1
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [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: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
2
|
Farzi A, Teymoor Davani A, Seyed A, Salehi O, Mosallanezhad Z. The effect of eight weeks of aerobic training with vitamin C on some apoptotic markers in the hippocampus tissue of rats with Alzheimer's disease; an experimental study. Neurol Res 2025; 47:77-86. [PMID: 39754544 DOI: 10.1080/01616412.2024.2448624] [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: 05/18/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
OBJECTIVES The aim of this study was to investigate the effect of eight weeks of aerobic training (AT) and vitamin C supplementation (VC) on apoptotic markers in hippocampus tissue of AD rats treated with trimethyltin (TMT). MATERIALS AND METHODS In this experimental study, 32 Sprague- Dawley rats (mean age: 14-18 months and mean weight 270-320 g) were treated with (10 mg/kg) TMT and divided into 4 groups including: 1) ADcontrol, 2) VC, 3) AT and 4) AT+VC groups. In order to investigate the effects of AD induction on research variables, 8 healthy rats selected as healthy control group (HC). Groups 3 and 4 trained for eight weeks, three sessions per week and each session lasted 15-48 minutes with an intensity of 10-24 m/min. Groups 2 and 4 received 4 mg/kg VC orally. One-way ANOVA with Tukey's post- hoc tests were used for statistical analysis of data (p ≤ 0.05). RESULTS The gene expression levels of Caspase 3, FasL, Cyt-C and AP-1 in the AT, VC and AT+VC groups were significantly lower than TMT group (p ≤ 0.05); Caspase 3, FasL and Cyt-C levels were significantly lower in the AT+VC group compare to VC and ET groups (p ≤ 0.05). CytC levels in AT group were significantly lower than VC group (p = 0.002). Also, AP-1 levels in AT+VC group were significantly lower than AT group (p = 0.01). CONCLUSIONS It seems that AT and VC, both alone and interactively, can probably induce their anti-apoptotic effects in the hippocampus tissue of rats with AD via a common signaling pathway.
Collapse
Affiliation(s)
- Azadeh Farzi
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Amin Teymoor Davani
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Asiye Seyed
- Department of Sport Physiology, Behbahan Branch, Islamic Azad University, Behbehan, Iran
| | - Omidreza Salehi
- Department of Physical Education and Sport Sciences, University of Kurdistan, Sanandaj, Iran
| | - Zahra Mosallanezhad
- Department of Sport Sciences, Zand Institute of Higher Education, Shiraz, Iran
| |
Collapse
|
3
|
Tan Q. The Beneficial Effects of Combined Exercise and Polyphenols in Alzheimer's Disease. Phytother Res 2025; 39:1020-1034. [PMID: 39716920 DOI: 10.1002/ptr.8422] [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: 04/21/2024] [Revised: 11/16/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Regular exercise enhances life quality, lowers the risk of cognitive damage, and slows the advancement of Alzheimer's disease (AD). Natural compounds rich in polyphenols have garnered attention as a non-pharmacological means of treating and preventing AD. The primary component of wine, grape seeds, and nuts is polyphenols. Research suggests that polyphenols slow down the rate of neurodegeneration in AD and lessen learning impairment. Furthermore, polyphenols lessen brain impairments related to cognition. Additionally, polyphenols can specifically restructure amyloid-β (Aβ) structures and soluble oligomers into non-toxic alternative species. They have also been revealed to increase brain-derived neurotrophic factors expression, suggesting that they have a positive impact on the creation of neurotrophins. The benefits of polyphenol supplementation and exercise, which can both provide neuroprotection, have not been well studied in AD patients. This review aimed to investigate the effects of combined exercise polyphenols on inflammation, neuroprotection, several conformational toxic species of Aβ, and Aβ-induced apoptosis in AD.
Collapse
Affiliation(s)
- Qinghua Tan
- Graduate School of Education in Physical Education, Sangmyung University, Seoul, Korea
| |
Collapse
|
4
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Effects and mechanisms of APP and its cleavage product Aβ in the comorbidity of sarcopenia and Alzheimer's disease. Front Aging Neurosci 2024; 16:1482947. [PMID: 39654807 PMCID: PMC11625754 DOI: 10.3389/fnagi.2024.1482947] [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: 08/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Sarcopenia and AD are both classic degenerative diseases, and there is growing epidemiological evidence of their comorbidity with aging; however, the mechanisms underlying the biology of their commonality have not yet been thoroughly investigated. APP is a membrane protein that is expressed in tissues and is expressed not only in the nervous system but also in the NMJ and muscle. Deposition of its proteolytic cleavage product, Aβ, has been described as a central component of AD pathogenesis. Recent studies have shown that excessive accumulation and aberrant expression of APP in muscle lead to pathological muscle lesions, but the pathogenic mechanism by which APP and its proteolytic cleavage products act in skeletal muscle is less well understood. By summarizing and analyzing the literature concerning the role, pathogenicity and pathological mechanisms of APP and its cleavage products in the nervous system and muscles, we aimed to explore the intrinsic pathological mechanisms of myocerebral comorbidities and to provide new perspectives and theoretical foundations for the prevention and treatment of AD and sarcopenia comorbidities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| |
Collapse
|
5
|
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
|
6
|
Kamatham PT, Shukla R, Khatri DK, Vora LK. Pathogenesis, diagnostics, and therapeutics for Alzheimer's disease: Breaking the memory barrier. Ageing Res Rev 2024; 101:102481. [PMID: 39236855 DOI: 10.1016/j.arr.2024.102481] [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: 05/06/2024] [Revised: 06/28/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for 60-70 % of all cases. It affects millions of people worldwide. AD poses a substantial economic burden on societies and healthcare systems. AD is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. As the prevalence of AD continues to increase, understanding its pathogenesis, improving diagnostic methods, and developing effective therapeutics have become paramount. This comprehensive review delves into the intricate mechanisms underlying AD, explores the current state of diagnostic techniques, and examines emerging therapeutic strategies. By revealing the complexities of AD, this review aims to contribute to the growing body of knowledge surrounding this devastating disease.
Collapse
Affiliation(s)
- Pushpa Tryphena Kamatham
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rashi Shukla
- Molecular and Cellular Neuroscience Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, Nims Institute of Pharmacy, Nims University Rajasthan, Jaipur, India.
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast, Northern Ireland BT9 7BL, UK.
| |
Collapse
|
7
|
Li D, Jia J, Zeng H, Zhong X, Chen H, Yi C. Efficacy of exercise rehabilitation for managing patients with Alzheimer's disease. Neural Regen Res 2024; 19:2175-2188. [PMID: 38488551 PMCID: PMC11034587 DOI: 10.4103/1673-5374.391308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/16/2023] [Accepted: 11/25/2023] [Indexed: 04/24/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive and degenerative neurological disease characterized by the deterioration of cognitive functions. While a definitive cure and optimal medication to impede disease progression are currently unavailable, a plethora of studies have highlighted the potential advantages of exercise rehabilitation for managing this condition. Those studies show that exercise rehabilitation can enhance cognitive function and improve the quality of life for individuals affected by AD. Therefore, exercise rehabilitation has been regarded as one of the most important strategies for managing patients with AD. Herein, we provide a comprehensive analysis of the currently available findings on exercise rehabilitation in patients with AD, with a focus on the exercise types which have shown efficacy when implemented alone or combined with other treatment methods, as well as the potential mechanisms underlying these positive effects. Specifically, we explain how exercise may improve the brain microenvironment and neuronal plasticity. In conclusion, exercise is a cost-effective intervention to enhance cognitive performance and improve quality of life in patients with mild to moderate cognitive dysfunction. Therefore, it can potentially become both a physical activity and a tailored intervention. This review may aid the development of more effective and individualized treatment strategies to address the challenges imposed by this debilitating disease, especially in low- and middle-income countries.
Collapse
Affiliation(s)
- Dan Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jinning Jia
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Haibo Zeng
- Department of Pathology, Huichang County People’s Hospital, Ganzhou, Jiangxi Province, China
| | - Xiaoyan Zhong
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong Province, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, Guangdong Province, China
| |
Collapse
|
8
|
Früholz I, Meyer-Luehmann M. The intricate interplay between microglia and adult neurogenesis in Alzheimer's disease. Front Cell Neurosci 2024; 18:1456253. [PMID: 39360265 PMCID: PMC11445663 DOI: 10.3389/fncel.2024.1456253] [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: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system, play a crucial role in regulating adult neurogenesis and contribute significantly to the pathogenesis of Alzheimer's disease (AD). Under physiological conditions, microglia support and modulate neurogenesis through the secretion of neurotrophic factors, phagocytosis of apoptotic cells, and synaptic pruning, thereby promoting the proliferation, differentiation, and survival of neural progenitor cells (NPCs). However, in AD, microglial function becomes dysregulated, leading to chronic neuroinflammation and impaired neurogenesis. This review explores the intricate interplay between microglia and adult neurogenesis in health and AD, synthesizing recent findings to provide a comprehensive overview of the current understanding of microglia-mediated regulation of adult neurogenesis. Furthermore, it highlights the potential of microglia-targeted therapies to modulate neurogenesis and offers insights into potential avenues for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Iris Früholz
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
Hu J, Huang B, Chen K. The impact of physical exercise on neuroinflammation mechanism in Alzheimer's disease. Front Aging Neurosci 2024; 16:1444716. [PMID: 39233828 PMCID: PMC11371602 DOI: 10.3389/fnagi.2024.1444716] [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: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Alzheimer's disease (AD), a major cause of dementia globally, imposes significant societal and personal costs. This review explores the efficacy of physical exercise as a non-pharmacological intervention to mitigate the impacts of AD. Methods This review draws on recent studies that investigate the effects of physical exercise on neuroinflammation and neuronal enhancement in individuals with AD. Results Consistent physical exercise alters neuroinflammatory pathways, enhances cognitive functions, and bolsters brain health among AD patients. It favorably influences the activation states of microglia and astrocytes, fortifies the integrity of the blood-brain barrier, and attenuates gut inflammation associated with AD. These changes are associated with substantial improvements in cognitive performance and brain health indicators. Discussion The findings underscore the potential of integrating physical exercise into comprehensive AD management strategies. Emphasizing the necessity for further research, this review advocates for the refinement of exercise regimens to maximize their enduring benefits in decelerating the progression of AD.
Collapse
Affiliation(s)
- Junhui Hu
- School of Physical Education, West Anhui University, Lu'an, China
| | - Baiqing Huang
- School of Physical Education, Yunnan Minzu University, Kunming, China
| | - Kang Chen
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
10
|
Shah J, Orosz T, Singh A, Laxma SP, Gross RE, Smith N, Vroegop S, Sudler S, Porter JT, Colon M, Jun L, Babu JR, Shim M, Broderick TL, Al-Nakkash L. Influence of Exercise and Genistein to Mitigate the Deleterious Effects of High-Fat High-Sugar Diet on Alzheimer's Disease-Related Markers in Male Mice. Int J Mol Sci 2024; 25:9019. [PMID: 39201705 PMCID: PMC11354341 DOI: 10.3390/ijms25169019] [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: 07/08/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
The prevalence of obesity and related consequences, including insulin resistance and Alzheimer's-like neuropathology, has increased dramatically. Contributing to this prevalence is the shift in lifestyle preference away from wholesome foods and exercise to the Western-style diet and sedentarism. Despite advances in drug development, a healthy diet and regular exercise remain the most effective approaches to mitigating the unwanted sequelae of diet-induced obesity on brain health. In this study, we used the high-fat high-sugar (HFHS) mouse model of neurodegeneration to examine the effects of exercise training (HFHS+Ex), genistein treatment (HFHS+Gen), and combination treatment (HFHS+Ex+Gen) on proteins relating to neurodegeneration in the brain of male mice. After a period of 12 weeks, as expected, HFHS feeding increased body weight, adipose tissue weight, and systemic plasma inflammation (TNF-α) compared to lean mice fed a standard diet. HFHS feeding also increased protein expression of brain markers of insulin resistance (pGSK-3β, p-IR), apoptosis (caspase 3), early neurofibrillary tangles (CP13), and amyloid-beta precursor (CT20). Compared to HFHS mice, Ex decreased body weight, plasma TNF-α, and expression of pGSK-3β, caspase 3, CP13, amyloid-β precursor (22c11), and ADAM10. Treatment with Gen was equally protective on these markers and decreased the expression of p-IR. Combination treatment with Ex and Gen afforded the greatest overall benefits, and this group exhibited the greatest reduction in body and adipose tissue weight and all brain markers, except for 22c11 and ADAM10, which were decreased compared to mice fed an HFHS diet. In addition, levels of 4G8, which detects protein levels of amyloid-β, were decreased with combination treatment. Our results indicate that exercise training, genistein supplementation, or combination treatment provide varying degrees of neuroprotection from HFHS feeding-induced Alzheimer's pathology. Future perspectives could include evaluating moderate exercise regimens in combination with dietary supplementation with genistein in humans to determine whether the same benefits translate clinically.
Collapse
Affiliation(s)
- Juhi Shah
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Tyler Orosz
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Avneet Singh
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Savan Parameshwar Laxma
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Rachel E. Gross
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Nicholas Smith
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Spencer Vroegop
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Sydney Sudler
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - James T. Porter
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce 00732, Puerto Rico; (J.T.P.); (M.C.)
| | - Maria Colon
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce 00732, Puerto Rico; (J.T.P.); (M.C.)
| | - Lauren Jun
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Jeganathan R. Babu
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Minsub Shim
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Thomas L. Broderick
- Department of Physiology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Layla Al-Nakkash
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
- Department of Physiology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| |
Collapse
|
11
|
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
|
12
|
Huang T, Gong XK, Liang Z, Yang R, Wu L, Yang C, Wu M, Wang XC, Shu XJ, Bao J. Exercised-enriched blood plasma rescues hippocampal impairments and cognitive deficits in an Alzheimer's disease model. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167197. [PMID: 38653353 DOI: 10.1016/j.bbadis.2024.167197] [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: 12/07/2023] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, and moderate exercise holds promise in ameliorating the ongoing neurodegeneration and cognitive decline. Here, we investigated whether exercise-enriched blood plasm could yield a beneficial therapeutic effect on AD pathologies and cognitive decline in transgenic AD (P301S) mice. In this investigation, a cohort of 2-month-old C57BL/6 mice were granted continuous access to either a running wheel or a fixed wheel for 6 weeks. After that, their plasmas were extracted and subsequently injected intravenously into 4.5-month-old P301S mice biweekly over a 6-week period. A comprehensive methodology was then employed, integrating behavioral tests, pathology assessments, and biochemical analyses to unveil the potential anti-dementia implications of exercise-enriched blood plasma in P301S mice. Upon systemic administration, the findings revealed a noteworthy attenuation of hippocampus-dependent behavioral impairments in P301S mice. Conversely, blood plasma from sedentary counterparts exhibited no discernible impact. These effects were intricately associated with the mitigation of neuroinflammation, the augmentation of hippocampal adult neurogenesis, and a reduction of synaptic impairments following the administration of exercise-enriched blood plasma. These findings advance the proposition that administering exercise-enriched blood plasma may serve as an effective prophylactic measure against AD, opening avenues for further exploration and potential therapeutic interventions.
Collapse
Affiliation(s)
- Tiantian Huang
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiao-Kang Gong
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Zheng Liang
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Rong Yang
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Liangwei Wu
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Chaoqing Yang
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Mengjuan Wu
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiao-Chuan Wang
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xi-Ji Shu
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China.
| | - Jian Bao
- Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
13
|
Numakawa T, Kajihara R. An Interaction between Brain-Derived Neurotrophic Factor and Stress-Related Glucocorticoids in the Pathophysiology of Alzheimer's Disease. Int J Mol Sci 2024; 25:1596. [PMID: 38338875 PMCID: PMC10855648 DOI: 10.3390/ijms25031596] [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: 12/13/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Both the brain-derived neurotrophic factor (BDNF) and glucocorticoids (GCs) play multiple roles in various aspects of neurons, including cell survival and synaptic function. BDNF and its receptor TrkB are extensively expressed in neurons of the central nervous system (CNS), and the contribution of the BDNF/TrkB system to neuronal function is evident; thus, its downregulation has been considered to be involved in the pathogenesis of Alzheimer's disease (AD). GCs, stress-related molecules, and glucocorticoid receptors (GRs) are also considered to be associated with AD in addition to mental disorders such as depression. Importantly, a growing body of evidence suggests a close relationship between BDNF/TrkB-mediated signaling and the GCs/GR system in the CNS. Here, we introduce the current studies on the interaction between the neurotrophic system and stress in CNS neurons and discuss their involvement in the pathophysiology of AD.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ryutaro Kajihara
- Department of Biomedical Laboratory Sciences, Faculty of Life Science, Kumamoto University, Kumamoto 862-0976, Japan
| |
Collapse
|
14
|
Martín-Rodríguez A, Gostian-Ropotin LA, Beltrán-Velasco AI, Belando-Pedreño N, Simón JA, López-Mora C, Navarro-Jiménez E, Tornero-Aguilera JF, Clemente-Suárez VJ. Sporting Mind: The Interplay of Physical Activity and Psychological Health. Sports (Basel) 2024; 12:37. [PMID: 38275986 PMCID: PMC10819297 DOI: 10.3390/sports12010037] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The symbiotic relationship between sports practice and psychological well-being has, in recent times, surged to the forefront of academic and public attention. The aim of this narrative review is to comprehensively explore the intricate pathways linking physical engagement in sports to its subsequent impacts on mental health and synthesize the multifarious effects of sports on psychological health, offering insights for integrating physical and psychological strategies to enhance well-being. From neurobiological underpinnings to therapeutic applications, this comprehensive manuscript provides an in-depth dive into the multifaceted world of sports and psychology. Highlighting evidence-based interventions, this review aspires to offer actionable insights for practitioners, athletes, and individuals alike, advocating for a holistic approach to mental well-being. This manuscript highlights the profound impact of sports on mental health, emphasizing its role in emotional regulation, resilience, cognitive function, and treating psychological conditions. It details how sports induce neurochemical changes, enhance brain functions like memory and learning, and aid against cognitive decline. This review also notes the benefits of regular exercise in mood improvement, stress management, and social skill enhancement, particularly when combined with mindfulness practices. It underscores the importance of considering cultural and gender perspectives in sports psychology, advocating for an integrated physical-psychological approach to promote overall well-being.
Collapse
Affiliation(s)
- Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Laura Augusta Gostian-Ropotin
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | | | - Noelia Belando-Pedreño
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Juan Antonio Simón
- Department Ciencias Sociales Act Fis Deporte & Ocio, Universidad Politécnica de Madrid, 28040 Madrid, Spain;
| | - Clara López-Mora
- Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Valencia, Pg. de l’Albereda, 7, 46010 València, Spain;
| | | | - José Francisco Tornero-Aguilera
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
| | - Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (A.M.-R.); (L.A.G.-R.); (N.B.-P.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| |
Collapse
|
15
|
Widjaya MA, Lee SD, Cheng WC, Wu BT. Effects of Exercise Training on Immune-Related Genes and Pathways in the Cortex of Animal Models of Alzheimer's Disease: A Systematic Review. J Alzheimers Dis 2024; 98:1219-1234. [PMID: 38578886 DOI: 10.3233/jad-230803] [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: 04/07/2024]
Abstract
Background Alzheimer's disease (AD) is a chronic neurodegenerative disease that affects the immune system due to the accumulation of amyloid-β (Aβ) and tau associated molecular pathology and other pathogenic processes. To address AD pathogenesis, various approaches had been conducted from drug development to lifestyle modification to reduce the prevalence of AD. Exercise is considered a prominent lifestyle modification to combat AD. Objective This observation prompted us to review the literature on exercise related to immune genes in the cortex of animal models of AD. We focused on animal model studies due to their prevalence in this domain. Methods The systematic review was conducted according to PRISMA standards using Web of Science (WoS) and PubMed databases. Any kind of genes, proteins, and molecular molecules were included in this systematic review. The list of these immune-related molecules was analyzed in the STRING database for functional enrichment analysis. Results We found that 17 research studies discussed immune-related molecules and 30 immune proteins. These studies showed that exercise had the ability to ameliorate dysfunction in AD-related pathways, which led to decreasing the expression of microglia-related pathways and Th17-related immune pathways. As a result of decreasing the expression of immune-related pathways, the expression of apoptosis-related pathways was also decreasing, and neuronal survival was increased by exercise activity. Conclusions Based on functional enrichment analysis, exercise not only could reduce apoptotic factors and immune components but also could increase cell survival and Aβ clearance in cortex samples. PROSPERO ID: CRD42022326093.
Collapse
Affiliation(s)
- Michael Anekson Widjaya
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, PhD program in Healthcare Science, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Bor-Tsang Wu
- Department of Senior Citizen Service Management, National Taichung University of Science and Technology, Taichung, Taiwan
| |
Collapse
|
16
|
Augusto-Oliveira M, Arrifano GP, Leal-Nazaré CG, Santos-Sacramento L, Lopes-Araújo A, Royes LFF, Crespo-Lopez ME. Exercise Reshapes the Brain: Molecular, Cellular, and Structural Changes Associated with Cognitive Improvements. Mol Neurobiol 2023; 60:6950-6974. [PMID: 37518829 DOI: 10.1007/s12035-023-03492-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023]
Abstract
Physical exercise is well known as a non-pharmacological and holistic therapy believed to prevent and mitigate numerous neurological conditions and alleviate ageing-related cognitive decline. To do so, exercise affects the central nervous system (CNS) at different levels. It changes brain physiology and structure, promoting cognitive improvements, which ultimately improves quality of life. Most of these effects are mediated by neurotrophins release, enhanced adult hippocampal neurogenesis, attenuation of neuroinflammation, modulation of cerebral blood flow, and structural reorganisation, besides to promote social interaction with beneficial cognitive outcomes. In this review, we discuss, based on experimental and human research, how exercise impacts the brain structure and function and how these changes contribute to cognitive improvements. Understanding the mechanisms by which exercise affects the brain is essential to understand the brain plasticity following exercise, guiding therapeutic approaches to improve the quality of life, especially in obesity, ageing, neurodegenerative disorders, and following traumatic brain injury.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil.
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Caio G Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Letícia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Amanda Lopes-Araújo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil
| | - Luiz Fernando Freire Royes
- Laboratório de Bioquímica Do Exercício, Centro de Educacão Física E Desportos, Universidade Federal de Santa Maria, Santa Maria, RGS, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, PA, Brazil.
| |
Collapse
|
17
|
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: 1.5] [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
|
18
|
Numakawa T, Kajihara R. Involvement of brain-derived neurotrophic factor signaling in the pathogenesis of stress-related brain diseases. Front Mol Neurosci 2023; 16:1247422. [PMID: 37781095 PMCID: PMC10537938 DOI: 10.3389/fnmol.2023.1247422] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
Neurotrophins including brain-derived neurotrophic factor, BDNF, have critical roles in neuronal differentiation, cell survival, and synaptic function in the peripheral and central nervous system. It is well known that a variety of intracellular signaling stimulated by TrkB, a high-affinity receptor for BDNF, is involved in the physiological and pathological neuronal aspects via affecting cell viability, synaptic function, neurogenesis, and cognitive function. As expected, an alteration of the BDNF/TrkB system is suspected to be one of the molecular mechanisms underlying cognitive decline in cognitive diseases and mental disorders. Recent evidence has also highlighted a possible link between the alteration of TrkB signaling and chronic stress. Furthermore, it has been demonstrated that downregulation of the BDNF/TrkB system and chronic stress have a role in the pathogenesis of Alzheimer's disease (AD) and mental disorders. In this review, we introduce current evidence showing a close relationship between the BDNF/TrkB system and the development of cognition impairment in stress-related disorders, and the possible contribution of the upregulation of the BDNF/TrkB system in a therapeutic approach against these brain diseases.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryutaro Kajihara
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
- Department of Biomedical Laboratory Sciences, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Perez Garcia G, Bicak M, Buros J, Haure-Mirande JV, Perez GM, Otero-Pagan A, Gama Sosa MA, De Gasperi R, Sano M, Gage FH, Barlow C, Dudley JT, Glicksberg BS, Wang Y, Readhead B, Ehrlich ME, Elder GA, Gandy S. Beneficial effects of physical exercise and an orally active mGluR2/3 antagonist pro-drug on neurogenesis and behavior in an Alzheimer's amyloidosis model. FRONTIERS IN DEMENTIA 2023; 2:1198006. [PMID: 39081972 PMCID: PMC11285632 DOI: 10.3389/frdem.2023.1198006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/31/2023] [Indexed: 08/02/2024]
Abstract
Background Modulation of physical activity represents an important intervention that may delay, slow, or prevent mild cognitive impairment (MCI) or dementia due to Alzheimer's disease (AD). One mechanism proposed to underlie the beneficial effect of physical exercise (PE) involves the apparent stimulation of adult hippocampal neurogenesis (AHN). BCI-838 is a pro-drug whose active metabolite BCI-632 is a negative allosteric modulator at group II metabotropic glutamate receptors (mGluR2/3). We previously demonstrated that administration of BCI-838 to a mouse model of brain accumulation of oligomeric AβE22Q (APP E693Q = "Dutch APP") reduced learning behavior impairment and anxiety, both of which are associated with the phenotype of Dutch APP mice. Methods 3-month-old mice were administered BCI-838 and/or physical exercise for 1 month and then tested in novel object recognition, neurogenesis, and RNAseq. Results Here we show that (i) administration of BCI-838 and a combination of BCI-838 and PE enhanced AHN in a 4-month old mouse model of AD amyloid pathology (APP KM670/671NL /PSEN1 Δexon9= APP/PS1), (ii) administration of BCI-838 alone or with PE led to stimulation of AHN and improvement in recognition memory, (iii) the hippocampal dentate gyrus transcriptome of APP/PS1 mice following BCI-838 treatment showed up-regulation of brain-derived neurotrophic factor (BDNF), PIK3C2A of the PI3K-mTOR pathway, and metabotropic glutamate receptors, and down-regulation of EIF5A involved in modulation of mTOR activity by ketamine, and (iv) validation by qPCR of an association between increased BDNF levels and BCI-838 treatment. Conclusion Our study points to BCI-838 as a safe and orally active compound capable of mimicking the beneficial effect of PE on AHN and recognition memory in a mouse model of AD amyloid pathology.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Mesude Bicak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jacqueline Buros
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Gissel M. Perez
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Alena Otero-Pagan
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Miguel A. Gama Sosa
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Rita De Gasperi
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mary Sano
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- BrainCells, Inc., La Jolla, CA, United States
| | - Carrolee Barlow
- BrainCells, Inc., La Jolla, CA, United States
- E-Scape Bio, South San Francisco, CA, United States
| | - Joel T. Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benjamin S. Glicksberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yanzhuang Wang
- Department of Developmental and Cell Biology, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin Readhead
- Arizona State University-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, United States
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Gregory A. Elder
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, United States
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Research and Development, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
- Department of Psychiatry and Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Santiago JA, Potashkin JA. Physical activity and lifestyle modifications in the treatment of neurodegenerative diseases. Front Aging Neurosci 2023; 15:1185671. [PMID: 37304072 PMCID: PMC10250655 DOI: 10.3389/fnagi.2023.1185671] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/03/2023] [Indexed: 06/13/2023] Open
Abstract
Neurodegenerative diseases have reached alarming numbers in the past decade. Unfortunately, clinical trials testing potential therapeutics have proven futile. In the absence of disease-modifying therapies, physical activity has emerged as the single most accessible lifestyle modification with the potential to fight off cognitive decline and neurodegeneration. In this review, we discuss findings from epidemiological, clinical, and molecular studies investigating the potential of lifestyle modifications in promoting brain health. We propose an evidence-based multidomain approach that includes physical activity, diet, cognitive training, and sleep hygiene to treat and prevent neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Judith A. Potashkin
- Center for Neurodegenerative Diseases and Therapeutics, Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
21
|
Jin Y, Kim T, Kang H. Forced treadmill running modifies gut microbiota with alleviations of cognitive impairment and Alzheimer's disease pathology in 3xTg-AD mice. Physiol Behav 2023; 264:114145. [PMID: 36889489 DOI: 10.1016/j.physbeh.2023.114145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
Physical exercise has been recommended as a non-pharmacologic treatment for delaying the onset or slowing the progression of Alzheimer's disease (AD). The therapeutic potential of exercise training-induced changes in symbiotic gut microbiota against AD neuropathology is not well understood, yet. This study investigated the effects of a 20-week forced treadmill exercise program on the makeup of the gut microbiota, the integrity of the blood-brain barrier (BBB), and the development of AD-like cognitive deficits and neuropathology in triple transgenic AD mice. Our findings show that forced treadmill running causes symbiotic changes in the gut microbiota, such as increased Akkermansia muciniphila and decreased Bacteroides species, as well as increased BBB-related protein expression and reduced AD-like cognitive impairments and neuropathology progression. The current findings of this animal study suggest that the interaction between the gut microbiota and the brain, possibly via the BBB, is responsible for exercise training-induced cognitive benefits and alleviation of AD pathology.
Collapse
Affiliation(s)
- Youngyun Jin
- College of Sport Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Taewan Kim
- College of Sport Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyunsik Kang
- College of Sport Science, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
22
|
Widjaya MA, Cheng YJ, Kuo YM, Liu CH, Cheng WC, Lee SD. Transcriptomic Analyses of Exercise Training in Alzheimer's Disease Cerebral Cortex. J Alzheimers Dis 2023; 93:349-363. [PMID: 36970901 DOI: 10.3233/jad-221139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
BACKGROUND Research reported exercise could reduce Alzheimer's disease (AD) symptoms in human and animals. However, the molecular mechanism of exercise training via transcriptomic analysis was unclear especially in AD in the cortex area. OBJECTIVE Investigate potential significant pathways in the cortex area that were affected by exercise during AD. METHODS RNA-seq analysis, differential expressed genes, functional enrichment analysis, and GSOAP clustering analysis were performed in the isolated cerebral cortex from eight 3xTg AD mice (12 weeks old) randomly and equally divided into control (AD) and exercise training (AD-EX) group. Swimming exercise training in AD-EX group was conducted 30 min/day for 1 month. RESULTS There were 412 genes significant differentially expressed in AD-EX group compared to AD group. Top 10 upregulated genes in AD-EX group against AD group mostly correlated with neuroinflammation, while top 10 downregulated genes mostly had connection with vascularization, membrane transport, learning memory, and chemokine signal. Pathway analysis revealed the upregulated interferon alpha beta signaling in AD-EX had association with cytokines delivery in microglia cells compared to AD and top 10 upregulated genes involved in interferon alpha beta were Usp18, Isg15, Mx1, Mx2, Stat1, Oas1a, and Irf9; The downregulated extracellular matrix organization in AD-EX had correlation with Aβ and neuron cells interaction and Vtn was one of the top 10 downregulated genes involved in this pathway. CONCLUSION Exercise training influenced 3xTg mice cortex through interferon alpha beta signaling upregulation and extracellular matrix organization downregulation based on transcriptomics analysis.
Collapse
Affiliation(s)
- Michael Anekson Widjaya
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung, Tainan, Taiwan
| | - Chia-Hsin Liu
- Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Research Center for Cancer Biology, China Medical University, Taichung, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
- School of Rehabilitation Medicine, Weifang Medical University, Weifang, China
| |
Collapse
|
23
|
Tarawneh R, Penhos E. The gut microbiome and Alzheimer's disease: Complex and bidirectional interactions. Neurosci Biobehav Rev 2022; 141:104814. [PMID: 35934087 PMCID: PMC9637435 DOI: 10.1016/j.neubiorev.2022.104814] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022]
Abstract
Structural and functional alterations to the gut microbiome, referred to as gut dysbiosis, have emerged as potential key mediators of neurodegeneration and Alzheimer disease (AD) pathogenesis through the "gut -brain" axis. Emerging data from animal and clinical studies support an important role for gut dysbiosis in mediating neuroinflammation, central and peripheral immune dysregulation, abnormal brain protein aggregation, and impaired intestinal and brain barrier permeability, leading to neuronal loss and cognitive impairment. Gut dysbiosis has also been shown to directly influence various mechanisms involved in neuronal growth and repair, synaptic plasticity, and memory and learning functions. Aging and lifestyle factors including diet, exercise, sleep, and stress influence AD risk through gut dysbiosis. Furthermore, AD is associated with characteristic gut microbial signatures which offer value as potential markers of disease severity and progression. Together, these findings suggest the presence of a complex bidirectional relationship between AD and the gut microbiome and highlight the utility of gut modulation strategies as potential preventative or therapeutic strategies in AD. We here review the current literature regarding the role of the gut-brain axis in AD pathogenesis and its potential role as a future therapeutic target in AD treatment and/or prevention.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, Alzheimer Disease Research Center, The University of New Mexico, Albuquerque, NM 87106, USA.
| | - Elena Penhos
- College of Medicine, The Ohio State University, Columbus, OH, USA 43210
| |
Collapse
|
24
|
Jullienne A, Quan R, Szu JI, Trinh MV, Behringer EJ, Obenaus A. Progressive Vascular Abnormalities in the Aging 3xTg-AD Mouse Model of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081967. [PMID: 36009514 PMCID: PMC9405684 DOI: 10.3390/biomedicines10081967] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular dysfunction and structural abnormalities in Alzheimer’s disease (AD) are known to contribute to the progression of the pathology, and studies have tended to ignore the role of the vasculature in AD progression. We utilized the 3xTg-AD mouse model of AD to examine individual cerebral vessels and the cortical vascular network across the lifespan. Our vessel painting approach was used to label the entire cortical vasculature, followed by epifluorescence microscopy. The middle cerebral artery (MCA) tree was assessed with confocal microscopy, and a new method was developed to assess branching patterns as a measure of aging-related changes. We found that vascular remodeling was profoundly altered at 4–6 months of age, when the 3xTg-AD mouse is known to transition to cognitive impairment and Aβ deposition in both sexes. Analysis of vascular features (density, junctions, length) of the MCA territory highlighted sex-dependent differences across the 3xTg-AD mouse lifespan, with no alterations in branching patterns. Our current cerebrovascular angioarchitectural analyses demonstrate progressive alterations in individual cortical vessels, as well as in the vascular network of the cortex. These new findings advance our understanding of brain anatomy and physiology in the 3xTg-AD mouse, while potentially identifying unique diagnostic signatures of AD progression.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Ryan Quan
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Jenny I. Szu
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Michelle V. Trinh
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Erik J. Behringer
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
- Correspondence:
| |
Collapse
|
25
|
Maroofi A, Moro T, Agrimi J, Safari F. Cognitive decline in heart failure: Biomolecular mechanisms and benefits of exercise. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166511. [PMID: 35932891 DOI: 10.1016/j.bbadis.2022.166511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
Abstract
By definition, heart failure (HF) is a human pathological condition affecting the structure and function of all organs in the body, and the brain is not an exception to that. Failure of the heart to pump enough blood centrally and peripherally is at the foundation of HF patients' inability to attend even the most ordinary daily activities and progressive deterioration of their cognitive capacity. What is more, between heart and brain exists a bidirectional relationship that goes well beyond hemodynamics and concerns bioelectric and endocrine signaling. This increasingly consolidated evidence makes the scenario even more complex. Studies have mainly chased how HF impairs cognition without focusing much on preventive measures, notably cardio-cerebral health proxies. Here, we aim to provide a brief account of known and hypothetical factors that may explain how exercise can help obviate cognitive dysfunction associated with HF in its different forms. As we shall see, there is a stringent need for a deeper grasp of such mechanisms. Indeed, gaining this new knowledge will automatically shed new light on the inner workings of HF itself, thus resulting in more effective prevention and treatment of this escalating syndrome.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Tatiana Moro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Jacopo Agrimi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy.
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
26
|
Lion's Mane ( Hericium erinaceus) Exerts Anxiolytic Effects in the rTg4510 Tau Mouse Model. Behav Sci (Basel) 2022; 12:bs12070235. [PMID: 35877305 PMCID: PMC9312024 DOI: 10.3390/bs12070235] [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: 06/11/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) significantly impairs the life of an individual both cognitively and behaviorally. Tau and beta-amyloid (Aβ) proteins are major contributors to the etiology of AD. This study used mice modeling AD through the presence of tau pathology to assess the effects of Hericium erinaceus (H. erinaceus), also known as Lion’s mane, on cognitive and non-cognitive behaviors. Despite neurocognitive and neurobiological effects of H. erinaceus being seen in both healthy and transgenic mice, no research to date has explored its effects on mice with solely tau pathology. In this study, mice were placed on a diet supplemented with H. erinaceus or a standard rodent diet for 4.5 months in order to determine the effect of this medicinal mushroom on behavior. Tau mice given H. erinaceus had significantly shorter latencies to enter the center of the open field (OF) (p < 0.05) and spent significantly more time in the open arms of the elevated zero maze (EZM) (p < 0.001) compared to tau control mice. Mice given H. erinaceus spent significantly more time in the open arms of and made more head dips in the elevated zero maze (EZM) (p < 0.05). While H. erinaceus had anxiolytic effects, no improvements were seen in spatial memory or activities of daily living. These findings provide additional support for the anxiolytic effects of H. erinaceus and point to its potential benefit as a therapeutic for anxiety in AD.
Collapse
|
27
|
Kim TA, Syty MD, Wu K, Ge S. Adult hippocampal neurogenesis and its impairment in Alzheimer's disease. Zool Res 2022; 43:481-496. [PMID: 35503338 PMCID: PMC9113964 DOI: 10.24272/j.issn.2095-8137.2021.479] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/25/2022] [Indexed: 11/07/2022] Open
Abstract
Adult neurogenesis is the creation of new neurons which integrate into the existing neural circuit of the adult brain. Recent evidence suggests that adult hippocampal neurogenesis (AHN) persists throughout life in mammals, including humans. These newborn neurons have been implicated to have a crucial role in brain functions such as learning and memory. Importantly, studies have also found that hippocampal neurogenesis is impaired in neurodegenerative and neuropsychiatric diseases. Alzheimer's disease (AD) is one of the most common forms of dementia affecting millions of people. Cognitive dysfunction is a common symptom of AD patients and progressive memory loss has been attributed to the degeneration of the hippocampus. Therefore, there has been growing interest in identifying how hippocampal neurogenesis is affected in AD. However, the link between cognitive decline and changes in hippocampal neurogenesis in AD is poorly understood. In this review, we summarized the recent literature on AHN and its impairments in AD.
Collapse
Affiliation(s)
- Thomas A Kim
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
- Medical Scientist Training Program (MSTP), Renaissance School of Medicine at SUNY, Stony Brook, Stony Brook, NY 11794, USA
| | - Michelle D Syty
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Kaitlyn Wu
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Shaoyu Ge
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA. E-mail:
| |
Collapse
|
28
|
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: 10] [Impact Index Per Article: 3.3] [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
|
29
|
Huuha AM, Norevik CS, Moreira JBN, Kobro-Flatmoen A, Scrimgeour N, Kivipelto M, Van Praag H, Ziaei M, Sando SB, Wisløff U, Tari AR. Can exercise training teach us how to treat Alzheimer's disease? Ageing Res Rev 2022; 75:101559. [PMID: 34999248 DOI: 10.1016/j.arr.2022.101559] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and there is currently no cure. Novel approaches to treat AD and curb the rapidly increasing worldwide prevalence and costs of dementia are needed. Physical inactivity is a significant modifiable risk factor for AD, estimated to contribute to 12.7% of AD cases worldwide. Exercise interventions in humans and animals have shown beneficial effects of exercise on brain plasticity and cognitive functions. In animal studies, exercise also improved AD pathology. The mechanisms underlying these effects of exercise seem to be associated mainly with exercise performance or cardiorespiratory fitness. In addition, exercise-induced molecules of peripheral origin seem to play an important role. Since exercise affects the whole body, there likely is no single therapeutic target that could mimic all the benefits of exercise. However, systemic strategies may be a viable means to convey broad therapeutic effects in AD patients. Here, we review the potential of physical activity and exercise training in AD prevention and treatment, shining light on recently discovered underlying mechanisms and concluding with a view on future development of exercise-free treatment strategies for AD.
Collapse
Affiliation(s)
- Aleksi M Huuha
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cecilie S Norevik
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - José Bianco N Moreira
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway; K.G. Jebsen Centre for Alzheimer's Disease, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan Scrimgeour
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Miia Kivipelto
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Stockholm, Sweden; Karolinska University Hospital, Theme Aging and Inflammation, Stockholm, Sweden
| | - Henriette Van Praag
- Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, United States
| | - Maryam Ziaei
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway; Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Sigrid Botne Sando
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Atefe R Tari
- Cardiac Exercise Research Group (CERG), Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
30
|
Park SS, Park HS, Kim CJ, Baek SS, Park SY, Anderson CP, Kim MK, Park IR, Kim TW. Combined effects of Aerobic exercise and 40Hz light flicker exposure on early cognitive impairments in Alzheimer's disease of 3xTg mice. J Appl Physiol (1985) 2022; 132:1054-1068. [PMID: 35201933 DOI: 10.1152/japplphysiol.00751.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative brain disease and the primary cause of dementia. At an early stage, AD is generally characterized by short-term memory impairment, owing to dysfunctions of the cortex and hippocampus. We previously reported that a combination of exercise and 40 Hz light flickering can protect against AD-related neuroinflammation, gamma oscillations, reduction in Aβ, and cognitive decline. Therefore, we sought to extend our previous findings to the 5-month-old 3xTg-AD mouse model to examine whether the same favorable effects occur in earlier stages of cognitive dysfunction. We investigated the effects of 12 weeks of exercise combined with 40-Hz light flickering on cognitive function by analyzing neuroinflammation, mitochondrial function, and neuroplasticity in the hippocampus in a 3xTg-AD mouse model. 5-month-old 3xTg-AD mice performed 12 weeks of exercise with 40-Hz light flickering administered independently and in combination. Spatial learning and memory, long-term memory, hippocampal Aβ, tau, neuroinflammation, pro-inflammatory cytokine expression, mitochondrial function, and neuroplasticity, were analyzed. Aβ and tau proteins levels were significantly reduced in the early stage of AD, resulting in protection against cognitive decline by reducing neuroinflammation and pro-inflammatory cytokines. Furthermore, mitochondrial function improved, apoptosis was reduced, and synapse-related protein expression increased. Overall, exercise with 40-Hz light flickering was significantly more effective than exercise or 40-Hz light flickering alone, and the improvement was comparable to the levels in the non-transgenic aged-match control group. Our results indicate a synergistic effect of exercise and 40-Hz light flickering on pathological improvements in the hippocampus during early AD associated cognitive impairment.
Collapse
Affiliation(s)
- Sang-Seo Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Nebraska, United States
| | - Hye-Sang Park
- Department of Physiology, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Seung-Soo Baek
- Department of Exercise and Health Science, Sangmyung University, Seoul, Republic of Korea
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Nebraska, United States
| | - Cody Philip Anderson
- School of Health and Kinesiology, University of Nebraska at Omaha, Nebraska, United States
| | - Myung-Ki Kim
- Division of Global Sport Studies, Korea University, Sejong, Republic of Korea
| | - Ik-Ryeul Park
- Department of Human Health care, Gyeongsang National University, Jinju, Republic of Korea
| | - Tae-Woon Kim
- Department of Human Health care, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
31
|
Bareiss SK, Johnston T, Lu Q, Tran TD. The effect of exercise on early sensorimotor performance alterations in the 3xTg-AD model of Alzheimer's disease. Neurosci Res 2022; 178:60-68. [PMID: 35033583 DOI: 10.1016/j.neures.2022.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 12/29/2021] [Accepted: 01/12/2022] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is characterized by a progressive decline in cognitive function; however, recent evidence suggests that non-cognitive sensorimotor and psychomotor symptoms accompany early stages of the disease in humans and AD models. Although exercise is emerging as an important therapeutic to combat AD progression, little is known about the effect of exercise on sensorimotor domain functions. The purpose of this study was to determine if early sensorimotor symptoms accompany deficits in Morris water maze (MWM) performance in the 3xTg-AD model, and investigate if exercise could protect against early behavioral decline. 3xTg-AD and wild-type (WT) control mice were subjected to 12 weeks of moderate intensity wheel running or remained sedentary. At 6 months of age, animals underwent a series of sensorimotor and MWM testing. 3xTg-AD mice displayed deficits in sensorimotor function (beam traversal, spontaneous activity, and adhesive removal) and MWM performance. Interestingly, 3xTg-AD animals exhibited increased freezing and unusual shaking/tremoring behaviors not displayed by WT controls. Exercise improved beam traversal, adhesive removal, and reduced the unusual motor-related behaviors in 3xTg-AD mice. Our study shows that sensorimotor symptoms coincide with deficits in MWM performance, and suggest that exercise may mitigate deficits associated with early disease in 3xTg-AD mice.
Collapse
Affiliation(s)
- Sonja K Bareiss
- Department of Physical Therapy, School of Rehabilitation and Movement Sciences, Bellarmine University, Louisville, KY 40205, United States; Department of Neurological Surgery, University of Louisville, United States; The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Diseases Research, East Carolina University, United States.
| | - Tyler Johnston
- Department of Physical Therapy, East Carolina University, Greenville, NC 27834, United States.
| | - Qun Lu
- Department of Anatomy and Cell Biology, Brody School of Medicine, United States; The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Diseases Research, East Carolina University, United States.
| | - Tuan D Tran
- The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Diseases Research, East Carolina University, United States; Department of Psychology East Carolina University, Greenville, NC 27834, United States.
| |
Collapse
|
32
|
Mu L, Cai J, Gu B, Yu L, Li C, Liu QS, Zhao L. Treadmill Exercise Prevents Decline in Spatial Learning and Memory in 3×Tg-AD Mice through Enhancement of Structural Synaptic Plasticity of the Hippocampus and Prefrontal Cortex. Cells 2022; 11:244. [PMID: 35053360 PMCID: PMC8774241 DOI: 10.3390/cells11020244] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/22/2021] [Accepted: 01/08/2022] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by deficits in learning and memory. A pathological feature of AD is the alterations in the number and size of synapses, axon length, dendritic complexity, and dendritic spine numbers in the hippocampus and prefrontal cortex. Treadmill exercise can enhance synaptic plasticity in mouse or rat models of stroke, ischemia, and dementia. The aim of this study was to examine the effects of treadmill exercise on learning and memory, and structural synaptic plasticity in 3×Tg-AD mice, a mouse model of AD. Here, we show that 12 weeks treadmill exercise beginning in three-month-old mice improves spatial working memory in six-month-old 3×Tg-AD mice, while non-exercise six-month-old 3×Tg-AD mice exhibited impaired spatial working memory. To investigate potential mechanisms for the treadmill exercise-induced improvement of spatial learning and memory, we examined structural synaptic plasticity in the hippocampus and prefrontal cortex of six-month-old 3×Tg-AD mice that had undergone 12 weeks of treadmill exercise. We found that treadmill exercise led to increases in synapse numbers, synaptic structural parameters, the expression of synaptophysin (Syn, a presynaptic marker), the axon length, dendritic complexity, and the number of dendritic spines in 3×Tg-AD mice and restored these parameters to similar levels of non-Tg control mice without treadmill exercise. In addition, treadmill exercise also improved these parameters in non-Tg control mice. Strengthening structural synaptic plasticity may represent a potential mechanism by which treadmill exercise prevents decline in spatial learning and memory and synapse loss in 3×Tg-AD mice.
Collapse
Affiliation(s)
- Lianwei Mu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Jiajia Cai
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Laikang Yu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| | - Cui Li
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
- School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou 450001, China
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA;
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing 100084, China; (L.M.); (J.C.); (B.G.); (L.Y.); (C.L.)
| |
Collapse
|
33
|
Penning A, Tosoni G, Abiega O, Bielefeld P, Gasperini C, De Pietri Tonelli D, Fitzsimons CP, Salta E. Adult Neural Stem Cell Regulation by Small Non-coding RNAs: Physiological Significance and Pathological Implications. Front Cell Neurosci 2022; 15:781434. [PMID: 35058752 PMCID: PMC8764185 DOI: 10.3389/fncel.2021.781434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
The adult neurogenic niches are complex multicellular systems, receiving regulatory input from a multitude of intracellular, juxtacrine, and paracrine signals and biological pathways. Within the niches, adult neural stem cells (aNSCs) generate astrocytic and neuronal progeny, with the latter predominating in physiological conditions. The new neurons generated from this neurogenic process are functionally linked to memory, cognition, and mood regulation, while much less is known about the functional contribution of aNSC-derived newborn astrocytes and adult-born oligodendrocytes. Accumulating evidence suggests that the deregulation of aNSCs and their progeny can impact, or can be impacted by, aging and several brain pathologies, including neurodevelopmental and mood disorders, neurodegenerative diseases, and also by insults, such as epileptic seizures, stroke, or traumatic brain injury. Hence, understanding the regulatory underpinnings of aNSC activation, differentiation, and fate commitment could help identify novel therapeutic avenues for a series of pathological conditions. Over the last two decades, small non-coding RNAs (sncRNAs) have emerged as key regulators of NSC fate determination in the adult neurogenic niches. In this review, we synthesize prior knowledge on how sncRNAs, such as microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), may impact NSC fate determination in the adult brain and we critically assess the functional significance of these events. We discuss the concepts that emerge from these examples and how they could be used to provide a framework for considering aNSC (de)regulation in the pathogenesis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Caterina Gasperini
- Neurobiology of miRNAs Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Carlos P. Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
34
|
Xu B, He Y, Liu L, Ye G, Chen L, Wang Q, Chen M, Chen Y, Long D. The Effects of Physical Running on Dendritic Spines and Amyloid-beta Pathology in 3xTg-AD Male Mice. Aging Dis 2022; 13:1293-1310. [PMID: 35855335 PMCID: PMC9286906 DOI: 10.14336/ad.2022.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/10/2022] [Indexed: 11/01/2022] Open
Abstract
Memory loss is the key symptom of Alzheimer's disease (AD). As successful drug treatments have not yet been identified, non-pharmaceutical interventions such as physical exercise and training have been employed to improve the memory function of people with dementia. We investigated the effect of prolonged physical running on hippocampal-dependent spatial memory and its underlying mechanisms using a well-established rodent model of AD. 3xTg-AD transgenic mice and non-transgenic mice were subjected to voluntary wheel running for 5 months (1 hour per day, 5 days per week), followed by spatial memory testing. After the behavioral testing, dendritic spines, synapses, and synaptic proteins as well as amyloid-beta (Aβ) pathology were analyzed in the dorsal hippocampi. Running improved hippocampal-dependent spatial memory in 3xTg-AD mice. This running strategy prevented both thin and mushroom-type spines on CA1 pyramidal cells in 3xTg-AD mice, whereas the effects of running in non-transgenic mice were limited to thin spines. The enormous effects of running on spines were accompanied by an increased number of synapses and upregulated expression of synaptic proteins. Notably, running downregulated the processing of amyloid precursor protein, decreasing intracellular APP expression and extracellular Aβ accumulation, and spatial memory performance correlated with levels of Aβ peptides Aβ1-40 and Aβ1-42. These data suggest that prolonged running may improve memory in preclinical AD via slowing down the amyloid pathology and preventing the loss of synaptic contacts.
Collapse
Affiliation(s)
- Benke Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Yun He
- Department of Human Anatomy, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Lian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Yangtze University, Hubei 434023, China.
| | - Guosheng Ye
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lulu Chen
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Qingning Wang
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Michael Chen
- University of California, Los Angeles, CA 90095, USA.
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA 92697, USA.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| | - Dahong Long
- Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
- Correspondence should be addressed to: Dr. Dahong Long, Key Lab of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China. E-mail: or Dr. Yuncai Chen, Department of Pediatrics, University of California-Irvine, Irvine, California 92697, USA. E-mail:
| |
Collapse
|
35
|
Leite ADOF, Bento Torres Neto J, dos Reis RR, Sobral LL, de Souza ACP, Trévia N, de Oliveira RB, Lins NADA, Diniz DG, Diniz JAP, Vasconcelos PFDC, Anthony DC, Brites D, Picanço Diniz CW. Unwanted Exacerbation of the Immune Response in Neurodegenerative Disease: A Time to Review the Impact. Front Cell Neurosci 2021; 15:749595. [PMID: 34744633 PMCID: PMC8570167 DOI: 10.3389/fncel.2021.749595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
The COVID-19 pandemic imposed a series of behavioral changes that resulted in increased social isolation and a more sedentary life for many across all age groups, but, above all, for the elderly population who are the most vulnerable to infections and chronic neurodegenerative diseases. Systemic inflammatory responses are known to accelerate neurodegenerative disease progression, which leads to permanent damage, loss of brain function, and the loss of autonomy for many aged people. During the COVID-19 pandemic, a spectrum of inflammatory responses was generated in affected individuals, and it is expected that the elderly patients with chronic neurodegenerative diseases who survived SARSCoV-2 infection, it will be found, sooner or later, that there is a worsening of their neurodegenerative conditions. Using mouse prion disease as a model for chronic neurodegeneration, we review the effects of social isolation, sedentary living, and viral infection on the disease progression with a focus on sickness behavior and on the responses of microglia and astrocytes. Focusing on aging, we discuss the cellular and molecular mechanisms related to immunosenescence in chronic neurodegenerative diseases and how infections may accelerate their progression.
Collapse
Affiliation(s)
- Amanda de Oliveira Ferreira Leite
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - João Bento Torres Neto
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Renata Rodrigues dos Reis
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Luciane Lobato Sobral
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Aline Cristine Passos de Souza
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Nonata Trévia
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Roseane Borner de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Nara Alves de Almeida Lins
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| | | | | | | | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| |
Collapse
|
36
|
Liang J, Wang C, Zhang H, Huang J, Xie J, Chen N. Exercise-Induced Benefits for Alzheimer's Disease by Stimulating Mitophagy and Improving Mitochondrial Function. Front Aging Neurosci 2021; 13:755665. [PMID: 34658846 PMCID: PMC8519401 DOI: 10.3389/fnagi.2021.755665] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Neurons are highly specialized post-mitotic cells that are inherently dependent on mitochondria due to their higher bioenergetic demand. Mitochondrial dysfunction is closely associated with a variety of aging-related neurological disorders, such as Alzheimer’s disease (AD), and the accumulation of dysfunctional and superfluous mitochondria has been reported as an early stage that significantly facilitates the progression of AD. Mitochondrial damage causes bioenergetic deficiency, intracellular calcium imbalance and oxidative stress, thereby aggravating β-amyloid (Aβ) accumulation and Tau hyperphosphorylation, and further leading to cognitive decline and memory loss. Although there is an intricate parallel relationship between mitochondrial dysfunction and AD, their triggering factors, such as Aβ aggregation and hyperphosphorylated Tau protein and action time, are still unclear. Moreover, many studies have confirmed abnormal mitochondrial biosynthesis, dynamics and functions will present once the mitochondrial quality control is impaired, thus leading to aggravated AD pathological changes. Accumulating evidence shows beneficial effects of appropriate exercise on improved mitophagy and mitochondrial function to promote mitochondrial plasticity, reduce oxidative stress, enhance cognitive capacity and reduce the risks of cognitive impairment and dementia in later life. Therefore, stimulating mitophagy and optimizing mitochondrial function through exercise may forestall the neurodegenerative process of AD.
Collapse
Affiliation(s)
- Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Cenyi Wang
- School of Physical Education and Sports Science, Soochow University, Suzhou, China
| | - Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, China
| | - Juying Xie
- Affiliated Hospital of Xiangnan University, Chenzhou, China
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan, China
| |
Collapse
|
37
|
Liu Y, Yao J, Song Z, Guo W, Sun B, Wei J, Estillore JP, Back TG, Chen SRW. Limiting RyR2 open time prevents Alzheimer's disease-related deficits in the 3xTG-AD mouse model. J Neurosci Res 2021; 99:2906-2921. [PMID: 34352124 DOI: 10.1002/jnr.24936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/11/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) progression is driven by a vicious cycle of soluble β-amyloid (Aβ)-induced neuronal hyperactivity. Thus, breaking this vicious cycle by suppressing neuronal hyperactivity may represent a logical approach to stopping AD progression. In support of this, we have recently shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevented neuronal hyperactivity, memory impairment, dendritic spine loss, and neuronal cell death in a rapid, early onset AD mouse model (5xFAD). Here, we assessed the impact of limiting RyR2 open time on AD-related deficits in a relatively late occurring, slow developing AD mouse model (3xTG-AD) that bears more resemblance (compared to 5xFAD) to that of human AD. Using behavioral tests, long-term potentiation recordings, and Golgi and Nissl staining, we found that the RyR2-E4872Q mutation, which markedly shortens the open duration of the RyR2 channel, prevented learning and memory impairment, defective long-term potentiation, dendritic spine loss, and neuronal cell death in the 3xTG-AD mice. Furthermore, pharmacologically shortening the RyR2 open time with R-carvedilol rescued these AD-related deficits in 3xTG mice. Therefore, limiting RyR2 open time may offer a promising, neuronal hyperactivity-targeted anti-AD strategy.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Wenting Guo
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Sun
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
38
|
Li B, Mao Q, Zhao N, Xia J, Zhao Y, Xu B. Treadmill exercise overcomes memory deficits related to synaptic plasticity through modulating ionic glutamate receptors. Behav Brain Res 2021; 414:113502. [PMID: 34331969 DOI: 10.1016/j.bbr.2021.113502] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/10/2021] [Accepted: 07/27/2021] [Indexed: 01/06/2023]
Abstract
Neuronal death and synaptic loss are major pathogensis of Alzheimer's disease (AD), which may be related to the ionic glutamate receptors abnormality. Ionic glutamate receptors are important postsynaptic membrane receptors that regulate excitatory synaptic transmission and are also major component of the postsynaptic density. Beta-Amyloid (Aβ) attacks ionic glutamate receptors to reduce synaptic efficacy and synaptic plasticity, resulting in neuronal death and synaptic loss. The current study aimed to investigate whether exercise-ameliorated AD was associated with changes in ionic glutamate receptors. Transgenic APP/PS1 mice (TgAPP/PS1) and age-matched littermate wild mice were divided into wild type control group, wild type exercise group, transgenic control group and transgenic exercise group. The mice in exercise groups were subjected to treadmill training for 12 weeks. The results showed that 12-week treadmill exercise improved the spatial learning and memory abilities of TgAPP/PS1 mice. Moreover, exercise decreased the contents of Aβ40, Aβ42 and amyloid plaque deposition in hippocampus of TgAPP/PS1 mice. The number of synapses and the length and thickness of postsynaptic densities (PSD) in the hippocampal CA1 region of TgAPP/PS1 mice were significantly increased after exercise. Concomitantly, TgAPP/PS1 displayed obstacles in synaptic plasticity as evidenced by significant decreases in the levels of synaptic structural plasticity-related proteins SYN, PSD95, MAP2 and NCAM, as well as ionic glutamate neuroreceptor subunit proteins GluN2B and GluA1. Interestingly, exercise alleviated these synaptic plasticity disorder in TgAPP/PS1 mice. Thus, this study demonstrates that 12-week treadmill exercise reduces Aβ levels in the hippocampus and mitigates cognitive decline in TgAPP/PS1 mice, which may be mediated by improvements in synaptic structural plasticity and excitatory neurotransmission.
Collapse
Affiliation(s)
- Baixia Li
- School of Physical Education and Health Care, East China Normal University, Shanghai, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Qian Mao
- School of Physical Education and Health Care, East China Normal University, Shanghai, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Na Zhao
- School of Physical Education and Health Care, East China Normal University, Shanghai, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Jie Xia
- School of Physical Education and Health Care, East China Normal University, Shanghai, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Yongcai Zhao
- School of Exercise and Health Science, Tianjin University of Sport, Tianjin, China
| | - Bo Xu
- School of Physical Education and Health Care, East China Normal University, Shanghai, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China.
| |
Collapse
|
39
|
Sagud M, Tudor L, Pivac N. Personalized treatment interventions: nonpharmacological and natural treatment strategies in Alzheimer's disease. Expert Rev Neurother 2021; 21:571-589. [PMID: 33749488 DOI: 10.1080/14737175.2021.1906223] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Introduction: Alzheimer's disease (AD) is a slow, irreversible, progressive, complex, and fatal neurodegenerative disorder. Available pharmacological treatment, known for almost two decades, does not cure the disease, but only alleviates the symptoms, with various efficacy and different side effects. Therefore, there is an unmet need to find other person-centered or personalized approaches to treat AD.Areas covered: This article describes the application of precision medicine-like approaches utilizing nonpharmacological treatment strategies and the use of natural products in personalized care for patients with AD.Expert opinion: Due to the heterogeneity of disease symptoms, somatic conditions, and patient preferences, there is definitely no "one size fits all" intervention. Therefore, individualized treatment choice is based on dementia stage, medical and psychiatric comorbidity, leading symptoms, patient preferences, and remaining capacity of the patient. In the absence of disease-modifying agents, a patient-centered, multidisciplinary team approach appears to be the best option to alleviate the heavy symptomatic burden in this unfortunate population. Hence, appropriate interventions can be offered along the AD continuum, while a better understanding of personal characteristics might help in establishing optimal individualized treatment, as well as its duration and intensity, to deliver interventions in the most effective ways.
Collapse
Affiliation(s)
- Marina Sagud
- Department of Psychiatry, Clinical Hospital Centre, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Lucija Tudor
- Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Nela Pivac
- Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| |
Collapse
|
40
|
Babcock KR, Page JS, Fallon JR, Webb AE. Adult Hippocampal Neurogenesis in Aging and Alzheimer's Disease. Stem Cell Reports 2021; 16:681-693. [PMID: 33636114 PMCID: PMC8072031 DOI: 10.1016/j.stemcr.2021.01.019] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022] Open
Abstract
Cognitive deficits associated with Alzheimer's disease (AD) severely impact daily life for the millions of affected individuals. Progressive memory impairment in AD patients is associated with degeneration of the hippocampus. The dentate gyrus of the hippocampus, a region critical for learning and memory functions, is a site of adult neurogenesis in mammals. Recent evidence in humans indicates that hippocampal neurogenesis likely persists throughout life, but declines with age and is strikingly impaired in AD. Our understanding of how neurogenesis supports learning and memory in healthy adults is only beginning to emerge. The extent to which decreased neurogenesis contributes to cognitive decline in aging and AD remains poorly understood. However, studies in rodent models of AD and other neurodegenerative diseases raise the possibility that targeting neurogenesis may ameliorate cognitive dysfunction in AD. Here, we review recent progress in understanding how adult neurogenesis is impacted in the context of aging and AD.
Collapse
Affiliation(s)
- Kelsey R Babcock
- Graduate Program in Neuroscience, Brown University, Providence, RI 02912, USA
| | - John S Page
- Warren Alpert Medical School of Brown University, Providence, RI 02912, USA; Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Justin R Fallon
- Department of Neuroscience, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA
| | - Ashley E Webb
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA; Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
41
|
Lauretta G, Ravalli S, Maugeri G, D'Agata V, Rosa MD, Musumeci G. The impact of physical exercise on hippocampus, in physiological condition and ageing-related decline: current evidence from animal and human studies. Curr Pharm Biotechnol 2021; 23:180-189. [PMID: 33820516 DOI: 10.2174/1389201022666210405142611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/19/2021] [Accepted: 02/14/2021] [Indexed: 11/22/2022]
Abstract
Physical exercise (PE), notoriously, promotes a state of general well-being, throughout the entire human lifespan. Moreover, maintaining an adequate and regular PE habit results to be a powerful preventive factor towards many diseases and may also help in managing existing pathological conditions. PE induces structural and functional changes in various districts of the body, determining biological and psychological benefits. Additionally, in elderly, PE might represent a remarkable tool reducing cognitive impairments related to the normal aging processes and it has also been found to have an impact in neurodegenerative diseases such as Alzheimer's disease. The present review aims to provide an overview about PE effects on hippocampus, since it is one of the brain regions most susceptible to aging and, therefore, involved in diseases characterized by cognitive impairment.
Collapse
Affiliation(s)
- Giovanni Lauretta
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Velia D'Agata
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia n°87, Catania. Italy
| |
Collapse
|
42
|
Büeler H. Mitochondrial and Autophagic Regulation of Adult Neurogenesis in the Healthy and Diseased Brain. Int J Mol Sci 2021; 22:ijms22073342. [PMID: 33805219 PMCID: PMC8036818 DOI: 10.3390/ijms22073342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis is a highly regulated process during which new neurons are generated from neural stem cells in two discrete regions of the adult brain: the subventricular zone of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. Defects of adult hippocampal neurogenesis have been linked to cognitive decline and dysfunction during natural aging and in neurodegenerative diseases, as well as psychological stress-induced mood disorders. Understanding the mechanisms and pathways that regulate adult neurogenesis is crucial to improving preventative measures and therapies for these conditions. Accumulating evidence shows that mitochondria directly regulate various steps and phases of adult neurogenesis. This review summarizes recent findings on how mitochondrial metabolism, dynamics, and reactive oxygen species control several aspects of adult neural stem cell function and their differentiation to newborn neurons. It also discusses the importance of autophagy for adult neurogenesis, and how mitochondrial and autophagic dysfunction may contribute to cognitive defects and stress-induced mood disorders by compromising adult neurogenesis. Finally, I suggest possible ways to target mitochondrial function as a strategy for stem cell-based interventions and treatments for cognitive and mood disorders.
Collapse
Affiliation(s)
- Hansruedi Büeler
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
43
|
Pairojana T, Phasuk S, Suresh P, Huang SP, Pakaprot N, Chompoopong S, Hsieh TC, Liu IY. Age and gender differences for the behavioral phenotypes of 3xTg alzheimer's disease mice. Brain Res 2021; 1762:147437. [PMID: 33753066 DOI: 10.1016/j.brainres.2021.147437] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/14/2021] [Accepted: 03/13/2021] [Indexed: 02/08/2023]
Abstract
The triple transgenic Alzheimer's disease (3xTg-AD) strain is a common mouse model used for studying the pathology and mechanism of Alzheimer's disease (AD). The 3xTg-AD strain exhibits two hallmarks of AD, amyloid beta (Aβ) and neurofibrillary tangles. Several studies using different gender and age of 3xTg-AD mice to investigate their behavior phenotypes under the influence of various treatments have reported mixed results. Therefore, a comprehensive investigation on the optimal gender, age, and training paradigms used for behavioral studies of 3xTg-AD is necessary. In the present study, we investigated the behavioral phenotypes for the two genders of 3xTg-AD mice at 3, 6, 9, and 12 months old and compared the results with age-, gender-matched C57BL/6N control strain. All mice were subjected to tail flick, pinprick, open field, elevated plus maze, passive avoidance, and trace fear conditioning (TFC) tests to evaluate their sensory, locomotor, anxiety, and learning/memory functions. The results showed that TFC on male 3xTg-AD mice is optimal for studying the memory performance in AD. The sensory and locomotor functions of 3xTg-AD mice for two genders appear to be normal before 6 months, decline in fear memory afterwards. The differences between control and 3xTg-AD male mice in contextual and cued memory are robust, thus they are ideal for evaluating the effect of a treatment. Since it is costly and time consuming to obtain wildtype littermates as controls, C57BL/6N strain is suggested to be used as control mice because their baseline performance of sensorimotor functions are similar to that of 3xTg-AD mice.
Collapse
Affiliation(s)
- Tanita Pairojana
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan; Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pavithra Suresh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supin Chompoopong
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Ingrid Y Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
44
|
Liang J, Wang H, Zeng Y, Qu Y, Liu Q, Zhao F, Duan J, Jiang Y, Li S, Ying J, Li J, Mu D. Physical exercise promotes brain remodeling by regulating epigenetics, neuroplasticity and neurotrophins. Rev Neurosci 2021; 32:615-629. [PMID: 33583156 DOI: 10.1515/revneuro-2020-0099] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023]
Abstract
Exercise has been shown to have beneficial effects on brain functions in humans and animals. Exercise can improve memory and learning in age-related neurodegenerative diseases. In animal models, physical exercise regulates epigenetics, promotes synaptic plasticity and hippocampal neurogenesis, regulates the expression levels of neurotrophic factors, and improves cognitive function. Therefore, exercise is very important for brain rehabilitation and remodeling. The purpose of this review is to explore the mechanisms by which exercise exerts positive effects on brain function. This knowledge implies that physical exercise can be used as a non-drug therapy for neurological diseases.
Collapse
Affiliation(s)
- Juan Liang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Yan Zeng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Qian Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Fengyan Zhao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Jianan Duan
- West China Hospital, Sichuan University, Chengdu610041, China
| | - Yin Jiang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), West China Second University Hospital, Sichuan University, Chengdu610041, China
| |
Collapse
|
45
|
Liu H, Zhang H, Ma Y. Molecular mechanisms of altered adult hippocampal neurogenesis in Alzheimer's disease. Mech Ageing Dev 2021; 195:111452. [PMID: 33556365 DOI: 10.1016/j.mad.2021.111452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia globally. AD is a progressive neurodegenerative disorder, eventually manifesting as severe cognitive impairment. Adult hippocampal neurogenesis (AHN) occurs throughout adulthood and plays an important role in hippocampus-dependent learning and memory. The stages of AHN, predominantly comprising the proliferation, differentiation, survival, and maturation of newborn neurons, are affected to varying degrees in AD. However, the exact molecular mechanisms remain to be elucidated. Recent evidence suggests that the molecules involved in AD pathology contribute to the compromised AHN in AD. Notably, various interventions may have common signaling pathways that, once identified, could be harnessed to enhance adult neurogenesis. This in turn could putatively rescue cognitive deficits associated with impaired neurogenesis as observed in animal models of AD. In this manuscript, we review the current knowledge concerning AHN under normal physiological and AD pathological conditions and highlight the possible role of specific molecules in AHN alteration in AD. In addition, we summarize in vivo experiments with emphasis on the effect of the activation of certain key signalings on AHN in AD rodent models. We propose that these signaling targets and corresponding interventions should be considered when developing novel therapies for AD.
Collapse
Affiliation(s)
- Hang Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Han Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Ying Ma
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
46
|
Li Puma DD, Piacentini R, Grassi C. Does Impairment of Adult Neurogenesis Contribute to Pathophysiology of Alzheimer's Disease? A Still Open Question. Front Mol Neurosci 2021; 13:578211. [PMID: 33551741 PMCID: PMC7862134 DOI: 10.3389/fnmol.2020.578211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Adult hippocampal neurogenesis is a physiological mechanism contributing to hippocampal memory formation. Several studies associated altered hippocampal neurogenesis with aging and Alzheimer's disease (AD). However, whether amyloid-β protein (Aβ)/tau accumulation impairs adult hippocampal neurogenesis and, consequently, the hippocampal circuitry, involved in memory formation, or altered neurogenesis is an epiphenomenon of AD neuropathology contributing negligibly to the AD phenotype, is, especially in humans, still debated. The detrimental effects of Aβ/tau on synaptic function and neuronal viability have been clearly addressed both in in vitro and in vivo experimental models. Until some years ago, studies carried out on in vitro models investigating the action of Aβ/tau on proliferation and differentiation of hippocampal neural stem cells led to contrasting results, mainly due to discrepancies arising from different experimental conditions (e.g., different cellular/animal models, different Aβ and/or tau isoforms, concentrations, and/or aggregation profiles). To date, studies investigating in situ adult hippocampal neurogenesis indicate severe impairment in most of transgenic AD mice; this impairment precedes by several months cognitive dysfunction. Using experimental tools, which only became available in the last few years, research in humans indicated that hippocampal neurogenesis is altered in cognitive declined individuals affected by either mild cognitive impairment or AD as well as in normal cognitive elderly with a significant inverse relationship between the number of newly formed neurons and cognitive impairment. However, despite that such information is available, the question whether impaired neurogenesis contributes to AD pathogenesis or is a mere consequence of Aβ/pTau accumulation is not definitively answered. Herein, we attempted to shed light on this complex and very intriguing topic by reviewing relevant literature on impairment of adult neurogenesis in mouse models of AD and in AD patients analyzing the temporal relationship between the occurrence of altered neurogenesis and the appearance of AD hallmarks and cognitive dysfunctions.
Collapse
Affiliation(s)
- Domenica Donatella Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberto Piacentini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
47
|
Krivanek TJ, Gale SA, McFeeley BM, Nicastri CM, Daffner KR. Promoting Successful Cognitive Aging: A Ten-Year Update. J Alzheimers Dis 2021; 81:871-920. [PMID: 33935078 PMCID: PMC8293659 DOI: 10.3233/jad-201462] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
A decade has passed since we published a comprehensive review in this journal addressing the topic of promoting successful cognitive aging, making this a good time to take stock of the field. Because there have been limited large-scale, randomized controlled trials, especially following individuals from middle age to late life, some experts have questioned whether recommendations can be legitimately offered about reducing the risk of cognitive decline and dementia. Despite uncertainties, clinicians often need to at least make provisional recommendations to patients based on the highest quality data available. Converging lines of evidence from epidemiological/cohort studies, animal/basic science studies, human proof-of-concept studies, and human intervention studies can provide guidance, highlighting strategies for enhancing cognitive reserve and preventing loss of cognitive capacity. Many of the suggestions made in 2010 have been supported by additional research. Importantly, there is a growing consensus among major health organizations about recommendations to mitigate cognitive decline and promote healthy cognitive aging. Regular physical activity and treatment of cardiovascular risk factors have been supported by all of these organizations. Most organizations have also embraced cognitively stimulating activities, a heart-healthy diet, smoking cessation, and countering metabolic syndrome. Other behaviors like regular social engagement, limiting alcohol use, stress management, getting adequate sleep, avoiding anticholinergic medications, addressing sensory deficits, and protecting the brain against physical and toxic damage also have been endorsed, although less consistently. In this update, we review the evidence for each of these recommendations and offer practical advice about behavior-change techniques to help patients adopt brain-healthy behaviors.
Collapse
Affiliation(s)
- Taylor J. Krivanek
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Hale Building for Transformative Medicine, Boston, MA, USA
| | - Seth A. Gale
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Hale Building for Transformative Medicine, Boston, MA, USA
| | - Brittany M. McFeeley
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Hale Building for Transformative Medicine, Boston, MA, USA
| | - Casey M. Nicastri
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Hale Building for Transformative Medicine, Boston, MA, USA
| | - Kirk R. Daffner
- Center for Brain/Mind Medicine, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Hale Building for Transformative Medicine, Boston, MA, USA
| |
Collapse
|
48
|
Esselun C, Dilberger B, Silaidos CV, Koch E, Schebb NH, Eckert GP. A Walnut Diet in Combination with Enriched Environment Improves Cognitive Function and Affects Lipid Metabolites in Brain and Liver of Aged NMRI Mice. Neuromolecular Med 2020; 23:140-160. [PMID: 33367957 PMCID: PMC7929966 DOI: 10.1007/s12017-020-08639-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022]
Abstract
This in vivo study aimed to test if a diet enriched with 6% walnuts alone or in combination with physical activity supports healthy ageing by changing the oxylipin profile in brain and liver, improving motor function, cognition, and cerebral mitochondrial function. Female NMRI mice were fed a 6% walnut diet starting at an age of 12 months for 24 weeks. One group was additionally maintained in an enriched environment, one group without intervention served as control. After three months, one additional control group of young mice (3 weeks old) was introduced. Motor and cognitive functions were measured using Open Field, Y-Maze, Rotarod and Passive Avoidance tests. Lipid metabolite profiles were determined using RP-LC-ESI(-)-MS/MS in brain and liver tissues of mice. Cerebral mitochondrial function was characterized by the determination of ATP levels, mitochondrial membrane potential and mitochondrial respiration. Expression of genes involved with mito- and neurogenesis, inflammation, and synaptic plasticity were determined using qRT-PCR. A 6% walnut-enriched diet alone improved spatial memory in a Y-Maze alternation test (p < 0.05) in mice. Additional physical enrichment enhanced the significance, although the overall benefit was virtually identical. Instead, physical enrichment improved motor performance in a Rotarod experiment (p* < 0.05) which was unaffected by walnuts alone. Bioactive oxylipins like hydroxy-polyunsaturated fatty acids (OH-PUFA) derived from linoleic acid (LA) were significantly increased in brain (p** < 0.01) and liver (p*** < 0.0001) compared to control mice, while OH-PUFA of α-linolenic acid (ALA) could only be detected in the brains of mice fed with walnuts. In the brain, walnuts combined with physical activity reduced arachidonic acid (ARA)-based oxylipin levels (p < 0.05). Effects of walnut lipids were not linked to mitochondrial function, as ATP production, mitochondrial membrane potential and mitochondrial respiration were unaffected. Furthermore, common markers for synaptic plasticity and neuronal growth, key genes in the regulation of cytoprotective response to oxidative stress and neuronal growth were unaffected. Taken together, walnuts change the oxylipin profile in liver and brain, which could have beneficial effects for healthy ageing, an effect that can be further enhanced with an active lifestyle. Further studies may focus on specific nutrient lipids that potentially provide preventive effects in the brain.
Collapse
Affiliation(s)
- Carsten Esselun
- Laboratory for Nutrition in Prevention and Therapy, Institute of Nutritional Sciences, Justus-Liebig-University, Biomedical Research Center Seltersberg (BFS), Schubertstr. 81, 35392, Giessen, Germany
| | - Benjamin Dilberger
- Laboratory for Nutrition in Prevention and Therapy, Institute of Nutritional Sciences, Justus-Liebig-University, Biomedical Research Center Seltersberg (BFS), Schubertstr. 81, 35392, Giessen, Germany
| | - Carmina V Silaidos
- Laboratory for Nutrition in Prevention and Therapy, Institute of Nutritional Sciences, Justus-Liebig-University, Biomedical Research Center Seltersberg (BFS), Schubertstr. 81, 35392, Giessen, Germany
| | - Elisabeth Koch
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, 42119, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaussstr. 20, 42119, Wuppertal, Germany
| | - Gunter P Eckert
- Laboratory for Nutrition in Prevention and Therapy, Institute of Nutritional Sciences, Justus-Liebig-University, Biomedical Research Center Seltersberg (BFS), Schubertstr. 81, 35392, Giessen, Germany.
| |
Collapse
|
49
|
Dungan CM, Valentino T, Vechetti IJ, Zdunek CJ, Murphy MP, Lin AL, McCarthy JJ, Peterson CA. Exercise-mediated alteration of hippocampal Dicer mRNA and miRNAs is associated with lower BACE1 gene expression and Aβ 1-42 in female 3xTg-AD mice. J Neurophysiol 2020; 124:1571-1577. [PMID: 33052800 DOI: 10.1152/jn.00503.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Changes to cerebral miRNA expression have been implicated in the progression of Alzheimer's disease (AD), as miRNAs that regulate the expression of gene products involved in amyloid beta (Aβ) processing, such as BACE1, are dysregulated in those that suffer from AD. Exercise training improves cognition and reduces BACE1 and Aβ-plaque burden; however, the mechanisms are not fully understood. Using our progressive weighted wheel running (PoWeR) exercise program, we assessed the effect of 20 wk of exercise training on changes in hippocampal miRNA expression in female 3xTg-AD (3xTg) mice. PoWeR was sufficient to promote muscle hypertrophy and increase myonuclear abundance. Furthermore, PoWeR elevated hippocampal Dicer gene expression in 3xTg mice, while altering miRNA expression toward a more wild-type profile. Specifically, miR-29, which is validated to target BACE1, was significantly lower in sedentary 3xTg mice when compared with wild-type but was elevated following PoWeR. Accordingly, BACE1 gene expression, along with detergent-soluble Aβ1-42, was lower in PoWeR-trained 3xTg mice. Our data suggest that PoWeR training upregulates Dicer gene expression to alter cerebral miRNA expression, which may contribute to reduced Aβ accumulation and delay AD progression.NEW & NOTEWORTHY Previous studies have outlined the beneficial effects of exercise on lowering BACE1 expression and reducing Aβ plaques. This study extends upon the work of others by outlining a new potential mechanism by which exercise elicits beneficial effects on Alzheimer's disease pathology, specifically through modulation of Dicer and miRNA expression. This is the first study to examine Dicer and miRNA expression in the hippocampus of the 3xTg model within the context of exercise.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky.,The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Taylor Valentino
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Ivan J Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | | | - Michael P Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky.,The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
50
|
Broderick TL, Rasool S, Li R, Zhang Y, Anderson M, Al-Nakkash L, Plochocki JH, Geetha T, Babu JR. Neuroprotective Effects of Chronic Resveratrol Treatment and Exercise Training in the 3xTg-AD Mouse Model of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21197337. [PMID: 33020412 PMCID: PMC7582460 DOI: 10.3390/ijms21197337] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
To date, there is no cure or effective treatment for Alzheimer’s disease (AD), a chronic neurodegenerative condition that affects memory, language, and behavior. AD is characterized by neuroinflammation, accumulation of brain amyloid-beta (Aβ) oligomers and neurofibrillary tangles, increased neuronal apoptosis, and loss of synaptic function. Promoting regular exercise and a diet containing polyphenols are effective non-pharmacological approaches that prevent the progression of neurodegenerative diseases. In this study, we measured various conformational toxic species of Aβ and markers of inflammation, apoptosis, endolysosomal degradation, and neuroprotection after 5 months of exercise training (ET), resveratrol (Resv) treatment, or combination treatment in the 3xTg-AD mouse model of AD. Our main results indicate that Resv decreased neuroinflammation and accumulation of Aβ oligomers, increased levels of neurotrophins, synaptic markers, silent information regulator, and decreased markers of apoptosis, autophagy, endolysosomal degradation and ubiquitination in the brains of 3xTg-AD mice. ET improved some markers related to neuroprotection, but when combined with Resv treatment, the benefits achieved were as effective as Resv treatment alone. Our results show that the neuroprotective effects of Resv, ET or Resv and ET are associated with reduced toxicity of Aβ oligomers, suppression of neuronal autophagy, decreased apoptosis, and upregulation of key growth-related proteins.
Collapse
Affiliation(s)
- Tom L. Broderick
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
- Laboratory of Diabetes and Exercise Metabolism, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
- Correspondence: (T.L.B.); (J.R.B.)
| | - Suhail Rasool
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (S.R.); (R.L.); (Y.Z.); (T.G.)
| | - Rongzi Li
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (S.R.); (R.L.); (Y.Z.); (T.G.)
| | - Yuxian Zhang
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (S.R.); (R.L.); (Y.Z.); (T.G.)
| | - Miranda Anderson
- Laboratory of Diabetes and Exercise Metabolism, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - Layla Al-Nakkash
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA;
| | - Jeffrey H. Plochocki
- Department of Medical Education, University of Central Florida, College of Medicine, 6850 Lake Nona Blvd, Orlando, FL 32827, USA;
| | - Thangiah Geetha
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (S.R.); (R.L.); (Y.Z.); (T.G.)
| | - Jeganathan Ramesh Babu
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849, USA; (S.R.); (R.L.); (Y.Z.); (T.G.)
- Correspondence: (T.L.B.); (J.R.B.)
| |
Collapse
|