with a focus on elucidating the molecular and cellular mechanisms underlying astrocyte remodeling. The review begins with an overview of the neuropathological changes observed in AD, followed by an examination of astrocyte dysfunction as a feature of the disease. Lastly, the review explores the potential therapeutic implications of exercise-induced astrocyte remodeling in the context of AD.
Collapse
Affiliation(s)
- Jie Shao
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Xiaocao Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Li C, Cui K, Zhu X, Wang S, Yang Q, Fang G. 8-weeks aerobic exercise ameliorates cognitive deficit and mitigates ferroptosis triggered by iron overload in the prefrontal cortex of APP Swe/ PSEN 1dE9 mice through Xc -/GPx4 pathway. Front Neurosci 2024; 18:1453582. [PMID: 39315073 PMCID: PMC11417105 DOI: 10.3389/fnins.2024.1453582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/16/2024] [Indexed: 09/25/2024] Open
Abstract
Background Alzheimer's disease (AD) is a degenerative disorder of the central nervous system characterized by notable pathological features such as neurofibrillary tangles and amyloid beta deposition. Additionally, the significant iron accumulation in the brain is another important pathological hallmark of AD. Exercise can play a positive role in ameliorating AD, but the mechanism is unclear. The purpose of the study is to explore the effect of regular aerobic exercise iron homeostasis and lipid antioxidant pathway regarding ferroptosis in the prefrontal cortex (PFC) of APP Swe/PSEN 1dE9 (APP/PS1) mice. Methods Eighty 6-month-old C57BL/6 J and APP/PS1 mice were divided equally into 8-weeks aerobic exercise groups and sedentary groups. Subsequently, Y-maze, Morris water maze test, iron ion detection by probe, Western Blot, ELISA, RT-qPCR, HE, Nissle, Prussian Blue, IHC, IF, and FJ-C staining experiments were conducted to quantitatively assess the behavioral performance, iron levels, iron-metabolism-related proteins, lipid antioxidant-related proteins and morphology in each group of mice. Results In APP/PS1 mice, the increase in heme input proteins and heme oxygenase lead to the elevated levels of free iron in the PFC. The decrease in ferritin content by ferritin autophagy fails to meet the storage needs for excess free iron within the nerve cells. Ultimately, the increase of free ferrous iron triggers the Fenton reaction, may lead to ferroptosis and resulting in cognitive impairment in APP/PS1 mice. However, 8-weeks aerobic exercise induce upregulation of the Xc-/GPx4 pathway, which can reverse the lipid peroxidation process, thereby inhibiting ferroptosis in APP/PS1 mice. Conclusion 8 weeks aerobic exercise can improve learning and memory abilities in AD, upregulate GPx4/Xc- pathway in PFC to reduce ferroptosis induced by AD.
Collapse
Affiliation(s)
- Chaoyang Li
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Kaiyin Cui
- Sport Science School, Beijing Sport University, Beijing, China
| | - Xinyuan Zhu
- Department of Medical Supervision, China National Institute of Sports Medicine, Beijing, China
| | - Shufan Wang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| | - Qing Yang
- National Fitness and Scientific Exercise Research Center, China Institute of Sport Science, Beijing, China
| | - Guoliang Fang
- Exercise Biology Research Center, China Institute of Sport Science, Beijing, China
| |
Collapse
|
4
|
Hasanabadi AJ, Beirami E, Kamaei M, Esfahani DE. Effect of imipramine on memory, adult neurogenesis, neuroinflammation, and mitochondrial biogenesis in a rat model of alzheimer's disease. Exp Gerontol 2024; 194:112517. [PMID: 38986856 DOI: 10.1016/j.exger.2024.112517] [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: 12/12/2023] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline and memory loss. Imipramine, a tricyclic antidepressant, has potent anti-inflammatory and antioxidant properties in the central nervous system. The aim of this study was to investigate the neuroprotective effects of imipramine on streptozotocin (STZ)-induced memory impairment. Male Wistar rats received an intracerebroventricular injection of STZ (3 mg/kg, 3 μl/ventricle) using the stereotaxic apparatus. The Morris water maze and passive avoidance tests were used to evaluate cognitive functions. 24 h after the STZ injection, imipramine was administered intraperitoneally at doses of 10 or 20 mg/kg for 14 consecutive days. The mRNA and protein levels of neurotrophic factors (BDNF and GDNF) and pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α) were measured in the hippocampus using real-time PCR and ELISA techniques, respectively. In addition, real-time PCR was used to evaluate the mRNA levels of markers associated with neurogenesis (Nestin, DCX, and Ki67) and mitochondrial biogenesis (PGC-1α, NRF-1, and TFAM). The results showed that imipramine, especially at a dose of 20 mg/kg, effectively improved STZ-induced memory impairment. This improvement was associated with an increase in neurogenesis and neurotrophic factors and a decrease in neuroinflammation and mitochondrial biogenesis dysfunction. Based on these results, imipramine appears to be a promising therapeutic option for improving cognitive functions in neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
| | - Elmira Beirami
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
| | - Mehdi Kamaei
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Delaram Eslimi Esfahani
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
5
|
Yang L, Lin W, Yan X, Zhang Z. Comparative effects of lifelong moderate-intensity continuous training and high-intensity interval training on blood lipid levels and mental well-being in naturally ageing mice. Exp Gerontol 2024; 194:112519. [PMID: 38992822 DOI: 10.1016/j.exger.2024.112519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/29/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVE This study aimed to investigate the impact of lifelong exercise, including both moderate-intensity continuous training and high-intensity interval training, on blood lipid levels and mental behaviour in naturally ageing mice to identify effective exercise strategies for ageing-related health issues. METHODS Six-week-old male BALB/c mice were randomly assigned to one of four groups: young control (YC), natural ageing control (OC), lifelong moderate-intensity continuous exercise (EM), and lifelong high-intensity interval exercise (EH) groups. The EM group was trained at a speed corresponding to 70 % of the maximum running speed, while the EH group was trained at a running speed alternating between 50 % of the maximum running speed, 70 % of the maximum running speed, and 90 % of the maximum running speed. All exercise sessions were conducted three times per week, with each session lasting 50 min. Behavioural tests and blood sample collection were conducted at 72 weeks of age. RESULTS Ageing in mice led to changes in muscle and fat mass. Both the EM and EH groups showed greater muscle mass and lower fat mass than did the OC group. Ageing was associated with elevated anxiety (fewer open arm entries, time spent in the central region) and depression (lower sucrose preference) indicators. However, these changes were reversed in both exercise groups, with no differences between the two exercise groups. Blood lipid levels, including total cholesterol (TC), total triglycerides (TGs), low-density lipoprotein (LDL), and free fatty acid (FFA) levels, were greater in the OC group than in the YC group. Additionally, the OC group exhibited lower high-density lipoprotein (HDL) levels. However, both the EM and EH groups exhibited improved lipid profiles compared to those of the YC group. CONCLUSION Lifelong exercise, whether moderate-intensity continuous or high-intensity interval training, can preserve body health during ageing, prevent anxiety and depression, and maintain stable blood lipid levels. Both exercise types are equally effective, suggesting that exercise intensity may not be the critical factor underlying these beneficial adaptations.
Collapse
Affiliation(s)
- Ling Yang
- School of Physical Education, Shaoguan University, Shaoguan 512000, Guangdong, China; Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia
| | - Wentao Lin
- School of Physical Education and Health, Zhuhai College of Science and Technology, Zhuhai 519090, Guangdong, China
| | - Xu Yan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia
| | - Zhishang Zhang
- Department of Physical Education, Guangdong Medical University, Dongguan 523808, Guangdong, China.
| |
Collapse
|
6
|
Wu C, Deng Q, Zhu L, Liu TCY, Duan R, Yang L. Methylene Blue Pretreatment Protects Against Repeated Neonatal Isoflurane Exposure-Induced Brain Injury and Memory Loss. Mol Neurobiol 2024; 61:5787-5801. [PMID: 38233687 DOI: 10.1007/s12035-024-03931-0] [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: 07/18/2023] [Accepted: 01/06/2024] [Indexed: 01/19/2024]
Abstract
Perioperative neurocognitive impairment (PND) is a common medical complication in the postoperative period. General anesthesia through volatile anesthetics poses a high risk of POCD. Moreover, the developing brain is especially vulnerable to anesthesia-induced neurotoxicity. Therefore, finding a practical approach to prevent or alleviate neonatal isoflurane (ISO) exposure-induced brain injury and cognitive decline is essential for reducing medical complications following major surgery during the early postnatal period. Using a repeated neonatal ISO exposure-induced PND rat model, we investigated the effects of methylene blue (MB) pretreatment on repeated neonatal isoflurane exposure-induced brain injury and memory loss. Intraperitoneal injection of low-dose MB (1 mg/kg) was conducted three times 24 h before each ISO exposure. The Barnes maze and novel objection test were conducted to assess learning and memory. Immunofluorescence staining, F-Jade C staining, TUNEL staining, and Western blot analysis were performed to determine mitochondrial fragmentation, neuronal injury, degeneration, and apoptosis. Evans blue extravasation assay, total antioxidant capacity assay, MDA assay kit, and related inflammatory assay kits were used to test blood-brain barrier (BBB) disruption, antioxidant capacity, and neuroinflammation. Behavioral tests revealed that MB pretreatment significantly ameliorated ISO exposure-induced cognitive deficits. In addition, MB pretreatment alleviates neuronal injury, apoptosis, and degeneration. Furthermore, the BBB integrity was preserved by MB pretreatment. Additional studies revealed that ISO-induced excessive mitochondrial fragmentation, oxidative stress, and neuroinflammation were significantly attenuated by MB pretreatment in the PND rat model. Our findings suggest that MB pretreatment alleviates ISO exposure-induced brain injury and memory loss for the first time, supporting MB pretreatment as a promising approach to protect the brain against neonatal ISO exposure-induced postoperative cognitive dysfunction.
Collapse
Affiliation(s)
- Chongyun Wu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
7
|
Deng Q, Wu C, Parker E, Liu TCY, Duan R, Yang L. Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances. Aging Dis 2024; 15:1537-1564. [PMID: 37815901 PMCID: PMC11272214 DOI: 10.14336/ad.2023.0907] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Alzheimer's disease, one of the most common forms of dementia, is characterized by a slow progression of cognitive impairment and neuronal loss. Currently, approved treatments for AD are hindered by various side effects and limited efficacy. Despite considerable research, practical treatments for AD have not been developed. Increasing evidence shows that glial cells, especially microglia and astrocytes, are essential in the initiation and progression of AD. During AD progression, activated resident microglia increases the ability of resting astrocytes to transform into reactive astrocytes, promoting neurodegeneration. Extensive clinical and molecular studies show the involvement of microglia and astrocyte-mediated neuroinflammation in AD pathology, indicating that microglia and astrocytes may be potential therapeutic targets for AD. This review will summarize the significant and recent advances of microglia and astrocytes in the pathogenesis of AD in three parts. First, we will review the typical pathological changes of AD and discuss microglia and astrocytes in terms of function and phenotypic changes. Second, we will describe microglia and astrocytes' physiological and pathological role in AD. These roles include the inflammatory response, "eat me" and "don't eat me" signals, Aβ seeding, propagation, clearance, synapse loss, synaptic pruning, remyelination, and demyelination. Last, we will review the pharmacological and non-pharmacological therapies targeting microglia and astrocytes in AD. We conclude that microglia and astrocytes are essential in the initiation and development of AD. Therefore, understanding the new role of microglia and astrocytes in AD progression is critical for future AD studies and clinical trials. Moreover, pharmacological, and non-pharmacological therapies targeting microglia and astrocytes, with specific studies investigating microglia and astrocyte-mediated neuronal damage and repair, may be a promising research direction for future studies regarding AD treatment and prevention.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Chittora R, Jain S, Roy A, Pandey S, Kochhar KP. Multifactorial effects of short duration early exposure low intensity magnetic field stimulation in Streptozotocin induced Alzheimer's disease rat model. Neurosci Lett 2024; 836:137878. [PMID: 38862088 DOI: 10.1016/j.neulet.2024.137878] [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: 01/28/2024] [Revised: 05/17/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024]
Abstract
Alzheimer's disease (AD) is an approaching, progressive public health crisis which presently lacks an effective treatment. Various non-invasive novel therapies like repetitive transcranial magnetic stimulation have shown potential to improve cognitive performance in AD patients. In the present study, the effect of extremely low intensity magnetic field (MF) stimulation on neurogenesis and cortical electrical activity was explored. Adult Wistar rats were divided into Sham, AD and AD + MF groups. Streptozotocin (STZ) was injected intracerebroventricularly, at a dose of 3 mg/kg body weight for developing AD model. The AD rats were then exposed to MF (17.96 µT) from 8th day of STZ treatment until 15th day, followed by cognitive assessments and electrocortical recording. In brain tissue samples, cresyl violet staining and BrdU immunohistochemistry were done. MF exposure, improved passive avoidance and recognition memory, attenuated neuronal degeneration and enhanced cell proliferation (BrdU positive cells) in comparison to AD rats. It also significantly restores delta wave power from frontal lobe. Our results suggest that early-stage MF exposure could be an asset for AD research and open new avenues in slowing down the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Reena Chittora
- Neurophysiology and Nanomedicine Laboratory, Department of Physiology, AIIMS, New Delhi, India
| | - Suman Jain
- Neurophysiology and Nanomedicine Laboratory, Department of Physiology, AIIMS, New Delhi, India.
| | - Avishek Roy
- Neurophysiology and Nanomedicine Laboratory, Department of Physiology, AIIMS, New Delhi, India; Centre Broca Nouvelle-Aquitaine, Bordeaux, France
| | - Shivam Pandey
- Department of Biostatistics, AIIMS, New Delhi, India
| | | |
Collapse
|
9
|
Zhao R. Can exercise benefits be harnessed with drugs? A new way to combat neurodegenerative diseases by boosting neurogenesis. Transl Neurodegener 2024; 13:36. [PMID: 39049102 PMCID: PMC11271207 DOI: 10.1186/s40035-024-00428-7] [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: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Adult hippocampal neurogenesis (AHN) is affected by multiple factors, such as enriched environment, exercise, ageing, and neurodegenerative disorders. Neurodegenerative disorders can impair AHN, leading to progressive neuronal loss and cognitive decline. Compelling evidence suggests that individuals engaged in regular exercise exhibit higher production of proteins that are essential for AHN and memory. Interestingly, specific molecules that mediate the effects of exercise have shown effectiveness in promoting AHN and cognition in different transgenic animal models. Despite these advancements, the precise mechanisms by which exercise mimetics induce AHN remain partially understood. Recently, some novel exercise molecules have been tested and the underlying mechanisms have been proposed, involving intercommunications between multiple organs such as muscle-brain crosstalk, liver-brain crosstalk, and gut-brain crosstalk. In this review, we will discuss the current evidence regarding the effects and potential mechanisms of exercise mimetics on AHN and cognition in various neurological disorders. Opportunities, challenges, and future directions in this research field are also discussed.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, 88 South Daxue Road, Yangzhou, 225009, China.
| |
Collapse
|
10
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2024; 0:revneuro-2024-0046. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [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: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
11
|
Almasi E, Heidarianpour A, Keshvari M. The interactive effects of different exercises and hawthorn consumption on the pain threshold of TMT-induced Alzheimer male rats. J Physiol Sci 2024; 74:36. [PMID: 39014320 PMCID: PMC11251243 DOI: 10.1186/s12576-024-00925-4] [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: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 07/18/2024]
Abstract
Exercise increases the pain threshold in healthy people. However, the pain threshold modulation effect of exercise and hawthorn is unclear because of its potential benefits in people with persistent pain, including those with Alzheimer's disease. Accordingly, after the induction of Alzheimer's disease by trimethyl chloride, male rats with Alzheimer's disease were subjected to a 12-week training regimen consisting of resistance training, swimming endurance exercises, and combined exercises. In addition, hawthorn extract was orally administered to the rats. Then, their pain threshold was evaluated using three Tail-flick, Hot-plate, and Formalin tests. Our results showed that Alzheimer's decreased the pain threshold in all three behavioral tests. Combined exercise with hawthorn consumption had the most statistically significant effect on Alzheimer's male rats' pain threshold in all three experiments. A combination of swimming endurance and resistance exercises with hawthorn consumption may modulate hyperalgesia in Alzheimer's rats. Future studies need to determine the effects of these factors on the treatment and/or management of painful conditions.
Collapse
Affiliation(s)
- Ensiyeh Almasi
- Department of Exercise Physiology, Faculty of Sport Sciences, Bu-Ali Sina University, Hamedan, Iran
| | - Ali Heidarianpour
- Department of Exercise Physiology, Faculty of Sport Sciences, Bu-Ali Sina University, Hamedan, Iran.
| | - Maryam Keshvari
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| |
Collapse
|
12
|
Guo J, Cao Y, Zhang T, Xu C, Liu Z, Li W, Wang Q. Multisensory Fusion Training and 7, 8-Dihydroxyflavone Improve Amyloid-β-Induced Cognitive Impairment, Anxiety, and Depression-Like Behavior in Mice Through Multiple Mechanisms. Neuropsychiatr Dis Treat 2024; 20:1247-1270. [PMID: 38883414 PMCID: PMC11180438 DOI: 10.2147/ndt.s459891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Background There is growing interest in the role of physical activity in patients with of Alzheimer's disease (AD), particularly regarding its impact of cognitive function, gut microbiota, metabolites, and neurotrophic factors. Objective To investigate the impact of multisensory fusion training (MSFT) combined with 7, 8-dihydroxyflavone (DHF) on the behavioral characteristics, protein expression, microbiome, and serum metabolome using the AD model in mice induced with amyloid-β (Aβ). Methods We assessed cognitive ability, anxiety-like and depression-like behaviors in Aβ mice using behavioral measures. Western blotting was employed to detect the expression of relevant proteins. The 16S rRNA gene sequencing and metabolomics were used to analyze changes in the intestinal microbial composition and serum metabolic profile, respectively, of Aβ mice. Results The behavioral outcomes indicated that a 4-week intervention combining DHF and MSFT yielded remarkable improvements in cognitive function and reduced anxiety and depression-like behaviors in Aβ mice. In the hippocampus of Aβ mice, the combined intervention increased the levels of BDNF, VGF, PSD-95, Nrf2, p-GSK3β and p-CREB proteins. Analyses of sequence and metabolomic data revealed that Bacteroides and Ruminococcaceae were remarkably more abundant following the combined intervention, influencing the expression of specific metabolites directly linked to the maintenance of neuronal and neurobehavioral functions. These metabolites play a crucial role in vital processes, such as amino acid metabolism, lipid metabolism, and neurotransmitter metabolism in mice. Conclusion Our study highlighted that MSFT combined with DHF improves cognitive impairment, anxiety, and depression-like behavior in Aβ mice through multiple mechanisms, and further validated the correlation between the gut microbiome and serum metabolome. These findings open up a promising avenue for future investigations into potential treatment strategies for AD.
Collapse
Affiliation(s)
- Jiejie Guo
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Yanzi Cao
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Ting Zhang
- Department of Clinical Laboratory, The First People's Hospital of Wenling, Taizhou, People's Republic of China
| | - Chunshuang Xu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Zhitao Liu
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
- Fujian Normal University, Fuzhou, People's Republic of China
| | - Wanyi Li
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, NBU Health Science Center, Ningbo University, Ningbo, People's Republic of China
| |
Collapse
|
13
|
Xu X, Zhang J, Xing H, Han L, Li X, Wu P, Tang J, Jing L, Luo J, Luo J, Liu L. Identification of metabolism-related key genes as potential biomarkers for pathogenesis of immune thrombocytopenia. Sci Rep 2024; 14:9040. [PMID: 38641637 PMCID: PMC11031595 DOI: 10.1038/s41598-024-59493-7] [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: 01/03/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
Immune thrombocytopenia (ITP), an acquired autoimmune disease, is characterized by immune-mediated platelet destruction. A biomarker is a biological entity that contributes to disease pathogenesis and reflects disease activity. Metabolic alterations are reported to be associated with the occurrence of various diseases. As metabolic biomarkers for ITP have not been identified. This study aimed to identify metabolism-related differentially expressed genes as potential biomarkers for pathogenesis of ITP using bioinformatic analyses.The microarray expression data of the peripheral blood mononuclear cells were downloaded from the Gene Expression Omnibus database (GSE112278 download link: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE112278 ). Key module genes were intersected with metabolism-related genes to obtain the metabolism-related key candidate genes. The hub genes were screened based on the degree function in the coytoscape sofware. The key ITP-related genes were subjected to functional enrichment analysis. Immune infiltration analysis was performed using a single-sample gene set enrichment analysis algorithm to evaluate the differential infiltration levels of immune cell types between ITP patient and control. Molecular subtypes were identified based on the expression of hub genes. The expression of hub genes in the ITP patients was validated using quantitative real-time polymerase chain reaction analysis. This study identified five hub genes (ADH4, CYP7A1, CYP1A2, CYP8B1, and NR1H4), which were be associated with the pathogenesis of ITP, and two molecular subtypes of ITP. Among these hub genes, CYP7A1 and CYP8B1 involved in cholesterol metabolism,were further verified in clinical samples.
Collapse
Affiliation(s)
- Xiangmei Xu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jiamin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Hongyun Xing
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Liying Han
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xiaoming Li
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Pengqiang Wu
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jirui Tang
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Li Jing
- Department of Hematology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jie Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Jing Luo
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
14
|
Mohanad M, Mohamed SK, Aboulhoda BE, Ahmed MAE. Neuroprotective effects of vitamin D in an Alzheimer's disease rat model: Improvement of mitochondrial dysfunction via calcium/calmodulin-dependent protein kinase kinase 2 activation of Sirtuin1 phosphorylation. Biofactors 2024; 50:371-391. [PMID: 37801071 DOI: 10.1002/biof.2013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/17/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial dysfunction is an early event in Alzheimer's disease (AD) pathogenesis. To assess the impact of vitamin D3 (Vit.D) on neurogenesis, we investigated its role in mitigating cognitive impairment and mitochondrial dysfunction through calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2)-mediated phosphorylation of Sirtuin1 (SIRT1) in an aluminum-chloride-D-galactose (AlCl3-D-gal)-induced AD rat model. Rats were distributed into four groups: control, AlCl3 + D-gal (10 + 60 mg/kg, ip), Vit.D (500 IU/kg, po), and AlCl3 + D-gal+Vit.D. Novel object recognition (NOR), Morris Water Maze, and passive avoidance (PA) tests were used to measure memory abilities. The hippocampal tissue was used to assess vitamin D3 receptor (VDR) and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α) expression by quantitative real-time polymerase chain reaction (qRT-PCR), CAMKK2, p-SIRT1, phosphorylated-AMP-activated protein kinase (p-AMPK), dynamin-related-protein-1 (Drp1), and mitofusin-1 (Mnf1) proteins by western blot and Ca2+ levels, endothelial nitic oxide synthase (eNOS), superoxide dismutase (SOD), amyloid beta (Aβ), and phospho tau (p-Tau) via enzyme-linked immunosorbent assay(ELISA) in addition to histological and ultrastructural examination of rat's brain tissue. Vit.D-attenuated hippocampal injury reversed the cognitive decline and Aβ aggregation, and elevated p-Tau levels in the AlCl3 + D-gal-induced AD rat model. In AlCl3 + D-gal-exposed rats, Vit.D induced VDR expression, normalized Ca2+ levels, elevated CAMKK2, p-AMPK, p-SIRT1, and PGC-1α expression. Vit.D reduced Drp1, induced Mnf1, increased mitochondrial membrane potential, preserved mitochondrial structure, restored normal mitochondrial function, and retained normal eNOS level and SOD activity in AlCl3 + D-gal rats. In conclusion, our findings proved that Vit.D may ameliorate cognitive deficits in AlCl3 + D-gal-induced AD by restoring normal mitochondrial function and reducing inflammatory and oxidative stress via CAMKK2-AMPK/SIRT1 pathway upregulation.
Collapse
Affiliation(s)
- Marwa Mohanad
- Department of Biochemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Shimaa K Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Basma E Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| |
Collapse
|
15
|
Lazarova M, Tsvetanova E, Georgieva A, Stefanova M, Uzunova D, Denev P, Vassileva V, Tasheva K. Extracts of Sideritis scardica and Clinopodium vulgare Alleviate Cognitive Impairments in Scopolamine-Induced Rat Dementia. Int J Mol Sci 2024; 25:1840. [PMID: 38339117 PMCID: PMC10855470 DOI: 10.3390/ijms25031840] [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: 01/04/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Sideritis scardica Griseb. and Clinopodium vulgare L., belonging to the Lamiaceae family, are rich in terpenoids and phenolics and exhibit various pharmacological effects, including antioxidant, anti-inflammatory and anti-cancer activities. While the memory-enhancing impacts of S. scardica are well documented, the cognitive benefits of C. vulgare remain unexplored. This study assessed the potential effect of C. vulgare on learning and memory in healthy and scopolamine (Sco)-induced memory-impaired male Wistar rats, comparing it with the effects of S. scardica. Over a 21-day period, rats orally received extracts of cultivated S. scardica (200 mg/kg) and C. vulgare (100 mg/kg), either individually or in combination, with administration starting 10 days before and continuing 11 days simultaneously with Sco injection at a dose of 2 mg/kg intraperitoneally. The results showed that both extracts effectively mitigated Sco-induced memory impairment. Their combination significantly improved recognition memory and maintained monoaminergic function. S. scardica excelled in preserving spatial working memory, while C. vulgare exhibited comparable retention of recognition memory, robust antioxidant activity and acetylcholinesterase inhibitory activity. The extracts alleviated Sco-induced downregulation of p-CREB/BDNF signaling, suggesting neuroprotective mechanisms. The extract combination positively affected most of the Sco-induced impairments, underscoring the potential for further investigation of these extracts for therapeutic development.
Collapse
Affiliation(s)
- Maria Lazarova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Science, 1113 Sofia, Bulgaria; (M.L.); (E.T.); (A.G.); (M.S.); (D.U.)
| | - Petko Denev
- Laboratory of Biologically Active Substances, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 4000 Plovdiv, Bulgaria
| | - Valya Vassileva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 21, 1113 Sofia, Bulgaria;
| | - Krasimira Tasheva
- Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. Georgi Bonchev Str., Block 21, 1113 Sofia, Bulgaria;
| |
Collapse
|
16
|
Zhao R. Exercise mimetics: a novel strategy to combat neuroinflammation and Alzheimer's disease. J Neuroinflammation 2024; 21:40. [PMID: 38308368 PMCID: PMC10837901 DOI: 10.1186/s12974-024-03031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), characterized by the stimulation of resident immune cells of the brain and the penetration of peripheral immune cells. These inflammatory processes facilitate the deposition of amyloid-beta (Aβ) plaques and the abnormal hyperphosphorylation of tau protein. Managing neuroinflammation to restore immune homeostasis and decrease neuronal damage is a therapeutic approach for AD. One way to achieve this is through exercise, which can improve brain function and protect against neuroinflammation, oxidative stress, and synaptic dysfunction in AD models. The neuroprotective impact of exercise is regulated by various molecular factors that can be activated in the same way as exercise by the administration of their mimetics. Recent evidence has proven some exercise mimetics effective in alleviating neuroinflammation and AD, and, additionally, they are a helpful alternative option for patients who are unable to perform regular physical exercise to manage neurodegenerative disorders. This review focuses on the current state of knowledge on exercise mimetics, including their efficacy, regulatory mechanisms, progress, challenges, limitations, and future guidance for their application in AD therapy.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, Yangzhou, China.
| |
Collapse
|
17
|
Savall ASP, de Mello JD, Fidelis EM, Comis-Neto AA, Nepomuceno MR, Pacheco CDO, Haas SE, Pinton S. Nanoencapsulated Curcumin: Enhanced Efficacy in Reversing Memory Loss in An Alzheimer Disease Model. Brain Sci 2024; 14:130. [PMID: 38391705 PMCID: PMC10886961 DOI: 10.3390/brainsci14020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Investigating new drugs or formulations that target Alzheimer disease (AD) is critical for advancing therapeutic interventions. Therefore, this study aimed to assess the effectiveness of nanoencapsulated curcumin (NC Curc) in alleviating memory impairment, oxidative stress, and neuroinflammation in a validated AD model. Male Wistar rats were given bilateral intracerebroventricular injections of either saline or streptozotocin (STZ) (3 mg/3 µL/site) to establish the AD model (day 0). On day 22, daily oral administrations of curcumin (6 mg/kg), NC Curc (6 mg/kg), or a vehicle (unloaded NC) were initiated and continued for 14 days. NC Curc significantly reversed memory deficits in object recognition and inhibitory avoidance tests induced by STZ. Both formulations of curcumin attenuated elevated acetylcholinesterase activity caused by STZ. Importantly, NC Curc alone effectively mitigated STZ-induced oxidative stress. Additionally, NC Curc treatment normalized GFAP levels, suggesting a potential reduction in neuroinflammation in STZ-treated rats. Our findings indicate that NC Curc improves memory in an AD rat model, highlighting its enhanced therapeutic effects compared to unencapsulated curcumin. This research significantly contributes to understanding the therapeutic and neurorestorative potential of NC Curc in AD, particularly in reversing pathophysiological changes.
Collapse
Affiliation(s)
- Anne Suély Pinto Savall
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Jhuly Dorneles de Mello
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Eduarda Monteiro Fidelis
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Antonio Alvenir Comis-Neto
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Maria Regina Nepomuceno
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Camila de Oliveira Pacheco
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Sandra Elisa Haas
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Simone Pinton
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| |
Collapse
|
18
|
Gelfo F, Petrosini L, Mandolesi L, Landolfo E, Caruso G, Balsamo F, Bonarota S, Bozzali M, Caltagirone C, Serra L. Land/Water Aerobic Activities: Two Sides of the Same Coin. A Comparative Analysis on the Effects in Cognition of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1181-1197. [PMID: 38552114 DOI: 10.3233/jad-231279] [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] [Indexed: 04/20/2024]
Abstract
Evidence in the literature indicates that aerobic physical activity may have a protective role in aging pathologies. However, it has not been clarified whether different types of aerobic exercise produce different effects. In particular, these potential differences have not been explored in patients with Alzheimer's disease (AD). The present narrative review has the specific aim of evaluating whether land (walking/running) and water (swimming) aerobic activities exert different effects on cognitive functions and neural correlates in AD patients. In particular, the investigation is carried out by comparing the evidence provided from studies on AD animal models and on patients. On the whole, we ascertained that both human and animal studies documented beneficial effects of land and water aerobic exercise on cognition in AD. Also, the modulation of numerous biological processes is documented in association with structural modifications. Remarkably, we found that aerobic activity appears to improve cognition per se, independently from the specific kind of exercise performed. Aerobic exercise promotes brain functioning through the secretion of molecular factors from skeletal muscles and liver. These molecular factors stimulate neuroplasticity, reduce neuroinflammation, and inhibit neurodegenerative processes leading to amyloid-β accumulation. Additionally, aerobic exercise improves mitochondrial activity, reducing oxidative stress and enhancing ATP production. Aerobic activities protect against AD, but implementing exercise protocols for patients is challenging. We suggest that health policies and specialized institutions should direct increasing attention on aerobic activity as lifestyle modifiable factor for successful aging and age-related conditions.
Collapse
Affiliation(s)
- Francesca Gelfo
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | | | - Laura Mandolesi
- Department of Humanities, Federico II University of Naples, Naples, Italy
| | | | | | - Francesca Balsamo
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | - Sabrina Bonarota
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Marco Bozzali
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Turin, Italy
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | | | | |
Collapse
|
19
|
Lei L, Luo Y, Kang D, Yang F, Meng D, Wang JZ, Liu R, Wang X, Li HL. Gypenoside IX restores Akt/GSK-3β pathway and alleviates Alzheimer's disease-like neuropathology and cognitive deficits. Aging (Albany NY) 2023; 15:14172-14191. [PMID: 38095632 PMCID: PMC10756109 DOI: 10.18632/aging.205295] [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: 08/16/2023] [Accepted: 11/06/2023] [Indexed: 12/21/2023]
Abstract
The main pathological changes of Alzheimer's disease (AD), a progressive neurodegenerative disorder, include senile plaque (deposited by amyloid beta), neurofibrillary tangle (formed by paired helical filaments composed of hyperphosphorylated tau), and massive loss of neurons. Currently there is a lack of ideal drugs to halt AD progression. Gypenosides (GPs), a kind of natural product, possesses potential therapeutic effects for neurodegenerative diseases, including AD. However, the specific role and mechanism of GPs for AD remain unclear. In the current study, we used staurosporine (STP), an inducer of apoptosis and causing tau hyperphosphorylation, to mimic AD models, and explored the role and mechanism of Gypenoside IX (one of the extracts of Gynostemma, GP for short name in our experiments) in STP treated primary hippocampal neurons and rats. We found STP not only increased apoptosis and tau hyperphosphorylation, but also significantly increased Aβ production, resulting in synaptic dysfunction and cognitive decline in mimic AD models by STP. GP was found to rescue apoptosis and cognitive impairments caused by STP treatment. Moreover, GP recovered the decreased synaptic proteins PSD95, Synaptophysin and GluR2, and blocked dendritic spine loss. Interestingly, GP decreased the STP induced tau hyperphosphorylation at different sites including S-199, S-202, T-205, T-231, S-262, S-396, and S-404, and at the same time decreased Aβ production through down-regulation of BACE1 and PS1. These effects in STP treated primary hippocampal neurons and rats were accompanied with a restoration of AKT/GSK-3β signaling axis with GP treatment, supporting that dysregulation of AKT/GSK-3β pathway might be involved in STP related AD pathogenesis. The results from our research proved that GP might be a potential candidate compound to reduce neuronal damage and prevent the cognitive decline in AD.
Collapse
Affiliation(s)
- Ling Lei
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yong Luo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongkun Kang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fumin Yang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongli Meng
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen 518000, China
| | - Hong-Lian Li
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
20
|
Morella I, Negro M, Dossena M, Brambilla R, D'Antona G. Gut-muscle-brain axis: Molecular mechanisms in neurodegenerative disorders and potential therapeutic efficacy of probiotic supplementation coupled with exercise. Neuropharmacology 2023; 240:109718. [PMID: 37774944 DOI: 10.1016/j.neuropharm.2023.109718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 10/01/2023]
Abstract
Increased longevity is often associated with age-related conditions. The most common neurodegenerative disorders in the older population are Alzheimer's disease (AD) and Parkinson's disease (PD), associated with progressive neuronal loss leading to functional and cognitive impairments. Although symptomatic treatments are available, there is currently no cure for these conditions. Gut dysbiosis has been involved in the pathogenesis of AD and PD, thus interventions targeting the "gut-brain axis" could potentially prevent or delay these pathologies. Recent evidence suggests that the skeletal muscle and the gut microbiota can affect each other via the "gut-muscle axis". Importantly, cognitive functions in AD and PD patients significantly benefit from physical activity. In this review, we aim to provide a comprehensive picture of the crosstalk between the brain, the skeletal muscle and the gut microbiota, introducing the concept of "gut-muscle-brain axis". Moreover, we discuss human and animal studies exploring the modulatory role of exercise and probiotics on cognition in AD and PD. Collectively, the findings presented here support the potential benefits of physical activity and probiotic supplementation in AD and PD. Further studies will be needed to develop targeted and multimodal strategies, including lifestyle changes, to prevent or delay the course of these pathologies.
Collapse
Affiliation(s)
- Ilaria Morella
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Massimo Negro
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Riccardo Brambilla
- Neuroscience and Mental Health Innovation Institute, School of Biosciences, Cardiff University, Cardiff, UK; Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Giuseppe D'Antona
- Centro di Ricerca Interdipartimentale Nelle Attività Motorie e Sportive (CRIAMS)-Sport Medicine Centre, University of Pavia, Voghera, Italy; Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
21
|
Zou P, Wu C, Liu TCY, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2023; 12:52. [PMID: 37964328 PMCID: PMC10644503 DOI: 10.1186/s40035-023-00385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
Collapse
Affiliation(s)
- Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
22
|
Khemka S, Reddy A, Garcia RI, Jacobs M, Reddy RP, Roghani AK, Pattoor V, Basu T, Sehar U, Reddy PH. Role of diet and exercise in aging, Alzheimer's disease, and other chronic diseases. Ageing Res Rev 2023; 91:102091. [PMID: 37832608 PMCID: PMC10842571 DOI: 10.1016/j.arr.2023.102091] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, characterized by memory loss and multiple cognitive impairments. Genetic mutations cause a small proportion (1-2%) of early-onset AD, with mutations in amyloid precursor protein (APP), presenilin 1 (PS1) and presenilin 2 (PS2). Major contributing factors of late-onset AD are ApoE4 genotype, traumatic brain injury, diabetes, obesity, hypertension, cardiovascular conditions, in addition to lifestyle factors, such as unhealthy diet and lack of physical exercise. Disease progression can be delayed and/or prevented to a greater extent by adopting healthy lifestyle with balanced and antioxidant enriched diet and daily exercise. The interaction and interplay of diet, exercise, age, and pharmacological interventions holds a crucial role in the progression, pathogenesis and management of AD and its comorbidities, including diabetes, obesity, hypertension and cardiovascular conditions. Antioxidant enriched diet contributes to brain health, glucose control, weight management, and cardiovascular well-being. Regular exercise removes toxins including free radicals and enhances insulin sensitivity, and supports cardiovascular function. In the current article, we discussed, the role of diet, and exercise in aging, AD and other conditions including diabetes, obesity, hypertension, cardiovascular conditions. This article also highlights the impact of medication, socioeconomic and lifestyle factors, and pharmacological interventions. These aspects were discussed in different races and ethnic groups in Texas, and the US.
Collapse
Affiliation(s)
- Sachi Khemka
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Aananya Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA
| | - Ricardo Isaiah Garcia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Micheal Jacobs
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Ruhananhad P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Lubbock High School, Lubbock, TX 79401, USA
| | - Aryan Kia Roghani
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Frenship High School, Lubbock, TX 79382, USA
| | - Vasanthkumar Pattoor
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; University of South Florida, Tampa, FL 33620, USA
| | - Tanisha Basu
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department pf Speech, Language and Hearing Services, School Health Professions, Texas Tech University Healthy Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
23
|
Andrade-Guerrero J, Rodríguez-Arellano P, Barron-Leon N, Orta-Salazar E, Ledesma-Alonso C, Díaz-Cintra S, Soto-Rojas LO. Advancing Alzheimer's Therapeutics: Exploring the Impact of Physical Exercise in Animal Models and Patients. Cells 2023; 12:2531. [PMID: 37947609 PMCID: PMC10648553 DOI: 10.3390/cells12212531] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Alzheimer's disease (AD) is the main neurodegenerative disorder characterized by several pathophysiological features, including the misfolding of the tau protein and the amyloid beta (Aβ) peptide, neuroinflammation, oxidative stress, synaptic dysfunction, metabolic alterations, and cognitive impairment. These mechanisms collectively contribute to neurodegeneration, necessitating the exploration of therapeutic approaches with multiple targets. Physical exercise has emerged as a promising non-pharmacological intervention for AD, with demonstrated effects on promoting neurogenesis, activating neurotrophic factors, reducing Aβ aggregates, minimizing the formation of neurofibrillary tangles (NFTs), dampening inflammatory processes, mitigating oxidative stress, and improving the functionality of the neurovascular unit (NVU). Overall, the neuroprotective effects of exercise are not singular, but are multi-targets. Numerous studies have investigated physical exercise's potential in both AD patients and animal models, employing various exercise protocols to elucidate the underlying neurobiological mechanisms and effects. The objective of this review is to analyze the neurological therapeutic effects of these exercise protocols in animal models and compare them with studies conducted in AD patients. By translating findings from different approaches, this review aims to identify opportune, specific, and personalized therapeutic windows, thus advancing research on the use of physical exercise with AD patients.
Collapse
Affiliation(s)
- Jesús Andrade-Guerrero
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico;
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Paola Rodríguez-Arellano
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Nayeli Barron-Leon
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Erika Orta-Salazar
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Sofía Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Queretaro 76230, Mexico; (P.R.-A.); (N.B.-L.); (E.O.-S.); (C.L.-A.)
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico;
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| |
Collapse
|
24
|
Zhang H, Tian Y, Yu W, Tong D, Ji Y, Qu X, Deng T, Li X, Xu Y. TMEM175 downregulation participates in impairment of the autophagy related lysosomal dynamics following neonatal hypoxic-ischemic brain injury. J Cell Physiol 2023; 238:2512-2527. [PMID: 37566721 DOI: 10.1002/jcp.31096] [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: 05/19/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023]
Abstract
The mechanism underlying long-term cognitive impairment caused by neonatal hypoxic-ischemic brain injury (HIBI) remains unclear. Autophagy is a closely related mechanism and may play a role in this process. We aimed to investigate the role of lysosomal transmembrane protein 175 (TMEM175) in the autophagy-lysosome pathway in neonatal rats with HIBI. A neonatal rat model of HIBI was established, hypoxia was induced, followed by left common carotid artery ligation. Expression levels of TMEM175 and the corresponding proteins involved in autophagy flux and the endolysosomal system fusion process were measured. Rats were administered TMEM175 plasmid via intracerebroventricular injection to induce overexpression. Brain damage and cognitive function were then assessed. TMEM175 was downregulated in the hippocampal tissue, and the autophagy-lysosome pathway was impaired following HIBI in neonatal rats. Overexpression of TMEM175 significantly mitigated neuronal injury and improved long-term cognitive and memory function in neonatal rats with HIBI. We found that improvement in the autophagy-lysosome pathway and endolysosomal system homeostasis, which are TMEM175 related, occurred via regulation of lysosomal membrane dynamic fusion. TMEM175 plays a critical role in maintaining the autophagy-lysosome pathway and endolysosomal homeostasis, contributing to the amelioration of neuronal injury and impaired long-term cognitive function following neonatal HIBI.
Collapse
Affiliation(s)
- Huiyi Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ye Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weiwei Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dongyi Tong
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yichen Ji
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinrui Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tianjiao Deng
- The First Clinical College, China Medical University, Shenyang, China
| | - Xinsheng Li
- The First Clinical College, China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Liu X, Yang L, Zhang G, Ling J. Neuroprotective Effects of Phenolic Antioxidant Tert-butylhydroquinone (tBHQ) in Brain Diseases. Mol Neurobiol 2023; 60:4909-4923. [PMID: 37191855 DOI: 10.1007/s12035-023-03370-3] [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: 10/18/2022] [Accepted: 04/25/2023] [Indexed: 05/17/2023]
Abstract
Human life and health are gravely threatened by brain diseases. The onset and progression of the illnesses are influenced by a variety of factors, including pathogenic causes, environmental factors, mental issues, etc. According to scientific studies, neuroinflammation and oxidative stress play a significant role in the development and incidence of brain diseases by producing pro-inflammatory cytokines and oxidative tissue damage to induce inflammation and apoptosis. Neuroinflammation, oxidative stress, and oxidative stress-related changes are inseparable factors in the etiology of several brain diseases. Numerous neurodegenerative diseases have undergone substantial research into the therapeutic alternatives that target oxidative stress, the function of oxidative stress, and the possible therapeutic use of antioxidants. Formerly, tBHQ is a synthetic phenolic antioxidant, which has been widely used as a food additive. According to recent researches, tBHQ can suppress the processes that lead to neuroinflammation and oxidative stress, which offers a fresh approach to treating brain diseases. In order to achieve the goal of decreasing inflammation and apoptosis, tBHQ is a specialized nuclear factor erythroid 2-related factor (Nrf2) activator that decreases oxidative stress and enhances antioxidant status by upregulating the Nrf2 gene and reducing nuclear factor kappa-B (NF-κB) activity. This article reviews the effects of tBHQ on neuroinflammation and oxidative stress in recent years and looks into how tBHQ inhibits neuroinflammation and oxidative stress through human, animal, and cell experiments to play a neuroprotective role in Alzheimer's disease (AD), stroke, depression, and Parkinson's disease (PD). It is anticipated that this article will be useful as a reference for upcoming research and the creation of drugs to treat brain diseases.
Collapse
Affiliation(s)
- Xiaojin Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
- Department of Pharmacy, Shandong Medical College, Linyi, 276000, China
| | - Luodan Yang
- College of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Guoying Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Jianya Ling
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| |
Collapse
|
26
|
Hao Z, Liu K, Zhou L, Chen P. Precious but convenient means of prevention and treatment: physiological molecular mechanisms of interaction between exercise and motor factors and Alzheimer's disease. Front Physiol 2023; 14:1193031. [PMID: 37362440 PMCID: PMC10285460 DOI: 10.3389/fphys.2023.1193031] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Disproportionate to the severity of Alzheimer's disease (AD) and the huge number of patients, the exact treatment and prevention of AD is still being explored. With increasing ageing, the search for means to prevent and treat AD has become a high priority. In the search for AD, it has been suggested that exercise may be one of the more effective and less costly means of preventing and treating AD, and therefore a large part of current research is aimed at exploring the effectiveness of exercise in the prevention and treatment of AD. However, due to the complexity of the specific pathogenesis of AD, there are multiple hypotheses and potential mechanisms for exercise interventions in AD that need to be explored. This review therefore specifically summarises the hypotheses of the interaction between exercise and AD from a molecular perspective, based on the available evidence from animal models or human experiments, and explores them categorised according to the pathologies associated with AD: exercise can activate a number of signalling pathways inhibited by AD (e.g., Wnt and PI3K/Akt signalling pathways) and reactivate the effects of downstream factors regulated by these signalling pathways, thus acting to alleviate autophagic dysfunction, relieve neuroinflammation and mitigate Aβ deposition. In addition, this paper introduces a new approach to regulate the blood-brain barrier, i.e., to restore the stability of the blood-brain barrier, reduce abnormal phosphorylation of tau proteins and reduce neuronal apoptosis. In addition, this paper introduces a new concept." Motor factors" or "Exerkines", which act on AD through autocrine, paracrine or endocrine stimulation in response to movement. In this process, we believe there may be great potential for research in three areas: (1) the alleviation of AD through movement in the brain-gut axis (2) the prevention and treatment of AD by movement combined with polyphenols (3) the continued exploration of movement-mediated activation of the Wnt signalling pathway and AD.
Collapse
Affiliation(s)
- Zikang Hao
- Department of Physical Education, Laoshan Campus, Ocean University of China, Qingdao, China
| | - Kerui Liu
- Department of Sports Medicine, Daiyue Campus, Shandong First Medical University, Tai’an, Shandong, China
| | - Lu Zhou
- Department of Sports Medicine, Daiyue Campus, Shandong First Medical University, Tai’an, Shandong, China
| | - Ping Chen
- Department of Physical Education, Laoshan Campus, Ocean University of China, Qingdao, China
| |
Collapse
|
27
|
Feng S, Wu C, Zou P, Deng Q, Chen Z, Li M, Zhu L, Li F, Liu TCY, Duan R, Yang L. High-intensity interval training ameliorates Alzheimer's disease-like pathology by regulating astrocyte phenotype-associated AQP4 polarization. Theranostics 2023; 13:3434-3450. [PMID: 37351177 PMCID: PMC10283053 DOI: 10.7150/thno.81951] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 05/23/2023] [Indexed: 06/24/2023] Open
Abstract
Background: Alzheimer's disease (AD), one of the most common forms of dementia, is a widely studied neurodegenerative disease characterized by Aβ accumulation and tau hyperphosphorylation. Currently, there is no effective cure available for AD. The astrocyte AQP4 polarized distribution-mediated glymphatic system is essential for Aβ and abnormal tau clearance and is a potential therapeutic target for AD. However, the role of exercise on the AQP4 polarized distribution and the association between the AQP4 polarized distribution and astrocyte phenotype polarization are poorly understood. Methods: Using a streptozotocin (STZ)-induced sporadic AD rat model, we investigated the effects of high-intensity interval training on AD pathologies. The Branes maze task was conducted to measure spatial learning and memory. Immunofluorescence staining of NeuN with TUNEL, Fluoro-Jade C, and relative neuronal damage markers was applied to measure neuronal apoptosis, neurodegeneration, and damage. Sholl analysis was carried out to analyze the morphology of microglia. Line-scan analysis, 3D rendering, and the orthogonal view were applied to analyze the colocalization. Western blot analysis and enzyme-linked immunosorbent assay (ELISA) analysis were conducted to examine AQP4 and Aβ, respectively. An APP/PS1 transgenic AD mice model was used to confirm the key findings. Results: High-intensity interval training (HIIT) alleviates cognitive dysfunction in STZ-induced AD-like rat models and provides neuroprotection against neurodegeneration, neuronal damage, and neuronal loss. Additionally, HIIT improved the drainage of abnormal tau and Aβ from the cortex and hippocampus via the glymphatic system to the kidney. Further mechanistic studies support that the beneficial effects of HIIT on AD might be due, in part, to the polarization of glial cells from a neurotoxic phenotype towards a neuroprotective phenotype. Furthermore, an intriguing finding of our study is that the polarized distribution of AQP4 was strongly correlated with astrocyte phenotype. We found A2 phenotype exhibited more evident AQP4 polarization than the A1 phenotype. Conclusion: Our findings indicate that HIIT ameliorates Alzheimer's disease-like pathology by regulating astrocyte phenotype and astrocyte phenotype-associated AQP4 polarization. These changes promote Aβ and p-tau clearance from the brain tissue through the glymphatic system and the kidney.
Collapse
Affiliation(s)
- Shu Feng
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Chongyun Wu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Peibin Zou
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Qianting Deng
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Zhe Chen
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Meng Li
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Ling Zhu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Fanghui Li
- School of Sport Sciences, Nanjing Normal University, Nanjing 210046, China
| | - Timon Cheng-Yi Liu
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Rui Duan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Luodan Yang
- School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| |
Collapse
|
28
|
Özdemir Ç, Arslan M, Küçük A, Yığman Z, Dursun AD. Therapeutic Efficacy of Boric Acid Treatment on Brain Tissue and Cognitive Functions in Rats with Experimental Alzheimer's Disease. Drug Des Devel Ther 2023; 17:1453-1462. [PMID: 37220543 PMCID: PMC10200114 DOI: 10.2147/dddt.s405963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/06/2023] [Indexed: 05/25/2023] Open
Abstract
Introduction Oxidative stress has an important role in the pathophysiology of Alzheimer's disease (AD), the most common type of dementia. Boric acid (BA) contributes significantly to the protection of the brain by reducing lipid peroxidation and supporting antioxidant defense. We aimed to evaluate the therapeutic potential of BA treatment in AD rats. Materials and Methods Four groups were formed as Control (C), Alzheimer's (A), Alzheimer's + Boric acid (ABA), Boric acid (BA). Intracerebroventricular injection of Streptozotocin (STZ) was preferred to create an AD. After 4 weeks, BA was applied 3 times every other day. The Radial Arm Maze Test (RAMT) was used to evaluate memory and learning abilities. Biochemical and histopathological evaluations were made in the hippocampus. Results Initial RAMT inlet/outlet (I/O) numbers were similar. Two weeks after STZ injection, I/O numbers decreased in group A and ABA compared to group C and BA (p<0.05). After the second BA application, I/O numbers increased in the ABA group compared to the A group (p<0.05). In group A, PON-1, TOS and OSI levels were higher and TAS levels were lower than in groups BA and C. After BA treatment, PON-1 and OSI levels were lower in the ABA group than in the A group (p<0.05). Although there was an increase in TAS value and a decrease in TOS, this did not make a statistical difference. The thickness of the pyramidal cell in CA1 and the granular cell layers in the dentate gyrus, and the number of intact and degenerated neurons in the pyramidal cell layer were similar between the groups. Discussion Significant improvement in learning and memory abilities after BA application is promising for AD. Conclusion These results show that BA application positively affects learning and memory abilities, and reduces oxidative stress. More extensive studies are required to evaluate histopathological efficacy.
Collapse
Affiliation(s)
- Çağrı Özdemir
- Mamak State Hospital Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Mustafa Arslan
- Gazi University Faculty of Medicine Department of Anesthesiology and Reanimation, Ankara, Turkey
- Gazi University, Life Sciences Application and Research Center, Ankara, Turkey
- Gazi University, Laboratory Animal Breeding and Experimental Research Center (GÜDAM), Ankara, Turkey
| | - Ayşegül Küçük
- Kutahya Health Sciences University Faculty of Medicine, Department of Physiology, Kutahya, Turkey
| | - Zeynep Yığman
- Gazi University Faculty of Medicine, Department of Histology and Embryology, Ankara, Turkey
- Gazi University Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey
| | - Ali Doğan Dursun
- Atılım University Faculty of Medicine Department of Physiology, Ankara, Turkey
| |
Collapse
|
29
|
Xiong J, Lv Y, Ma X, Peng G, Wu C, Hou J, Zhang Y, Wu C, Chen-Yi Liu T, Yang L. Neuroprotective Effect of Sub-lethal Hyperthermia Preconditioning in a Rat Model of Repeated Closed Head Injury. Neuroscience 2023; 522:57-68. [PMID: 37164305 DOI: 10.1016/j.neuroscience.2023.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/28/2023] [Accepted: 04/29/2023] [Indexed: 05/12/2023]
Abstract
Repeated mild traumatic brain injury (rTBI), one of the most common forms of traumatic brain injury, is a worldwide severe public health concern. rTBI induces cumulative neuronal injury, neurological dysfunction, and cognitive deficits. Although there are clinical treatment methods, there is still an urgent need to develop preventive approaches for susceptible populations. Using a repeated closed head injury (rCHI) rat model, we interrogate the effect of sub-lethal hyperthermia preconditioning (SHP) on rCHI-induced neuronal injury and behavioral changes. Our study applied the repeated weight-drop model to induce the rCHI. According to the changes of heat shock protein 70 (HSP 70) in the cortex and hippocampus following a single SHP treatment in normal rats, the SHP was delivered to the rats 18 hours before rCHI. We found that HSP significantly alleviated rCHI-induced anxiety-like behaviors and impairments in motor abilities and spatial memory. SHP exerts significant neuroprotection against rCHI-induced neuronal damage, apoptosis, and neuroinflammation. Our findings support the potential use of SHP as a preventative approach for alleviating rCHI-induced brain damage.
Collapse
Affiliation(s)
- Jing Xiong
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China; Guangzhou Cadre Health Management Center, Guangzhou, China 510006, China
| | - Ying Lv
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Xu Ma
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Guangcong Peng
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Chunyi Wu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Jun Hou
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Yulan Zhang
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China
| | - Chongyun Wu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| | - Timon Chen-Yi Liu
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| | - Luodan Yang
- Collage of Physical Education and Sport Science, South China Normal University, Guangzhou, China 510006, China.
| |
Collapse
|
30
|
Wu C, Zou P, Feng S, Zhu L, Li F, Liu TCY, Duan R, Yang L. Molecular Hydrogen: an Emerging Therapeutic Medical Gas for Brain Disorders. Mol Neurobiol 2023; 60:1749-1765. [PMID: 36567361 DOI: 10.1007/s12035-022-03175-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022]
Abstract
Oxidative stress and neuroinflammation are the main physiopathological changes involved in the initiation and progression of various neurodegenerative disorders or brain injuries. Since the landmark finding reported in 2007 found that hydrogen reduced the levels of peroxynitrite anions and hydroxyl free radicals in ischemic stroke, molecular hydrogen's antioxidative and anti-inflammatory effects have aroused widespread interest. Due to its excellent antioxidant and anti-inflammatory properties, hydrogen therapy via different routes of administration exhibits great therapeutic potential for a wide range of brain disorders, including Alzheimer's disease, neonatal hypoxic-ischemic encephalopathy, depression, anxiety, traumatic brain injury, ischemic stroke, Parkinson's disease, and multiple sclerosis. This paper reviews the routes for hydrogen administration, the effects of hydrogen on the previously mentioned brain disorders, and the primary mechanism underlying hydrogen's neuroprotection. Finally, we discuss hydrogen therapy's remaining issues and challenges in brain disorders. We conclude that understanding the exact molecular target, finding novel routes, and determining the optimal dosage for hydrogen administration is critical for future studies and applications.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Fanghui Li
- School of Sports Science, Nanjing Normal University, Nanjing, 210046, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
31
|
Zhu C, Zhang T, Li Q, Chen X, Wang K. Depression and Anxiety During the COVID-19 Pandemic: Epidemiology, Mechanism, and Treatment. Neurosci Bull 2023; 39:675-684. [PMID: 36411394 PMCID: PMC9685018 DOI: 10.1007/s12264-022-00970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic has had an adverse impact on the physical and mental health of the public worldwide. In addition to illness in patients with COVID-19, isolated people and the general population have experienced mental health problems due to social distancing policies, mandatory lockdown, and other psychosocial factors, and the prevalence of depression and anxiety significantly increased during the pandemic. The purpose of this review is to elucidate the epidemiology, contributing factors, and pathogenesis of depression and anxiety. during the pandemic. These findings indicate that physicians and psychiatrists should pay more attention to and identify those with a high risk for mental problems, such as females, younger people, unmarried people, and those with a low educational level. In addition, researchers should focus on identifying the neural and neuroimmune mechanisms involved in depression and anxiety, and assess the intestinal microbiome to identify effective biomarkers. We also provide an overview of various intervention methods, including pharmacological treatment, psychological therapy, and physiotherapy, to provide a reference for different populations to guide the development of optimized intervention methods.
Collapse
Affiliation(s)
- Chunyan Zhu
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
| | - Ting Zhang
- Department of Psychiatry, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qianqian Li
- Department of Psychiatry, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xingui Chen
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Kai Wang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230032, China.
| |
Collapse
|
32
|
Wang M, Zhang H, Liang J, Huang J, Chen N. Exercise suppresses neuroinflammation for alleviating Alzheimer's disease. J Neuroinflammation 2023; 20:76. [PMID: 36935511 PMCID: PMC10026496 DOI: 10.1186/s12974-023-02753-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 02/28/2023] [Indexed: 03/21/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, with the characteristics of neurofibrillary tangle (NFT) and senile plaque (SP) formation. Although great progresses have been made in clinical trials based on relevant hypotheses, these studies are also accompanied by the emergence of toxic and side effects, and it is an urgent task to explore the underlying mechanisms for the benefits to prevent and treat AD. Herein, based on animal experiments and a few clinical trials, neuroinflammation in AD is characterized by long-term activation of pro-inflammatory microglia and the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes. Damaged signals from the periphery and within the brain continuously activate microglia, thus resulting in a constant source of inflammatory responses. The long-term chronic inflammatory response also exacerbates endoplasmic reticulum oxidative stress in microglia, which triggers microglia-dependent immune responses, ultimately leading to the occurrence and deterioration of AD. In this review, we systematically summarized and sorted out that exercise ameliorates AD by directly and indirectly regulating immune response of the central nervous system and promoting hippocampal neurogenesis to provide a new direction for exploring the neuroinflammation activity in AD.
Collapse
Affiliation(s)
- Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| | - Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| | - Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
33
|
Ayari S, Abellard A, Carayol M, Guedj É, Gavarry O. A systematic review of exercise modalities that reduce pro-inflammatory cytokines in humans and animals' models with mild cognitive impairment or dementia. Exp Gerontol 2023; 175:112141. [PMID: 36898593 DOI: 10.1016/j.exger.2023.112141] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/21/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
PURPOSE To investigate which type, frequency, duration, intensity, and volume of chronic exercise might more strongly reduce pro-inflammatory cytokines and enhance anti-inflammatory cytokines in human and animal models with Mild Cognitive Impairment (MCI) or dementia. DESIGN A systematic review. DATA SOURCE English-language search of 13 electronic databases: Web of Science, PubMed/Medline, Sport Discus, Scopus, Cochrane, Psych Net, Springer, ScienceDirect, Pascal & Francis, Sage journals, Pedro, Google Scholar, and Sage. INCLUSION CRITERIA (i) human and animal studies that included exercise, physical activity, or fitness training as an experimental intervention, (ii) studies that addressed MCI, dementia, or AD, (iii) studies that focused on measuring cytokines and/or other inflammatory and/or neuroinflammatory immune markers, (iii) studies that examined inflammatory indicators in blood, CSF (Cerebrospinal Fluid), and brain tissue. RESULTS Of the 1290 human and animal studies found, 38 were included for qualitative analysis, 11 human articles, 27 animal articles, and two articles addressing both human and animal protocols. In the animal model, physical exercise decreased pro-inflammatory markers in 70.8 % of the articles and anti-inflammatory cytokines: IL -4, IL -10, IL-4β, IL -10β, and TGF-β in 26 % of articles. Treadmill running, resistance exercise, and swimming exercise reduce pro-inflammatory cytokines and increase anti-inflammatory cytokines. In the human model, 53.9 % of items reduced pro-inflammatory proteins and 23 % increased anti-inflammatory proteins. Cycling exercise, multimodal, and resistance training effectively decreased pro-inflammatory cytokines. CONCLUSION In rodent animal models with AD phenotype, treadmill, swimming, and resistance training remain good interventions that can delay various mechanisms of dementia progression. In the human model, aerobic, multimodal, and resistance training are beneficial in both MCI and AD. Multimodal training of moderate to high intensity multimodal exercise is effective for MCI. Voluntary cycling training, moderate- or high-intensity aerobic exercise is effective in mild AD patients.
Collapse
Affiliation(s)
- Sawsen Ayari
- Research Unit "Impact of Physical Activity on Health" (IAPS n°201723207F), University of Toulon, Toulon, France.
| | - Alexandre Abellard
- Mediterranean Institute of Information and Communication Sciences, Toulon, France.
| | - Marion Carayol
- Research Unit "Impact of Physical Activity on Health" (IAPS n°201723207F), University of Toulon, Toulon, France.
| | - Éric Guedj
- APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, CERIMED, Nuclear Medicine Department, Aix-Marseille University, Marseille, France.
| | - Olivier Gavarry
- Research Unit "Impact of Physical Activity on Health" (IAPS n°201723207F), University of Toulon, Toulon, France.
| |
Collapse
|
34
|
Kumar H, Green R, Cornfeld DM, Condron P, Emsden T, Elsayed A, Zhao D, Gilbert K, Nash MP, Clark AR, Tawhai MH, Burrowes K, Murphy R, Tayebi M, McGeown J, Kwon E, Shim V, Wang A, Choisne J, Carman L, Besier T, Handsfield G, Babarenda Gamage TP, Shen J, Maso Talou G, Safaei S, Maller JJ, Taylor D, Potter L, Holdsworth SJ, Wilson GA. Roadmap for an imaging and modelling paediatric study in rural NZ. Front Physiol 2023; 14:1104838. [PMID: 36969588 PMCID: PMC10036853 DOI: 10.3389/fphys.2023.1104838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/30/2023] [Indexed: 03/12/2023] Open
Abstract
Our study methodology is motivated from three disparate needs: one, imaging studies have existed in silo and study organs but not across organ systems; two, there are gaps in our understanding of paediatric structure and function; three, lack of representative data in New Zealand. Our research aims to address these issues in part, through the combination of magnetic resonance imaging, advanced image processing algorithms and computational modelling. Our study demonstrated the need to take an organ-system approach and scan multiple organs on the same child. We have pilot tested an imaging protocol to be minimally disruptive to the children and demonstrated state-of-the-art image processing and personalized computational models using the imaging data. Our imaging protocol spans brain, lungs, heart, muscle, bones, abdominal and vascular systems. Our initial set of results demonstrated child-specific measurements on one dataset. This work is novel and interesting as we have run multiple computational physiology workflows to generate personalized computational models. Our proposed work is the first step towards achieving the integration of imaging and modelling improving our understanding of the human body in paediatric health and disease.
Collapse
Affiliation(s)
- Haribalan Kumar
- Mātai Medical Research Institute, Gisborne, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- GE Healthcare (Australia & New Zealand), Auckland, New Zealand
| | - Robby Green
- Mātai Medical Research Institute, Gisborne, New Zealand
| | - Daniel M. Cornfeld
- Mātai Medical Research Institute, Gisborne, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Paul Condron
- Mātai Medical Research Institute, Gisborne, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Taylor Emsden
- Mātai Medical Research Institute, Gisborne, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Ayah Elsayed
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Auckland University of Technology, Auckland, New Zealand
| | - Debbie Zhao
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Kat Gilbert
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Martyn P. Nash
- Mātai Medical Research Institute, Gisborne, New Zealand
- Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | - Alys R. Clark
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Merryn H. Tawhai
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Kelly Burrowes
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Rinki Murphy
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Maryam Tayebi
- Mātai Medical Research Institute, Gisborne, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Josh McGeown
- Mātai Medical Research Institute, Gisborne, New Zealand
| | - Eryn Kwon
- Mātai Medical Research Institute, Gisborne, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alan Wang
- Mātai Medical Research Institute, Gisborne, New Zealand
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Julie Choisne
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Laura Carman
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Thor Besier
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Geoffrey Handsfield
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | | | - Jiantao Shen
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Gonzalo Maso Talou
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jerome J. Maller
- GE Healthcare (Australia & New Zealand), Auckland, New Zealand
- Monash Alfred Psychiatry Research Centre, Melbourne, VIC, Australia
| | | | - Leigh Potter
- Mātai Medical Research Institute, Gisborne, New Zealand
| | - Samantha J. Holdsworth
- Mātai Medical Research Institute, Gisborne, New Zealand
- Faculty of Medical and Health Sciences, Centre for Brain Research, University of Auckland, Auckland, New Zealand
- *Correspondence: Samantha J. Holdsworth,
| | | |
Collapse
|
35
|
Guo L, Yang X, Zhang Y, Xu X, Li Y. Effect of exercise on cognitive function and synaptic plasticity in Alzheimer's disease models: A systematic review and meta-analysis. Front Aging Neurosci 2023; 14:1077732. [PMID: 36704501 PMCID: PMC9872519 DOI: 10.3389/fnagi.2022.1077732] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023] Open
Abstract
Introduction Cognitive decline is a central manifestation of Alzheimer's disease (AD), and its process is inseparable from changes in synaptic plasticity. The aim of this review was to summarize and evaluate the effectiveness of exercise on cognitive function and synaptic plasticity in AD animal models. Materials and methods Eligible studies were searched from PubMed, MEDLINE, EMBASE, Web of Science, and Cochrane Library from April to May 2022. The risk of bias was evaluated by Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE). The Morris water maze (MWM) test and synaptic plasticity were considered outcome measures. Data were analyzed using random-effects meta-analyses using the software Stata. Heterogeneity was examined by using I2 test. Sensitivity analysis and publication bias were also assessed. Results A total of 20 randomized controlled studies were eligible for study inclusion. Compared with controls, exercise decreased escape latency (SMD = -0.86, 95% CI: -1.21 to -0.50, P < 0.001), increased platform crossover numbers (SMD = 1.34, 95% CI: 0.57-2.11, P = 0.001) and time in the target quadrant (SMD = 1.65, 95% CI: 0.95-2.36, P < 0.001) and the expression of PSD95 (SMD = 0.73, 95% CI: 0.25-1.21, P = 0.003) in AD animals. The results of the subgroup analysis showed that exercise before AD had a greater effect on escape latency (SMD = -0.88, 95% CI: -1.25 to -0.52, P < 0.001), platform crossover numbers (SMD = 1.71, 95% CI: 1.23-2.18, P < 0.001), time in the target quadrant (SMD = 2.03, 95% CI: 1.19-2.87, P < 0.001) and the expression of PSD95 (SMD = 0.94, 95% CI: 0.19-1.69, P = 0.014) than exercise after AD. The results of the subgroup analysis also showed that treadmill running might be an appropriate exercise type. Conclusion Our findings suggested that exercise had a potential effect on improving cognitive function and synaptic plasticity. It can play a better neuroprotective role before AD. Systematic review registration PROSPERO, identifier: CRD42022328438.
Collapse
Affiliation(s)
- Linlin Guo
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Xinxin Yang
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Zhang
- College of Nursing, Hebei Medical University, Shijiazhuang, China
| | - Xinyi Xu
- College of Nursing, Hebei Medical University, Shijiazhuang, China,Postdoctoral Research Station in Basic Medicine, Hebei Medical University, Shijiazhuang, China,*Correspondence: Xinyi Xu ✉
| | - Yan Li
- College of Nursing, Hebei Medical University, Shijiazhuang, China,Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China,*Correspondence: Yan Li ✉
| |
Collapse
|
36
|
Azevedo CV, Hashiguchi D, Campos HC, Figueiredo EV, Otaviano SFSD, Penitente AR, Arida RM, Longo BM. The effects of resistance exercise on cognitive function, amyloidogenesis, and neuroinflammation in Alzheimer's disease. Front Neurosci 2023; 17:1131214. [PMID: 36937673 PMCID: PMC10017453 DOI: 10.3389/fnins.2023.1131214] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
With the increasing prevalence of Alzheimer's disease (AD) and difficulties in finding effective treatments, it is essential to discover alternative therapies through new approaches. In this regard, non-pharmacological therapies, such as physical exercise, have been proposed and explored for the treatment of AD. Recent studies have suggested that resistance exercise (RE) is an effective strategy for promoting benefits in memory and cognitive function, producing neuroprotective and anti-inflammatory effects, and reducing amyloid load and plaques, thereby reducing the risk, and alleviating the neurodegeneration process of AD and other types of dementia in the elderly. In addition, RE is the exercise recommended by the World Health Organization for the elderly due to its benefits in improving muscle strength and balance, and increasing autonomy and functional capacity, favoring improvements in the quality of life of the elderly population, who is more likely to develop AD and other types of dementia. In this mini-review, we discuss the impact of RE on humans affected by MCI and AD, and animal models of AD, and summarize the main findings regarding the effects of RE program on memory and cognitive functions, neurotrophic factors, Aβ deposition and plaque formation, as well as on neuroinflammation. Overall, the present review provides clinical and preclinical evidence that RE plays a role in alleviating AD symptoms and may help to understand the therapeutic potential of RE, thereby continuing the advances in AD therapies.
Collapse
Affiliation(s)
| | - Debora Hashiguchi
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte (UFRN), Natal, Brazil
| | | | | | | | - Arlete Rita Penitente
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Escola de Medicina, Departamento de Ginecologia Obstetrícia e Propedêutica da, Universidade Federal de Ouro Preto (UFOP), Minas Gerais, Brazil
| | - Ricardo Mario Arida
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz Monteiro Longo
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Beatriz Monteiro Longo,
| |
Collapse
|
37
|
Zhou R, Wang Z, Zhou B, Yu Z, Wu C, Hou J, Cheng K, Liu TC. Estrogen receptors mediate the antidepressant effects of aerobic exercise: A possible new mechanism. Front Aging Neurosci 2022; 14:1040828. [PMID: 36570542 PMCID: PMC9780551 DOI: 10.3389/fnagi.2022.1040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose This study aimed to examine whether aerobic exercise exerts mood-modulating effects through an estrogen signaling mechanism. Method The experiment was divided into two parts. The first part is to compare the three modeling methods to obtain the most obvious method of depression-like phenotype for further study in the second part. The first part of ovariectomized rats (age, 13 weeks) was tested when rats were 14 or 22 weeks old or in the sixth week after 3 weeks of chronic restraint stress. The second part was to treat the animals with the most obvious depression-like phenotype in different ways, placebo treatment or estradiol (E2) replacement therapy was administered, aerobic training, or estrogen receptor antagonist treatment. The cognitive (Barnes maze and 3-chamber social tests), anxiety-like (open-field and elevated plus maze tests) and depression-like (sucrose preference and forced swim tests) behaviors of rats in both parts were analyzed to study the effects of estrogen depletion and aerobic exercise. Results Rats did not develop depressive symptoms immediately after ovariectomy, however, the symptoms became more pronounced with a gradual decrease in ovarian hormone levels. Compared with the placebo or control groups, the exercise and E2 groups showed improved performance in all behavioral test tasks, and the antidepressant effects of aerobic exercise were comparable to those of estrogen. Moreover, the estrogen receptor antagonist has markedly inhibited the antidepressant effects of aerobic exercise. Conclusion Estrogen receptors may mediate the antidepressant effects of aerobic exercise. In addition, an increasingly fragile ovarian hormonal environment may underlies chronic restraint stress-induced depression.
Collapse
|
38
|
Performance of the intracerebroventricularly injected streptozotocin Alzheimer's disease model in a translationally relevant, aged and experienced rat population. Sci Rep 2022; 12:20247. [PMID: 36424423 PMCID: PMC9691696 DOI: 10.1038/s41598-022-24292-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
The intracerebroventricularly (icv) injected streptozotocin (STZ) induced brain state is a widely used model of sporadic Alzheimer-disease (AD). However, data have been generated in young, naive albino rats. We postulate that the translationally most relevant animal population of an AD model should be that of aged rats with substantial learning history. The objective of the study was thus to probe the model in old rats with knowledge in various cognitive domains. Long-Evans rats of 23 and 10 months age with acquired knowledge in five-choice serial reaction time task (5-CSRTT), a cooperation task, Morris water-maze (MWM) and "pot-jumping" exercise were treated with 3 × 1.5 mg/kg icv. STZ and their performance were followed for 3 months in the above and additional behavioral assays. Both STZ-treated age groups showed significant impairment in the MWM (spatial learning) and novel object recognition test (recognition memory) but not in passive avoidance and fear conditioning paradigms (fear memory). In young STZ treated rats, significant differences were also found in the 5CSRTT (attention) and pot jumping test (procedural learning) while in old rats a significant increase in hippocampal phospho-tau/tau protein ratio was observed. No significant difference was found in the cooperation (social cognition) and pairwise discrimination (visual memory) assays and hippocampal β-amyloid levels. STZ treated old animals showed impulsivity-like behavior in several tests. Our results partly coincide with partly deviate from those published on young, albino, unexperienced rats. Beside the age, strain and experience level of the animals differences can also be attributed to the increased dose of STZ, and the applied food restriction regime. The observed cognitive and non-cognitive activity pattern of icv. STZ in aged experienced rats call for more extensive studies with the STZ model to further strengthen and specify its translational validity.
Collapse
|
39
|
Huang Z, Lin HW(K, Zhang Q, Zong X. Targeting Alzheimer's Disease: The Critical Crosstalk between the Liver and Brain. Nutrients 2022; 14:nu14204298. [PMID: 36296980 PMCID: PMC9609624 DOI: 10.3390/nu14204298] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disorder, is currently incurable. Imbalanced amyloid-beta (Aβ) generation and clearance are thought to play a pivotal role in the pathogenesis of AD. Historically, strategies targeting Aβ clearance have typically focused on central clearance, but with limited clinical success. Recently, the contribution of peripheral systems, particularly the liver, to Aβ clearance has sparked an increased interest. In addition, AD presents pathological features similar to those of metabolic syndrome, and the critical involvement of brain energy metabolic disturbances in this disease has been recognized. More importantly, the liver may be a key regulator in these abnormalities, far beyond our past understanding. Here, we review recent animal and clinical findings indicating that liver dysfunction represents an early event in AD pathophysiology. We further propose that compromised peripheral Aβ clearance by the liver and aberrant hepatic physiological processes may contribute to AD neurodegeneration. The role of a hepatic synthesis product, fibroblast growth factor 21 (FGF21), in the management of AD is also discussed. A deeper understanding of the communication between the liver and brain may lead to new opportunities for the early diagnosis and treatment of AD.
Collapse
|
40
|
Wu C, Yang L, Feng S, Zhu L, Yang L, Liu TCY, Duan R. Therapeutic non-invasive brain treatments in Alzheimer's disease: recent advances and challenges. Inflamm Regen 2022; 42:31. [PMID: 36184623 PMCID: PMC9527145 DOI: 10.1186/s41232-022-00216-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) is one of the major neurodegenerative diseases and the most common form of dementia. Characterized by the loss of learning, memory, problem-solving, language, and other thinking abilities, AD exerts a detrimental effect on both patients' and families' quality of life. Although there have been significant advances in understanding the mechanism underlying the pathogenesis and progression of AD, there is no cure for AD. The failure of numerous molecular targeted pharmacologic clinical trials leads to an emerging research shift toward non-invasive therapies, especially multiple targeted non-invasive treatments. In this paper, we reviewed the advances of the most widely studied non-invasive therapies, including photobiomodulation (PBM), transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), and exercise therapy. Firstly, we reviewed the pathological changes of AD and the challenges for AD studies. We then introduced these non-invasive therapies and discussed the factors that may affect the effects of these therapies. Additionally, we review the effects of these therapies and the possible mechanisms underlying these effects. Finally, we summarized the challenges of the non-invasive treatments in future AD studies and clinical applications. We concluded that it would be critical to understand the exact underlying mechanisms and find the optimal treatment parameters to improve the translational value of these non-invasive therapies. Moreover, the combined use of non-invasive treatments is also a promising research direction for future studies and sheds light on the future treatment or prevention of AD.
Collapse
Affiliation(s)
- Chongyun Wu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luoman Yang
- Department of Anesthesiology, Peking University Third Hospital (PUTH), Beijing, 100083, China
| | - Shu Feng
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Ling Zhu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
41
|
Yang L, Feng S, Wu C, Yang L. Microglia-Mediated Aβ Propagation in Alzheimer's Disease. Neurosci Bull 2022; 38:1274-1276. [PMID: 35729454 PMCID: PMC9554111 DOI: 10.1007/s12264-022-00907-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022] Open
Affiliation(s)
- Luoman Yang
- Department of Anesthesiology, Peking University Third Hospital, Beijing, 100083, China
| | - Shu Feng
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA.
| |
Collapse
|
42
|
de Almeida EJR, Ibrahim HJ, Chitolina Schetinger MR, de Andrade CM, Cardoso AM. Modulation of Inflammatory Mediators and Microglial Activation Through Physical Exercise in Alzheimer's and Parkinson's Diseases. Neurochem Res 2022; 47:3221-3240. [PMID: 35962936 DOI: 10.1007/s11064-022-03713-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/19/2022]
Abstract
Neuroinflammation is an inflammatory process in the central nervous system (CNS), in addition to being one of the main features of Alzheimer's disease (AD) and Parkinson's disease (PD). Microglia are known for their immune functions and have multiple reactive phenotypes related to the types of stages involving neurodegenerative diseases. Depending on the state of activation of microglia in the CNS, it can be neuroprotective or neurotoxic. In this context, AD is a neurodegenerative and neuroinflammatory disease characterized by the deposition of beta-amyloid plaques, formation of fibrillar tangles of tau protein, and loss of neurons due to neurotoxic activation of microglia. However, PD is characterized by the loss of dopaminergic neurons in the substantia nigra and accumulation of alpha-synuclein in the cortical regions, spinal cord, and brain stem, which occurs by microglial activation, contributing to the neuroinflammatory process. In this aspect, the activation of microglia in both pathologies triggers high levels of inflammatory markers, such as interleukins, and causes the neuroinflammatory process of the diseases. Thus, physical exercise is pointed out as neuroprotective, as it can act to strengthen neurogenesis and reduce the inflammatory process. Therefore, the present review addresses the neuroprotective effect of microglia after different types of physical exercise protocols and evaluates the activity and effects of inflammatory and anti-inflammatory parameters and mechanisms of AD and PD. This review will discuss the anti-inflammatory effects of physical exercise through microglia activation with neuroprotective activity and the role of pro-and anti-inflammatory cytokines in AD and PD.
Collapse
Affiliation(s)
| | | | | | - Cinthia Melazzo de Andrade
- Department of Small Animal Clinic, Center of Rural Sciences, Federal University of Santa Maria-RS, Room 121, Veterinary Hospital Building, Avenue Roraima No. 1000, Santa Maria, RS, 97105-900, Brazil
| | - Andréia Machado Cardoso
- Graduate Program in Physical Education From Federal University of Santa Maria, Santa Maria, RS, Brazil. .,Graduate Program in Biomedical Sciences From Federal University of Fronteira Sul, Campus Chapecó, Rodovia SC 484 - Km 02, Fronteira Sul, SC, 89815-899, Brazil.
| |
Collapse
|
43
|
Wu CC, Xiong HY, Zheng JJ, Wang XQ. Dance movement therapy for neurodegenerative diseases: A systematic review. Front Aging Neurosci 2022; 14:975711. [PMID: 36004000 PMCID: PMC9394857 DOI: 10.3389/fnagi.2022.975711] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe proportion of the world's elderly population continues to rise, and the treatment and improvement of neurodegenerative diseases have become issue of public health importance as people live longer and many countries have aging populations. This systematic review aims to discuss the effects of dance movement therapy (DMT) on motor function, cognitive deficit, mood, and quality of life in people with neurodegenerative diseases, such as Parkinson's disease (PD), mild cognitive impairment (MCI), Alzheimer's disease (AD).MethodsTwo reviewers independently conducted systematic search on the Cochrane library, PubMed database, Web of Science Core Collection database, and Physiotherapy Evidence database until February 1, 2022. Only systematic analyses and randomized controlled trials were included and further analyzed.ResultsThirty-three studies on PD, 16 studies on MCI, 4 studies on AD were obtained. This systematic review found that DMT substantially improved the global cognitive function, memory, and executive function on the population with MCI. Compared with the non-dance group, DMT remarkably improved general disease condition, balance, and gait for individuals with PD. The evidence of the efficacy of DMT on AD is insufficient, and further research is needed.ConclusionDMT can effectively improve the motor function and cognitive deficits in neurodegenerative diseases. Positive effects of DMT on the mood and quality of life in ND patients are controversial and require further evidence. Future research on the effects of DMT on AD requires scientific design, large sample size, long-term comprehensive intervention, and clear reporting standards.Systematic review registrationwww.osf.io/wktez, identifier: 10.17605/OSF.IO/UYBKT.
Collapse
Affiliation(s)
- Cheng-Cheng Wu
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Huan-Yu Xiong
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Jie-Jiao Zheng
- Huadong Hospital, Shanghai, China
- *Correspondence: Jie-Jiao Zheng
| | - Xue-Qiang Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China
- Xue-Qiang Wang
| |
Collapse
|
44
|
Effects of an Exercise Program Combining Aerobic and Resistance Training on Protein Expressions of Neurotrophic Factors in Obese Rats Injected with Beta-Amyloid. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19137921. [PMID: 35805580 PMCID: PMC9266049 DOI: 10.3390/ijerph19137921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022]
Abstract
In this study, the effects of a 12-week exercise program combining aerobic and resistance training on high-fat diet-induced obese Sprague Dawley (SD) rats after the injection of beta-amyloid into the cerebral ventricle were investigated. Changes in physical fitness, cognitive function, blood levels of beta-amyloid and metabolic factors, and protein expressions of neurotrophic factors related to brain function such as BDNF (brain-derived neurotrophic factor) in the quadriceps femoris, hippocampus, and cerebral cortex were analyzed. The subjects were thirty-two 10-week-old SD rats (DBL Co., Ltd., Seoul, Korea). The rats were randomized into four groups: β-Non-Ex group (n = 8) with induced obesity and βA25-35 injection into the cerebral ventricle through stereotactic biopsy; β-Ex group (n = 8) with induced obesity, βA25-35 injection, and exercise; S-Non-Ex group (n = 8) with an injection of saline in lieu of βA25-35 as the control; and S-Ex group (n = 8) with saline injection and exercise. The 12-week exercise program combined aerobic training and resistance training. As for protein expressions of the factors related to brain function, the combined exercise program was shown to have a clear effect on activating the following factors: PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha), FNDC5 (fibronectin type III domain-containing protein 5), and BDNF in the quadriceps femoris; TrkB (Tropomyosin receptor kinase B), FNDC5, and BDNF in the hippocampus; PGC-1α, FNDC5, and BDNF in the cerebral cortex. The protein expression of β-amyloid in the cerebral cortex was significantly lower in the β-Ex group than in the β-Non-Ex group (p < 0.05). The 12-week intervention with the combined exercise program of aerobic and resistance training was shown to improve cardiopulmonary function, muscular endurance, and short-term memory. The results demonstrate a set of positive effects of the combined exercise program, which were presumed to have arisen mainly due to its alleviating effect on β-amyloid plaques, the main cause of reduced brain function, as well as the promotion of protein expressions of PGC-1α, FNDC5, and BDNF in the quadriceps femoris, hippocampus, and cerebral cortex.
Collapse
|
45
|
Prophylactic Zinc Administration Combined with Swimming Exercise Prevents Cognitive-Emotional Disturbances and Tissue Injury following a Transient Hypoxic-Ischemic Insult in the Rat. Behav Neurol 2022; 2022:5388944. [PMID: 35637877 PMCID: PMC9146809 DOI: 10.1155/2022/5388944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
Exercise performance and zinc administration individually yield a protective effect on various neurodegenerative models, including ischemic brain injury. Therefore, this work was aimed at evaluating the combined effect of subacute prophylactic zinc administration and swimming exercise in a transient cerebral ischemia model. The prophylactic zinc administration (2.5 mg/kg of body weight) was provided every 24 h for four days before a 30 min common carotid artery occlusion (CCAO), and 24 h after reperfusion, the rats were subjected to swimming exercise in the Morris Water Maze (MWM). Learning was evaluated daily for five days, and memory on day 12 postreperfusion; anxiety or depression-like behavior was measured by the elevated plus maze and the motor activity by open-field test. Nitrites, lipid peroxidation, and the activity of superoxide dismutase (SOD) and catalase (CAT) were assessed in the temporoparietal cortex and hippocampus. The three nitric oxide (NO) synthase isoforms, chemokines, and their receptor levels were measured by ELISA. Nissl staining evaluated hippocampus cytoarchitecture and Iba-1 immunohistochemistry activated the microglia. Swimming exercise alone could not prevent ischemic damage but, combined with prophylactic zinc administration, reversed the cognitive deficit, decreased NOS and chemokine levels, prevented tissue damage, and increased Iba-1 (+) cell number. These results suggest that the subacute prophylactic zinc administration combined with swimming exercise, but not the individual treatment, prevents the ischemic damage on day 12 postreperfusion in the transient ischemia model.
Collapse
|
46
|
Physical-Exercise-Induced Antioxidant Effects on the Brain and Skeletal Muscle. Antioxidants (Basel) 2022; 11:antiox11050826. [PMID: 35624690 PMCID: PMC9138070 DOI: 10.3390/antiox11050826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Erythroid-related nuclear factor 2 (NRF2) and the antioxidant-responsive-elements (ARE) signaling pathway are the master regulators of cell antioxidant defenses, playing a key role in maintaining cellular homeostasis, a scenario in which proper mitochondrial function is essential. Increasing evidence indicates that the regular practice of physical exercise increases cellular antioxidant defenses by activating NRF2 signaling. This manuscript reviewed classic and ongoing research on the beneficial effects of exercise on the antioxidant system in both the brain and skeletal muscle.
Collapse
|
47
|
Effects of Involuntary and Voluntary Exercise in Combination with Acousto-Optic Stimulation on Adult Neurogenesis in an Alzheimer's Mouse Model. Mol Neurobiol 2022; 59:3254-3279. [PMID: 35297012 DOI: 10.1007/s12035-022-02784-9] [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: 11/07/2021] [Accepted: 02/10/2022] [Indexed: 10/18/2022]
Abstract
Single-factor intervention, such as physical exercise and auditory and visual stimulation, plays a positive role on the prevention and treatment of Alzheimer's disease (AD); however, the therapeutic effects of single-factor intervention are limited. The beneficial effects of these multifactor combinations on AD and its molecular mechanism have yet to be elucidated. Here, we investigated the effect of multifactor intervention, voluntary wheel exercise, and involuntary treadmill running in combination with acousto-optic stimulation, on adult neurogenesis and behavioral phenotypes in a mouse model of AD. We found that 4 weeks of multifactor intervention can significantly increase the production of newborn cells (BrdU+ cells) and immature neurons (DCX+ cells) in the hippocampus and lateral ventricle of Aβ oligomer-induced mice. Importantly, the multifactor intervention could promote BrdU+ cells to differentiate into neurons (BrdU+ DCX+ cells or BrdU+ NeuN+ cells) and astrocytes (BrdU+GFAP+ cells) in the hippocampus and ameliorate Aβ oligomer-induced cognitive impairment and anxiety- and depression-like behaviors in mice evaluated by novel object recognition, Morris water maze tests, elevated zero maze, forced swimming test, and tail suspension test, respectively. Moreover, multifactor intervention could lead to an increase in the protein levels of PSD-95, SYP, DCX, NeuN, GFAP, Bcl-2, BDNF, TrkB, and pSer473-Akt and a decrease in the protein levels of BAX and caspase-9 in the hippocampal lysates of Aβ oligomer-induced mice. Furthermore, sequencing analysis of serum metabolites revealed that aberrantly expressed metabolites modulated by multifactor intervention were highly enriched in the biological process associated with keeping neurons functioning and neurobehavioral function. Additionally, the intervention-mediated serum metabolites mainly participated in glutamate metabolism, glucose metabolism, and the tricarboxylic acid cycle in mice. Our findings suggest the potential of multifactor intervention as a non-invasive therapeutic strategy for AD to anti-Aβ oligomer neurotoxicity.
Collapse
|
48
|
Liu ZT, Ma YT, Pan ST, Xie K, Shen W, Lin SY, Gao JY, Li WY, Li GY, Wang QW, Li LP. Effects of involuntary treadmill running in combination with swimming on adult neurogenesis in an Alzheimer's mouse model. Neurochem Int 2022; 155:105309. [PMID: 35276288 DOI: 10.1016/j.neuint.2022.105309] [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: 10/28/2021] [Revised: 01/24/2022] [Accepted: 02/15/2022] [Indexed: 10/18/2022]
Abstract
Physical exercise plays a role on the prevention and treatment of Alzheimer's disease (AD), but the exercise mode and the mechanism for these positive effects is still ambiguous. Here, we investigated the effect of an aerobic interval exercise, running in combination with swimming, on behavioral dysfunction and associated adult neurogenesis in a mouse model of AD. We demonstrate that 4 weeks of the exercise could ameliorate Aβ42 oligomer-induced cognitive impairment in mice utilizing Morris water maze tests. Additionally, the exercised Aβ42 oligomer-induced mice exhibited a significant reduction of anxiety- and depression-like behaviors compared to the sedentary Aβ42 oligomer-induced mice utilizing an Elevated zero maze and a Tail suspension test. Moreover, by utilizing 5'-bromodeoxyuridine (BrdU) as an exogenous cell tracer, we found that the exercised Aβ42 oligomer-induced mice displayed a significant increase in newborn cells (BrdU+ cells), which differentiated into a majority of neurons (BrdU+ DCX+ cells or BrdU+NeuN+ cells) and a few of astrocytes (BrdU+GFAP+ cells). Likewise, the exercised Aβ42 oligomer-induced mice also displayed the higher levels of NeuN, PSD95, synaptophysin, Bcl-2 and lower level of GFAP protein. Furthermore, alteration of serum metabolites in transgenic AD mice between the exercised and sedentary group were significantly associated with lipid metabolism, amino acid metabolism, and neurotransmitters. These findings suggest that combined aerobic interval exercise-mediated metabolites and proteins contributed to improving adult neurogenesis and behavioral performance after AD pathology, which might provide a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Zhi-Tao Liu
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Rehabilitative Department, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315211, PR China; Faculty of Sports Science, Ningbo University, Ningbo, 315211, China
| | - Yu-Tao Ma
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China
| | - Shao-Tao Pan
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China
| | - Kai Xie
- Rehabilitative Department, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315211, PR China
| | - Wei Shen
- Rehabilitative Department, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315211, PR China
| | - Su-Yang Lin
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China
| | - Jun-Yan Gao
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China
| | - Wan-Yi Li
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China
| | - Guang-Yu Li
- Faculty of Sports Science, Ningbo University, Ningbo, 315211, China
| | - Qin-Wen Wang
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China.
| | - Li-Ping Li
- Department of Physiology and Pharmacology, Ningbo University School of Medicine, Ningbo, Zhejiang, 315211, PR China; Rehabilitative Department, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315211, PR China; Key Laboratory of Addiction Research of Zhejiang Province, Ningbo, Zhejiang, 315010, PR China.
| |
Collapse
|
49
|
Yang L, Wu C, Li Y, Dong Y, Wu CYC, Lee RHC, Brann DW, Lin HW, Zhang Q. Long-term exercise pre-training attenuates Alzheimer's disease-related pathology in a transgenic rat model of Alzheimer's disease. GeroScience 2022; 44:1457-1477. [PMID: 35229257 DOI: 10.1007/s11357-022-00534-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite enormous efforts around the world, there remains no effective cure for AD. This study was performed to investigate the effects of long-term exercise pretreatment on the typical pathology of AD in a novel transgenic AD rat model. Male 2-month-old animals were divided into the following groups: wild-type (WT) rats, AD rats, and AD rats with treadmill exercise pretreatment (AD-Exe). After exercise pretreatment, the Barnes maze task, passive avoidance task, and cued fear conditioning test were performed to test learning and memory function. The elevated plus maze, open field test, sucrose preference test, and forced swim test were conducted to measure anxious-depressive-like behavior. Immunofluorescence staining, Golgi staining, transmission electron microscopy, Western blot analysis, F-Jade C staining, TUNEL staining, and related assay kits were conducted to measure Aβ plaques, tau hyperphosphorylation, neuronal damage, neuronal degeneration, dendritic spine density, synapses, synaptic vesicles, mitochondrial morphology, mitochondrial dynamic, oxidative stress, and neuroinflammation. Behavioral tests revealed that long-term exercise pretreatment significantly alleviated learning and memory dysfunction and anxious-depressive-like behaviors in AD animals. In addition, exercise pretreatment attenuated amyloid-β deposition and tau hyperphosphorylation and preserved spine density, synapses, and presynaptic vesicles. Exercise also inhibited neuronal damage, neuronal apoptosis, and neuronal degeneration. Additional studies revealed the imbalance of mitochondrial dynamics was significantly inhibited by exercise pretreatment accompanied by a remarkable suppression of oxidative stress and neuroinflammation. Our findings suggest that long-term exercise pretreatment alleviated behavioral deficits and typical pathologies of the AD rat model, supporting long-term exercise pretreatment as a potential approach to delay the progression of AD.
Collapse
Affiliation(s)
- Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Chongyun Wu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yong Li
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA.
| |
Collapse
|
50
|
Lee EC, Hong DY, Lee DH, Park SW, Lee JY, Jeong JH, Kim EY, Chung HM, Hong KS, Park SP, Lee MR, Oh JS. Inflammation and Rho-Associated Protein Kinase-Induced Brain Changes in Vascular Dementia. Biomedicines 2022; 10:biomedicines10020446. [PMID: 35203655 PMCID: PMC8962349 DOI: 10.3390/biomedicines10020446] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Patients with vascular dementia, caused by cerebral ischemia, experience long-term cognitive impairment due to the lack of effective treatment. The mechanisms of and treatments for vascular dementia have been investigated in various animal models; however, the insufficient information on gene expression changes that define pathological conditions hampers progress. To investigate the underlying mechanism of and facilitate treatment development for vascular dementia, we established a mouse model of chronic cerebral hypoperfusion, including bilateral carotid artery stenosis, by using microcoils, and elucidated the molecular pathway underlying vascular dementia development. Rho-associated protein kinase (ROCK) 1/2, which regulates cellular structure, and inflammatory cytokines (IL-1 and IL-6) were upregulated in the vascular dementia model. However, expression of claudin-5, which maintains the blood–brain barrier, and MAP2 as a nerve cell-specific factor, was decreased in the hippocampal region of the vascular dementia model. Thus, we revealed that ROCK pathway activation loosens the tight junction of the blood–brain barrier and increases the influx of inflammatory cytokines into the hippocampal region, leading to neuronal death and causing cognitive and emotional dysfunction. Our vascular dementia model allows effective study of the vascular dementia mechanism. Moreover, the ROCK pathway may be a target for vascular dementia treatment development in the future.
Collapse
Affiliation(s)
- Eun Chae Lee
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
| | - Dong-Yong Hong
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
| | - Dong-Hun Lee
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
| | - Sang-Won Park
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
| | - Ji Young Lee
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
| | - Ji Hun Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
| | - Eun-Young Kim
- Mireacellbio Co., Ltd., Seoul 04795, Korea; (E.-Y.K.); (H.-M.C.); (K.-S.H.); (S.-P.P.)
| | - Hyung-Min Chung
- Mireacellbio Co., Ltd., Seoul 04795, Korea; (E.-Y.K.); (H.-M.C.); (K.-S.H.); (S.-P.P.)
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea
| | - Ki-Sung Hong
- Mireacellbio Co., Ltd., Seoul 04795, Korea; (E.-Y.K.); (H.-M.C.); (K.-S.H.); (S.-P.P.)
| | - Se-Pill Park
- Mireacellbio Co., Ltd., Seoul 04795, Korea; (E.-Y.K.); (H.-M.C.); (K.-S.H.); (S.-P.P.)
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju 63243, Korea
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
- Correspondence: (M.R.L.); (J.S.O.)
| | - Jae Sang Oh
- Department of Neurosurgery, College of Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea; (E.C.L.); (D.-Y.H.); (D.-H.L.); (S.-W.P.); (J.Y.L.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea;
- Correspondence: (M.R.L.); (J.S.O.)
| |
Collapse
|