1
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
2
|
Gluck L, Gerstein B, Kaunzner UW. Repair mechanisms of the central nervous system: From axon sprouting to remyelination. Neurotherapeutics 2025:e00583. [PMID: 40348704 DOI: 10.1016/j.neurot.2025.e00583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
The central nervous system (CNS), comprising the brain, spinal cord, and optic nerve, has limited regenerative capacity, posing significant challenges in treating neurological disorders. Recent advances in neuroscience and neurotherapeutics have introduced promising strategies to stimulate CNS repair, particularly in the context of neurodegenerative diseases such as multiple sclerosis. This review explores the complex interplay between inflammation, demyelination, and remyelination possibilities. Glial cells, including oligodendrocyte precursors, oligodendrocytes, astrocytes and microglia play dual roles in injury response, with reactive gliosis promoting repair but also potentially inhibiting recovery through glial scar formation. There is also an emphasis on axonal regeneration, axonal sprouting and stem cell therapies. We highlight the role of neuroplasticity in recovery post-injury and the limited regenerative potential of axons in the CNS due to inhibitory factors such as myelin-associated inhibitors. Moreover, neurotrophic factors support neuronal survival and axonal growth, while stem cell-based approaches offer promise for replacing lost neurons and glial cells. However, challenges such as stem cell survival, integration, and risk of tumor formation remain. Furthermore, we examine the role of neurogenesis in CNS repair and the remodeling of the extracellular matrix, which can facilitate regeneration. Through these diverse mechanisms, ongoing research aims to overcome the intrinsic and extrinsic barriers to CNS repair and advance therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lauren Gluck
- Montefiore Medical Center, 1250 Waters Place Tower 2, Bronx, NY 10461, USA.
| | - Brittany Gerstein
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| | - Ulrike W Kaunzner
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| |
Collapse
|
3
|
Cagigas ML, De Ciutiis I, Masedunskas A, Fontana L. Dietary and pharmacological energy restriction and exercise for healthspan extension. Trends Endocrinol Metab 2025:S1043-2760(25)00076-1. [PMID: 40318928 DOI: 10.1016/j.tem.2025.04.001] [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: 02/23/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
Extending healthspan - the years lived in optimal health - holds transformative potential to reduce chronic diseases and healthcare costs. Dietary restriction (DR), particularly when combined with nutrient-rich diets and exercise, is among the most effective, evidence-based strategies for enhancing metabolic health and longevity. By targeting fundamental pathways, it mitigates the onset and progression of obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), neurodegeneration, and cancer. This review synthesizes human data on the impact of DR and exercise on metabolic and age-related diseases, while emphasizing key biological mechanisms such as nutrient sensing, insulin sensitivity, inflammation, mitochondrial function, and gut microbiota. We also examine the emerging role of pharmacologically induced DR, focusing on glucagon-like peptide 1 (GLP-1) receptor agonists (RAs) that partially mimic DR and present opportunities for chronic disease prevention.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Schmanske N, Ngo JM, Kalra K, Nanna MG, Damluji AA. Healthy ageing in older adults with cardiovascular disease. Eur Heart J 2025:ehaf231. [PMID: 40296653 DOI: 10.1093/eurheartj/ehaf231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/20/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
As life expectancy continues to increase due to advancements in medical technology, public health, nutrition, and socioeconomic progress, the population of older adults in the USA and Europe is rapidly growing. By 2050, individuals aged 65 and older are projected to constitute over 20% of the US population and 29% of the European population, leading to a higher prevalence of chronic diseases, including cardiovascular disease. Cardiovascular disease, the leading cause of death in the USA, poses significant challenges to healthy ageing by contributing to accelerated biological ageing and the development of geriatric syndromes. This state-of-the-art review aims to (i) define healthy ageing for older patients living with cardiovascular disease; (ii) compare chronological vs biological ageing as it pertains to cardiovascular disease; (iii) describe the impact of geriatric syndromes and provide an approach to management and prevention; and (iv) address the gaps in knowledge and future directions for potential interventions that could promote healthy ageing.
Collapse
Affiliation(s)
- Nathalie Schmanske
- Cardiovascular Center on Aging, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jonathan M Ngo
- Cardiovascular Center on Aging, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kriti Kalra
- MedStar Washington Hospital Center, Washington, DC, USA
| | | | - Abdulla A Damluji
- Cardiovascular Center on Aging, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, 3300 Gallows Road, Falls Church, Baltimore, MD 22042, USA
| |
Collapse
|
5
|
Zhang Q, Schultz J, Simmering J, Kirkpatrick BQ, Weber MA, Skuodas S, Hicks T, Pierce G, Laughlin M, Bertolli AX, Larson T, Thangavel R, Oya M, Meyerholz D, Aldridge G, Fassler J, Narayanan NS. Glycolysis-enhancing α1-adrenergic antagonists are neuroprotective in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647018. [PMID: 40236185 PMCID: PMC11996510 DOI: 10.1101/2025.04.03.647018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Terazosin (TZ) is an α 1 -adrenergic receptor antagonist that enhances glycolysis by activating the enzyme phosphoglycerate kinase 1 (PGK1). Epidemiological data suggest that TZ may be neuroprotective in Parkinson's disease and in dementia with Lewy bodies and that glycolysis-enhancing drugs might be protective in other neurodegenerative diseases involving protein aggregation, such as Alzheimer's disease (AD). We investigated TZ in AD and report four main results. First, we found that TZ increased ATP levels in a Saccharomyces cerevisiae mutant with impaired energy homeostasis and reduced the aggregation of the AD-associated protein, amyloid beta (Aβ) 42. Second, in an AD transgenic mouse model (5xFAD) we found that TZ attenuated amyloid pathology in the hippocampus and rescued cognitive impairments in spatial memory and interval timing behavioral assays. Third, using the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, we found that AD patients newly started on TZ or related glycolysis-enhancing drugs had a slower progression of both cognitive dysfunction and neuroimaging biomarkers, such as 18 F-fluorodeoxyglucose positron emission tomography (FDG-PET), a measure of brain metabolism. Finally, in a large human administrative dataset, we found that patients taking TZ or related glycolysis-enhancing drugs had a lower hazard of being diagnosed with AD compared to those taking tamsulosin or 5-alpha reductase inhibitors. These data further implicate metabolism in neurodegenerative diseases and suggest that glycolysis-enhancing drugs may be neuroprotective in AD.
Collapse
|
6
|
Popescu I, Lenain M, Rovini E, Ruthsatz M. Lifestyle Interventions and Innovative Approaches for the Management of Neurodegenerative Disorders in Older Adults-State-Of-The-Art and Future Directions. Am J Lifestyle Med 2025:15598276251330170. [PMID: 40161281 PMCID: PMC11954138 DOI: 10.1177/15598276251330170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Aging increases the risk of neurodegenerative diseases (NDs) like Alzheimer's (AD) and Parkinson's (PD), characterized by neuronal loss and cognitive decline. Potential preventive/therapeutic interventions also include lifestyle changes like nutrition & diet, exercise, leisure and social engagement. Here, we discuss several lifestyle interventions for healthy brain aging, and in older adults with NDs. Balanced diets like the Mediterranean and MIND diets can reduce cognitive decline during aging. Long-term use of specific nutrient combinations in medical food may also exert benefits on memory. Metabolic interventions like calorie restriction (CR) and intermittent fasting (IF) have shown potential benefits in aging. However, their effects on cognitive function in older adults were modestly explored and remain unclear. Clinical trials are ongoing to test the cognitive and health outcomes of CR/IF variants in older adults with AD or PD (associated or not with metabolic disorders). We also highlight the role of cognitive reserve (CoR) in delaying dementia symptoms. Engaging in diverse leisure activities like music and bilingualism may enhance CoR and reduce AD risk. Finally, we outline future directions for promoting healthy brain aging through lifestyle interventions (e.g., personalized diets, precision nutrition, synergistic lifestyle approaches, AI-based technologies to monitor the effectiveness of lifestyle practices).
Collapse
Affiliation(s)
- Iuliana Popescu
- Barnstable Brown Diabetes Research Centre, University of Kentucky, Lexington, KY, USA (IP)
| | - Manon Lenain
- Centre de Prévention Santé Longévité (CPSL), Institut Pasteur de Lille, Sciences Cognitives et Sciences Affectives (SCALab), Université de Lille, Lille, France (ML)
| | - Erika Rovini
- Department of Industrial Engineering, University of Florence, Florence, Italy (ER)
| | | |
Collapse
|
7
|
Yang X, Miao X, Schweiggart F, Großmann S, Rauss K, Hallschmid M, Born J, Lutz ND. The effect of fasting on human memory consolidation. Neurobiol Learn Mem 2025; 218:108034. [PMID: 39938634 DOI: 10.1016/j.nlm.2025.108034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/17/2024] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
The consolidation of long-term memory is thought to critically rely on sleep. However, first evidence from a study in Drosophila suggests that hunger, as another brain state, can benefit memory consolidation as well. Here, we report two human (within-subjects crossover) experiments examining the effects of fasting (versus satiated conditions) during a 10-hour post-encoding consolidation period on subsequent recall of declarative and procedural memories in healthy men. In Experiment 1, participants (n = 16), after an 18.5-hour fasting period, encoded 3 memory tasks (word paired associates, a visual version of the Deese-Roediger-McDermott task, finger tapping) and subsequently either continued to fast or received standardized meals. Recall was tested 48 h later in a satiated state. Experiment 2 (n = 16 participants) differed from Experiment 1 in that a What-Where-When episodic memory task replaced the Deese-Roediger-McDermott task and recall was tested only 24 h later in a fasted state. Compared with the satiated state, fasting enhanced cued recall of word paired associates (more correct and faster responses) and item recognition in the What-Where-When task. By contrast, fasting impaired recall of episodic context memory, i.e., spatial context in the Deese-Roediger-McDermott task, and temporal-spatial context in the What-Where-When task. Procedural memory (finger tapping) remained unaffected. This pattern suggests a differential effect of fasting selectively promoting consolidation of semantic-like representations in cortical networks whereas hippocampal representations of episodic context are weakened. We speculate that hunger strengthens cortical representations by suppressing hippocampal interference during wake consolidation. Yet, the underlying mechanism remains to be clarified.
Collapse
Affiliation(s)
- Xuefeng Yang
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Graduate School of Neural & Behavioural Science, International Max Planck Research School, Tübingen, Germany
| | - Xiu Miao
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Graduate School of Neural & Behavioural Science, International Max Planck Research School, Tübingen, Germany
| | - Franziska Schweiggart
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Sophia Großmann
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Karsten Rauss
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
| | - Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research & Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Germany; German Center for Mental Health (DZPG), Tübingen, Germany
| | - Jan Born
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), Tübingen, Germany; Institute for Diabetes Research & Metabolic Diseases of the Helmholtz Center Munich at the University Tübingen (IDM), Germany; German Center for Mental Health (DZPG), Tübingen, Germany; Werner Reichert Center for Integrative Neuroscience, University of Tübingen, Tübingen, Germany.
| | - Nicolas D Lutz
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology, LMU Munich, Munich, Germany
| |
Collapse
|
8
|
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
|
9
|
Yassin LK, Nakhal MM, Alderei A, Almehairbi A, Mydeen AB, Akour A, Hamad MIK. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat 2025; 19:1504065. [PMID: 40012737 PMCID: PMC11860919 DOI: 10.3389/fnana.2025.1504065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a significant role in the maintenance of brain structure and function. The MGBA serves as a conduit between the CNS and the ENS, facilitating communication between the emotional and cognitive centers of the brain via diverse pathways. In the initial stages of this review, we will examine the way how MGBA affects neurogenesis, neuronal dendritic morphology, axonal myelination, microglia structure, brain blood barrier (BBB) structure and permeability, and synaptic structure. Furthermore, we will review the potential mechanistic pathways of neuroplasticity through MGBA influence. The short-chain fatty acids (SCFAs) play a pivotal role in the MGBA, where they can modify the BBB. We will therefore discuss how SCFAs can influence microglia, neuronal, and astrocyte function, as well as their role in brain disorders such as Alzheimer's disease (AD), and Parkinson's disease (PD). Subsequently, we will examine the technical strategies employed to study MGBA interactions, including using germ-free (GF) animals, probiotics, fecal microbiota transplantation (FMT), and antibiotics-induced dysbiosis. Finally, we will examine how particular bacterial strains can affect brain structure and function. By gaining a deeper understanding of the MGBA, it may be possible to facilitate research into microbial-based pharmacological interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
10
|
Ma R, Zhou Y, Huang W, Kong X. Icariin maintaining TMEM119-positive microglial population improves hippocampus-associated memory in senescent mice in relation to R-3-hydroxybutyric acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119287. [PMID: 39736348 DOI: 10.1016/j.jep.2024.119287] [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: 11/21/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium Tourn. ex L. is a traditional Chinese medicine used for thousands of years in China to treat forgetfulness. Icariin is a principal component of the genus Epimedium. AIM OF THE STUDY The metabolic mechanism of icariin treating forgetfulness is explored. MATERIALS AND METHODS A D-galactose-induced senescent mouse model was employed. The cognitive performance of mice was assessed in the fear conditioning test. Hippocampal pathology was assessed in the immunohistochemistry assay. Plasma metabolome was analyzed using GC-MS method, and the differential metabolites were further identified by UPLC-MS/MS or GC-MS method. The liver function, including ALT and AST, was assessed by enzyme reaction. Icariin was administered intraperitoneally at 50 and 100 mg/kg. Mice were administered five consecutive days per week for 8 weeks. RESULTS Icariin treatment improved hippocampus-related fear memory but not amygdala-related memory, whereas Pexidartinib (PLX3397), a microglial scavenger, did not. Icariin treatment maintained the TMEM119-positive microglial population and decreased the accumulation of the senescent biomarker p16 in the dorsal hippocampus in senescent mouse brains, whereas PLX3397 did not. Notably, p16 in the CA2 subregion significantly decreased in icariin-treated mice than the other hippocampal subregions. The senescent mice exhibited the circulating metabolic characteristics of mild ketoacidosis, active tricarboxylic acid (TCA) cycle, lactic acidosis, hyperglycemia, active detoxification, active cis-oleic acid metabolism, and inhibitory GABA shut. R-3-Hydroxybutyric acid primarily produced in the liver was selectively and robustly decreased by icariin treatment, which was not observed with PLX3397 treatment. The TCA cycle was rescued in senescent mice by icariin treatment. Icariin also protected liver function (plasma ALT) in D-gal-induced senescent mice. CONCLUSIONS Icariin may protect mouse hippocampal cognition from D-gal-induced senescence by protecting microglial homeostasis, and facilitating the utilization of R-3-hydroxybutyric acid is one of the underpins.
Collapse
Affiliation(s)
- Rong Ma
- Central Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yuge Zhou
- Central Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Weifan Huang
- Central Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiaoni Kong
- Central Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
11
|
Liu Y, Ding X, Jia S, Gu X. Current understanding and prospects for targeting neurogenesis in the treatment of cognitive impairment. Neural Regen Res 2025; 21:01300535-990000000-00655. [PMID: 39820472 PMCID: PMC12094536 DOI: 10.4103/nrr.nrr-d-24-00802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 07/24/2024] [Accepted: 10/31/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT Adult hippocampal neurogenesis is linked to memory formation In the adult brain, with new neurons in the hippocampus exhibiting greater plasticity during their immature stages compared to mature neurons. Abnormal adult hippocampal neurogenesis is closely associated with cognitive impairment in central nervous system diseases. Targeting and regulating adult hippocampal neurogenesis have been shown to improve cognitive deficits. This review aims to expand the current understanding and prospects of targeting neurogenesis in the treatment of cognitive impairment. Recent research indicates the presence of abnormalities in AHN in several diseases associated with cognitive impairment, including cerebrovascular diseases, Alzheimer's disease, aging-related conditions, and issues related to anesthesia and surgery. The role of these abnormalities in the cognitive deficits caused by these diseases has been widely recognized, and targeting AHN is considered a promising approach for treating cognitive impairment. However, the underlying mechanisms of this role are not yet fully understood, and the effectiveness of targeting abnormal adult hippocampal neurogenesis for treatment remains limited, with a need for further development of treatment methods and detection techniques.By reviewing recent studies, we classify the potential mechanisms of adult hippocampal neurogenesis abnormalities into four categories: immunity, energy metabolism, aging, and pathological states. In immunity-related mechanisms, abnormalities in meningeal, brain, and peripheral immunity can disrupt normal adult hippocampal neurogenesis.Lipid metabolism and mitochondrial function disorders are significant energy metabolism factors that lead to abnormal adult hippocampal neurogenesis. During aging, the inflammatory state of the neurogenic niche and the expression of aging-related microRNAs contribute to reduced adult hippocampal neurogenesis and cognitive impairment in older adult patients. Pathological states of the body and emotional disorders may also result in abnormal adult hippocampal neurogenesis. Among the current strategies used to enhance this form of neurogenesis, physical therapies such as exercise, transcutaneous electrical nerve stimulation, and enriched environments have proven effective. Dietary interventions, including energy intake restriction and nutrient optimization, have shown efficacy in both basic research and clinical trials. However, drug treatments, such as antidepressants and stem cell therapy, are primarily reported in basic research, with limited clinical application. The relationship between abnormal adult hippocampal neurogenesis and cognitive impairment has garnered widespread attention, and targeting the former may be an important strategy for treating the latter. However, the mechanisms underlying abnormal adult hippocampal neurogenesis remain unclear, and treatments are lacking. This highlights the need for greater focus on translating research findings into clinical practice.
Collapse
Affiliation(s)
- Ye Liu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xibing Ding
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shushan Jia
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| | - Xiyao Gu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| |
Collapse
|
12
|
O’Leary J, Georgeaux-Healy C, Serpell L. The impact of continuous calorie restriction and fasting on cognition in adults without eating disorders. Nutr Rev 2025; 83:146-159. [PMID: 38263325 PMCID: PMC11632361 DOI: 10.1093/nutrit/nuad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024] Open
Abstract
Research into the effects of calorie restriction continues to intrigue those interested in whether it may allow humans to live longer and healthier lives. Animal studies of continuous calorie restriction (CCR) and fasting have demonstrated substantial advantages to health and longevity. However, concerns remain about the impact of restricting calorie intake on human health and cognition. Given the emerging evidence of cognitive impairments in eating disorders, studies investigating restricted calorie intake in healthy humans (in an ethical way) may also have implications for understanding restrictive eating disorders. In this review, the published literature on the impact of CCR and fasting on cognitive function in healthy human participants is synthesized. Of the 33 studies of CCR and fasting in humans identified, 23 demonstrated significant changes in cognition. Despite variation across the cognitive domains, results suggest CCR benefits inhibition, processing speed, and working memory, but may lead to impairments in cognitive flexibility. The results of fasting studies suggest fasting is associated with impairments in cognitive flexibility and psychomotor abilities. Overall, the results of these studies suggest the degree (ie, the severity) of calorie restriction is what most likely predicts cognitive improvements as opposed to impairments. For individuals engaging in sustained restriction, this may have serious, irreversible consequences. However, there are mixed findings regarding the impact of CCR and fasting on this aspect of human functioning, suggesting further research is required to understand the costs and benefits of different types of calorie restriction.
Collapse
Affiliation(s)
- John O’Leary
- North East London NHS Foundation Trust, CEME Centre, West Wing Marsh Way, Rainham, Essex RM13 8GQ, UK
| | - Chloé Georgeaux-Healy
- Research Department of Clinical, Educational and Health Psychology, University College London, London WC1E 6BT, UK
| | - Lucy Serpell
- North East London NHS Foundation Trust, CEME Centre, West Wing Marsh Way, Rainham, Essex RM13 8GQ, UK
- Research Department of Clinical, Educational and Health Psychology, University College London, London WC1E 6BT, UK
| |
Collapse
|
13
|
Ngah WZW, Ahmad HF, Ankasha SJ, Makpol S, Tooyama I. Dietary Strategies to Mitigate Alzheimer's Disease: Insights into Antioxidant Vitamin Intake and Supplementation with Microbiota-Gut-Brain Axis Cross-Talk. Antioxidants (Basel) 2024; 13:1504. [PMID: 39765832 PMCID: PMC11673287 DOI: 10.3390/antiox13121504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease (AD), which is characterized by deterioration in cognitive function and neuronal death, is the most prevalent age-related progressive neurodegenerative disease. Clinical and experimental research has revealed that gut microbiota dysbiosis may be present in AD patients. The changed gut microbiota affects brain function and behavior through several mechanisms, including tau phosphorylation and increased amyloid deposits, neuroinflammation, metabolic abnormalities, and persistent oxidative stress. The lack of effective treatments to halt or reverse the progression of this disease has prompted a search for non-pharmaceutical tools. Modulation of the gut microbiota may be a promising strategy in this regard. This review aims to determine whether specific dietary interventions, particularly antioxidant vitamins, either obtained from the diet or as supplements, may support the formation of beneficial microbiota in order to prevent AD development by contributing to the systemic reduction of chronic inflammation or by acting locally in the gut. Understanding their roles would be beneficial as it may have the potential to be used as a future therapy option for AD patients.
Collapse
Affiliation(s)
- Wan Zurinah Wan Ngah
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| | - Hajar Fauzan Ahmad
- Department of Industrial Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Gambang 26300, Pahang, Malaysia;
| | - Sheril June Ankasha
- Unisza Science and Medicine Foundation Centre, Universiti Sultan Zainal Abidin, Gong Badak Campus, Kuala Nerus 21300, Terengganu, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Ikuo Tooyama
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| |
Collapse
|
14
|
Mishra A, Sobha D, Patel D, Suresh PS. Intermittent fasting in health and disease. Arch Physiol Biochem 2024; 130:755-767. [PMID: 37828854 DOI: 10.1080/13813455.2023.2268301] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
CONTEXT Intermittent fasting, a new-age dietary concept derived from an age-old tradition, involves repetitive cycles of fasting/calorie restriction and eating. OBJECTIVE We aim to take a deep dive into the biological responses to intermittent fasting, delineate the disease-modifying and cognitive effects of intermittent fasting, and also shed light on the possible side effects. METHODS Numerous in vitro and in vivo studies were reviewed, followed by an in-depth analysis, and compilation of their implications in health and disease. RESULTS Intermittent fasting improves the body's stress tolerance, which is further amplified with exercise. It impacts various pathological conditions like cancer, obesity, diabetes, cardiovascular disease, and neurodegenerative diseases. CONCLUSION During dietary restriction, the human body experiences a metabolic switch due to the depletion of liver glycogen, which promotes a shift towards utilising fatty acids and ketones in the system, thereby significantly impacting adiposity, ageing and the immune response to various diseases.
Collapse
Affiliation(s)
- Anubhav Mishra
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Devika Sobha
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Dimple Patel
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| |
Collapse
|
15
|
Wang J, Rang Y, Liu C. Effects of Caloric Restriction and Intermittent Fasting and Their Combined Exercise on Cognitive Functioning: A Review. Curr Nutr Rep 2024; 13:691-700. [PMID: 39240488 DOI: 10.1007/s13668-024-00570-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE OF REVIEW The impact of dietary habits on cognitive function is increasingly gaining attention. The review is to discuss how caloric restriction (CR) and intermittent fasting (IF) can enhance cognitive function in healthy states through multiple pathways that interact with one another. Secondly, to explore the effects of CR and IF on cognitive function in conditions of neurodegenerative diseases, obesity diabetes and aging, as well as potential synergistic effects in combination with exercise to prevent cognitively related neurodegenerative diseases. RECENT FINDINGS With age, the human brain ages and develops corresponding neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and epilepsy, which in turn trigger cognitive impairment. Recent research indicates that the impact of diet and exercise on cognitive function is increasingly gaining attention. The benefits of exercise for cognitive function and brain plasticity are numerous, and future research can examine the efficacy of particular dietary regimens during physical activity when combined with diet which can prevent cognitive decline.
Collapse
Affiliation(s)
- Junming Wang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
- The Key Laboratory of Food Quality and Safety of Guangdong Province, Guangzhou, 510642, China
| | - Yifeng Rang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
- The Key Laboratory of Food Quality and Safety of Guangdong Province, Guangzhou, 510642, China
| | - Chunhong Liu
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
- The Key Laboratory of Food Quality and Safety of Guangdong Province, Guangzhou, 510642, China.
| |
Collapse
|
16
|
Waheed A, Ghaffar M, Mustafa S, Abbas A, Khan S, Waheed A, Naz H. Nutrigenomics and neurological disorders: exploring diet-brain interactions for cognitive health. Neurogenetics 2024; 26:10. [PMID: 39589612 DOI: 10.1007/s10048-024-00791-7] [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: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 11/27/2024]
Abstract
This review article investigates the intricate relationship between nutrigenomics and neurological disorders, highlighting how genetic variations affect an individual's response to nutrients. The study delves into the role of diet-related oxidative stress and the gut-brain axis in the progression and management of brain disorders such as Parkinson's disease, Alzheimer's disease, epilepsy, stroke, migraines, and depression. The review encompasses various clinical trials and introduces new trends and techniques, including omics and artificial intelligence, in identifying and managing neurological disorders. The main findings emphasize that personalized diet recommendations, tailored to an individual's genetic makeup, can significantly improve cognitive health and manage neurological conditions. The study concludes that further research in the field of nutrigenomics is essential to advancing personalized nutrition strategies for better neurological functioning, ultimately linking diet, genes, and brain health.
Collapse
Affiliation(s)
- Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Maliha Ghaffar
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan.
| | - Samavia Mustafa
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Anam Abbas
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Sana Khan
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| |
Collapse
|
17
|
Ghosh P, Fontanella RA, Scisciola L, Taktaz F, Pesapane A, Basilicata MG, Tortorella G, Matacchione G, Capuano A, Vietri MT, Selvaggi F, Paolisso G, Barbieri M. Obesity-induced neuronal senescence: Unraveling the pathophysiological links. Ageing Res Rev 2024; 101:102533. [PMID: 39368666 DOI: 10.1016/j.arr.2024.102533] [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: 06/21/2024] [Revised: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Obesity is one of the most prevalent and increasing metabolic disorders and is considered one of the twelve risk factors for dementia. Numerous studies have demonstrated that obesity induces pathophysiological changes leading to cognitive decline; however, the underlying molecular mechanisms are yet to be fully elucidated. Various biochemical processes, including chronic inflammation, oxidative stress, insulin resistance, dysregulation of lipid metabolism, disruption of the blood-brain barrier, and the release of adipokines have been reported to contribute to the accumulation of senescent neurons during obesity. These senescent cells dysregulate neuronal health and function by exhibiting a senescence-associated secretory phenotype, inducing neuronal inflammation, deregulating cellular homeostasis, causing mitochondrial dysfunction, and promoting microglial infiltration. These factors act as major risks for the occurrence of neurodegenerative diseases and cognitive decline. This review aims to focus on how obesity upregulates neuronal senescence and explores both pharmacological and non-pharmacological interventions for preventing cognitive impairments, thus offering new insights into potential therapeutic strategies.
Collapse
Affiliation(s)
- Puja Ghosh
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fatemeh Taktaz
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Ada Pesapane
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Tortorella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Annalisa Capuano
- Department of Experimental Medicine - Section of Pharmacology "L. Donatelli", University of Campania "Luigi Vanvitelli", Naples 80138, Italy
| | - Maria Teresa Vietri
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio, Naples 80138, Italy; UOC Clinical and Molecular Pathology, AOU University of Campania "Luigi Vanvitelli", Naple 80138, Italy
| | - Francesco Selvaggi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; UniCamillus, International Medical University, Rome, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
18
|
Babygirija R, Han JH, Sonsalla MM, Matoska R, Calubag MF, Green CL, Tobon A, Yeh CY, Vertein D, Schlorf S, Illiano J, Liu Y, Grunow I, Rigby MJ, Puglielli L, Harris DA, Denu JM, Lamming DW. Fasting is required for many of the benefits of calorie restriction in the 3xTg mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613904. [PMID: 39386545 PMCID: PMC11463641 DOI: 10.1101/2024.09.19.613904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Caloric restriction (CR) is a widely recognized geroprotective intervention that slows or prevents Alzheimer's disease (AD) in animal models. CR is typically implemented via feeding mice a single meal per day; as CR mice rapidly consume their food, they are subject to a prolonged fast between meals. While CR has been shown to improve metabolic and cognitive functions and suppress pathological markers in AD mouse models, the specific contributions of fasting versus calorie reduction remains unclear. Here, we investigated the contribution of fasting and energy restriction to the beneficial effects of CR on AD progression. To test this, we placed 6-month-old 3xTg mice on one of several diet regimens, allowing us to dissect the effects of calories and fasting on metabolism, AD pathology, and cognition. We find that energy restriction alone, without fasting, was sufficient to improve glucose tolerance and reduce adiposity in both sexes, and to reduce Aβ plaques and improve aspects of cognitive performance in females. However, we find that a prolonged fast between meals is necessary for many of the benefits of CR, including improved insulin sensitivity, reduced phosphorylation of tau, decreased neuroinflammation, inhibition of mTORC1 signaling, and activation of autophagy, as well as for the full cognitive benefits of CR. Finally, we find that fasting is essential for the benefits of CR on survival in male 3xTg mice. Overall, our results demonstrate that fasting is required for the full benefits of a CR diet on the development and progression of AD in 3xTg mice, and suggest that both when and how much we eat influences the development and progress of AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Diana Vertein
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Sophia Schlorf
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Julia Illiano
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yang Liu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Endocrinology and Reproductive Physiology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael J. Rigby
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - David A. Harris
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Laboratory for Surgical Metabolism, Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, Madison, WI 53705, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Comprehensive Diabetes Center, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI 53705, USA
| |
Collapse
|
19
|
Huston CA, Milan M, Vance ML, Bickel MA, Miller LR, Negri S, Hibbs C, Vaden H, Hayes L, Csiszar A, Ungvari Z, Yabluchanskiy A, Tarantini S, Conley SM. The effects of time restricted feeding on age-related changes in the mouse retina. Exp Gerontol 2024; 194:112510. [PMID: 38964431 PMCID: PMC11425985 DOI: 10.1016/j.exger.2024.112510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Dietary modifications such as caloric restriction (CR) and intermittent fasting (IF) have gained popularity due to their proven health benefits in aged populations. In time restricted feeding (TRF), a form of intermittent fasting, the amount of time for food intake is regulated without restricting the caloric intake. TRF is beneficial for the central nervous system to support brain health in the context of aging. Therefore, we here ask whether TRF also exerts beneficial effects in the aged retina. We compared aged mice (24 months) on a TRF paradigm (access to food for six hours per day) for either 6 or 12 months against young control mice (8 months) and aged control mice on an ad libitum diet. We examined changes in the retina at the functional (electroretinography), structural (histology and fluorescein angiograms) and molecular (gene expression) level. TRF treatment showed amelioration of age-related reductions in both scotopic and photopic b-wave amplitudes suggesting benefits for retinal interneuron signaling. TRF did not affect age-related signs of retinal inflammation or microglial activation at either the molecular or histological level. Our data indicate that TRF helps preserve some aspects of retinal function that are decreased with aging, adding to our understanding of the health benefits that altered feeding patterns may confer.
Collapse
Affiliation(s)
- Cade A Huston
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madison Milan
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neuroscience and Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lauren R Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sharon Negri
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Clara Hibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lindsay Hayes
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neuroscience and Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neuroscience and Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neuroscience and Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neuroscience and Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
20
|
Sproten R, Nohr D, Guseva D. Nutritional strategies modulating the gut microbiome as a preventative and therapeutic approach in normal and pathological age-related cognitive decline: a systematic review of preclinical and clinical findings. Nutr Neurosci 2024; 27:1042-1057. [PMID: 38165747 DOI: 10.1080/1028415x.2023.2296727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
CONTEXT The proportion of the elderly population is on the rise across the globe, and with it the prevalence of age-related neurodegenerative diseases. The gut microbiota, whose composition is highly regulated by dietary intake, has emerged as an exciting research field in neurology due to its pivotal role in modulating brain functions via the gut-brain axis. OBJECTIVES We aimed at conducting a systematic review of preclinical and clinical studies investigating the effects of dietary interventions on cognitive ageing in conjunction with changes in gut microbiota composition and functionality. METHODS PubMed and Scopus were searched using terms related to ageing, cognition, gut microbiota and dietary interventions. Studies were screened, selected based on previously determined inclusion and exclusion criteria, and evaluated for methodological quality using recommended risk of bias assessment tools. RESULTS A total of 32 studies (18 preclinical and 14 clinical) were selected for inclusion. We found that most of the animal studies showed significant positive intervention effects on cognitive behavior, while outcomes on cognition, microbiome features, and health parameters in humans were less pronounced. The effectiveness of dietary interventions depended markedly on the age, gender, degree of cognitive decline and baseline microbiome composition of participants. CONCLUSION To harness the full potential of microbiome-inspired nutrition for cognitive health, one of the main challenges remains to better understand the interplay between host, his microbiome, dietary exposures, whilst also taking into account environmental influences. Future research should aim toward making use of host-specific microbiome data to guide the development of personalized therapies.
Collapse
Affiliation(s)
- Rieke Sproten
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Donatus Nohr
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Daria Guseva
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| |
Collapse
|
21
|
Borrego-Ruiz A, Borrego JJ. Influence of human gut microbiome on the healthy and the neurodegenerative aging. Exp Gerontol 2024; 194:112497. [PMID: 38909763 DOI: 10.1016/j.exger.2024.112497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The gut microbiome plays a crucial role in host health throughout the lifespan by influencing brain function during aging. The microbial diversity of the human gut microbiome decreases during the aging process and, as a consequence, several mechanisms increase, such as oxidative stress, mitochondrial dysfunction, inflammatory response, and microbial gut dysbiosis. Moreover, evidence indicates that aging and neurodegeneration are closely related; consequently, the gut microbiome may serve as a novel marker of lifespan in the elderly. In this narrative study, we investigated how the changes in the composition of the gut microbiome that occur in aging influence to various neuropathological disorders, such as mild cognitive impairment (MCI), dementia, Alzheimer's disease (AD), and Parkinson's disease (PD); and which are the possible mechanisms that govern the relationship between the gut microbiome and cognitive impairment. In addition, several studies suggest that the gut microbiome may be a potential novel target to improve hallmarks of brain aging and to promote healthy cognition; therefore, current and future therapeutic interventions have been also reviewed.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain.
| |
Collapse
|
22
|
Brogi S, Tabanelli R, Puca S, Calderone V. Intermittent Fasting: Myths, Fakes and Truth on This Dietary Regimen Approach. Foods 2024; 13:1960. [PMID: 38998465 PMCID: PMC11241639 DOI: 10.3390/foods13131960] [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: 05/25/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Intermittent fasting (IF) has been indicated as a valuable alternative to the classical caloric restriction dietary regimen for lowering body weight and preventing obesity-related complications, such as metabolic syndrome and type II diabetes. However, is it effective? In this review article, we analyzed over 50 clinical studies in which IF, conducted by alternate day fasting (ADF) or time-restricted feeding (TRF), was compared with the caloric restriction approach. We evaluated the different roles of IF in treating and preventing human disorders such as metabolic syndrome, type II diabetes, and some types of cancer, as well as the usefulness of IF in reducing body weight and cardiovascular risk factors such as hypertension. Furthermore, we explored the cellular pathways targeted by IF to exert their beneficial effects by activating effector proteins that modulate cell functions and resistance to oxidative stress. In contrast, we investigated concerns regarding human health related to the adoption of IF dietary regimens, highlighting the profound debate surrounding weight loss regimens. We examined and compared several clinical trials to formulate an updated concept regarding IF and its therapeutic potential.
Collapse
Affiliation(s)
- Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (S.P.); (V.C.)
- Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Rita Tabanelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (S.P.); (V.C.)
| | - Sara Puca
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (S.P.); (V.C.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (S.P.); (V.C.)
| |
Collapse
|
23
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of pathology in a mouse model of Alzheimer's disease. Nat Commun 2024; 15:5217. [PMID: 38890307 PMCID: PMC11189507 DOI: 10.1038/s41467-024-49589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and dietary protein restriction extends the lifespan and healthspan of mice. In this study, we examined the effect of protein restriction (PR) on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. Here, we show that PR promotes leanness and glycemic control in 3xTg mice, specifically rescuing the glucose intolerance of 3xTg females. PR induces sex-specific alterations in circulating and brain metabolites, downregulating sphingolipid subclasses in 3xTg females. PR also reduces AD pathology and mTORC1 activity, increases autophagy, and improves the cognition of 3xTg mice. Finally, PR improves the survival of 3xTg mice. Our results suggest that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jessica H Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Dominique A Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Natalie M Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
24
|
Islam MA, Sehar U, Sultana OF, Mukherjee U, Brownell M, Kshirsagar S, Reddy PH. SuperAgers and centenarians, dynamics of healthy ageing with cognitive resilience. Mech Ageing Dev 2024; 219:111936. [PMID: 38657874 DOI: 10.1016/j.mad.2024.111936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Graceful healthy ageing and extended longevity is the most desired goal for human race. The process of ageing is inevitable and has a profound impact on the gradual deterioration of our physiology and health since it triggers the onset of many chronic conditions like dementia, osteoporosis, diabetes, arthritis, cancer, and cardiovascular disease. However, some people who lived/live more than 100 years called 'Centenarians" and how do they achieve their extended lifespans are not completely understood. Studying these unknown factors of longevity is important not only to establish a longer human lifespan but also to manage and treat people with shortened lifespans suffering from age-related morbidities. Furthermore, older adults who maintain strong cognitive function are referred to as "SuperAgers" and may be resistant to risk factors linked to cognitive decline. Investigating the mechanisms underlying their cognitive resilience may contribute to the development of therapeutic strategies that support the preservation of cognitive function as people age. The key to a long, physically, and cognitively healthy life has been a mystery to scientists for ages. Developments in the medical sciences helps us to a better understanding of human physiological function and greater access to medical care has led us to an increase in life expectancy. Moreover, inheriting favorable genetic traits and adopting a healthy lifestyle play pivotal roles in promoting longer and healthier lives. Engaging in regular physical activity, maintaining a balanced diet, and avoiding harmful habits such as smoking contribute to overall well-being. The synergy between positive lifestyle choices, access to education, socio-economic factors, environmental determinants and genetic supremacy enhances the potential for a longer and healthier life. Our article aims to examine the factors associated with healthy ageing, particularly focusing on cognitive health in centenarians. We will also be discussing different aspects of ageing including genomic instability, metabolic burden, oxidative stress and inflammation, mitochondrial dysfunction, cellular senescence, immunosenescence, and sarcopenia.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
25
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
26
|
Babygirija R, Sonsalla MM, Mill J, James I, Han JH, Green CL, Calubag MF, Wade G, Tobon A, Michael J, Trautman MM, Matoska R, Yeh CY, Grunow I, Pak HH, Rigby MJ, Baldwin DA, Niemi NM, Denu JM, Puglielli L, Simcox J, Lamming DW. Protein restriction slows the development and progression of Alzheimer's disease in mice. RESEARCH SQUARE 2024:rs.3.rs-3342413. [PMID: 37790423 PMCID: PMC10543316 DOI: 10.21203/rs.3.rs-3342413/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Dietary protein is a critical regulator of metabolic health and aging. Low protein diets are associated with healthy aging in humans, and many independent groups of researchers have shown that dietary protein restriction (PR) extends the lifespan and healthspan of mice. Here, we examined the effect of PR on metabolic health and the development and progression of Alzheimer's disease (AD) in the 3xTg mouse model of AD. We found that PR has metabolic benefits for 3xTg mice and non-transgenic controls of both sexes, promoting leanness and glycemic control in 3xTg mice and rescuing the glucose intolerance of 3xTg females. We found that PR induces sex-specific alterations in circulating metabolites and in the brain metabolome and lipidome, downregulating sphingolipid subclasses including ceramides, glucosylceramides, and sphingomyelins in 3xTg females. Consumption of a PR diet starting at 6 months of age reduced AD pathology in conjunction with reduced mTORC1 activity, increased autophagy, and had cognitive benefits for 3xTg mice. Finally, PR improved the survival of 3xTg mice. Our results demonstrate that PR slows the progression of AD at molecular and pathological levels, preserves cognition in this mouse model of AD, and suggests that PR or pharmaceutical interventions that mimic the effects of this diet may hold promise as a treatment for AD.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Michelle M. Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Isabella James
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jessica H. Han
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara L. Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F. Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Anna Tobon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Michael
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michaela M. Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ryan Matoska
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac Grunow
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heidi H. Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael J. Rigby
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dominique A. Baldwin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Natalie M. Niemi
- Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John M. Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Judith Simcox
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
- Comparative Biomedical Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Hansen B, Roomp K, Ebid H, Schneider JG. Perspective: The Impact of Fasting and Caloric Restriction on Neurodegenerative Diseases in Humans. Adv Nutr 2024; 15:100197. [PMID: 38432589 PMCID: PMC10997874 DOI: 10.1016/j.advnut.2024.100197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/29/2023] [Accepted: 02/23/2024] [Indexed: 03/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by the progressive functional and structural denaturation of neurons in the central and peripheral nervous systems. Despite the wide range of genetic predispositions, the increased emergence of these disorders has been associated with a variety of modifiable risk factors, including lifestyle factors. Diet has been shown to influence cognitive alterations in the elderly population with age-related brain pathologies, and specific dietary interventions might, therefore, confer preservatory protection to neural structures. Although Mediterranean and ketogenic diets have been studied, no clear guidelines have been implemented for the prevention or treatment of ND in clinical practice. Murine models have shown that intermittent fasting and caloric restriction (CR) can counteract disease processes in various age-related disorders, including NDs. The objective of this perspective is to provide a comprehensive, comparative overview of the available primary intervention studies on fasting and CR in humans with ND and to elucidate possible links between the mechanisms underlying the effects of fasting, CR, and the neuropathology of ND. We also included all currently available studies in older adults (with and without mild cognitive impairment) in which the primary endpoint was cognitive function to provide further insights into the feasibility and outcomes of such interventions. Overall, we conclude that nutritional intervention trials focusing on fasting and CR in humans with ND have been neglected, and more high-quality studies, including longitudinal clinical intervention trials, are urgently needed to elucidate the underlying immune-metabolic mechanisms in diet and ND.
Collapse
Affiliation(s)
- Bérénice Hansen
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kirsten Roomp
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Hebah Ebid
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jochen G Schneider
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg; Departments of Internal Medicine II and Psychiatry, Saarland University Medical Center, Homburg, Germany.
| |
Collapse
|
28
|
Iyer SH, Yeh MY, Netzel L, Lindsey MG, Wallace M, Simeone KA, Simeone TA. Dietary and Metabolic Approaches for Treating Autism Spectrum Disorders, Affective Disorders and Cognitive Impairment Comorbid with Epilepsy: A Review of Clinical and Preclinical Evidence. Nutrients 2024; 16:553. [PMID: 38398876 PMCID: PMC10893388 DOI: 10.3390/nu16040553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Epilepsy often occurs with other neurological disorders, such as autism, affective disorders, and cognitive impairment. Research indicates that many neurological disorders share a common pathophysiology of dysfunctional energy metabolism, neuroinflammation, oxidative stress, and gut dysbiosis. The past decade has witnessed a growing interest in the use of metabolic therapies for these disorders with or without the context of epilepsy. Over one hundred years ago, the high-fat, low-carbohydrate ketogenic diet (KD) was formulated as a treatment for epilepsy. For those who cannot tolerate the KD, other diets have been developed to provide similar seizure control, presumably through similar mechanisms. These include, but are not limited to, the medium-chain triglyceride diet, low glycemic index diet, and calorie restriction. In addition, dietary supplementation with ketone bodies, polyunsaturated fatty acids, or triheptanoin may also be beneficial. The proposed mechanisms through which these diets and supplements work to reduce neuronal hyperexcitability involve normalization of aberrant energy metabolism, dampening of inflammation, promotion of endogenous antioxidants, and reduction of gut dysbiosis. This raises the possibility that these dietary and metabolic therapies may not only exert anti-seizure effects, but also reduce comorbid disorders in people with epilepsy. Here, we explore this possibility and review the clinical and preclinical evidence where available.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Timothy A. Simeone
- Department of Pharmacology & Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA; (S.H.I.); (K.A.S.)
| |
Collapse
|
29
|
Neudorf H, Little JP. Impact of fasting & ketogenic interventions on the NLRP3 inflammasome: A narrative review. Biomed J 2024; 47:100677. [PMID: 37940045 PMCID: PMC10821592 DOI: 10.1016/j.bj.2023.100677] [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: 06/29/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
Overactivation of the NLRP3 inflammasome is implicated in chronic low-grade inflammation associated with various disease states, including obesity, type 2 diabetes, atherosclerosis, Alzheimer's disease, and Parkinson's disease. Emerging evidence, mostly from cell and animal models of disease, supports a role for ketosis in general, and the main circulating ketone body beta-hydroxybutyrate (BHB) in particular, in reducing NLRP3 inflammasome activation to improve chronic inflammation. As a result, interventions that can induce ketosis (e.g., fasting, intermittent fasting, time-restricted feeding/eating, very low-carbohydrate high-fat ketogenic diets) and/or increase circulating BHB (e.g., exogenous ketone supplementation) have garnered increasing interest for their therapeutic potential. The purpose of the present review is to summarize our current understanding of the literature on how ketogenic interventions impact the NLRP3 inflammasome across human, rodent and cell models. Overall, there is convincing evidence that ketogenic interventions, likely acting through multiple interacting mechanisms in a cell-, disease- and context-specific manner, can reduce NLRP3 inflammasome activation. The evidence supports a direct effect of BHB, although it is important to consider the myriad of other metabolic responses to fasting or ketogenic diet interventions (e.g., elevated lipolysis, low insulin, stable glucose, negative energy balance) that may also impact innate immune responses. Future research is needed to translate promising findings from discovery science to clinical application.
Collapse
Affiliation(s)
- Helena Neudorf
- University of British Columbia, Okanagan Campus, Kelowna, BC, Canada
| | - Jonathan P Little
- University of British Columbia, Okanagan Campus, Kelowna, BC, Canada.
| |
Collapse
|
30
|
Hashimoto T, Hotta R, Kawashima R. Enhanced memory and hippocampal connectivity in humans 2 days after brief resistance exercise. Brain Behav 2024; 14:e3436. [PMID: 38383042 PMCID: PMC10881282 DOI: 10.1002/brb3.3436] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Exercise has significant health benefits and can enhance learning. A single bout of high-intensity resistance training may be sufficient to improve memory. This study aimed to assess memory enhancement by a single bout of high-intensity resistance training and to examine the neural underpinnings using resting-state functional magnetic resonance imaging (MRI). METHODS Sixty young adults (34 men and 26 women), divided into the training and control groups, participated. The first session included verbal memory recall tests (cued- and free-recall), resting-state functional MRI (rs-fMRI), and a single-bout high-intensity resistance training for the training group. Two days later, they underwent post-intervention memory tests and rs-fMRI. The study design was 2 groups × 2 sessions for memory tests, and within training group comparisons for rs-fMRI. RESULTS Compared to the control group without resistance training, the training group showed higher cued-recall performance 2 days after the brief resistance training (training: +0.27, control: -0.13, interaction: p = .01), and their free-recall scores were associated with enhanced left posterior hippocampal connectivity (r = .64, p < .001). CONCLUSIONS These results suggest that brief high-intensity resistance exercise/strength training could enhance memory without repeated exercising. The quick effect of resistance training on memory and hippocampal connectivity could be revealed. A focused and one-shot exercise may be sufficient to enhance memory performance and neural plasticity in a few days.
Collapse
Affiliation(s)
- Teruo Hashimoto
- Department of Functional Brain Imaging, Institute Development, Aging and CancerTohoku UniversitySendaiJapan
| | | | - Ryuta Kawashima
- Department of Functional Brain Imaging, Institute Development, Aging and CancerTohoku UniversitySendaiJapan
| |
Collapse
|
31
|
Cogut V, McNeely TL, Bussian TJ, Graves SI, Baker DJ. Caloric Restriction Improves Spatial Learning Deficits in Tau Mice. J Alzheimers Dis 2024; 98:925-940. [PMID: 38517786 PMCID: PMC11068089 DOI: 10.3233/jad-231117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Background Caloric restriction (CR) has been recognized for its benefits in delaying age-related diseases and extending lifespan. While its effects on amyloid pathology in Alzheimer's disease (AD) mouse models are well-documented, its effects on tauopathy, another hallmark of AD, are less explored. Objective To assess the impact of a short-term 30% CR regimen on age-dependent spatial learning deficits and pathological features in a tauopathy mouse model. Methods We subjected male PS19 tau P301S (hereafter PS19) and age-matched wildtype mice from two age cohorts (4.5 and 7.5 months old) to a 6-week 30% CR regimen. Spatial learning performance was assessed using the Barnes Maze test. Tau pathology, neuroinflammation, hippocampal cell proliferation, and neurogenesis were evaluated in the older cohort by immunohistochemical staining and RT-qPCR. Results CR mitigated age-dependent spatial learning deficits in PS19 mice but exhibited limited effects on tau pathology and the associated neuroinflammation. Additionally, we found a decrease in hippocampal cell proliferation, predominantly of Iba1+ cells. Conclusions Our findings reinforce the cognitive benefits conferred by CR despite its limited modulation of disease pathology. Given the pivotal role of microglia in tau-driven pathology, the observed reduction in Iba1+ cells under CR suggests potential therapeutic implications, particularly if CR would be introduced early in disease progression.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Taylor L. McNeely
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Tyler J. Bussian
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Sara I. Graves
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Darren J. Baker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Paul F. Glenn Center for Biology of Aging Research at Mayo Clinic, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| |
Collapse
|
32
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 67] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
33
|
Kueck PJ, Morris JK, Stanford JA. Current Perspectives: Obesity and Neurodegeneration - Links and Risks. Degener Neurol Neuromuscul Dis 2023; 13:111-129. [PMID: 38196559 PMCID: PMC10774290 DOI: 10.2147/dnnd.s388579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/21/2023] [Indexed: 01/11/2024] Open
Abstract
Obesity is increasing in prevalence across all age groups. Long-term obesity can lead to the development of metabolic and cardiovascular diseases through its effects on adipose, skeletal muscle, and liver tissue. Pathological mechanisms associated with obesity include immune response and inflammation as well as oxidative stress and consequent endothelial and mitochondrial dysfunction. Recent evidence links obesity to diminished brain health and neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Both AD and PD are associated with insulin resistance, an underlying syndrome of obesity. Despite these links, causative mechanism(s) resulting in neurodegenerative disease remain unclear. This review discusses relationships between obesity, AD, and PD, including clinical and preclinical findings. The review then briefly explores nonpharmacological directions for intervention.
Collapse
Affiliation(s)
- Paul J Kueck
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jill K Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - John A Stanford
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Landon Center on Aging, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| |
Collapse
|
34
|
Marzola P, Melzer T, Pavesi E, Gil-Mohapel J, Brocardo PS. Exploring the Role of Neuroplasticity in Development, Aging, and Neurodegeneration. Brain Sci 2023; 13:1610. [PMID: 38137058 PMCID: PMC10741468 DOI: 10.3390/brainsci13121610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 12/24/2023] Open
Abstract
Neuroplasticity refers to the ability of the brain to reorganize and modify its neural connections in response to environmental stimuli, experience, learning, injury, and disease processes. It encompasses a range of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in the structure and function of neurons, and the generation of new neurons. Neuroplasticity plays a crucial role in developing and maintaining brain function, including learning and memory, as well as in recovery from brain injury and adaptation to environmental changes. In this review, we explore the vast potential of neuroplasticity in various aspects of brain function across the lifespan and in the context of disease. Changes in the aging brain and the significance of neuroplasticity in maintaining cognitive function later in life will also be reviewed. Finally, we will discuss common mechanisms associated with age-related neurodegenerative processes (including protein aggregation and accumulation, mitochondrial dysfunction, oxidative stress, and neuroinflammation) and how these processes can be mitigated, at least partially, by non-invasive and non-pharmacologic lifestyle interventions aimed at promoting and harnessing neuroplasticity.
Collapse
Affiliation(s)
- Patrícia Marzola
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Thayza Melzer
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Eloisa Pavesi
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
| | - Patricia S. Brocardo
- Department of Morphological Sciences and Graduate Neuroscience Program, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis 88040-900, SC, Brazil; (P.M.); (T.M.); (E.P.)
| |
Collapse
|
35
|
Hu X, Peng J, Tang W, Xia Y, Song P. A circadian rhythm-restricted diet regulates autophagy to improve cognitive function and prolong lifespan. Biosci Trends 2023; 17:356-368. [PMID: 37722875 DOI: 10.5582/bst.2023.01221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Diet and circadian rhythms have been found to have a profound impact on health, disease, and aging. Skipping breakfast, eating late, and overeating have adverse effects on the body's metabolism and increase the risk of cardiovascular and metabolic diseases. Disturbance of circadian rhythms has been associated with increased risk of atherosclerosis, Alzheimer's disease, Parkinson's disease, and other diseases. Abnormal deposition of amyloid β (Aβ) and tau proteins in the brain and impaired synaptic function are linked to cognitive dysfunction. A restrictive diet following the circadian rhythm can affect the metabolism of lipids, glucose, and amino acids such as branched chain amino acids and cysteine. These metabolic changes contribute to autophagy through molecular mechanisms such as adenosine monophosphate-activated protein kinase (AMPK), rapamycin (mTOR), D-β-hydroxybutyrate (D-BHB), and neuropeptide Y (NPY). Autophagy, in turn, promotes the removal of abnormally deposited proteins and damaged organelles and improves cognitive function, ultimately prolonging lifespan. In addition, a diet restricted to the circadian rhythm induces increased expression of brain-derived neurotrophic factor (BDNF) in the forebrain region, regulating autophagy and increasing synaptic plasticity, thus enhancing cognitive function. Consequently, circadian rhythm-restricted diets could serve as a promising non-pharmacological treatment for preventing and improving cognitive dysfunction and prolonging lifespan.
Collapse
Affiliation(s)
- Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Wei Tang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
- International Health Care Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Peipei Song
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Alharbi M, Stephan BC, Shannon OM, Siervo M. Does dietary nitrate boost the effects of caloric restriction on brain health? Potential physiological mechanisms and implications for future research. Nutr Metab (Lond) 2023; 20:45. [PMID: 37880786 PMCID: PMC10599060 DOI: 10.1186/s12986-023-00766-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/07/2023] [Indexed: 10/27/2023] Open
Abstract
Dementia is a highly prevalent and costly disease characterised by deterioration of cognitive and physical capacity due to changes in brain function and structure. Given the absence of effective treatment options for dementia, dietary and other lifestyle approaches have been advocated as potential strategies to reduce the burden of this condition. Maintaining an optimal nutritional status is vital for the preservation of brain function and structure. Several studies have recognised the significant role of nutritional factors to protect and enhance metabolic, cerebrovascular, and neurocognitive functions. Caloric restriction (CR) positively impacts on brain function via a modulation of mitochondrial efficiency, endothelial function, neuro-inflammatory, antioxidant and autophagy responses. Dietary nitrate, which serves as a substrate for the ubiquitous gasotransmitter nitric oxide (NO), has been identified as a promising nutritional intervention that could have an important role in improving vascular and metabolic brain regulation by affecting oxidative metabolism, ROS production, and endothelial and neuronal integrity. Only one study has recently tested the combined effects of both interventions and showed preliminary, positive outcomes cognitive function. This paper explores the potential synergistic effects of a nutritional strategy based on the co-administration of CR and a high-nitrate diet as a potential and more effective (than either intervention alone) strategy to protect brain health and reduce dementia risk.
Collapse
Affiliation(s)
- Mushari Alharbi
- School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, 22252, Saudi Arabia
| | - Blossom Cm Stephan
- Curtin Dementia Centre of Excellence, EnAble Institute, Curtin University, Kent Street, Bentley, WA, 6102, Australia
| | - Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Mario Siervo
- Curtin Dementia Centre of Excellence, EnAble Institute, Curtin University, Kent Street, Bentley, WA, 6102, Australia.
| |
Collapse
|
37
|
Elias A, Padinjakara N, Lautenschlager NT. Effects of intermittent fasting on cognitive health and Alzheimer's disease. Nutr Rev 2023; 81:1225-1233. [PMID: 37043764 PMCID: PMC10413426 DOI: 10.1093/nutrit/nuad021] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
OBJECTIVE Caloric restriction by intermittent fasting produces several metabolic changes, such as increased insulin sensitivity and use of ketone bodies as energy sources. In humans, intermittent fasting has been studied in hypertension, diabetes, and related conditions, but, to date, not as a strategy to reduce the risk of emergent dementia. In this scoping review, the relevance of intermittent fasting as a potential preventive intervention for Alzheimer's dementia is explored. BACKGROUND The beneficial effects of calorie restriction have been documented in animals and humans. Decreased oxidative stress damage and attenuated inflammatory responses are associated with intermittent fasting. These changes have a favorable impact on the vascular endothelium and stress-induced cellular adaptation. RESULTS Physiological alterations associated with fasting have profound implications for pathological mechanisms associated with dementias, particularly Alzheimer's disease. Compared with ad libitum feeding, caloric restriction in animals was associated with a reduction in β-amyloid accumulation, which is the cardinal pathological marker of Alzheimer's disease. Animal studies have demonstrated synaptic adaptations in the hippocampus and enhanced cognitive function after fasting, consistent with these theoretical frameworks. Furthermore, vascular dysfunction plays a crucial role in Alzheimer's disease pathology, and intermittent fasting promotes vascular health. CONCLUSIONS These observations lead to a hypothesis that intermittent fasting over the years will potentially reverse or delay the pathological process in Alzheimer's disease.
Collapse
Affiliation(s)
- Alby Elias
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, The University of Melbourne, North-Western Mental Health, Melbourne Health, Victoria, Australia
| | - Noushad Padinjakara
- Department of Endocrinology and Metabolic Medicine, South Warwickshire University NHS Foundation Trust, Coventry, United Kingdom
| | - Nicola T Lautenschlager
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, The University of Melbourne, North-Western Mental Health, Melbourne Health, Victoria, Australia
| |
Collapse
|
38
|
McLeod A, Bernabe BP, Xia Y, Sanchez-Flack J, Lamar M, Schiffer L, Hemphill NON, Fantuzzi G, Maki P, Fitzgibbon M, Tussing-Humphreys L. Exploring the Effects of a Mediterranean Diet and Weight Loss on the Gut Microbiome and Cognitive Performance in Older, African American Obese Adults: A Post Hoc Analysis. Nutrients 2023; 15:3332. [PMID: 37571270 PMCID: PMC10420801 DOI: 10.3390/nu15153332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
African American adults have a higher prevalence of Alzheimer's dementia (AD) than non-Hispanic Whites. The impact of a Mediterranean Diet (Med Diet) and intentional weight loss (IWL) on the gut microbiome may alter AD risk. A post hoc analysis of the Building Research in Diet and Cognition (BRIDGE) trial was performed to determine whether participation in an 8-month Med Diet lifestyle intervention with (n = 35) or without IWL (n = 31) was associated with changes in gut microbiota structure, abundance, and function and whether these changes were related to changes in cognitive performance. The results showed that family and genus alpha diversity increased significantly in both groups combined (p = 0.0075 and p = 0.024, respectively). However, there were no other significant microbially related within- or between-group changes over time. Also, an increase in Med Diet adherence was significantly associated with a decrease in alpha diversity at the phylum level only (p = 0.049). Increasing alpha diversity was associated with decreasing cognitive performance, but this association was attenuated after controlling for Med Diet adherence. In sum, an 8-month Med Diet lifestyle intervention with or without IWL did not appreciably alter the gut microbiome.
Collapse
Affiliation(s)
- Andrew McLeod
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA; (G.F.); (L.T.-H.)
- Institute for Health Research and Policy, University of Illinois Chicago, Chicago, IL 60608, USA; (J.S.-F.); (L.S.); (M.F.)
| | | | - Yinglin Xia
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (Y.X.); (M.L.)
| | - Jennifer Sanchez-Flack
- Institute for Health Research and Policy, University of Illinois Chicago, Chicago, IL 60608, USA; (J.S.-F.); (L.S.); (M.F.)
- Department of Pediatrics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Melissa Lamar
- Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (Y.X.); (M.L.)
- Rush Alzheimer’s Disease Center, Rush University, Chicago, IL 60612, USA
| | - Linda Schiffer
- Institute for Health Research and Policy, University of Illinois Chicago, Chicago, IL 60608, USA; (J.S.-F.); (L.S.); (M.F.)
| | | | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA; (G.F.); (L.T.-H.)
| | - Pauline Maki
- Departments of Psychology and Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Marian Fitzgibbon
- Institute for Health Research and Policy, University of Illinois Chicago, Chicago, IL 60608, USA; (J.S.-F.); (L.S.); (M.F.)
- Department of Pediatrics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA; (G.F.); (L.T.-H.)
- Institute for Health Research and Policy, University of Illinois Chicago, Chicago, IL 60608, USA; (J.S.-F.); (L.S.); (M.F.)
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
39
|
Boccardi V, Pigliautile M, Guazzarini AG, Mecocci P. The Potential of Fasting-Mimicking Diet as a Preventive and Curative Strategy for Alzheimer's Disease. Biomolecules 2023; 13:1133. [PMID: 37509169 PMCID: PMC10377404 DOI: 10.3390/biom13071133] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/05/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
This review examines the potential of fasting-mimicking diets (FMDs) in preventing and treating Alzheimer's disease (AD). FMDs are low-calorie diets that mimic the physiological and metabolic effects of fasting, including the activation of cellular stress response pathways and autophagy. Recent studies have shown that FMDs can reduce amyloid-beta accumulation, tau phosphorylation, and inflammation, as well as improve cognitive function in animal models of AD. Human studies have also reported improvements in AD biomarkers, cognitive functions, and subjective well-being measures following FMDs. However, the optimal duration and frequency of FMDs and their long-term safety and efficacy remain to be determined. Despite these uncertainties, FMDs hold promise as a non-pharmacological approach to AD prevention and treatment, and further research in this area is warranted.
Collapse
Affiliation(s)
- Virginia Boccardi
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Martina Pigliautile
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Anna Giulia Guazzarini
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
| | - Patrizia Mecocci
- Department of Medicine and Surgery, Institute of Gerontology and Geriatrics, University of Perugia, Piazzale Gambuli 1, 06132 Perugia, Italy
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
40
|
Doroszkiewicz J, Mroczko J, Rutkowski P, Mroczko B. Molecular Aspects of a Diet as a New Pathway in the Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:10751. [PMID: 37445928 PMCID: PMC10341644 DOI: 10.3390/ijms241310751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease is the most common cause of dementia in the world. Lack of an established pathology makes it difficult to develop suitable approaches and treatment for the disease. Besides known hallmarks, including amyloid β peptides cumulating in plaques and hyperphosphorylated tau forming NFTs, inflammation also plays an important role, with known connections to the diet. In AD, adhering to reasonable nutrition according to age-related principles is recommended. The diet should be high in neuroprotective foods, such as polyunsaturated fatty acids, antioxidants, and B vitamins. In addition, foods capable of rising BDNF should be considered because of the known profitable results of this molecule in AD. Adhering to beneficial diets might result in improvements in memory, cognition, and biomarkers and might even reduce the risk of developing AD. In this review, we discuss the effects of various diets, foods, and nutrients on brain health and possible connections to Alzheimer's disease.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
| | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-089 Bialystok, Poland; (J.M.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-089 Bialystok, Poland
| |
Collapse
|
41
|
Reiss AB, Muhieddine D, Jacob B, Mesbah M, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Alzheimer's Disease Treatment: The Search for a Breakthrough. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1084. [PMID: 37374288 PMCID: PMC10302500 DOI: 10.3390/medicina59061084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
As the search for modalities to cure Alzheimer's disease (AD) has made slow progress, research has now turned to innovative pathways involving neural and peripheral inflammation and neuro-regeneration. Widely used AD treatments provide only symptomatic relief without changing the disease course. The recently FDA-approved anti-amyloid drugs, aducanumab and lecanemab, have demonstrated unclear real-world efficacy with a substantial side effect profile. Interest is growing in targeting the early stages of AD before irreversible pathologic changes so that cognitive function and neuronal viability can be preserved. Neuroinflammation is a fundamental feature of AD that involves complex relationships among cerebral immune cells and pro-inflammatory cytokines, which could be altered pharmacologically by AD therapy. Here, we provide an overview of the manipulations attempted in pre-clinical experiments. These include inhibition of microglial receptors, attenuation of inflammation and enhancement of toxin-clearing autophagy. In addition, modulation of the microbiome-brain-gut axis, dietary changes, and increased mental and physical exercise are under evaluation as ways to optimize brain health. As the scientific and medical communities work together, new solutions may be on the horizon to slow or halt AD progression.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Dalia Muhieddine
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Berlin Jacob
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Michael Mesbah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | | | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, NYU School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
42
|
Sonsalla MM, Lamming DW. Geroprotective interventions in the 3xTg mouse model of Alzheimer's disease. GeroScience 2023; 45:1343-1381. [PMID: 37022634 PMCID: PMC10400530 DOI: 10.1007/s11357-023-00782-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disease. As the population ages, the increasing prevalence of AD threatens massive healthcare costs in the coming decades. Unfortunately, traditional drug development efforts for AD have proven largely unsuccessful. A geroscience approach to AD suggests that since aging is the main driver of AD, targeting aging itself may be an effective way to prevent or treat AD. Here, we discuss the effectiveness of geroprotective interventions on AD pathology and cognition in the widely utilized triple-transgenic mouse model of AD (3xTg-AD) which develops both β-amyloid and tau pathologies characteristic of human AD, as well as cognitive deficits. We discuss the beneficial impacts of calorie restriction (CR), the gold standard for geroprotective interventions, and the effects of other dietary interventions including protein restriction. We also discuss the promising preclinical results of geroprotective pharmaceuticals, including rapamycin and medications for type 2 diabetes. Though these interventions and treatments have beneficial effects in the 3xTg-AD model, there is no guarantee that they will be as effective in humans, and we discuss the need to examine these interventions in additional animal models as well as the urgent need to test if some of these approaches can be translated from the lab to the bedside for the treatment of humans with AD.
Collapse
Affiliation(s)
- Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, 2500 Overlook Terrace, VAH C3127 Research 151, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
43
|
Lü W, Yu T, Kuang W. Effects of dietary restriction on cognitive function: a systematic review and meta-analysis. Nutr Neurosci 2023; 26:540-550. [PMID: 35469542 DOI: 10.1080/1028415x.2022.2068876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Potential positive link between cognitive function and dietary restriction has been widely concerned. The present study describes a systematic review and preliminary meta-analysis to assess the efficacy of dietary restriction on cognitive function. We classified dietary restrictions as Calories Restriction (CR) and Intermittent Fasting (IF). METHOD This systematic review and meta-analysis conducted following the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Statement (PRISMA) Checklist, Databases including PubMed, Embase, Web of Science, and the Cochrane Library were searched for randomized controlled trials (RCTs) assessing the cognition effectiveness of dietary restriction from database inception to Sep 2021. RESULT Eleven RCTs met the inclusion criteria in the systematic review and meta-analysis. The overall effect of dietary restriction on cognitive function is SMD = 0.22 (95% CI: 0.09-0.34, p < 0.01). The effect on global function SMD = 0.35 (95% CI: 0.04-0.65, p < 0.05) and memory SMD = 0.18 (95% CI: 0.00-0.35, p = 0.05) is significant. MCI showed the best effectiveness SMD = 0.36 (95% CI: 0.05-0.68, p < 0.05), followed by the normal weight population SMD = 0.28 (95% CI: 0.03-0.52, p < 0.05) and overweight population SMD = 0.20 (95% CI: 0.06-0.34, p < 0.01). No statistically significant difference showed between IF and CR (p > 0.05). CONCLUSION Our study demonstrated that dietary restriction has varying degrees of positive effect on cognitive function in overweight/normal-weight people and MCI. However, it should be cautious when generalizing to other populations. Additional high-quality, large-scale, cohort and intervention studies are needed to further assess the effectiveness of dietary restriction on cognition.
Collapse
Affiliation(s)
- Wenqi Lü
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tingting Yu
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Weihong Kuang
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
44
|
Rad MS, Ansarinia M, Shafir E. Temporary Self-Deprivation Can Impair Cognitive Control: Evidence From the Ramadan Fast. PERSONALITY AND SOCIAL PSYCHOLOGY BULLETIN 2023; 49:415-428. [PMID: 35094597 DOI: 10.1177/01461672211070385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
During Ramadan, people of Muslim faith fast by not eating or drinking between sunrise and sunset. This is likely to have physiological and psychological consequences for fasters, and societal and economic impacts on the wider population. We investigate whether, during this voluntary and temporally limited fast, reminders of food can impair the fasters' reaction time and accuracy on a non-food-related test of cognitive control. Using a repeated measures design in a sample of Ramadan fasters (N = 190), we find that when food is made salient, fasters are slower and less accurate during Ramadan compared with after Ramadan. Control participants perform similarly across time. Furthermore, during Ramadan performances vary by how recently people had their last meal. Potential mechanisms are suggested, grounded in research on resource scarcity, commitment, and thought suppression, as well as the psychology of rituals and self-regulation, and implications for people who fast for religious or health reasons are discussed.
Collapse
Affiliation(s)
- Mostafa Salari Rad
- Kahneman -Treistamn Center for Behavioral Science and Public Policy, Princeton School of Public and International Affairs, Princeton University, NJ, USA.,Department of Psychology, New School for Social Research, New York, USA
| | - Morteza Ansarinia
- University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Eldar Shafir
- Kahneman -Treistamn Center for Behavioral Science and Public Policy, Princeton School of Public and International Affairs, Princeton University, NJ, USA
| |
Collapse
|
45
|
Blood-to-brain communication in aging and rejuvenation. Nat Neurosci 2023; 26:379-393. [PMID: 36646876 DOI: 10.1038/s41593-022-01238-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 11/21/2022] [Indexed: 01/18/2023]
Abstract
Aging induces molecular, cellular and functional changes in the adult brain that drive cognitive decline and increase vulnerability to dementia-related neurodegenerative diseases. Leveraging systemic and lifestyle interventions, such as heterochronic parabiosis, administration of 'young blood', exercise and caloric restriction, has challenged prevalent views of brain aging as a rigid process and has demonstrated that aging-associated cognitive and cellular impairments can be restored to more youthful levels. Technological advances in proteomic and transcriptomic analyses have further facilitated investigations into the functional impact of intertissue communication on brain aging and have led to the identification of a growing number of pro-aging and pro-youthful factors in blood. In this review, we discuss blood-to-brain communication from a systems physiology perspective with an emphasis on blood-derived signals as potent drivers of both age-related brain dysfunction and brain rejuvenation.
Collapse
|
46
|
Caloric Restriction (CR) Plus High-Nitrate Beetroot Juice Does Not Amplify CR-Induced Metabolic Adaptation and Improves Vascular and Cognitive Functions in Overweight Adults: A 14-Day Pilot Randomised Trial. Nutrients 2023; 15:nu15040890. [PMID: 36839248 PMCID: PMC9962072 DOI: 10.3390/nu15040890] [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: 01/17/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Caloric restriction (CR) and dietary nitrate supplementation are nutritional interventions with pleiotropic physiological functions. This pilot study investigates the combined effects of CR and nitrate-rich beetroot juice (BRJ) on metabolic, vascular, and cognitive functions in overweight and obese middle-aged and older adults. This was a two-arm, parallel randomized clinical trial including 29 participants allocated to CR + BRJ (n = 15) or CR alone (n = 14) for 14 days. Body composition, resting energy expenditure (REE), and hand-grip strength were measured. Resting blood pressure (BP) and microvascular endothelial function were measured, and Trail-Making Test A and B were used to assess cognitive function. Salivary nitrate and nitrite, and urinary nitrate and 8-isoprostane concentrations were measured. Changes in body composition, REE, and systolic and diastolic BP were similar between the two interventions (p > 0.05). The CR + BRJ intervention produced greater changes in average microvascular flux (p = 0.03), NO-dependent endothelial activity (p = 0.02), and TMT-B cognitive scores (p = 0.012) compared to CR alone. Changes in urinary 8-isoprostane were greater in the CR + BRJ group (p = 0.02), and they were inversely associated with changes in average microvascular flux (r = -0.53, p = 0.003). These preliminary findings suggest that greater effects on vascular and cognitive functions could be achieved by combining CR with dietary nitrate supplementation.
Collapse
|
47
|
Abdulghani A, Poghosyan M, Mehren A, Philipsen A, Anderzhanova E. Neuroplasticity to autophagy cross-talk in a therapeutic effect of physical exercises and irisin in ADHD. Front Mol Neurosci 2023; 15:997054. [PMID: 36776770 PMCID: PMC9909442 DOI: 10.3389/fnmol.2022.997054] [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: 07/18/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023] Open
Abstract
Adaptive neuroplasticity is a pivotal mechanism for healthy brain development and maintenance, as well as its restoration in disease- and age-associated decline. Management of mental disorders such as attention deficit hyperactivity disorder (ADHD) needs interventions stimulating adaptive neuroplasticity, beyond conventional psychopharmacological treatments. Physical exercises are proposed for the management of ADHD, and also depression and aging because of evoked brain neuroplasticity. Recent progress in understanding the mechanisms of muscle-brain cross-talk pinpoints the role of the myokine irisin in the mediation of pro-cognitive and antidepressant activity of physical exercises. In this review, we discuss how irisin, which is released in the periphery as well as derived from brain cells, may interact with the mechanisms of cellular autophagy to provide protein recycling and regulation of brain-derived neurotrophic factor (BDNF) signaling via glia-mediated control of BDNF maturation, and, therefore, support neuroplasticity. We propose that the neuroplasticity associated with physical exercises is mediated in part by irisin-triggered autophagy. Since the recent findings give objectives to consider autophagy-stimulating intervention as a prerequisite for successful therapy of psychiatric disorders, irisin appears as a prototypic molecule that can activate autophagy with therapeutic goals.
Collapse
Affiliation(s)
- Alhasan Abdulghani
- C. and O. Vogt Institute for Brain Research, Medical Faculty and University Hospital Düsseldorf, Henrich Heine University, Düsseldorf, Düsseldorf, Germany,*Correspondence: Alhasan Abdulghani,
| | - Mikayel Poghosyan
- Institute for Biology-Neurobiology, Freie University of Berlin, Berlin, Germany
| | - Aylin Mehren
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Elmira Anderzhanova
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
48
|
Zonneveld MH, Noordam R, Sabayan B, Stott DJ, Mooijaart SP, Blauw GJ, Jukema JW, Sattar N, Trompet S. Weight loss, visit-to-visit body weight variability and cognitive function in older individuals. Age Ageing 2023; 52:6974853. [PMID: 36626325 PMCID: PMC9990986 DOI: 10.1093/ageing/afac312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 08/01/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE to investigate the association between variability and loss of body weight with subsequent cognitive performance and activities of daily living in older individuals. DESIGN cross-sectional cohort study. SETTING PROspective Study of Pravastatin in the Elderly at Risk, multicentre trial with participants from Scotland, Ireland and the Netherlands. SUBJECTS 4,309 participants without severe cognitive dysfunction (mean age 75.1 years, standard deviation (SD) = 3.3), at higher risk for cardiovascular disease (CVD). METHODS body weight was measured every 3 months for 2.5 years. Weight loss was defined as an average slope across all weight measurements and as ≥5% decrease in baseline body weight during follow-up. Visit-to-visit variability was defined as the SD of weight measurements (kg) between visits. Four tests of cognitive function were examined: Stroop test, letter-digit coding test (LDCT), immediate and delayed picture-word learning tests. Two measures of daily living activities: Barthel Index (BI) and instrumental activities of daily living (IADL). All tests were examined at month 30. RESULTS both larger body weight variability and loss of ≥5% of baseline weight were independently associated with worse scores on all cognitive tests, but minimally with BI and IADL. Compared with participants with stable weight, participants with significant weight loss performed 5.83 seconds (95% CI 3.74; 7.92) slower on the Stroop test, coded 1.72 digits less (95% CI -2.21; -1.13) on the LDCT and remembered 0.71 pictures less (95% CI -0.93; -0.48) on the delayed picture-word learning test. CONCLUSION in older people at higher risk for CVD, weight loss and variability are independent risk-factors for worse cognitive function.
Collapse
Affiliation(s)
- Michelle H Zonneveld
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Behnam Sabayan
- HealthPartners Institute, Neuroscience Center, Bloomington, MN, USA and University of Minnesota, School of Public Health, Division of Epidemiology and Community Health
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerard J Blauw
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Pal D, Sahu P, Mishra AK, Hagelgans A, Sukocheva O. Histone Deacetylase Inhibitors as Cognitive Enhancers and Modifiers of Mood and Behavior. Curr Drug Targets 2023; 24:728-750. [PMID: 36475351 DOI: 10.2174/1389450124666221207090108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Epigenetic regulation of gene signalling is one of the fundamental molecular mechanisms for the generation and maintenance of cellular memory. Histone acetylation is a common epigenetic mechanism associated with increased gene transcription in the central nervous system (CNS). Stimulation of gene transcription by histone acetylation is important for the development of CNS-based long-term memory. Histone acetylation is a target for cognitive enhancement via the application of histone deacetylase (HDAC) inhibitors. The promising potential of HDAC inhibitors has been observed in the treatment of several neurodevelopmental and neurodegenerative diseases. OBJECTIVE This study assessed the current state of HDAC inhibition as an approach to cognitive enhancement and treatment of neurodegenerative diseases. Our analysis provides insights into the mechanism of action of HDAC inhibitors, associated epigenetic priming, and describes the therapeutic success and potential complications after unsupervised use of the inhibitors. RESULTS AND CONCLUSION Several chromatin-modifying enzymes play key roles in the regulation of cognitive processes. The importance of HDAC signaling in the brain is highlighted in this review. Recent advancements in the field of cognitive epigenetics are supported by the successful development of various HDAC inhibitors, demonstrating effective treatment of mood-associated disorders. The current review discusses the therapeutic potential of HDAC inhibition and observed complications after mood and cognitive enhancement therapies.
Collapse
Affiliation(s)
- Dilipkumar Pal
- Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495 009, India
| | - Pooja Sahu
- Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495 009, India
| | | | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Olga Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, 5042, SA, Australia
| |
Collapse
|
50
|
Santos AL, Sinha S. Ageing, Metabolic Dysfunction, and the Therapeutic Role of Antioxidants. Subcell Biochem 2023; 103:341-435. [PMID: 37120475 DOI: 10.1007/978-3-031-26576-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The gradual ageing of the world population has been accompanied by a dramatic increase in the prevalence of obesity and metabolic diseases, especially type 2 diabetes. The adipose tissue dysfunction associated with ageing and obesity shares many common physiological features, including increased oxidative stress and inflammation. Understanding the mechanisms responsible for adipose tissue dysfunction in obesity may help elucidate the processes that contribute to the metabolic disturbances that occur with ageing. This, in turn, may help identify therapeutic targets for the treatment of obesity and age-related metabolic disorders. Because oxidative stress plays a critical role in these pathological processes, antioxidant dietary interventions could be of therapeutic value for the prevention and/or treatment of age-related diseases and obesity and their complications. In this chapter, we review the molecular and cellular mechanisms by which obesity predisposes individuals to accelerated ageing. Additionally, we critically review the potential of antioxidant dietary interventions to counteract obesity and ageing.
Collapse
Affiliation(s)
- Ana L Santos
- IdISBA - Fundación de Investigación Sanitaria de las Islas Baleares, Palma, Spain.
| | | |
Collapse
|