1
|
Wang Y, Zhang X, Guo H, Qian S, Fang H, Wu X, Shen Y, Xu C, Zhou B, Guo C, Lu X, Zhang X, Jin X. Aging-Induced Episodic-Like Memory Impairment Could be Alleviated by Melatonin Treatment via Preserving Blood-Brain Barrier Integrity and Upregulating CRTC1. CNS Neurosci Ther 2025; 31:e70412. [PMID: 40285336 PMCID: PMC12031892 DOI: 10.1111/cns.70412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/19/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Aging is accompanied by impairments in stimulus recognition, and decreased melatonin levels have been shown in aged mice and humans. These age-related changes are associated with an increased risk of neurological diseases. In the present study, our aim is to investigate whether melatonin supplementation could ameliorate age-related cognitive decline in aged mice. METHODS Mice were treated with melatonin or saline. The novel object recognition (NOR) task was used to provide a simultaneous assessment of object and object location memory, which is a component of episodic-like memory. Blood-brain barrier (BBB) leakage was assessed using an Immunoglobulin G (IgG) leakage assay. Immunofluorescence and Western blot analyses were employed to investigate changes in protein levels. RESULTS We demonstrate that aging impairs memory in the NOR task, with concomitant decreases in the levels of synaptophysin (SYP), CREB-regulated transcription coactivator 1 (CRTC1), and phosphorylated AMP-activated protein kinase (p-AMPK) levels within the prefrontal cortex (PFC) and hippocampus. Moreover, alongside compromised BBB integrity, aging results in the degradation of occludin in both the PFC and hippocampus. Our findings demonstrate that aging impairs memory performance in the NOR task, accompanied by reductions in SYP, CRTC1, and p-AMPK levels within the PFC and hippocampus. Furthermore, alongside compromised BBB integrity, aging results in the degradation of occludin in both the PFC and hippocampus. More importantly, PDZ and LIM domain 5 (Pldim5) was upregulated in melatonin-treated mice, and aging-related memory impairment in the NOR task was significantly reduced in Pdlim5-/- mice. Notably, 1 week of melatonin (10 mg/kg) treatment significantly improved memory, along with enhanced BBB integrity, Pdlim5 downregulation, and CRTC1 and p-AMPK upregulation. CONCLUSIONS Taken together, our findings suggest that melatonin ameliorates aging-related memory decline in the NOR task by downregulating Pdlim5, maintaining BBB integrity, and upregulating CRTC1 and p-AMPK in aged mice.
Collapse
Affiliation(s)
- Yanping Wang
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xinyu Zhang
- Institute of NeuroscienceThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hui Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingPeople's Republic of China
| | - Shuxia Qian
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Hailun Fang
- Department of NeurologyBengbu Medical CollegeBengbuChina
| | - Xiaoqiang Wu
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Yufei Shen
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Congying Xu
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Beiqun Zhou
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Chun Guo
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | - Xudong Lu
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xiaoling Zhang
- Department of NeurologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xinchun Jin
- Institute of NeuroscienceThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingPeople's Republic of China
| |
Collapse
|
2
|
Lv R, Liu B, Jiang Z, Zhou R, Liu X, Lu T, Bao Y, Huang C, Zou G, Zhang Z, Lu L, Yin Q. Intermittent fasting and neurodegenerative diseases: Molecular mechanisms and therapeutic potential. Metabolism 2025; 164:156104. [PMID: 39674569 DOI: 10.1016/j.metabol.2024.156104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Neurodegenerative disorders are straining public health worldwide. During neurodegenerative disease progression, aberrant neuronal network activity, bioenergetic impairment, adaptive neural plasticity impairment, dysregulation of neuronal Ca2+ homeostasis, oxidative stress, and immune inflammation manifest as characteristic pathological changes in the cellular milieu of the brain. There is no drug for the treatment of neurodegenerative disorders, and therefore, strategies/treatments for the prevention or treatment of neurodegenerative disorders are urgently needed. Intermittent fasting (IF) is characterized as an eating pattern that alternates between periods of fasting and eating, requiring fasting durations that vary depending on the specific protocol implemented. During IF, depletion of liver glycogen stores leads to the production of ketone bodies from fatty acids derived from adipocytes, thereby inducing an altered metabolic state accompanied by cellular and molecular adaptive responses within neural networks in the brain. At the cellular level, adaptive responses can promote the generation of synapses and neurons. At the molecular level, IF triggers the activation of associated transcription factors, thereby eliciting the expression of protective proteins. Consequently, this regulatory process governs central and peripheral metabolism, oxidative stress, inflammation, mitochondrial function, autophagy, and the gut microbiota, all of which contribute to the amelioration of neurodegenerative disorders. Emerging evidence suggests that weight regulation significantly contributes to the neuroprotective effects of IF. By alleviating obesity-related factors such as blood-brain barrier dysfunction, neuroinflammation, and β-amyloid accumulation, IF enhances metabolic flexibility and insulin sensitivity, further supporting its potential in mitigating neurodegenerative disorders. The present review summarizes animal and human studies investigating the role and underlying mechanisms of IF in physiology and pathology, with an emphasis on its therapeutic potential. Furthermore, we provide an overview of the cellular and molecular mechanisms involved in regulating brain energy metabolism through IF, highlighting its potential applications in neurodegenerative disorders. Ultimately, our findings offer novel insights into the preventive and therapeutic applications of IF for neurodegenerative disorders.
Collapse
Affiliation(s)
- Renjun Lv
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Bin Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan 250014, China
| | - Ziying Jiang
- Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological Disorders, Beijing, 100053, China
| | - Runfa Zhou
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehlstr. 13-17, Mannheim 68167, Germany
| | - Xiaoxing Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China
| | - Tangsheng Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Chunxia Huang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China
| | - Zongyong Zhang
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, 250117 Jinan, Shandong, China.
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), 100191 Beijing, China; National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871 Beijing, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
3
|
Nas JSB, Medina PMB. Evaluating the effects of sodium metabisulfite on the cognitive and motor function in Drosophila melanogaster. NARRA J 2024; 4:e1338. [PMID: 39816056 PMCID: PMC11731670 DOI: 10.52225/narra.v4i3.1338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025]
Abstract
Sodium metabisulfite is widely used as a preservative in many food and beverage products, yet its potential effects on cognitive and motor functions at low concentrations remain poorly understood. Evaluating learning, short-term memory, and motor activity is essential, as these functions are critical indicators of neurological health and could be impacted by low-level exposure to sodium metabisulfite. The aim of this study was to investigate the effects of sublethal concentrations of sodium metabisulfite on cognitive and motor functions using Drosophila melanogaster (fruit flies) as the model organism. Different levels of sodium metabisulfite were administered to male and female fruit flies, and their learning and short-term memory were observed. Additionally, their climbing activity with and without stressors (heat shock, ultraviolet A exposure, or energy deprivation) was examined. Our findings indicated that sodium metabisulfite did not impair learning, short-term memory, or motor activity. Furthermore, sodium metabisulfite did not affect the motor activity of fruit flies under heat, ultraviolet A, and energy-deprived conditions. In conclusion, our results suggested that the sublethal concentration of sodium metabisulfite did not harm cognitive and motor functions and did not exacerbate the effects of environmental stressors.
Collapse
Affiliation(s)
- John SB. Nas
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila, Philippines
| | - Paul MB. Medina
- Biological Models Laboratory, Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
4
|
Zhao R. Can exercise benefits be harnessed with drugs? A new way to combat neurodegenerative diseases by boosting neurogenesis. Transl Neurodegener 2024; 13:36. [PMID: 39049102 PMCID: PMC11271207 DOI: 10.1186/s40035-024-00428-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Adult hippocampal neurogenesis (AHN) is affected by multiple factors, such as enriched environment, exercise, ageing, and neurodegenerative disorders. Neurodegenerative disorders can impair AHN, leading to progressive neuronal loss and cognitive decline. Compelling evidence suggests that individuals engaged in regular exercise exhibit higher production of proteins that are essential for AHN and memory. Interestingly, specific molecules that mediate the effects of exercise have shown effectiveness in promoting AHN and cognition in different transgenic animal models. Despite these advancements, the precise mechanisms by which exercise mimetics induce AHN remain partially understood. Recently, some novel exercise molecules have been tested and the underlying mechanisms have been proposed, involving intercommunications between multiple organs such as muscle-brain crosstalk, liver-brain crosstalk, and gut-brain crosstalk. In this review, we will discuss the current evidence regarding the effects and potential mechanisms of exercise mimetics on AHN and cognition in various neurological disorders. Opportunities, challenges, and future directions in this research field are also discussed.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, 88 South Daxue Road, Yangzhou, 225009, China.
| |
Collapse
|
5
|
Dong H, Wang S, Hu C, Wang M, Zhou T, Zhou Y. Neuroprotective Effects of Intermittent Fasting in the Aging Brain. ANNALS OF NUTRITION & METABOLISM 2024; 80:175-185. [PMID: 38631305 DOI: 10.1159/000538782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 04/06/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND A major risk factor for neurodegenerative disorders is old age. Nutritional interventions that delay aging, such as calorie restriction (CR) and intermittent fasting (IF), as well as pharmaceuticals that affect the pathways linking nutrition and aging processes, have been developed in recent decades and have been shown to alleviate the effects of aging on the brain. SUMMARY CR is accomplished by alternating periods of ad libitum feeding and fasting. In animal models, IF has been shown to increase lifespan and slow the progression and severity of age-related pathologies such as cardiovascular and neurodegenerative diseases and cancer. According to recent research, dietary changes can help older people with dementia retain brain function. However, the mechanisms underlying the neuroprotective effect of IF on the aging brain and related questions in this area of study (i.e., the potential of IF to treat neurodegenerative disorders) remain to be examined. KEY MESSAGES This review addresses the hypothesis that IF may have translational potential in protecting the aged brain while summarizing the research supporting the putative neuroprotective mechanisms of IF in animal models. Additionally, given the emerging understanding of the connection between aging and dementia, our investigations may offer a fresh perspective on the use of dietary interventions for enhancing brain function and preventing dementia in elderly individuals. Finally, the absence of guidelines regarding the application of IF in patients hampers its broad utilization in clinical practice, and further studies are needed to improve our knowledge of the long-term effects of IF on dementia before it can be widely prescribed. In conclusion, IF may be an ancillary intervention for preserving memory and cognition in elderly individuals.
Collapse
Affiliation(s)
- Hao Dong
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhou
- Department of Pharmaceutical and Medical Equipment, Ba Yi Orthopedic Hospital, Chengdu, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
6
|
Zhao R. Exercise mimetics: a novel strategy to combat neuroinflammation and Alzheimer's disease. J Neuroinflammation 2024; 21:40. [PMID: 38308368 PMCID: PMC10837901 DOI: 10.1186/s12974-024-03031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024] Open
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), characterized by the stimulation of resident immune cells of the brain and the penetration of peripheral immune cells. These inflammatory processes facilitate the deposition of amyloid-beta (Aβ) plaques and the abnormal hyperphosphorylation of tau protein. Managing neuroinflammation to restore immune homeostasis and decrease neuronal damage is a therapeutic approach for AD. One way to achieve this is through exercise, which can improve brain function and protect against neuroinflammation, oxidative stress, and synaptic dysfunction in AD models. The neuroprotective impact of exercise is regulated by various molecular factors that can be activated in the same way as exercise by the administration of their mimetics. Recent evidence has proven some exercise mimetics effective in alleviating neuroinflammation and AD, and, additionally, they are a helpful alternative option for patients who are unable to perform regular physical exercise to manage neurodegenerative disorders. This review focuses on the current state of knowledge on exercise mimetics, including their efficacy, regulatory mechanisms, progress, challenges, limitations, and future guidance for their application in AD therapy.
Collapse
Affiliation(s)
- Renqing Zhao
- College of Physical Education, Yangzhou University, Yangzhou, China.
| |
Collapse
|
7
|
Cho SY, Kim EW, Park SJ, Phillips BU, Jeong J, Kim H, Heath CJ, Kim D, Jang Y, López-Cruz L, Saksida LM, Bussey TJ, Lee DY, Kim E. Reconsidering repurposing: long-term metformin treatment impairs cognition in Alzheimer's model mice. Transl Psychiatry 2024; 14:34. [PMID: 38238285 PMCID: PMC10796941 DOI: 10.1038/s41398-024-02755-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Metformin, a primary anti-diabetic medication, has been anticipated to provide benefits for Alzheimer's disease (AD), also known as "type 3 diabetes". Nevertheless, some studies have demonstrated that metformin may trigger AD pathology and even elevate AD risk in humans. Despite this, limited research has elucidated the behavioral outcomes of metformin treatment, which would hold significant translational value. Thus, we aimed to perform thorough behavioral research on the prolonged administration of metformin to mice: We administered metformin (300 mg/kg/day) to transgenic 3xTg-AD and non-transgenic (NT) C57BL/6 mice over 1 and 2 years, respectively, and evaluated their behaviors across multiple domains via touchscreen operant chambers, including motivation, attention, memory, visual discrimination, and cognitive flexibility. We found metformin enhanced attention, inhibitory control, and associative learning in younger NT mice (≤16 months). However, chronic treatment led to impairments in memory retention and discrimination learning at older age. Furthermore, metformin caused learning and memory impairment and increased levels of AMPKα1-subunit, β-amyloid oligomers, plaques, phosphorylated tau, and GSK3β expression in AD mice. No changes in potential confounding factors on cognition, including levels of motivation, locomotion, appetite, body weight, blood glucose, and serum vitamin B12, were observed in metformin-treated AD mice. We also identified an enhanced amyloidogenic pathway in db/db mice, as well as in Neuro2a-APP695 cells and a decrease in synaptic markers, such as PSD-95 and synaptophysin in primary neurons, upon metformin treatment. Our findings collectively suggest that the repurposing of metformin should be carefully reconsidered when this drug is used for individuals with AD.
Collapse
Affiliation(s)
- So Yeon Cho
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Metabolism-Dementia Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Eun Woo Kim
- Graduate School of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Nursing, Seoyeong University, Gwangju, 61268, Republic of Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Benjamin U Phillips
- Department of Psychology, The University of Cambridge, Cambridge, CB2 3EB, UK
| | - Jihyeon Jeong
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Metabolism-Dementia Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Metabolism-Dementia Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Christopher J Heath
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, MK7 6AA, UK
| | - Daehwan Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yurim Jang
- Interdisciplinary Program in Agricultural Genomics, Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Laura López-Cruz
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, MK7 6AA, UK
| | - Lisa M Saksida
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, N6A 5K8, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Timothy J Bussey
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, N6A 5K8, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, N6A 5C1, Canada
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program in Agricultural Genomics, Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eosu Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Metabolism-Dementia Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Graduate School of Medicine, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
8
|
Mas-Bargues C. Mitochondria pleiotropism in stem cell senescence: Mechanisms and therapeutic approaches. Free Radic Biol Med 2023; 208:657-671. [PMID: 37739140 DOI: 10.1016/j.freeradbiomed.2023.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Aging is a complex biological process characterized by a progressive decline in cellular and tissue function, ultimately leading to organismal aging. Stem cells, with their regenerative potential, play a crucial role in maintaining tissue homeostasis and repair throughout an organism's lifespan. Mitochondria, the powerhouses of the cell, have emerged as key players in the aging process, impacting stem cell function and contributing to age-related tissue dysfunction. Here are discuss the mechanisms through which mitochondria influence stem cell fate decisions, including energy production, metabolic regulation, ROS signalling, and epigenetic modifications. Therefore, this review highlights the role of mitochondria in driving stem cell senescence and the subsequent impact on tissue function, leading to overall organismal aging and age-related diseases. Finally, we explore potential anti-aging therapies targeting mitochondrial health and discuss their implications for promoting healthy aging. This comprehensive review sheds light on the critical interplay between mitochondrial function, stem cell senescence, and organismal aging, offering insights into potential strategies for attenuating age-related decline and promoting healthy longevity.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010, Valencia, Spain.
| |
Collapse
|
9
|
Townsend LK, Steinberg GR. AMPK and the Endocrine Control of Metabolism. Endocr Rev 2023; 44:910-933. [PMID: 37115289 DOI: 10.1210/endrev/bnad012] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Complex multicellular organisms require a coordinated response from multiple tissues to maintain whole-body homeostasis in the face of energetic stressors such as fasting, cold, and exercise. It is also essential that energy is stored efficiently with feeding and the chronic nutrient surplus that occurs with obesity. Mammals have adapted several endocrine signals that regulate metabolism in response to changes in nutrient availability and energy demand. These include hormones altered by fasting and refeeding including insulin, glucagon, glucagon-like peptide-1, catecholamines, ghrelin, and fibroblast growth factor 21; adipokines such as leptin and adiponectin; cell stress-induced cytokines like tumor necrosis factor alpha and growth differentiating factor 15, and lastly exerkines such as interleukin-6 and irisin. Over the last 2 decades, it has become apparent that many of these endocrine factors control metabolism by regulating the activity of the AMPK (adenosine monophosphate-activated protein kinase). AMPK is a master regulator of nutrient homeostasis, phosphorylating over 100 distinct substrates that are critical for controlling autophagy, carbohydrate, fatty acid, cholesterol, and protein metabolism. In this review, we discuss how AMPK integrates endocrine signals to maintain energy balance in response to diverse homeostatic challenges. We also present some considerations with respect to experimental design which should enhance reproducibility and the fidelity of the conclusions.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
10
|
Axelrod CL, Dantas WS, Kirwan JP. Sarcopenic obesity: emerging mechanisms and therapeutic potential. Metabolism 2023; 146:155639. [PMID: 37380015 PMCID: PMC11448314 DOI: 10.1016/j.metabol.2023.155639] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/08/2023] [Accepted: 06/17/2023] [Indexed: 06/30/2023]
Abstract
Sarcopenic obesity, or the loss of muscle mass and function associated with excess adiposity, is a largely untreatable medical condition associated with diminished quality of life and increased risk of mortality. To date, it remains somewhat paradoxical and mechanistically undefined as to why a subset of adults with obesity develop muscular decline, an anabolic stimulus generally associated with retention of lean mass. Here, we review evidence surrounding the definition, etiology, and treatment of sarcopenic obesity with an emphasis on emerging regulatory nodes with therapeutic potential. We review the available clinical evidence largely focused on diet, lifestyle, and behavioral interventions to improve quality of life in patients with sarcopenic obesity. Based upon available evidence, relieving consequences of energy burden, such as oxidative stress, myosteatosis, and/or mitochondrial dysfunction, is a promising area for therapeutic development in the treatment and management of sarcopenic obesity.
Collapse
Affiliation(s)
- Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
11
|
Gim JA, Lee SY, Kim SC, Baek KW, Seo SH, Yoo JI. Relationship between DNA methylation changes and skeletal muscle mass. BMC Genom Data 2023; 24:48. [PMID: 37653517 PMCID: PMC10472633 DOI: 10.1186/s12863-023-01152-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Sarcopenia is a disease diagnosed in the elderly. In patients with sarcopenia, the muscle mass decreases every year. The occurrence of sarcopenia is greatly affected by extrinsic factors such as eating habits, exercise, and lifestyle. The present study aimed to determine the relationship between muscle mass traits and genes affected by epigenetic factors with three different adjustment methods using Korean Genome and Epidemiology Study (KOGES) data. RESULTS We conducted a demographic study and DNA methylation profiling by three studies according to the muscle mass index (MMI) adjustment methods: appendicular skeletal muscle mass divided by body weight (MMI1); appendicular skeletal muscle mass divided by square of height (MMI2); appendicular skeletal muscle mass divided by BMI (MMI3). We analyzed differentially methylated regions (DMRs) for each group. We then restricted our subjects to be top 30% (T30) and bottom 30% (B30) based on each MMI adjustment method. Additionally, we performed enrichment analysis using PathfindR to evaluate the relationship between identified DMRs and sarcopenia. A total of 895 subjects were included in the demographic study. The values of BMI, waist, and hip showed a significant difference in all three groups. Among 446 participants, 44 subjects whose DNA methylation profiles were investigated were included for DNA methylation analysis. The results of enrichment analysis showed differences between groups. In the women group through MMI1 method, only the glutamatergic synapse pathway showed a significant result. In the men group through MMI2 method, the adherens junction pathway was the most significant. Women group through MMI2 method showed similar results, having an enriched Rap1 signaling pathway. In men group through MMI3 method, the Fc epsilon RI signaling pathway was the most enriched. Particularly, the notch signaling pathway was significantly enriched in women group through MMI3 method. CONCLUSION This study presents results about which factor should be concerned first in muscle mass index (MMI) adjustment. The present study suggested that GAB2 and JPH3 in MMI1 method, HLA-DQB1 and TBCD in MMI2 method, GAB2, NDUFB4 and ISPD in MMI3 method are potential genes that can have an impact on muscle mass. It could enable future epigenetic studies of genes based on annotation results. The present study is a nationwide study in Korea with the largest size up to date that compares adjustment indices for MMI in epigenetic research.
Collapse
Affiliation(s)
- Jeong-An Gim
- Department of Medical Science Research Center, College of Medicine, Korea University, Seoul, South Korea
| | - Sang-Yeob Lee
- Department of Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, South Korea
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Seung Chan Kim
- Department of Biostatistics Cooperation Center, Gyeongsang National University Hospital, Jinju, South Korea
| | - Kyung-Wan Baek
- Department of Physical Education, Gyeongsang National University, Jinju, South Korea
- Department of Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju, South Korea
| | - Sung Hyo Seo
- Department of Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, South Korea
| | - Jun-Il Yoo
- Department of Orthopaedic Surgery, Inha University Hospital, 27 Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
12
|
Salminen A. The plasticity of fibroblasts: A forgotten player in the aging process. Ageing Res Rev 2023; 89:101995. [PMID: 37391015 DOI: 10.1016/j.arr.2023.101995] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Tissue-resident fibroblasts are mesenchymal cells which possess an impressive plasticity in their ability to modify their properties according to the requirements of the microenvironment. There are diverse subgroups of fibroblast phenotypes associated with different tissue pathological conditions, e.g., cancers, wound healing, and many fibrotic and inflammatory conditions. The heterogeneous phenotypes can be subdivided into fibrogenic and non-fibrogenic, inflammatory and immunosuppressive subtypes as well as cellular senescent subsets. A major hallmark of activated fibroblasts is that they contain different amounts of stress fibers combined with α-smooth muscle actin (α-SMA) protein, i.e., commonly this phenotype has been called the myofibroblast. Interestingly, several stresses associated with the aging process are potent inducers of myofibroblast differentiation, e.g., oxidative and endoplasmic reticulum stresses, extracellular matrix (ECM) disorders, inflammatory mediators, and telomere shortening. Accordingly, anti-aging treatments with metformin and rapamycin inhibited the differentiation of myofibroblasts in tissues. There is evidence that the senescent phenotype induced in cultured fibroblasts does not represent the phenotype of fibroblasts in aged tissues. Considering the versatile plasticity of fibroblasts as well as their frequency and structural importance in tissues, it does seem that fibroblasts are overlooked players in the aging process.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
13
|
Wang Y, Du W, Sun Y, Zhang J, Ma C, Jin X. CRTC1 is a potential target to delay aging-induced cognitive deficit by protecting the integrity of the blood-brain barrier via inhibiting inflammation. J Cereb Blood Flow Metab 2023; 43:1042-1059. [PMID: 37086081 PMCID: PMC10291461 DOI: 10.1177/0271678x231169133] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
Aging can cause attenuation in the functioning of multiple organs, and blood-brain barrier (BBB) breakdown could promote the occurrence of disorders of the central nervous system during aging. Since inflammation is considered to be an important factor underlying BBB injury during aging, vascular endothelial cell senescence serves as a critical pathological basis for the destruction of BBB integrity. In the current review, we have first introduced the concepts related to aging-induced cognitive deficit and BBB integrity damage. Thereafter, we reviewed the potential relationship between disruption of BBB integrity and cognition deficit and the role of inflammation, vascular endothelial cell senescence, and BBB injury. We have also briefly introduced the function of CREB-regulated transcription co-activator 1 (CRTC1) in cognition and aging-induced CRTC1 changes as well as the critical roles of CRTC1/cyclooxygenase-2 (COX-2) in regulating inflammation, endothelial cell senescence, and BBB injury. Finally, the underlying mechanisms have been summarized and we propose that CRTC1 could be a promising target to delay aging-induced cognitive deficit by protecting the integrity of BBB through promoting inhibition of inflammation-mediated endothelial cell senescence.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurology, the Second Hospital of Jiaxing City, Jiaxing, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- Department of Physiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Chaolin Ma
- School of Life Science and Institute of Life Science, Nanchang University, Nanchang, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Reddy I, Yadav Y, Dey CS. Cellular and Molecular Regulation of Exercise-A Neuronal Perspective. Cell Mol Neurobiol 2023; 43:1551-1571. [PMID: 35986789 PMCID: PMC11412429 DOI: 10.1007/s10571-022-01272-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
The beneficial effects of exercise on the proper functioning of the body have been firmly established. Multi-systemic metabolic regulation of exercise is the consequence of multitudinous changes that occur at the cellular level. The exercise responsome comprises all molecular entities including exerkines, miRNA species, growth factors, signaling proteins that are elevated and activated by physical exercise. Exerkines are secretory molecules released by organs such as skeletal muscle, adipose tissue, liver, and gut as a function of acute/chronic exercise. Exerkines such as FNDC5/irisin, Cathepsin B, Adiponectin, and IL-6 circulate through the bloodstream, cross the blood-brain barrier, and modulate the expression of important signaling molecules such as AMPK, SIRT1, PGC1α, BDNF, IGF-1, and VEGF which further contribute to improved energy metabolism, glucose homeostasis, insulin sensitivity, neurogenesis, synaptic plasticity, and overall well-being of the body and brain. These molecules are also responsible for neuroprotective adaptations that exercise confers on the brain and potentially ameliorate neurodegeneration. This review aims to detail important cellular and molecular species that directly or indirectly mediate exercise-induced benefits in the body, with an emphasis on the central nervous system.
Collapse
Affiliation(s)
- Ishitha Reddy
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Yamini Yadav
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India
| | - Chinmoy Sankar Dey
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, 110016, India.
| |
Collapse
|
15
|
Rice-memolin, a novel peptide derived from rice bran, improves cognitive function after oral administration in mice. Sci Rep 2023; 13:2887. [PMID: 36807368 PMCID: PMC9938899 DOI: 10.1038/s41598-023-30021-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/14/2023] [Indexed: 02/20/2023] Open
Abstract
Many people eat polished rice, while rice bran, a by-product known to be rich in protein and expected to have potential functions for health benefits, has not been effectively utilized. In this study, we determined that orally administered Val-Tyr-Thr-Pro-Gly (VYTPG) derived from rice bran protein improved cognitive decline in mice fed a high-fat diet (HFD). It was demonstrated that VYTPG was released from model peptides corresponding to fragment sequences of original rice proteins (Os01g0941500, Os01g0872700, and allergenic protein) after treatment with thermolysin, a microorganism-derived enzyme often used in industrial scale processes. The thermolysin digest also improved cognitive decline after oral administration in mice. Because VYTPG (1.0 mg/kg) potently improved cognitive decline and is enzymatically produced from the rice bran, we named it rice-memolin. Next, we investigated the mechanisms underlying the cognitive decline improvement associated with rice-memolin. Methyllycaconitine, an antagonist for α7 nicotinic acetylcholine receptor, suppressed the rice-memolin-induced effect, suggesting that rice-memolin improved cognitive decline coupled to the acetylcholine system. Rice-memolin increased the number of 5-bromo-2'-deoxyuridine (BrdU)-positive cells and promoted the mRNA expression of EGF and FGF-2 in the hippocampus, implying that these neurotropic factors play a role in hippocampal neurogenesis after rice-memolin administration. Epidemiologic studies demonstrated that diabetes is a risk factor for dementia; therefore, we also examined the effect of rice-memolin on glucose metabolism. Rice-memolin improved glucose intolerance. In conclusion, we identified a novel rice-derived peptide that can improve cognitive decline. The mechanisms are associated with acetylcholine and hippocampal neurogenesis. Rice-memolin is the first rice-brain-derived peptide able to improve cognitive decline.
Collapse
|
16
|
Ji HZ, Bai CJ, Yun Y, Dong WX, Yi QZ, Ping WD, Yi T, Xia ZH, Min ZZ, Long XJ, Fei ZT, Ji WJ, Zhen L, Zhuang G, Ke LY. Relationships between athletic ability and academic performance in primary school students: A 3-year follow-up study. Front Public Health 2023; 10:1012757. [PMID: 36743154 PMCID: PMC9895930 DOI: 10.3389/fpubh.2022.1012757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
Background The aim of this study was to examine whether academic performance is associated with students' athletic ability in primary school. Methods A 3-year follow-up study was conducted among 1,136 Chinese students. Sit-up and jump rope testers were used to measure 1-min sit-ups and 1-min jump ropes, respectively. Meanwhile, the Pittsburgh Sleep Quality Scale and the Beck Depression Inventory were used to estimate sleep quality and depression levels. The end-of-semester examinations were used to evaluate students' academic performance during the follow-up period. Results After adjusting for confounders, the mean change in Chinese language performance for participants stratified by 1-min sit-ups at baseline was 0.35 (95% CI: -0.37 to 0.76) for level 1 (slowest), 0.52 (95% CI: -0.54 to 1.08) for level 2, and 1.72 (95% CI: 1.14 to 2.30) for level 3 (fastest) (P for trend = 0.003); the mean change in math scores was 0.28 (95% CI: -0.50 to 0.95) for level 1 (slowest), 0.95 (95% CI: 0.38 to 1.52) for level 2, and 1.41 (95% CI: 0.82 to 1.99) for level 3 (fastest) (P for trend = 0.048). The mean change in foreign language scores was -0.45 (95% CI: -0.99 to -0.93) for level 1 (slowest), -0.14 (95% CI: -0.44 to 0.41) for level 2, and 0.69 (95% CI: 0.25 to 1.13) for level 3 (fastest) (P for trend = 0.004). The mean change in Chinese language performance for participants stratified by 1-min jump ropes at the baseline was 0.30 (95% CI: -0.16 to 0.76) for level 1 (slowest), 1.09 (95% CI: 0.42 to 1.76) for level 2, and 1.74 (95% CI: 1.14 to 2.35) for level 3 (fastest) (P for trend = 0.001). The mean change in math scores was 0.41 (95% CI: -0.11 to 0.92) for level 1 (slowest), 1.44 (95% CI: 0.69 to 2.19) for level 2, and 1.43 (95% CI: 0.76 to 2.10) for level 3 (fastest) (P for trend = 0.019). The mean change in foreign language performance was -0.71 (95% CI: -1.08 to -0.33) for level 1 (slowest), 0.95 (95% CI: -0.40 to 1.50) for level 2, and 0.91 (95% CI: 0.41 to 1.41) for level 3 (fastest) (P for trend < 0.001). Conclusion This study suggests that participation in jump rope and sit-up exercises may positively affect students' academic performance.
Collapse
Affiliation(s)
- Hao Zong Ji
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China,*Correspondence: Hao Zong Ji ✉
| | - Cui Jin Bai
- School of Physical Education, Chinese Center of Exercise Epidemiology, Northeast Normal University, Changchun, China
| | - Yang Yun
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Wu Xiang Dong
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Qian Ze Yi
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Wu Dan Ping
- Physical Education Teaching and Research Group, Chengdu Caotang Primary School West Branch, Chengdu, China
| | - Tang Yi
- English Teaching and Research Group, Chongqing Bishan Yuhu Primary School, Chongqing, China
| | - Zheng Hong Xia
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Zhang Zhi Min
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Xu Jing Long
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Zhang Teng Fei
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Wang Jun Ji
- Physical Education Teaching and Research Group, Chongqing Liangjiang Yucai Middle School, Chongqing, China
| | - Li Zhen
- Sports Teaching and Research Group of Xingchen School, Affiliated Middle School of West University, Chongqing, China
| | - Gao Zhuang
- College of Physical Education, Yunnan University, Kunming, China
| | - Li Ying Ke
- School of Physical Education, Southwest University, Chongqing, China
| |
Collapse
|
17
|
Neuronal lack of PDE7a disrupted working memory, spatial learning, and memory but facilitated cued fear memory in mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110655. [PMID: 36220621 DOI: 10.1016/j.pnpbp.2022.110655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND PDEs regulate cAMP levels which is critical for PKA activity-dependent activation of CREB-mediated transcription in learning and memory. Inhibitors of PDEs like PDE4 and Pde7 improve learning and memory in rodents. However, the role of PDE7 in cognition or learning and memory has not been reported yet. METHODS Therefore, we aimed to explore the cognitive effects of a PDE7 subtype, PDE7a, using combined pharmacological and genetic approaches. RESULTS PDE7a-nko mice showed deficient working memory, impaired novel object recognition, deficient spatial learning & memory, and contextual fear memory, contrary to enhanced cued fear memory, highlighting the potential opposite role of PDE7a in the hippocampal neurons. Further, pharmacological inhibition of PDE7 by AGF2.20 selectively strengthens cued fear memory in C57BL/6 J mice, decreasing its extinction but did not affect cognitive processes assessed in other behavioral tests. The further biochemical analysis detected deficient cAMP in neural cell culture with genetic excision of the PDE7a gene, as well as in the hippocampus of PDE7a-nko mice in vivo. Importantly, we found overexpression of PKA-R and the reduced level of pPKA-C in the hippocampus of PDE7a-nko mice, suggesting a novel mechanism of the cAMP regulation by PDE7a. Consequently, the decreased phosphorylation of CREB, CAMKII, eif2a, ERK, and AMPK, and reduced total level of NR2A have been found in the brain of PDE7a-nko animals. Notably, genetic excision of PDE7a in neurons was not able to change the expression of NR2B, BDNF, synapsin1, synaptophysin, or snap25. CONCLUSION Altogether, our current findings demonstrated, for the first time, the role of PDE7a in cognitive processes. Future studies will untangle PDE7a-dependent neurobiological and molecular-cellular mechanisms related to cAMP-associated disorders.
Collapse
|
18
|
Stock AJ, McDevitt RA, Puligilla C, Wang Y, Zhang Y, Wang K, Sun C, Becker KG, Lehrmann E, Wood WH, Gong Y, Aqdas M, Sung MH, Hoffmann V, Liu C, Gorospe M, Harrington L, Ferrucci L, Liu Y. Aberrant expression and localization of the RAP1 shelterin protein contribute to age-related phenotypes. PLoS Genet 2022; 18:e1010506. [PMID: 36441670 PMCID: PMC9704629 DOI: 10.1371/journal.pgen.1010506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/02/2022] [Indexed: 11/29/2022] Open
Abstract
Short telomeres induce a DNA damage response (DDR) that evokes apoptosis and senescence in human cells. An extant question is the contribution of telomere dysfunction-induced DDR to the phenotypes observed in aging and telomere biology disorders. One candidate is RAP1, a telomere-associated protein that also controls transcription at extratelomeric regions. To distinguish these roles, we generated a knockin mouse carrying a mutated Rap1, which was incapable of binding telomeres and did not result in eroded telomeres or a DDR. Primary Rap1 knockin embryonic fibroblasts showed decreased RAP1 expression and re-localization away from telomeres, with an increased cytosolic distribution akin to that observed in human fibroblasts undergoing telomere erosion. Rap1 knockin mice were viable, but exhibited transcriptomic alterations, proinflammatory cytokine/chemokine signaling, reduced lifespan, and decreased healthspan with increased body weight/fasting blood glucose levels, spontaneous tumor incidence, and behavioral deficits. Taken together, our data present mechanisms distinct from telomere-induced DDR that underlie age-related phenotypes.
Collapse
Affiliation(s)
- Amanda J. Stock
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Ross A. McDevitt
- Comparative Medicine Section, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Chandrakala Puligilla
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yajun Wang
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Kun Wang
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Chongkui Sun
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Kevin G. Becker
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - William H. Wood
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yi Gong
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Mohammad Aqdas
- Laboratory of Molecular Biology and Immunology, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Myong-Hee Sung
- Laboratory of Molecular Biology and Immunology, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Victoria Hoffmann
- Division of Veterinary Resources, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Chengyu Liu
- Transgenic Core Facility, National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, Maryland, United States of America
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Lea Harrington
- Institute for Research in Immunology & Cancer, Marcelle-Coutu Pavilion, Université de Montréal, Montreal, Quebec, Canada
| | - Luigi Ferrucci
- Translational Gerontology Branch, Biomedical Research Center, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yie Liu
- Laboratory of Genetics and Genomics, National Institute on Aging/National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Caponio D, Veverová K, Zhang SQ, Shi L, Wong G, Vyhnalek M, Fang EF. Compromised autophagy and mitophagy in brain ageing and Alzheimer's diseases. AGING BRAIN 2022; 2:100056. [PMID: 36908880 PMCID: PMC9997167 DOI: 10.1016/j.nbas.2022.100056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most persistent and devastating neurodegenerative disorders of old age, and is characterized clinically by an insidious onset and a gradual, progressive deterioration of cognitive abilities, ranging from loss of memory to impairment of judgement and reasoning. Despite years of research, an effective cure is still not available. Autophagy is the cellular 'garbage' clearance system which plays fundamental roles in neurogenesis, neuronal development and activity, and brain health, including memory and learning. A selective sub-type of autophagy is mitophagy which recognizes and degrades damaged or superfluous mitochondria to maintain a healthy and necessary cellular mitochondrial pool. However, emerging evidence from animal models and human samples suggests an age-dependent reduction of autophagy and mitophagy, which are also compromised in AD. Upregulation of autophagy/mitophagy slows down memory loss and ameliorates clinical features in animal models of AD. In this review, we give an overview of autophagy and mitophagy and their link to the progression of AD. We also summarize approaches to upregulate autophagy/mitophagy. We hypothesize that age-dependent compromised autophagy/mitophagy is a cause of brain ageing and a risk factor for AD, while restoration of autophagy/mitophagy to more youthful levels could return the brain to health.
Collapse
Affiliation(s)
- Domenica Caponio
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Kateřina Veverová
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Shi-qi Zhang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
- Novo Nordisk Research Centre Oxford (NNRCO)
| | - Garry Wong
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Martin Vyhnalek
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
20
|
Rai M, Demontis F. Muscle-to-Brain Signaling Via Myokines and Myometabolites. Brain Plast 2022; 8:43-63. [PMID: 36448045 PMCID: PMC9661353 DOI: 10.3233/bpl-210133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle health and function are important determinants of systemic metabolic homeostasis and organism-wide responses, including disease outcome. While it is well known that exercise protects the central nervous system (CNS) from aging and disease, only recently this has been found to depend on the endocrine capacity of skeletal muscle. Here, we review muscle-secreted growth factors and cytokines (myokines), metabolites (myometabolites), and other unconventional signals (e.g. bioactive lipid species, enzymes, and exosomes) that mediate muscle-brain and muscle-retina communication and neuroprotection in response to exercise and associated processes, such as the muscle unfolded protein response and metabolic stress. In addition to impacting proteostasis, neurogenesis, and cognitive functions, muscle-brain signaling influences complex brain-dependent behaviors, such as depression, sleeping patterns, and biosynthesis of neurotransmitters. Moreover, myokine signaling adapts feeding behavior to meet the energy demands of skeletal muscle. Contrary to protective myokines induced by exercise and associated signaling pathways, inactivity and muscle wasting may derange myokine expression and secretion and in turn compromise CNS function. We propose that tailoring muscle-to-CNS signaling by modulating myokines and myometabolites may combat age-related neurodegeneration and brain diseases that are influenced by systemic signals.
Collapse
Affiliation(s)
- Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
21
|
McFaline-Figueroa J, Schifino AG, Nichenko AS, Lord MN, Hunda ET, Winders EA, Noble EE, Greising SM, Call JA. Pharmaceutical Agents for Contractile-Metabolic Dysfunction After Volumetric Muscle Loss. Tissue Eng Part A 2022; 28:795-806. [PMID: 35620911 PMCID: PMC9634984 DOI: 10.1089/ten.tea.2022.0036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/21/2022] [Indexed: 11/12/2022] Open
Abstract
Volumetric muscle loss (VML) injuries represent a majority of military service member casualties and are common in civilian populations following blunt and/or penetrating traumas. Characterized as a skeletal muscle injury with permanent functional impairments, there is currently no standard for rehabilitation, leading to lifelong disability. Toward developing rehabilitative strategies, previous research demonstrates that the remaining muscle after a VML injury lacks similar levels of plasticity or adaptability as healthy, uninjured skeletal muscle. This may be due, in part, to impaired innervation and vascularization of the remaining muscle, as well as disrupted molecular signaling cascades commonly associated with muscle adaptation. The primary objective of this study was to assess the ability of four pharmacological agents with a strong record of modulating muscle contractile and metabolic function to improve functional deficits in a murine model of VML injury. Male C57BL/6 mice underwent a 15% multimuscle VML injury of the posterior hindlimb and were randomized into drug treatment groups (formoterol [FOR], 5-aminoimidazole-4-carboxamide riboside [AICAR], pioglitazone [PIO], or sildenafil [SIL]) or untreated VML group. At the end of 60 days, the injury model was first validated by comparison to age-matched injury-naive mice. Untreated VML mice had 22% less gastrocnemius muscle mass, 36% less peak-isometric torque, and 27% less maximal mitochondrial oxygen consumption rate compared to uninjured mice (p < 0.01). Experimental drug groups were, then, compared to VML untreated, and there was minimal evidence of efficacy for AICAR, PIO, or SIL in improving contractile and metabolic functional outcomes. However, FOR-treated VML mice had 18% greater peak isometric torque (p < 0.01) and permeabilized muscle fibers had 36% greater State III mitochondrial oxygen consumption rate (p < 0.01) compared to VML untreated mice, suggesting an overall improvement in muscle condition. There was minimal evidence that these benefits came from greater mitochondrial biogenesis and/or mitochondrial complex protein content, but could be due to greater enzyme activity levels for complex I and complex II. These findings suggest that FOR treatment is candidate to pair with a rehabilitative approach to maximize functional improvements in VML-injured muscle. Impact statement Volumetric muscle loss (VML) injuries result in deficiencies in strength and mobility, which have a severe impact on patient quality of life. Despite breakthroughs in tissue engineering, there are currently no treatments available that can restore function to the affected limb. Our data show that treatment of VML injuries with clinically available and FDA-approved formoterol (FOR), a beta-agonist, significantly improves strength and metabolism of VML-injured muscle. FOR is therefore a promising candidate for combined therapeutic approaches (i.e., regenerative rehabilitation) such as pairing FOR with structured rehabilitation or cell-seeded biomaterials as it may provide greater functional improvements than either strategy alone.
Collapse
Affiliation(s)
- Jennifer McFaline-Figueroa
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, USA
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Albino G. Schifino
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, USA
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Anna S. Nichenko
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, USA
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | - Magen N. Lord
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Edward T. Hunda
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| | | | - Emily E. Noble
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Sarah M. Greising
- School of Kinesiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jarrod A. Call
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, USA
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
22
|
NADPH and Mitochondrial Quality Control as Targets for a Circadian-Based Fasting and Exercise Therapy for the Treatment of Parkinson's Disease. Cells 2022; 11:cells11152416. [PMID: 35954260 PMCID: PMC9367803 DOI: 10.3390/cells11152416] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Dysfunctional mitochondrial quality control (MQC) is implicated in the pathogenesis of Parkinson's disease (PD). The improper selection of mitochondria for mitophagy increases reactive oxygen species (ROS) levels and lowers ATP levels. The downstream effects include oxidative damage, failure to maintain proteostasis and ion gradients, and decreased NAD+ and NADPH levels, resulting in insufficient energy metabolism and neurotransmitter synthesis. A ketosis-based metabolic therapy that increases the levels of (R)-3-hydroxybutyrate (BHB) may reverse the dysfunctional MQC by partially replacing glucose as an energy source, by stimulating mitophagy, and by decreasing inflammation. Fasting can potentially raise cytoplasmic NADPH levels by increasing the mitochondrial export and cytoplasmic metabolism of ketone body-derived citrate that increases flux through isocitrate dehydrogenase 1 (IDH1). NADPH is an essential cofactor for nitric oxide synthase, and the nitric oxide synthesized can diffuse into the mitochondrial matrix and react with electron transport chain-synthesized superoxide to form peroxynitrite. Excessive superoxide and peroxynitrite production can cause the opening of the mitochondrial permeability transition pore (mPTP) to depolarize the mitochondria and activate PINK1-dependent mitophagy. Both fasting and exercise increase ketogenesis and increase the cellular NAD+/NADH ratio, both of which are beneficial for neuronal metabolism. In addition, both fasting and exercise engage the adaptive cellular stress response signaling pathways that protect neurons against the oxidative and proteotoxic stress implicated in PD. Here, we discuss how intermittent fasting from the evening meal through to the next-day lunch together with morning exercise, when circadian NAD+/NADH is most oxidized, circadian NADP+/NADPH is most reduced, and circadian mitophagy gene expression is high, may slow the progression of PD.
Collapse
|
23
|
Luo SS, Zou KX, Zhu H, Cheng Y, Yan YS, Sheng JZ, Huang HF, Ding GL. Integrated Multi-Omics Analysis Reveals the Effect of Maternal Gestational Diabetes on Fetal Mouse Hippocampi. Front Cell Dev Biol 2022; 10:748862. [PMID: 35237591 PMCID: PMC8883435 DOI: 10.3389/fcell.2022.748862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/19/2022] [Indexed: 11/25/2022] Open
Abstract
Growing evidence suggests that adverse intrauterine environments could affect the long-term health of offspring. Recent evidence indicates that gestational diabetes mellitus (GDM) is associated with neurocognitive changes in offspring. However, the mechanism remains unclear. Using a GDM mouse model, we collected hippocampi, the structure critical to cognitive processes, for electron microscopy, methylome and transcriptome analyses. Reduced representation bisulfite sequencing (RRBS) and RNA-seq in the GDM fetal hippocampi showed altered methylated modification and differentially expressed genes enriched in common pathways involved in neural synapse organization and signal transmission. We further collected fetal mice brains for metabolome analysis and found that in GDM fetal brains, the metabolites displayed significant changes, in addition to directly inducing cognitive dysfunction, some of which are important to methylation status such as betaine, fumaric acid, L-methionine, succinic acid, 5-methyltetrahydrofolic acid, and S-adenosylmethionine (SAM). These results suggest that GDM affects metabolites in fetal mice brains and further affects hippocampal DNA methylation and gene regulation involved in cognition, which is a potential mechanism for the adverse neurocognitive effects of GDM in offspring.
Collapse
Affiliation(s)
- Si-Si Luo
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ke-Xin Zou
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hong Zhu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yi Cheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yi-Shang Yan
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Jian-Zhong Sheng
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - He-Feng Huang
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Guo-Lian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
24
|
Iwabu M, Okada-Iwabu M, Kadowaki T, Yamauchi T. Elucidating exercise-induced skeletal muscle signaling pathways and applying relevant findings to preemptive therapy for lifestyle-related diseases. Endocr J 2022; 69:1-8. [PMID: 34511589 DOI: 10.1507/endocrj.ej21-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
While it is well recognized that exercise represents a radical preventive and therapeutic measure for lifestyle-related diseases, it is clear that contemporary lifestyles abound with situations where exercise may be found difficult to implement on a continuous basis. Indeed, this has led to global expectations for elucidation of the exercise-activated skeletal muscle signaling pathways as well as for development of exercise mimics that effectively activate such pathways. It is shown that exercise activates the transcriptional coactivator PGC-1α via AMPK/SIRT1 in muscle, thereby not only enhancing mitochondrial function and muscle endurance but upregulating energy metabolism. Further, adipocyte-derived adiponectin is also shown to activate AMPK/SIRT1/PGC-1α via its receptor AdipoR1 in skeletal muscles. Thus, adiponectin/AdipoR1 signaling is thought to constitute exercise-mimicking signaling. Indeed, it has become clear that AMPK, SIRT1 and AdipoR activators act as exercise mimetics. With the crystal structures of AdipoR elucidated and humanized AdipoR mice generated toward optimization of candidate AdipoR-activators for human use, expectations are mounting for the clinical application in the near future of AdipoR activators as exercise mimetics in humans. This review provides an overview of molecules activated by exercise and compounds activating these molecules, with a focus on the therapeutic potential of AdipoR activators as exercise mimetics.
Collapse
Affiliation(s)
- Masato Iwabu
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Miki Okada-Iwabu
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Advanced Research on Pathophysiology of Metabolic Diseases, The University of Tokyo, Tokyo, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Madhu LN, Kodali M, Shetty AK. Promise of metformin for preventing age-related cognitive dysfunction. Neural Regen Res 2022; 17:503-507. [PMID: 34380878 PMCID: PMC8504370 DOI: 10.4103/1673-5374.320971] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The expanded lifespan of people, while a positive advance, has also amplified the prevalence of age-related disorders, which include mild cognitive impairment, dementia, and Alzheimer's disease. Therefore, competent therapies that could improve the healthspan of people have great significance. Some of the dietary and pharmacological approaches that augment the lifespan could also preserve improved cognitive function in old age. Metformin, a drug widely used for treating diabetes, is one such candidate that could alleviate age-related cognitive dysfunction. However, the possible use of metformin to alleviate age-related cognitive dysfunction has met with conflicting results in human and animal studies. While most clinical studies have suggested the promise of metformin to maintain better cognitive function and reduce the risk for developing dementia and Alzheimer's disease in aged diabetic people, its efficacy in the nondiabetic population is still unclear. Moreover, a previous animal model study implied that metformin could adversely affect cognitive function in the aged. However, a recent animal study using multiple behavioral tests has reported that metformin treatment in late middle age improved cognitive function in old age. The study also revealed that cognition-enhancing effects of metformin in aged animals were associated with the activation of the energy regulator adenosine monophosphate-activated protein kinase, diminished neuroinflammation, inhibition of the mammalian target of rapamycin signaling, and augmented autophagy in the hippocampus. The proficiency of metformin to facilitate these favorable modifications in the aged hippocampus likely underlies its positive effect on cognitive function. Nonetheless, additional studies probing the outcomes of different doses and durations of metformin treatment at specific windows in the middle and old age across sex in nondiabetic and non-obese prototypes are required to substantiate the promise of metformin to maintain better cognitive function in old age.
Collapse
Affiliation(s)
- Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, USA
| |
Collapse
|
26
|
Abstract
Physical exercise can be effective in preventing or ameliorating various diseases, including diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. However, not everyone may be able to participate in exercise due to illnesses, age-related frailty, or difficulty in long-term behavior change. An alternative option is to utilize pharmacological interventions that mimic the positive effects of exercise training. Recent studies have identified signaling pathways associated with the benefits of physical activity and discovered exercise mimetics that can partially simulate the systemic impact of exercise. This review describes the molecular targets for exercise mimetics and their effect on skeletal muscle and other tissues. We will also discuss the potential advantages of using natural products as a multi-targeting agent for mimicking the health-promoting effects of exercise.
Collapse
Affiliation(s)
- Young Jin Jang
- Major of Food Science & Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Sanguine Byun
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
27
|
Jang YJ, Byun S. Molecular targets of exercise mimetics and their natural activators. BMB Rep 2021; 54:581-591. [PMID: 34814977 PMCID: PMC8728540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 02/21/2025] Open
Abstract
Physical exercise can be effective in preventing or ameliorating various diseases, including diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. However, not everyone may be able to participate in exercise due to illnesses, age-related frailty, or difficulty in long-term behavior change. An alternative option is to utilize pharmacological interventions that mimic the positive effects of exercise training. Recent studies have identified signaling pathways associated with the benefits of physical activity and discovered exercise mimetics that can partially simulate the systemic impact of exercise. This review describes the molecular targets for exercise mimetics and their effect on skeletal muscle and other tissues. We will also discuss the potential advantages of using natural products as a multitargeting agent for mimicking the health-promoting effects of exercise. [BMB Reports 2021; 54(12): 581-591].
Collapse
Affiliation(s)
- Young Jin Jang
- Major of Food Science & Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Sanguine Byun
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
28
|
The effect of AMP kinase activation on differentiation and maturation of osteoblast cultured on titanium plate. J Dent Sci 2021; 17:1225-1231. [PMID: 35784162 PMCID: PMC9236888 DOI: 10.1016/j.jds.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/02/2021] [Indexed: 12/01/2022] Open
Abstract
Background/purpose 5′ Adenosine monophosphate-activated protein kinase (AMPK) is known as an enzyme that maintains intracellular homeostasis and has various biological activity. The purpose of this study is evaluation effect of AMPK activation on implant prognosis. Materials & methods MC3T3-E1 osteoblast-like cells were cultured on titanium using a 24-well plate. The experimental group was divided into the following 3 groups: (1) the normal culture group (control group), (2) the osteogenic induction group, and (3) the osteogenic induction + AMPK activation group. The cell counts were measured; real-time PCR was used to assess the expression of ALP and Osterix as osteogenic related genes at Day 0,7,14 and 21 after experiments. Additionally, ALP activity and calcification were assessed. Results The results of the real-time PCR assessments revealed that the expression of ALP, which is a marker for the initial stages of calcification, was significantly increased by AMPK activation compared to the normal culture or osteogenic induction. A significant increase was also observed in the expression of Osterix, which is a marker for the later stages of calcification. Because significant increases were observed in ALP activity and calcification potential, this suggested that AMPK activation could elicit an increase in osteoblast calcification potential. Conclusion AMPK activation promotes implant peripheral osteoblast differentiation and maturation and enhances calcification. Our results suggest that AMPK activation may help to maintain implant stability.
Collapse
|
29
|
Exercise mimetics: harnessing the therapeutic effects of physical activity. Nat Rev Drug Discov 2021; 20:862-879. [PMID: 34103713 DOI: 10.1038/s41573-021-00217-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
Exercise mimetics are a proposed class of therapeutics that specifically mimic or enhance the therapeutic effects of exercise. Increased physical activity has demonstrated positive effects in preventing and ameliorating a wide range of diseases, including brain disorders such as Alzheimer disease and dementia, cancer, diabetes and cardiovascular disease. This article discusses the molecular mechanisms and signalling pathways associated with the beneficial effects of physical activity, focusing on effects on brain function and cognitive enhancement. Emerging therapeutic targets and strategies for the development of exercise mimetics, particularly in the field of central nervous system disorders, as well as the associated opportunities and challenges, are discussed.
Collapse
|
30
|
Gudden J, Arias Vasquez A, Bloemendaal M. The Effects of Intermittent Fasting on Brain and Cognitive Function. Nutrients 2021; 13:nu13093166. [PMID: 34579042 PMCID: PMC8470960 DOI: 10.3390/nu13093166] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
The importance of diet and the gut-brain axis for brain health and cognitive function is increasingly acknowledged. Dietary interventions are tested for their potential to prevent and/or treat brain disorders. Intermittent fasting (IF), the abstinence or strong limitation of calories for 12 to 48 h, alternated with periods of regular food intake, has shown promising results on neurobiological health in animal models. In this review article, we discuss the potential benefits of IF on cognitive function and the possible effects on the prevention and progress of brain-related disorders in animals and humans. We do so by summarizing the effects of IF which through metabolic, cellular, and circadian mechanisms lead to anatomical and functional changes in the brain. Our review shows that there is no clear evidence of a positive short-term effect of IF on cognition in healthy subjects. Clinical studies show benefits of IF for epilepsy, Alzheimer’s disease, and multiple sclerosis on disease symptoms and progress. Findings from animal studies show mechanisms by which Parkinson’s disease, ischemic stroke, autism spectrum disorder, and mood and anxiety disorders could benefit from IF. Future research should disentangle whether positive effects of IF hold true regardless of age or the presence of obesity. Moreover, variations in fasting patterns, total caloric intake, and intake of specific nutrients may be relevant components of IF success. Longitudinal studies and randomized clinical trials (RCTs) will provide a window into the long-term effects of IF on the development and progress of brain-related diseases.
Collapse
Affiliation(s)
- Jip Gudden
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.G.); (A.A.V.)
| | - Alejandro Arias Vasquez
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.G.); (A.A.V.)
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Mirjam Bloemendaal
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.G.); (A.A.V.)
- Correspondence:
| |
Collapse
|
31
|
Townsend LK, MacPherson REK, Wright DC. New Horizon: Exercise and a Focus on Tissue-Brain Crosstalk. J Clin Endocrinol Metab 2021; 106:2147-2163. [PMID: 33982072 DOI: 10.1210/clinem/dgab333] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Indexed: 01/03/2023]
Abstract
The world population is aging, leading to increased rates of neurodegenerative disorders. Exercise has countless health benefits and has consistently been shown to improve brain health and cognitive function. The purpose of this review is to provide an overview of exercise-induced adaptations in the brain with a focus on crosstalk between peripheral tissues and the brain. We highlight recent investigations into exercise-induced circulating factors, or exerkines, including irisin, cathepsin B, GPLD1, and ketones and the mechanisms mediating their effects in the brain.
Collapse
Affiliation(s)
- Logan K Townsend
- Department of Medicine, McMaster University, Hamilton, L8S 4L8, Canada
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, N1G 2W1, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences and Centre for Neuroscience, Brock University, St. Catharines, L2S 3A1, Canada
| | - David C Wright
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, N1G 2W1, Canada
| |
Collapse
|
32
|
Gupta R, Khan R, Cortes CJ. Forgot to Exercise? Exercise Derived Circulating Myokines in Alzheimer's Disease: A Perspective. Front Neurol 2021; 12:649452. [PMID: 34276532 PMCID: PMC8278015 DOI: 10.3389/fneur.2021.649452] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Regular exercise plays an essential role in maintaining healthy neurocognitive function and central nervous system (CNS) immuno-metabolism in the aging CNS. Physical activity decreases the risk of developing Alzheimer's Disease (AD), is associated with better AD prognosis, and positively affects cognitive function in AD patients. Skeletal muscle is an important secretory organ, communicating proteotoxic and metabolic stress to distant tissues, including the CNS, through the secretion of bioactive molecules collectively known as myokines. Skeletal muscle undergoes significant physical and metabolic remodeling during exercise, including alterations in myokine expression profiles. This suggests that changes in myokine and myometabolite secretion may underlie the well-documented benefits of exercise in AD. However, to date, very few studies have focused on specific alterations in skeletal muscle-originating secreted factors and their potential neuroprotective effects in AD. In this review, we discuss exercise therapy for AD prevention and intervention, and propose the use of circulating myokines as novel therapeutic tools for modifying AD progression.
Collapse
Affiliation(s)
- Rajesh Gupta
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rizwan Khan
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Constanza J Cortes
- Department of Cell, Developmental and Integrative Biology (CDIB), School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Neurodegeneration and Experimental Therapeutics (CNET), University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,UAB Nathan Shock Center for the Excellence in the Study of Aging, University of Alabama at Birmingman, Birmingham, AL, United States
| |
Collapse
|
33
|
Swimming Exercise Promotes Post-injury Axon Regeneration and Functional Restoration through AMPK. eNeuro 2021; 8:ENEURO.0414-20.2021. [PMID: 34031101 PMCID: PMC8211466 DOI: 10.1523/eneuro.0414-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/16/2021] [Accepted: 04/25/2021] [Indexed: 12/11/2022] Open
Abstract
Restoration of lost function following a nervous system injury is limited in adulthood as the regenerative capacity of nervous system declines with age. Pharmacological approaches have not been very successful in alleviating the consequences of nervous system injury. On the contrary, physical activity and rehabilitation interventions are often beneficial to improve the health conditions in the patients with neuronal injuries. Using touch neuron circuit of Caenorhabditis elegans, we investigated the role of physical exercise in the improvement of functional restoration after axotomy. We found that a swimming session of 90 min following the axotomy of posterior lateral microtubule (PLM) neuron can improve functional recovery in larval and adult stage animals. In older age, multiple exercise sessions were required to enhance the functional recovery. Genetic analysis of axon regeneration mutants showed that exercise-mediated enhancement of functional recovery depends on the ability of axon to regenerate. Exercise promotes early initiation of regrowth, self-fusion of proximal and distal ends, as well as postregrowth enhancement of function. We further found that the swimming exercise promotes axon regeneration through the activity of cellular energy sensor AAK-2/AMPK in both muscle and neuron. Our study established a paradigm where systemic effects of exercise on functional regeneration could be addressed at the single neuron level.
Collapse
|
34
|
Qiang Q, Manalo JM, Sun H, Zhang Y, Song A, Wen AQ, Wen YE, Chen C, Liu H, Cui Y, Nemkov T, Reisz JA, Edwards III G, Perreira FA, Kellems RE, Soto C, D’Alessandro A, Xia Y. Erythrocyte adenosine A2B receptor prevents cognitive and auditory dysfunction by promoting hypoxic and metabolic reprogramming. PLoS Biol 2021; 19:e3001239. [PMID: 34138843 PMCID: PMC8211187 DOI: 10.1371/journal.pbio.3001239] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022] Open
Abstract
Hypoxia drives aging and promotes age-related cognition and hearing functional decline. Despite the role of erythrocytes in oxygen (O2) transport, their role in the onset of aging and age-related cognitive decline and hearing loss (HL) remains undetermined. Recent studies revealed that signaling through the erythrocyte adenosine A2B receptor (ADORA2B) promotes O2 release to counteract hypoxia at high altitude. However, nothing is known about a role for erythrocyte ADORA2B in age-related functional decline. Here, we report that loss of murine erythrocyte-specific ADORA2B (eAdora2b-/-) accelerates early onset of age-related impairments in spatial learning, memory, and hearing ability. eAdora2b-/- mice display the early aging-like cellular and molecular features including the proliferation and activation of microglia and macrophages, elevation of pro-inflammatory cytokines, and attenuation of hypoxia-induced glycolytic gene expression to counteract hypoxia in the hippocampus (HIP), cortex, or cochlea. Hypoxia sufficiently accelerates early onset of cognitive and cochlear functional decline and inflammatory response in eAdora2b-/- mice. Mechanistically, erythrocyte ADORA2B-mediated activation of AMP-activated protein kinase (AMPK) and bisphosphoglycerate mutase (BPGM) promotes hypoxic and metabolic reprogramming to enhance production of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific metabolite triggering O2 delivery. Significantly, this finding led us to further discover that murine erythroblast ADORA2B and BPGM mRNA levels and erythrocyte BPGM activity are reduced during normal aging. Overall, we determined that erythrocyte ADORA2B-BPGM axis is a key component for anti-aging and anti-age-related functional decline.
Collapse
Affiliation(s)
- Qingfen Qiang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Jeanne M. Manalo
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Hong Sun
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yujin Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Anren Song
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Alexander Q. Wen
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of California at San Diego, La Jolla, California, United States of America
| | - Y. Edward Wen
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas Southwestern Medical School, Dallas, Texas, United States of America
| | - Changhan Chen
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Hong Liu
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Cui
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - George Edwards III
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Fred A. Perreira
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rodney E. Kellems
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| | - Claudio Soto
- Department of Neurology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School, Houston, Texas, United States of America
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas, United States of America
| |
Collapse
|
35
|
Nikbakhtzadeh M, Shaerzadeh F, Ashabi G. Highlighting the protective or degenerative role of AMPK activators in dementia experimental models. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:786-801. [PMID: 34042039 DOI: 10.2174/1871527320666210526160214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/02/2020] [Accepted: 12/21/2020] [Indexed: 11/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a serine/threonine kinase and a driving or deterrent factor in the development of neurodegenerative diseases and dementia. AMPK affects intracellular proteins like the mammalian target of rapamycin (mTOR). Peroxisome proliferator-activated receptor-γ coactivator 1-α (among others) contributes to a wide range of intracellular activities based on its downstream molecules such as energy balancing (ATP synthesis), extracellular inflammation, cell growth, and neuronal cell death (such as apoptosis, necrosis, and necroptosis). Several studies have looked at the dual role of AMPK in neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington disease (HD) but the exact effect of this enzyme on dementia, stroke, and motor neuron dysfunction disorders has not been elucidated yet. In this article, we review current research on the effects of AMPK on the brain to give an overview of the relationship. More specifically, we review the neuroprotective or neurodegenerative effects of AMPK or AMPK activators like metformin, resveratrol, and 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside on neurological diseases and dementia, which exert through the intracellular molecules involved in neuronal survival or death.
Collapse
Affiliation(s)
- Marjan Nikbakhtzadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Shaerzadeh
- Department of Neuroscience, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, United States
| | - Ghorbangol Ashabi
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Jiang J, Chang X, Nie Y, Shen Y, Liang X, Peng Y, Chang M. Peripheral Administration of a Cell-Penetrating MOTS-c Analogue Enhances Memory and Attenuates Aβ 1-42- or LPS-Induced Memory Impairment through Inhibiting Neuroinflammation. ACS Chem Neurosci 2021; 12:1506-1518. [PMID: 33861582 DOI: 10.1021/acschemneuro.0c00782] [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] [Indexed: 12/13/2022] Open
Abstract
MOTS-c is a 16-amino acid mitochondrial derivative peptide reported to be involved in regulating insulin and metabolic homeostasis via the AMP activated protein kinase (AMPK). AMPK agonist AICAR has been reported to improve cognition. Previous reports also pointed out that MOTS-c may be effective as a therapeutic option toward the prevention of the aging processes. Therefore, we investigated the roles of MOTS-c in the memory recognition process. The results showed that central MOTS-c not only enhanced object and location recognition memory formation and consolidation but also ameliorated the memory deficit induced by Aβ1-42 or LPS. The memory-ameliorating effects of MOTS-c could be blocked by AMPK inhibitor dorsomorphin. Moreover, MOTS-c treatment significantly increased the phosphorylation of AMPK but not ERK, JNK, and p38 in the hippocampus. The underlying mechanism of MOTS-c neuroprotection may involve inhibiting the activation of astrocytes and microglia and production of proinflammatory cytokines. In addition, we found that peripheral administration of MOTS-c does not cross the blood-brain barrier (BBB) and plays an effect. In order to improve the brain intake of MOTS-c, we screen out (PRR)5, a cell penetrating peptides, as a carrier for MOTS-c into the brain. Then in the NOR task, intranasal or intravenous MP (cell-penetrating MOTS-c analogue) showed good memory performance on memory formation, memory consolidation, and memory impairment. Near-infrared fluorescent experiments showed the real-time biodistribution in brain after intranasal or intravenous infusion of MP. These results suggested that MOTS-c might be a new potential target for treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- JinHong Jiang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
- Jiangsu Province Key Laboratory in Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Xin Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | - YaoYan Nie
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | - YuXuan Shen
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | - XueYa Liang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | - YaLi Peng
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| | - Min Chang
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu 730000, China
| |
Collapse
|
37
|
Višnjić D, Lalić H, Dembitz V, Tomić B, Smoljo T. AICAr, a Widely Used AMPK Activator with Important AMPK-Independent Effects: A Systematic Review. Cells 2021. [PMID: 34064363 DOI: 10.3390/cellsl0051095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
5-Aminoimidazole-4-carboxamide ribonucleoside (AICAr) has been one of the most commonly used pharmacological modulators of AMPK activity. The majority of early studies on the role of AMPK, both in the physiological regulation of metabolism and in cancer pathogenesis, were based solely on the use of AICAr as an AMPK-activator. Even with more complex models of AMPK downregulation and knockout being introduced, AICAr remained a regular starting point for many studies focusing on AMPK biology. However, there is an increasing number of studies showing that numerous AICAr effects, previously attributed to AMPK activation, are in fact AMPK-independent. This review aims to give an overview of the present knowledge on AMPK-dependent and AMPK-independent effects of AICAr on metabolism, hypoxia, exercise, nucleotide synthesis, and cancer, calling for caution in the interpretation of AICAr-based studies in the context of understanding AMPK signaling pathway.
Collapse
Affiliation(s)
- Dora Višnjić
- Laboratory of Cell Biology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Physiology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Hrvoje Lalić
- Laboratory of Cell Biology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Physiology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Vilma Dembitz
- Laboratory of Cell Biology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Physiology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Barbara Tomić
- Laboratory of Cell Biology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Physiology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Tomislav Smoljo
- Laboratory of Cell Biology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
- Department of Physiology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
38
|
AICAr, a Widely Used AMPK Activator with Important AMPK-Independent Effects: A Systematic Review. Cells 2021; 10:cells10051095. [PMID: 34064363 PMCID: PMC8147799 DOI: 10.3390/cells10051095] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/21/2021] [Accepted: 05/01/2021] [Indexed: 12/24/2022] Open
Abstract
5-Aminoimidazole-4-carboxamide ribonucleoside (AICAr) has been one of the most commonly used pharmacological modulators of AMPK activity. The majority of early studies on the role of AMPK, both in the physiological regulation of metabolism and in cancer pathogenesis, were based solely on the use of AICAr as an AMPK-activator. Even with more complex models of AMPK downregulation and knockout being introduced, AICAr remained a regular starting point for many studies focusing on AMPK biology. However, there is an increasing number of studies showing that numerous AICAr effects, previously attributed to AMPK activation, are in fact AMPK-independent. This review aims to give an overview of the present knowledge on AMPK-dependent and AMPK-independent effects of AICAr on metabolism, hypoxia, exercise, nucleotide synthesis, and cancer, calling for caution in the interpretation of AICAr-based studies in the context of understanding AMPK signaling pathway.
Collapse
|
39
|
Abstract
One of the best strategies for healthy brain aging is regular aerobic exercise. Commonly studied "anti-aging" compounds may mimic some effects of exercise on the brain, but novel approaches that target energy-sensing pathways similar to exercise probably will be more effective in this context. We review evidence in support of this hypothesis by focusing on biological hallmarks of brain aging.
Collapse
|
40
|
Trajano GS, Blazevich AJ. Static Stretching Reduces Motoneuron Excitability: The Potential Role of Neuromodulation. Exerc Sport Sci Rev 2021; 49:126-132. [PMID: 33720914 PMCID: PMC7967995 DOI: 10.1249/jes.0000000000000243] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Prolonged static muscle stretching transiently reduces maximal muscle force, and this force loss has a strong neural component. In this review, we discuss the evidence suggesting that stretching reduces the motoneuron's ability to amplify excitatory drive. We propose a hypothetical model in which stretching causes physiological relaxation, reducing the brainstem-derived neuromodulatory drive necessary to maximize motoneuron discharge rates.
Collapse
Affiliation(s)
- Gabriel S Trajano
- School of Exercise and Nutrition Sciences, Faculty of Health, Queensland University of Technology, Brisbane
| | - Anthony J Blazevich
- Centre for Exercise and Sports Science Research, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| |
Collapse
|
41
|
Kodali M, Attaluri S, Madhu LN, Shuai B, Upadhya R, Gonzalez JJ, Rao X, Shetty AK. Metformin treatment in late middle age improves cognitive function with alleviation of microglial activation and enhancement of autophagy in the hippocampus. Aging Cell 2021; 20:e13277. [PMID: 33443781 PMCID: PMC7884047 DOI: 10.1111/acel.13277] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/29/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022] Open
Abstract
Metformin, a drug widely used for treating diabetes, can prolong the lifespan in several species. Metformin also has the promise to slow down age‐related cognitive impairment. However, metformin's therapeutic use as an anti‐aging drug is yet to be accepted because of conflicting animal and human studies results. We examined the effects of metformin treatment in late middle age on cognitive function in old age. Eighteen‐month‐old male C57BL6/J mice received metformin or no treatment for 10 weeks. A series of behavioral tests revealed improved cognitive function in animals that received metformin. Such findings were evident from a better ability for pattern separation, object location, and recognition memory function. Quantification of microglia revealed that metformin treatment reduced the incidence of pathological microglial clusters with alternative activation of microglia into an M2 phenotype, displaying highly ramified processes in the hippocampus. Metformin treatment also seemed to reduce astrocyte hypertrophy. Additional analysis demonstrated that metformin treatment in late middle age increased adenosine monophosphate‐activated protein kinase activation, reduced proinflammatory cytokine levels, and the mammalian target of rapamycin signaling, and enhanced autophagy in the hippocampus. However, metformin treatment did not alter neurogenesis or synapses in the hippocampus, implying that improved cognitive function with metformin did not involve enhanced neurogenesis or neosynaptogenesis. The results provide new evidence that metformin treatment commencing in late middle age has promise for improving cognitive function in old age. Modulation of microglia, proinflammatory cytokines, and autophagy appear to be the mechanisms by which metformin facilitated functional benefits in the aged brain.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Leelavathi N. Madhu
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Bing Shuai
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Jenny Jaimes Gonzalez
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine Department of Molecular and Cellular Medicine Texas A&M University College of Medicine College Station TX USA
| |
Collapse
|
42
|
Alò R, Fazzari G, Zizza M, Avolio E, Di Vito A, Bruno R, Cuda G, Barni T, Canonaco M, Facciolo RM. Daidzein Pro-cognitive Effects Coincided with Changes of Brain Neurotensin1 Receptor and Interleukin-10 Expression Levels in Obese Hamsters. Neurotox Res 2021; 39:645-657. [PMID: 33428179 DOI: 10.1007/s12640-020-00328-4] [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: 09/26/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 10/22/2022]
Abstract
At present, concerns are pointing to "tasteful" high-fat diets as a cause of conditioning physical-social states that through alterations of some key emotional- and nutritional-related limbic circuits such as hypothalamic and amygdalar areas lead to obesity states. Feeding and energetic homeostatic molecular mechanisms are part of a complex neuronal circuit accounting for this metabolic disorder. In an attempt to exclude conventional drugs for treating obesity, daidzein, a natural glycosidic isoflavone, which mimics estrogenic neuroprotective properties against increased body weight, is beginning to be preferred. In this study, evident anxiolytic-like behaviors were detected following treatment of high-fat diet hamsters with daidzein as shown by extremely evident (p < 0.001) exploration tendencies in novel object recognition test and a notably greater amount of time spent (p < 0.01) in open arms of elevated plus maze. Moreover, the isoflavone promoted a protective role against neurodegeneration processes as shown by few, if any, amino cupric silver granules in amygdalar, hypothalamic and hippocampal neuronal fields when compared with obese hamsters. Interestingly, elevated expression levels of the anorexic neuropeptide receptor neurotensin1 in the above limbic areas of obese hamsters were extremely reduced by daidzein, especially during recovery of cognitive events. Contextually, such effects were strongly paralleled by increased levels of the anti-neuroinflammatory cytokine, interleukin-10. Our results corroborate a neuroprotective ability of this natural glycosidic isoflavone, which through its interaction with the receptor neurotensin1 and interleukin-10 pathways is correlated not only to improved feeding states, and subsequently obesity conditions, but above all to cognitive performances.
Collapse
Affiliation(s)
- Raffaella Alò
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy
| | - Gilda Fazzari
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy
| | - Merylin Zizza
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy
| | - Ennio Avolio
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy
| | - Anna Di Vito
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", Viale Europa, 88100, Catanzaro, Italy
| | - Rosalinda Bruno
- Department of Pharmacy and Science of Health and Nutrition, Polyfunctional Building, University of Calabria, Arcavacata Di Rende, 87036, Cosenza, Italy
| | - Giovanni Cuda
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", Viale Europa, 88100, Catanzaro, Italy
| | - Tullio Barni
- Department of Clinical and Experimental Medicine, University of Catanzaro "Magna Græcia", Viale Europa, 88100, Catanzaro, Italy
| | - Marcello Canonaco
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy.
| | - Rosa Maria Facciolo
- Comparative Neuroanatomy Laboratory of Biology, Ecology and Earth Science Department (DiBEST), University of Calabria, Arcavacata Di Rende, Ponte P. Bucci 4B, 87036, Cosenza, Italy
| |
Collapse
|
43
|
Yu Z, Ling Z, Lu L, Zhao J, Chen X, Xu P, Zou X. Regulatory Roles of Bone in Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:610581. [PMID: 33408628 PMCID: PMC7779400 DOI: 10.3389/fnagi.2020.610581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoporosis and neurodegenerative diseases are two kinds of common disorders of the elderly, which often co-occur. Previous studies have shown the skeletal and central nervous systems are closely related to pathophysiology. As the main structural scaffold of the body, the bone is also a reservoir for stem cells, a primary lymphoid organ, and an important endocrine organ. It can interact with the brain through various bone-derived cells, mostly the mesenchymal and hematopoietic stem cells (HSCs). The bone marrow is also a place for generating immune cells, which could greatly influence brain functions. Finally, the proteins secreted by bones (osteokines) also play important roles in the growth and function of the brain. This article reviews the latest research studying the impact of bone-derived cells, bone-controlled immune system, and bone-secreted proteins on the brain, and evaluates how these factors are implicated in the progress of neurodegenerative diseases and their potential use in the diagnosis and treatment of these diseases.
Collapse
Affiliation(s)
- Zhengran Yu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lin Lu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Zhao
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Orthopaedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Hor YY, Ooi CH, Lew LC, Jaafar MH, Lau ASY, Lee BK, Azlan A, Choi SB, Azzam G, Liong MT. The molecular mechanisms of probiotic strains in improving ageing bone and muscle of d-galactose-induced ageing rats. J Appl Microbiol 2020; 130:1307-1322. [PMID: 32638482 DOI: 10.1111/jam.14776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/05/2020] [Accepted: 07/02/2020] [Indexed: 12/31/2022]
Abstract
AIM The aim of this study was to evaluate the molecular mechanisms of Lactobacillus strains in improving ageing of the musculoskeletal system. METHODS AND RESULTS The anti-ageing mechanism of three probiotics strains Lactobacillus fermentum DR9, Lactobacillus paracasei OFS 0291 and L. helveticus OFS 1515 were evaluated on gastrocnemius muscle and tibia of d-galactose-induced ageing rats. Upon senescence induction, aged rats demonstrated reduced antioxidative genes CAT and SOD expression in both bone and muscle compared to the young rats (P < 0·05). Strain L. fermentum DR9 demonstrated improved expression of SOD in bone and muscle compared to the aged rats (P < 0·05). In the evaluation of myogenesis-related genes, L. paracasei OFS 0291 and L. fermentum DR9 increased the mRNA expression of IGF-1; L. helveticus OFS 1515 and L. fermentum DR9 reduced the expression of MyoD, in contrast to the aged controls (P < 0·05). Protective effects of L. fermentum DR9 on ageing muscle were believed to be contributed by increased AMPK-α2 expression. Among the osteoclastogenesis genes studied, TNF-α expression was highly elevated in tibia of aged rats, while all three probiotics strains ameliorated the expression. Lactobacillus fermentum DR9 also reduced the expression of IL-6 and TRAP in tibia when compared to the aged rats (P < 0·05). All probiotics treatment resulted in declined proinflammatory cytokines IL-1β in muscle and bone. CONCLUSIONS Lactobacillus fermentum DR9 appeared to be the strongest strain in modulation of musculoskeletal health during ageing. SIGNIFICANCE AND IMPACT OF THE STUDY The study demonstrated the protective effects of the bacteria on muscle and bone through antioxidative and anti-inflammatory actions. Therefore, L. fermentum DR9 may serve as a promising targeted anti-ageing therapy.
Collapse
Affiliation(s)
- Y-Y Hor
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | - C-H Ooi
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - L-C Lew
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | - M H Jaafar
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | - A S-Y Lau
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia
| | - B-K Lee
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia
| | - A Azlan
- School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - S-B Choi
- School of Data Sciences, Perdana University, Selangor, Malaysia
| | - G Azzam
- USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia.,School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - M-T Liong
- School of Industrial Technology, Universiti Sains Malaysia, Penang, Malaysia.,USM-RIKEN International Centre for Ageing Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
45
|
Rajpal A, Ismail-Beigi F. Intermittent fasting and 'metabolic switch': Effects on metabolic syndrome, prediabetes and type 2 diabetes. Diabetes Obes Metab 2020; 22:1496-1510. [PMID: 32372521 DOI: 10.1111/dom.14080] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/22/2020] [Accepted: 05/01/2020] [Indexed: 12/14/2022]
Abstract
Various intermittent fasting (IF) dietary plans have gained popularity among obese individuals in recent years as a means of achieving weight loss. However, studies evaluating the effect of IF regimens in people with metabolic syndrome, prediabetes and type 2 diabetes (T2D) are limited. The aim of the present review was to briefly elucidate the biochemical and physiological mechanisms underlying the positive effects of IF, especially the effect of the proposed 'metabolic switch' on metabolism. Next, we examined the efficacy and safety of IF regimens in individuals with metabolic syndrome, prediabetes and T2D. To achieve this, we performed a MEDLINE PubMed search using combinations of various IF terms, including trials in which participants met the additional criteria for metabolic syndrome, prediabetes or T2D. We found four studies in individuals with metabolic syndrome, one study in people with prediabetes, and eight studies in people with T2D evaluating the effects of different IF regimens. The limited available evidence, with small sample sizes and short trial durations, suggests that IF regimens have a similar effectiveness compared with calorie-restriction diets for weight loss and improvement in glycaemic variables. In general, most IF regimens are effective and safe. However, there is an increased risk of hypoglycaemia in patients with T2D who are treated with insulin or sulphonylureas. Moreover, long-term adherence to these regimens appears uncertain. There is a need for large controlled randomized trials to evaluate the efficacy of IF regimens, especially in individuals with metabolic syndrome and prediabetes. If proven to be sustainable and efficacious for prolonged periods, IF could offer a promising approach to improving health at the population level, and would result in multiple public health benefits.
Collapse
Affiliation(s)
- Aman Rajpal
- Department of Medicine, Case Western Reserve University and Cleveland VA Medical Center, Cleveland, Ohio, USA
| | - Faramarz Ismail-Beigi
- Department of Medicine, Case Western Reserve University and Cleveland VA Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Li W, Wu M, Zhang Y, Wei X, Zang J, Liu Y, Wang Y, Gong CX, Wei W. Intermittent fasting promotes adult hippocampal neuronal differentiation by activating GSK-3β in 3xTg-AD mice. J Neurochem 2020; 155:697-713. [PMID: 32578216 DOI: 10.1111/jnc.15105] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/26/2022]
Abstract
Moderate dietary restriction can ameliorate age-related chronic diseases such as Alzheimer's disease (AD) by increasing the expression of neurotrophic factors and promoting neurogenesis in the brain. Glycogen synthase kinase-3β (GSK-3β) signaling is essential for the coordination of progenitor cell proliferation and differentiation during brain development. The mechanisms by which GSK-3β is involved in dietary restriction-induced neurogenesis and cognitive improvement remain unclear. Six-month-old male 3xTg-AD and wild-type mice were fed on alternate days (intermittent fasting, IF) or ad libitum (AL) for 3 months. GSK-3β activity was regulated by bilaterally infusing lentiviral vectors carrying siRNA targeting GSK-3β into the dentate gyrus region of the hippocampus. Intermittent fasting promoted neuronal differentiation and maturation in the dentate gyrus and ameliorated recognized dysfunction in 3xTg-AD mice. These effects were reversed by siRNA targeting GSK-3β. After intermittent fasting, the insulin and protein kinase A signaling pathways were inhibited, while the adenosine monophosphate-activated protein kinase and brain-derived neurotrophic factor pathways were activated. These findings suggest that intermittent fasting can promote neuronal differentiation and maturation in the hippocampus by activating GSK-3β, thus improving learning and memory.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China.,Department of Pathology, The first people's hospital of foshan, Foshan, Guangdong, P. R. China
| | - Meijian Wu
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yilin Zhang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Xuemin Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Jiankun Zang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Yinghua Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, P. R. China
| | - Yanping Wang
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wei Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
47
|
Endosomal-lysosomal dysfunction in metabolic diseases and Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:303-324. [PMID: 32739009 DOI: 10.1016/bs.irn.2020.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The endosomal-lysosomal pathways and related autophagic processes are responsible for proteostasis, involving complexes between lysosomes and autophagosomes. Lysosomes are a key component of homeostasis, involved in cell signaling, metabolism, and quality control, and they experience functional compromise in metabolic diseases, aging, and neurodegenerative diseases. Many genetic mutations and risk factor genes associated with proteinopathies, as well as with metabolic diseases like diabetes, negatively influence endocytic trafficking and autophagic clearance. In contrast, health-improving exercise induces autophagy-lysosomal degradation, perhaps promoting efficient digestion of injured organelles so that undamaged organelles ensure cellular healthiness. Reductions in lysosomal hydrolases are implicated in Alzheimer's, Parkinson's, and lysosomal storage diseases, as well as obesity-related pathology, and members of the cathepsin enzyme family are involved in clearing both Aβ42 and α-synuclein. Upregulation of cathepsin hydrolases improves synaptic and memory functions in models of dementia and in exercising humans, thus identifying lysosomal-related systems as vital for healthy cognitive aging.
Collapse
|
48
|
Shu H, Wang M, Song M, Sun Y, Shen X, Zhang J, Jin X. Acute Nicotine Treatment Alleviates LPS-Induced Impairment of Fear Memory Reconsolidation Through AMPK Activation and CRTC1 Upregulation in Hippocampus. Int J Neuropsychopharmacol 2020; 23:687-699. [PMID: 32516360 PMCID: PMC7727489 DOI: 10.1093/ijnp/pyaa043] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Fear memory is a fundamental capability for animals and humans to survive. Its impairment results in the disability to avoid danger. When memory is reactivated, a reconsolidation process, which can be disrupted by various stimuli, including inflammation, is required to become permanent. Nicotine has been shown to improve cognitive deficits induced by inflammation and other stimuli. Therefore, in the present study, we investigated the effect of nicotine on lipopolysaccharide (LPS)-induced impairment of fear memory reconsolidation and the underlying mechanism. METHODS Step-through inhibitory avoidance task was recruited to study fear memory of rat, i.p. LPS (0.5 mg/kg) treatment was used to induce inflammation, and western blot and immunostaining were applied to detect protein expression and distribution in medial prefrontal cortex and hippocampus. RESULTS Our data showed that LPS induced fear memory reconsolidation impairment without affecting retrieval. In addition, LPS significantly increased inflammation factors tumor necrosis factor-α and interleukin-1 beta and decreased CREB-regulated transcription coactivator 1 (CRTC1) expression and adenosine monophosphate-activated protein kinase (AMPK) activation in hippocampus. More importantly, LPS significantly decreased CRTC1 expression and AMPK activation in neurons by activating microglia cells. Of note, either nicotine treatment or activation of AMPK by intracerebroventricular infusion of metformin reduced LPS-induced impairment of fear memory reconsolidation and ameliorated inflammation factor tumor necrosis factor-α and interleukin-1 beta as well as the expression of CRTC1. CONCLUSIONS In conclusion, our results showed that acute nicotine treatment alleviates LPS-induced impairment of fear memory reconsolidation through activation of AMPK and upregulation of CRTC1 in hippocampus.
Collapse
Affiliation(s)
- Hui Shu
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China,Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou, China
| | - Mengwei Wang
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Song
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianzhi Shen
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- School of Medicine, Ningbo University, Ningbo, China,Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo, China,Correspondence: Xinchun Jin, PhD, School of Basic Medical Sciences, Capital Medical University, Beijing, 100054, China () or Junfang Zhang, PhD, Ningbo University, Ningbo, 315211, China ()
| | - Xinchun Jin
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China,Correspondence: Xinchun Jin, PhD, School of Basic Medical Sciences, Capital Medical University, Beijing, 100054, China () or Junfang Zhang, PhD, Ningbo University, Ningbo, 315211, China ()
| |
Collapse
|
49
|
Mollo N, Cicatiello R, Aurilia M, Scognamiglio R, Genesio R, Charalambous M, Paladino S, Conti A, Nitsch L, Izzo A. Targeting Mitochondrial Network Architecture in Down Syndrome and Aging. Int J Mol Sci 2020; 21:E3134. [PMID: 32365535 PMCID: PMC7247689 DOI: 10.3390/ijms21093134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are organelles that mainly control energy conversion in the cell. In addition, they also participate in many relevant activities, such as the regulation of apoptosis and calcium levels, and other metabolic tasks, all closely linked to cell viability. Functionality of mitochondria appears to depend upon their network architecture that may dynamically pass from an interconnected structure with long tubular units, to a fragmented one with short separate fragments. A decline in mitochondrial quality, which presents itself as an altered structural organization and a function of mitochondria, has been observed in Down syndrome (DS), as well as in aging and in age-related pathologies. This review provides a basic overview of mitochondrial dynamics, from fission/fusion mechanisms to mitochondrial homeostasis. Molecular mechanisms determining the disruption of the mitochondrial phenotype in DS and aging are discussed. The impaired activity of the transcriptional co-activator PGC-1α/PPARGC1A and the hyperactivation of the mammalian target of rapamycin (mTOR) kinase are emerging as molecular underlying causes of these mitochondrial alterations. It is, therefore, likely that either stimulating the PGC-1α activity or inhibiting mTOR signaling could reverse mitochondrial dysfunction. Evidence is summarized suggesting that drugs targeting either these pathways or other factors affecting the mitochondrial network may represent therapeutic approaches to improve and/or prevent the effects of altered mitochondrial function. Overall, from all these studies it emerges that the implementation of such strategies may exert protective effects in DS and age-related diseases.
Collapse
Affiliation(s)
- Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Cicatiello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Miriam Aurilia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Roberta Scognamiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Genesio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Charalambous
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy
| | - Simona Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Conti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council, 80131 Naples, Italy
| | - Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
50
|
Petr MA, Tulika T, Carmona-Marin LM, Scheibye-Knudsen M. Protecting the Aging Genome. Trends Cell Biol 2020; 30:117-132. [DOI: 10.1016/j.tcb.2019.12.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022]
|