1
|
Qu S, Guan X, Li H, Yang J, Sun P, Song C, Surzenko N, Wang Y. Dietary Intake of Octanoic Acid Restores UBE3A Expression and Improves the Behavioral Phenotypes in a Mouse Model of Angelman Syndrome. FASEB J 2025; 39:e70559. [PMID: 40277210 PMCID: PMC12023718 DOI: 10.1096/fj.202403130rr] [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: 12/07/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder with no effective therapies. Most of the behavioral deficits observed in AS patients arise from the absence of ubiquitin protein ligase E3A (UBE3A) in the brain during development, driven by the loss of maternally expressed UBE3A and silencing of the paternal copy of this gene through imprinting. Safe and effective therapies aiming at restoring the expression of the paternal UBE3A gene early in human life are currently lacking. In this study, we investigated whether octanoic acid (OA), a medium-chain fatty acid, could unsilence the paternal Ube3a allele in neurons and ameliorate the behavioral defects in a murine model of AS. To this end, Ube3am-/p+ and Ube3am+/pYFP mice, as well as their wild-type littermates, were fed either a control or OA-supplemented diet from postnatal day 0 through adulthood, and the improvements in AS-related cellular and behavioral deficits were characterized. We demonstrate that dietary intake of OA activates the expression of the silenced, paternal Ube3a in neurons and improves select AS behavioral phenotypes in mice. We further show that downregulation of topoisomerase II beta and restoration of dendritic spine development may underlie the unsilencing of Ube3a and the behavioral improvements in OA-supplemented animals, respectively. Together, our findings suggest that dietary supplementation with OA could serve as an early, safe, and clinically feasible therapeutic strategy for reactivation of the paternal UBE3A allele in patients with AS.
Collapse
Affiliation(s)
- Song Qu
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xingying Guan
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Hongyan Li
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Jian Yang
- Undergraduate Student Brigade, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| | - Pu Sun
- Maternal and Child Health Hospital of Yongchuan DistrictChongqingChina
| | - Cui Song
- Department of Endocrinology and Genetic Metabolism Disease, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of PediatricsChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | | | - Yanyan Wang
- Department of Medical Genetics, College of Basic Medical ScienceArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
2
|
Huang SY, Yang ZJ, Cheng J, Li HY, Chen S, Huang ZH, Chen JD, Xiong RG, Yang MT, Wang C, Li MC, Song S, Huang WG, Wang DL, Li HB, Lan QY. Choline alleviates cognitive impairment in sleep-deprived young mice via reducing neuroinflammation and altering phospholipidomic profile. Redox Biol 2025; 81:103578. [PMID: 40056720 PMCID: PMC11930228 DOI: 10.1016/j.redox.2025.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025] Open
Abstract
Cognitive impairment resulting from insufficient sleep poses a significant public health concern, particularly in children. The effects and mechanisms of choline on cognitive impairment caused by sleep deprivation are unknown. Chronic sleep deprivation is induced in young mice in this study, followed by feeding diet containing 11.36 g/kg choline bitartrate. Choline supplementation significantly improves spatial learning ability. Functional MRI results reveal the hippocampus as a key region affected by sleep deprivation, where choline supplementation notably preserves hippocampal structural integrity and enhanced connectivity. Additionally, choline ameliorates hippocampal pathological injury, reduces blood-brain barrier permeability and serum brain injury biomarkers. Choline also reduces inflammation and oxidative stress biomarkers, and mitigates microglial activation in the hippocampus, which preserves synaptic plasticity. A key finding is the changes of hippocampal phospholipidomic profile along with cognitive function, and a total of 313 phospholipid molecules are identified. Choline increases the levels of total phospholipid and sub-classes (particularly PC), which are strongly correlated with reduced neuroinflammation and oxidative stress biomarkers, as well as improved cognitive outcomes. Furthermore, there are similar findings in some phospholipid molecules such as PC 36:1, PC O-33:0, PC p-38:3, PE 36:3, PE p-42:4 and PS 44:12. These findings highlight that choline alleviates cognitive impairment in sleep deprivation via reducing neuroinflammation and oxidative stress as well as altering phospholipidomic profile. This study suggests that choline could develop into functional food or medicine ingredient to prevent and treat cognitive impairment by sleep disturbances, particularly children and adolescents.
Collapse
Affiliation(s)
- Si-Yu Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhi-Jun Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jin Cheng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hang-Yu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Si Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zi-Hui Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jie-Dong Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ruo-Gu Xiong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Tao Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chen Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Chu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Wen-Ge Huang
- Center of Experimental Animals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong-Liang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hua-Bin Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Qiu-Ye Lan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China; School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Al-Abbas NS, Shaer NA. Gut microbiome synthesizes important core metabolites to prevent cognitive decline and mitigate onset and progression of Alzheimer's disease. J Alzheimers Dis Rep 2024; 8:1705-1721. [PMID: 40034366 PMCID: PMC11863740 DOI: 10.1177/25424823241309024] [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: 09/13/2024] [Accepted: 11/24/2024] [Indexed: 03/05/2025] Open
Abstract
Background This study explores how gut metabolites, produced through bacterial metabolism in the gut, influence neurological conditions like Alzheimer's disease (AD). Key metabolites such as succinate and short-chain fatty acids signal through the autonomic nervous system and can cross the blood-brain barrier, impacting central nervous system functions. Objective The aim is to examine the role of the gut microbiota in compensating for metabolic deficiencies in AD. By analyzing wild-type (WT) and APP/PS1 mice, the study investigates how the microbiome affects key metabolic processes and whether it can slow AD progression. Methods High-throughput sequencing data from the gut microbiomes of APP/PS1 transgenic AD model mice and age-matched WT C57BL/6 male mice were analyzed for microbial and metabolite profiles. Results Alpha and beta diversity analyses showed differences in microbial composition between groups. Partial least squares discriminant analysis and Anosim confirmed distinct microbiome profiles in WT and APP/PS1 mice. At the genus level, Vescimonas was more abundant in WT mice, while Odoribacter, Lacrimispora, Helicobacter, Bacteroides, and Alloprevotella were more prevalent in APP/PS1 mice. Conclusions While taxonomic differences did not directly link specific microorganisms to AD, functional analysis identified key metabolites-acetyl-CoA, glucose, succinate, lipids, choline, and acetylcholine-that may alleviate energy deficits and synaptic dysfunction. This study suggests that the microbiome may help compensate for AD-related impairments, opening avenues for microbiome-based therapies.
Collapse
Affiliation(s)
- Nouf S Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nehad A Shaer
- Department of Chemistry, Al Lieth University College, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
4
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [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: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
5
|
Eslami M, Alibabaei F, Babaeizad A, Banihashemian SZ, Mazandarani M, Hoseini A, Ramezankhah M, Oksenych V, Yousefi B. The Importance of Gut Microbiota on Choline Metabolism in Neurodegenerative Diseases. Biomolecules 2024; 14:1345. [PMID: 39595522 PMCID: PMC11591558 DOI: 10.3390/biom14111345] [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: 09/21/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
The gut microbiota is a complex ecosystem that influences digestion, immune response, metabolism, and has been linked to health and well-being. Choline is essential for neurotransmitters, lipid transport, cell-membrane signaling, methyl-group metabolism and is believed to have neuroprotective properties. It is found in two forms, water-soluble and lipid-soluble, and its metabolism is different. Long-term choline deficiency is associated with many diseases, and supplements are prescribed for improved health. Choline supplements can improve cognitive function in adults but not significantly. Choline is a precursor of phospholipids and an acetylcholine neurotransmitter precursor and can be generated de novo from phosphatidylcholine via phosphatidylethanolamine-N-methyltransferase and choline oxidase. Choline supplementation has been found to have a beneficial effect on patients with neurodegenerative diseases, such as Alzheimer's disease (AD), by increasing amyloid-β, thioflavin S, and tau hyper-phosphorylation. Choline supplementation has been shown to reduce amyloid-plaque load and develop spatial memory in an APP/PS1 mice model of AD. Choline is necessary for normative and improved function of brain pathways and can reduce amyloid-β deposition and microgliosis. Clinical research suggests that early neurodegenerative diseases (NDs) can benefit from a combination of choline supplements and the drugs currently used to treat NDs in order to improve memory performance and synaptic functioning.
Collapse
Affiliation(s)
- Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan 35134, Iran;
| | - Farnaz Alibabaei
- Student Research Committee, School of Medicine, Semnan University of Medical Sciences, Semnan 35134, Iran;
| | - Ali Babaeizad
- School of Medicine, Semnan University of Medical Sciences, Semnan 35134, Iran; (A.B.); (S.Z.B.)
| | | | - Mahdi Mazandarani
- Endocrinology and Metabolism Research Center, Faculty of Medicine, Tehran University of Medical Sciences, Tehran 11369, Iran;
| | - Aref Hoseini
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari 49414, Iran;
| | - Mohammad Ramezankhah
- Student Research Committee, Faculty of Medicine, Babol University of Medical Sciences, Babol 47134, Iran;
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Bahman Yousefi
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan 35134, Iran
| |
Collapse
|
6
|
Chartampila E, Elayouby KS, Leary P, LaFrancois JJ, Alcantara-Gonzalez D, Jain S, Gerencer K, Botterill JJ, Ginsberg SD, Scharfman HE. Choline supplementation in early life improves and low levels of choline can impair outcomes in a mouse model of Alzheimer's disease. eLife 2024; 12:RP89889. [PMID: 38904658 PMCID: PMC11192536 DOI: 10.7554/elife.89889] [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: 06/22/2024] Open
Abstract
Maternal choline supplementation (MCS) improves cognition in Alzheimer's disease (AD) models. However, the effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated the effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice are generalized EEG spikes (interictal spikes [IIS]). IIS also are common in other mouse models and occur in AD patients. In mouse models, hyperexcitability is also reflected by elevated expression of the transcription factor ∆FosB in the granule cells (GCs) of the dentate gyrus (DG), which are the principal cell type. Therefore, we studied ΔFosB expression in GCs. We also studied the neuronal marker NeuN within hilar neurons of the DG because reduced NeuN protein expression is a sign of oxidative stress or other pathology. This is potentially important because hilar neurons regulate GC excitability. Tg2576 breeding pairs received a diet with a relatively low, intermediate, or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ∆FosB expression was reduced, and hilar NeuN expression was restored. Using the novel object location task, spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. They had the weakest hilar NeuN immunoreactivity and greatest GC ΔFosB protein expression. However, their IIS frequency was low, which was surprising. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have mixed effects. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, ΔFosB, and spatial memory in an animal model of AD.
Collapse
Affiliation(s)
- Elissavet Chartampila
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Karim S Elayouby
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Paige Leary
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
| | - John J LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Justin J Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
| | - Stephen D Ginsberg
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- NYU Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Helen E Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric ResearchOrangeburgUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Departments of Child and Adolescent Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- NYU Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
7
|
Chartampila E, Elayouby KS, Leary P, LaFrancois JJ, Alcantara-Gonzalez D, Jain S, Gerencer K, Botterill JJ, Ginsberg SD, Scharfman HE. Choline supplementation in early life improves and low levels of choline can impair outcomes in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.12.540428. [PMID: 37214805 PMCID: PMC10197642 DOI: 10.1101/2023.05.12.540428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Maternal choline supplementation (MCS) improves cognition in Alzheimer's disease (AD) models. However, effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice are generalized EEG spikes (interictal spikes; IIS). IIS also are common in other mouse models and occur in AD patients. Im mouse models, hyperexcitability is also reflected by elevated expression of the transcription factor ΔFosB in the granule cells (GCs) of the dentate gyrus (DG), which are the principal cell type. Therefore we studied ΔFosB expression in GCs. We also studied the the neuronal marker NeuN within hilar neurons of the DG because other studies have reduced NeuN protein expression is a sign of oxidative stress or other pathology. This is potentially important because hilar neurons regulate GC excitability. Tg2576 breeding pairs received a diet with a relatively low, intermediate or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ΔFosB expression was reduced, and NeuN expression was restored. Using the novel object location task, spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. They had the weakest hilar NeuN immunoreactivity and greatest GC ΔFosB protein expression. However, their IIS frequency was low, which was surprising. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have mixed effects. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, ΔFosB and spatial memory in an animal model of AD.
Collapse
Affiliation(s)
- Elissavet Chartampila
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address:Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27510
| | - Karim S. Elayouby
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Neurology, Mount Sinai School of Medicine, New York, NY 10029
| | - Paige Leary
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
| | - John J. LaFrancois
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
| | - David Alcantara-Gonzalez
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
| | - Swati Jain
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
| | - Kasey Gerencer
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Psychology, University of Maine, Orono, ME 04469
| | - Justin J. Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Current address: Department of Anatomy, Physiology, & Pharmacology, College of Medicine, Saskatoon, SK S7N 5E5
| | - Stephen D. Ginsberg
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
- Department of Psychiatry, New York University Grossman School of Medicine New York, NY 10016
- NYU Neuroscience Institute,, New York University Grossman School of Medicine, New York, NY 10016
| | - Helen E. Scharfman
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY 100016
- Department of Child and Adolescent Psychiatry , New York University Grossman School of Medicine, New York, NY 10016
- Department of Psychiatry, New York University Grossman School of Medicine New York, NY 10016
- NYU Neuroscience Institute,, New York University Grossman School of Medicine, New York, NY 10016
| |
Collapse
|
8
|
Bellio TA, Laguna-Torres JY, Campion MS, Chou J, Yee S, Blusztajn JK, Mellott TJ. Perinatal choline supplementation prevents learning and memory deficits and reduces brain amyloid Aβ42 deposition in AppNL-G-F Alzheimer's disease model mice. PLoS One 2024; 19:e0297289. [PMID: 38315685 PMCID: PMC10843108 DOI: 10.1371/journal.pone.0297289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive and memory impairments and neuropathological abnormalities. AD has no cure, inadequate treatment options, and a limited understanding of possible prevention measures. Previous studies have demonstrated that AD model mice that received a diet high in the essential nutrient choline had reduced amyloidosis, cholinergic deficits, and gliosis, and increased neurogenesis. In this study, we investigated the lifelong effects of perinatal choline supplementation on behavior, cognitive function, and amyloidosis in AppNL-G-F AD model mice. Pregnant and lactating mice were given a diet containing either 1.1 g/kg (control) or 5 g/kg (supplemented) of choline chloride until weaning and subsequently, all offspring received the control diet throughout their life. At 3, 6, 9, and 12 months of age, animals were behaviorally tested in the Open Field Test, Elevated Plus Maze, Barnes Maze, and in a contextual fear conditioning paradigm. Immunohistochemical analysis of Aβ42 was also conducted on the brains of these mice. AppNL-G-F mice displayed hippocampal-dependent spatial learning deficits starting at 3-months-old that persisted until 12-months-old. These spatial learning deficits were fully prevented by perinatal choline supplementation at young ages (3 and 6 months) but not in older mice (12 months). AppNL-G-F mice also had impaired fearful learning and memory at 9- and 12-months-old that were diminished by choline supplementation. Perinatal choline supplementation reduced Aβ42 deposition in the amygdala, cortex, and hippocampus of AppNL-G-F mice. Together, these results demonstrate that perinatal choline supplementation is capable of preventing cognitive deficits and dampening amyloidosis in AppNL-G-F mice and suggest that ensuring adequate choline consumption during early life may be a valuable method to prevent or reduce AD dementia and neuropathology.
Collapse
Affiliation(s)
- Thomas A. Bellio
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jessenia Y. Laguna-Torres
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Mary S. Campion
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jay Chou
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Sheila Yee
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Jan K. Blusztajn
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| | - Tiffany J. Mellott
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
9
|
Judd JM, Jasbi P, Winslow W, Serrano GE, Beach TG, Klein-Seetharaman J, Velazquez R. Inflammation and the pathological progression of Alzheimer's disease are associated with low circulating choline levels. Acta Neuropathol 2023; 146:565-583. [PMID: 37548694 PMCID: PMC10499952 DOI: 10.1007/s00401-023-02616-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/08/2023]
Abstract
Deficiency of dietary choline, an essential nutrient, is observed worldwide, with ~ 90% of Americans being deficient. Previous work highlights a relationship between decreased choline intake and an increased risk for cognitive decline and Alzheimer's disease (AD). The associations between blood circulating choline and the pathological progression in both mild cognitive impairment (MCI) and AD remain unknown. Here, we examined these associations in a cohort of patients with MCI with presence of either sparse or high neuritic plaque density and Braak stage and a second cohort with either moderate AD (moderate to frequent neuritic plaques, Braak stage = IV) or severe AD (frequent neuritic plaques, Braak stage = VI), compared to age-matched controls. Metabolomic analysis was performed on serum from the AD cohort. We then assessed the effects of dietary choline deficiency (Ch-) in 3xTg-AD mice and choline supplementation (Ch+) in APP/PS1 mice, two rodent models of AD. The levels of circulating choline were reduced while pro-inflammatory cytokine TNFα was elevated in serum of both MCI sparse and high pathology cases. Reduced choline and elevated TNFα correlated with higher neuritic plaque density and Braak stage. In AD patients, we found reductions in choline, its derivative acetylcholine (ACh), and elevated TNFα. Choline and ACh levels were negatively correlated with neuritic plaque load, Braak stage, and TNFα, but positively correlated with MMSE, and brain weight. Metabolites L-Valine, 4-Hydroxyphenylpyruvic, Methylmalonic, and Ferulic acids were significantly associated with circuiting choline levels. In 3xTg-AD mice, the Ch- diet increased amyloid-β levels and tau phosphorylation in cortical tissue, and TNFα in both blood and cortical tissue, paralleling the severe human-AD profile. Conversely, the Ch+ diet increased choline and ACh while reducing amyloid-β and TNFα levels in brains of APP/PS1 mice. Collectively, low circulating choline is associated with AD-neuropathological progression, illustrating the importance of adequate dietary choline intake to offset disease.
Collapse
Affiliation(s)
- Jessica M Judd
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, 85014, USA
| | - Paniz Jasbi
- School of Molecular Sciences, Arizona State University, Phoenix, AZ, 85287, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Tempe, AZ, 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, 85014, USA
| | - Geidy E Serrano
- Arizona Alzheimer's Consortium, Phoenix, AZ, 85014, USA
- Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Thomas G Beach
- Arizona Alzheimer's Consortium, Phoenix, AZ, 85014, USA
- Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | | | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Tempe, AZ, 85287, USA.
- Arizona Alzheimer's Consortium, Phoenix, AZ, 85014, USA.
- School of Life Sciences, Arizona State University, 797 E Tyler St, Tempe, AZ, 85287, USA.
| |
Collapse
|
10
|
Aguree S, Zolnoori M, Atwood TP, Owora A. Association between choline supplementation and Alzheimer's disease risk: a systematic review protocol. Front Aging Neurosci 2023; 15:1242853. [PMID: 37700814 PMCID: PMC10493398 DOI: 10.3389/fnagi.2023.1242853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023] Open
Abstract
Background and aims There is growing evidence suggesting choline intake might have beneficial effects on cognitive function in the elderly. However, some studies report no relationship between choline intake and cognitive function or improvement in Alzheimer's disease patients. This protocol is for a systematic review of choline intake and Alzheimer's disease that aims to assess the comparative clinical effectiveness of choline supplementation on Alzheimer's disease risk. Methods and analysis literature search will be performed in PubMed, MEDLINE, EMBASE, CINAHL, Scopus, Cochrane, and the Web of Science electronic databases from inception until October 2023. We will follow the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Studies will be included if they compared two different time points of choline biomarkers measures in men or women (65+) with Alzheimer's Disease. The risk of bias in the included studies will be assessed within the Covidence data-management software. Results This review will summarize the clinical trial and quasi-experimental evidence of choline intake on Alzheimer's disease risk for adults aged 65+. The results from all eligible studies included in the analysis will be presented in tables, text, and figures. A descriptive synthesis will present the characteristics of included studies (e.g., age, sex of participants, type, length of intervention and comparator, and outcome measures), critical appraisal results, and descriptions of the main findings. Discussion This systematic review will summarize the existing evidence on the association between Choline intake and AD and to make recommendations if appropriate. The results of this review will be considered with respect to whether there is enough evidence of benefit to merit a more definitive randomized controlled trial. The results will be disseminated through peer-reviewed journals population. Conclusion This protocol outlines the methodology for a systematic review of choline intake and AD. The resulting systematic review from this protocol will form an evidence-based foundation to advance nutrition care for individuals with AD or poor cognitive function. Systematic review registration http://www.crd.york.ac.uk/PROSPERO, identifier CRD42023395004.
Collapse
Affiliation(s)
- Sixtus Aguree
- Department of Applied Health Science, School of Public Health, Indiana University Bloomington, Bloomington, IN, United States
| | - Maryam Zolnoori
- Columbia University Irving Medical Center, New York, NY, United States
| | - Thea Patricia Atwood
- Department of Chemistry, Indiana University Bloomington, Bloomington, IN, United States
| | - Arthur Owora
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
11
|
Judd JM, Jasbi P, Winslow W, Serrano GE, Beach TG, Klein-Seetharaman J, Velazquez R. Low circulating choline, a modifiable dietary factor, is associated with the pathological progression and metabolome dysfunction in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.06.539713. [PMID: 37214864 PMCID: PMC10197582 DOI: 10.1101/2023.05.06.539713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Most Americans (∼90%) are deficient in dietary choline, an essential nutrient. Associations between circulating choline and pathological progression in Alzheimer's disease (AD) remain unknown. Here, we examined these associations and performed a metabolomic analysis in blood serum from severe AD, moderate AD, and healthy controls. Additionally, to gain mechanistic insight, we assessed the effects of dietary choline deficiency (Ch-) in 3xTg-AD mice and choline supplementation (Ch+) in APP/PS1 mice. In humans, we found AD-associated reductions in choline, it's derivative acetylcholine (ACh), and elevated pro-inflammatory cytokine TNFα. Choline and ACh were negatively correlated with Plaque density, Braak stage, and TNFα, but positively correlated with MMSE and brain weight. Metabolites L-Valine, 4-Hydroxyphenylpyruvic, Methylmalonic, and Ferulic acids were associated with choline levels. In mice, Ch-paralleled AD severe, but Ch+ was protective. In conclusion, low circulating choline is associated with AD-neuropathological progression, illustrating the importance of dietary choline consumption to offset disease.
Collapse
|
12
|
Chiarini A, Gui L, Viviani C, Armato U, Dal Prà I. NLRP3 Inflammasome’s Activation in Acute and Chronic Brain Diseases—An Update on Pathogenetic Mechanisms and Therapeutic Perspectives with Respect to Other Inflammasomes. Biomedicines 2023; 11:biomedicines11040999. [PMID: 37189617 DOI: 10.3390/biomedicines11040999] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Increasingly prevalent acute and chronic human brain diseases are scourges for the elderly. Besides the lack of therapies, these ailments share a neuroinflammation that is triggered/sustained by different innate immunity-related protein oligomers called inflammasomes. Relevant neuroinflammation players such as microglia/monocytes typically exhibit a strong NLRP3 inflammasome activation. Hence the idea that NLRP3 suppression might solve neurodegenerative ailments. Here we review the recent Literature about this topic. First, we update conditions and mechanisms, including RNAs, extracellular vesicles/exosomes, endogenous compounds, and ethnic/pharmacological agents/extracts regulating NLRP3 function. Second, we pinpoint NLRP3-activating mechanisms and known NLRP3 inhibition effects in acute (ischemia, stroke, hemorrhage), chronic (Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, MS, ALS), and virus-induced (Zika, SARS-CoV-2, and others) human brain diseases. The available data show that (i) disease-specific divergent mechanisms activate the (mainly animal) brains NLRP3; (ii) no evidence proves that NLRP3 inhibition modifies human brain diseases (yet ad hoc trials are ongoing); and (iii) no findings exclude that concurrently activated other-than-NLRP3 inflammasomes might functionally replace the inhibited NLRP3. Finally, we highlight that among the causes of the persistent lack of therapies are the species difference problem in disease models and a preference for symptomatic over etiologic therapeutic approaches. Therefore, we posit that human neural cell-based disease models could drive etiological, pathogenetic, and therapeutic advances, including NLRP3’s and other inflammasomes’ regulation, while minimizing failure risks in candidate drug trials.
Collapse
|
13
|
Bekdash RA. Methyl Donors, Epigenetic Alterations, and Brain Health: Understanding the Connection. Int J Mol Sci 2023; 24:ijms24032346. [PMID: 36768667 PMCID: PMC9917111 DOI: 10.3390/ijms24032346] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Methyl donors such as choline, betaine, folic acid, methionine, and vitamins B6 and B12 are critical players in the one-carbon metabolism and have neuroprotective functions. The one-carbon metabolism comprises a series of interconnected chemical pathways that are important for normal cellular functions. Among these pathways are those of the methionine and folate cycles, which contribute to the formation of S-adenosylmethionine (SAM). SAM is the universal methyl donor of methylation reactions such as histone and DNA methylation, two epigenetic mechanisms that regulate gene expression and play roles in human health and disease. Epigenetic mechanisms have been considered a bridge between the effects of environmental factors, such as nutrition, and phenotype. Studies in human and animal models have indicated the importance of the optimal levels of methyl donors on brain health and behavior across the lifespan. Imbalances in the levels of these micronutrients during critical periods of brain development have been linked to epigenetic alterations in the expression of genes that regulate normal brain function. We present studies that support the link between imbalances in the levels of methyl donors, epigenetic alterations, and stress-related disorders. Appropriate levels of these micronutrients should then be monitored at all stages of development for a healthier brain.
Collapse
Affiliation(s)
- Rola A Bekdash
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
14
|
Chakrovorty A, Bhattacharjee B, Saxena A, Samadder A, Nandi S. Current Naturopathy to Combat Alzheimer's Disease. Curr Neuropharmacol 2023; 21:808-841. [PMID: 36173068 PMCID: PMC10227918 DOI: 10.2174/1570159x20666220927121022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/13/2022] [Accepted: 07/18/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegeneration is the progressive loss of structure or function of neurons, which may ultimately involve cell death. The most common neurodegenerative disorder in the brain happens with Alzheimer's disease (AD), the most common cause of dementia. It ultimately leads to neuronal death, thereby impairing the normal functionality of the central or peripheral nervous system. The onset and prevalence of AD involve heterogeneous etiology, either in terms of genetic predisposition, neurometabolomic malfunctioning, or lifestyle. The worldwide relevancies are estimated to be over 45 million people. The rapid increase in AD has led to a concomitant increase in the research work directed towards discovering a lucrative cure for AD. The neuropathology of AD comprises the deficiency in the availability of neurotransmitters and important neurotrophic factors in the brain, extracellular betaamyloid plaque depositions, and intracellular neurofibrillary tangles of hyperphosphorylated tau protein. Current pharmaceutical interventions utilizing synthetic drugs have manifested resistance and toxicity problems. This has led to the quest for new pharmacotherapeutic candidates naturally prevalent in phytochemicals. This review aims to provide an elaborative description of promising Phyto component entities having activities against various potential AD targets. Therefore, naturopathy may combine with synthetic chemotherapeutics to longer the survival of the patients.
Collapse
Affiliation(s)
- Arnob Chakrovorty
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Banani Bhattacharjee
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Aaruni Saxena
- Department of Cardiovascular Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Asmita Samadder
- Department of Zoology, Cytogenetics and Molecular Biology Lab., University of Kalyani, Kalyani, 741235, India
| | - Sisir Nandi
- Department of Pharmaceutical Chemistry, Global Institute of Pharmaceutical Education and Research, Affiliated to Uttarakhand Technical University, Kashipur, 244713, India
| |
Collapse
|
15
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial and neuronal changes in Alzheimer's disease, and the related controversies II: gliotherapies and multimodal therapy. J Cent Nerv Syst Dis 2022; 14:11795735221123896. [PMID: 36407561 PMCID: PMC9666878 DOI: 10.1177/11795735221123896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 08/05/2022] [Indexed: 08/30/2023] Open
Abstract
Since the original description of Alzheimer´s disease (AD), research into this condition has mainly focused on assessing the alterations to neurons associated with dementia, and those to the circuits in which they are involved. In most of the studies on human brains and in many models of AD, the glial cells accompanying these neurons undergo concomitant alterations that aggravate the course of neurodegeneration. As a result, these changes to neuroglial cells are now included in all the "pathogenic cascades" described in AD. Accordingly, astrogliosis and microgliosis, the main components of neuroinflammation, have been integrated into all the pathogenic theories of this disease, as discussed in this part of the two-part monograph that follows an accompanying article on gliopathogenesis and glioprotection. This initial reflection verified the implication of alterations to the neuroglia in AD, suggesting that these cells may also represent therapeutic targets to prevent neurodegeneration. In this second part of the monograph, we will analyze the possibilities of acting on glial cells to prevent or treat the neurodegeneration that is the hallmark of AD and other pathologies. Evidence of the potential of different pharmacological, non-pharmacological, cell and gene therapies (widely treated) to prevent or treat this disease is now forthcoming, in most cases as adjuncts to other therapies. A comprehensive AD multimodal therapy is proposed in which neuronal and neuroglial pharmacological treatments are jointly considered, as well as the use of new cell and gene therapies and non-pharmacological therapies that tend to slow down the progress of dementia.
Collapse
|
16
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
17
|
Tag SH, Kim B, Bae J, Chang KA, Im HI. Neuropathological and behavioral features of an APP/PS1/MAPT (6xTg) transgenic model of Alzheimer’s disease. Mol Brain 2022; 15:51. [PMID: 35676711 PMCID: PMC9175339 DOI: 10.1186/s13041-022-00933-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/14/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease is associated with various brain dysfunctions, including memory impairment, neuronal loss, astrocyte activation, amyloid-β plaques, and neurofibrillary tangles. Transgenic animal models of Alzheimer's disease have proven to be invaluable for the basic research of Alzheimer's disease. However, Alzheimer's disease mouse models developed so far do not fully recapitulate the pathological and behavioral features reminiscent of Alzheimer's disease in humans. Here, we investigated the neurobehavioral sequelae in the novel 6xTg mouse model of Alzheimer's disease, which was developed by incorporating human tau containing P301L mutation in the widely used 5xFAD mouse model of Alzheimer's disease. At 11-months-old, 6xTg mice displayed the core pathological processes found in Alzheimer's disease, including accumulation of amyloid-β plaque, extensive neuronal loss, elevated level of astrocyte activation, and abnormal tau phosphorylation in the brain. At 9 to 11-months-old, 6xTg mice exhibited both cognitive and non-cognitive behavioral impairments relevant to Alzheimer’s disease, including memory loss, hyperlocomotion, anxiety-like behavior, depression-like behavior, and reduced sensorimotor gating. Our data suggest that the aged 6xTg mouse model of Alzheimer's disease presents pathological and cognitive-behavioral features reminiscent of Alzheimer's disease in humans. Thus, the 6xTg mouse model of Alzheimer's disease may be a valuable model for studying Alzheimer’s disease-relevant non-cognitive behaviors.
Collapse
|
18
|
Han G, Zhen W, Dai Y, Yu H, Li D, Ma T. Dihuang-Yinzi Alleviates Cognition Deficits via Targeting Energy-Related Metabolism in an Alzheimer Mouse Model as Demonstrated by Integration of Metabolomics and Network Pharmacology. Front Aging Neurosci 2022; 14:873929. [PMID: 35431901 PMCID: PMC9011333 DOI: 10.3389/fnagi.2022.873929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Energy metabolism disturbance and the consequent reactive oxygen species (ROS) overproduction play a key and pathogenic role in the onset and progression of Alzheimer’s disease (AD). Dihuang-Yinzi (DHYZ) is a traditional Chinese herbal prescription clinically applied to treat AD and other neurodegenerative diseases for a long time. However, the systematical metabolic mechanism of DHYZ against AD remains largely unclear. Here we aimed to explore the mechanism of DHYZ in the treatment of AD comprehensively in an in vivo metabolic context by performing metabolomics analysis coupled with network pharmacology study and experimental validation. The network pharmacology was applied to dig out the potential target of DHYZ against AD. The metabolomics analysis based on UPLC-HRMS was carried out to profile the urine of 2× Tg-AD mice treated with DHYZ. By integrating network pharmacology and metabolomics, we found DHYZ could ameliorate 4 key energy-related metabolic pathways, including glycerophospholipid metabolism, nicotinate/nicotinamide metabolism, glycolysis, and tricarboxylic acid cycle. Besides, we identified 5 potential anti-AD targets of DHYZ, including DAO, HIF1A, PARP1, ALDH3B2, and ACHE, and 14 key differential metabolites involved in the 4 key energy-related metabolic pathways. Furthermore, DHYZ depressed the mitochondrial dysfunction and the resultant ROS overproduction through ameliorating glycerophospholipid metabolism disturbance. Thereby DHYZ increased nicotinamide adenine dinucleotide (NAD+) content and promoted glycolysis and tricarboxylic acid (TCA) cycle, and consequently improved oxidative phosphorylation and energy metabolism. In the present study, we provided a novel, comprehensive and systematic insight into investigating the therapeutic efficacy of DHYZ against AD via ameliorating energy-related metabolism.
Collapse
Affiliation(s)
- Guanghui Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weizhe Zhen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Dai
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongni Yu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyue Li
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Ma
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Tao Ma,
| |
Collapse
|
19
|
Modulatory Properties of Food and Nutraceutical Components Targeting NLRP3 Inflammasome Activation. Nutrients 2022; 14:nu14030490. [PMID: 35276849 PMCID: PMC8840562 DOI: 10.3390/nu14030490] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 12/27/2022] Open
Abstract
Inflammasomes are key intracellular multimeric proteins able to initiate the cellular inflammatory signaling pathway. NLRP3 inflammasome represents one of the main protein complexes involved in the development of inflammatory events, and its activity has been largely demonstrated to be connected with inflammatory or autoinflammatory disorders, including diabetes, gouty arthritis, liver fibrosis, Alzheimer’s disease, respiratory syndromes, atherosclerosis, and cancer initiation. In recent years, it has been demonstrated how dietary intake and nutritional status represent important environmental elements that can modulate metabolic inflammation, since food matrices are an important source of several bioactive compounds. In this review, an updated status of knowledge regarding food bioactive compounds as NLRP3 inflammasome modulators is discussed. Several chemical classes, namely polyphenols, organosulfurs, terpenes, fatty acids, proteins, amino acids, saponins, sterols, polysaccharides, carotenoids, vitamins, and probiotics, have been shown to possess NLRP3 inflammasome-modulating activity through in vitro and in vivo assays, mainly demonstrating an anti-NLRP3 inflammasome activity. Plant foods are particularly rich in important bioactive compounds, each of them can have different effects on the pathway of inflammatory response, confirming the importance of the nutritional pattern (food model) as a whole rather than any single nutrient or functional compound.
Collapse
|
20
|
Chaudhary S, Zhornitsky S, Chao HH, van Dyck CH, Li CSR. Emotion Processing Dysfunction in Alzheimer's Disease: An Overview of Behavioral Findings, Systems Neural Correlates, and Underlying Neural Biology. Am J Alzheimers Dis Other Demen 2022; 37:15333175221082834. [PMID: 35357236 PMCID: PMC9212074 DOI: 10.1177/15333175221082834] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
We described behavioral studies to highlight emotional processing deficits in Alzheimer's disease (AD). The findings suggest prominent deficit in recognizing negative emotions, pronounced effect of positive emotion on enhancing memory, and a critical role of cognitive deficits in manifesting emotional processing dysfunction in AD. We reviewed imaging studies to highlight morphometric and functional markers of hippocampal circuit dysfunction in emotional processing deficits. Despite amygdala reactivity to emotional stimuli, hippocampal dysfunction conduces to deficits in emotional memory. Finally, the reviewed studies implicating major neurotransmitter systems in anxiety and depression in AD supported altered cholinergic and noradrenergic signaling in AD emotional disorders. Overall, the studies showed altered emotions early in the course of illness and suggest the need of multimodal imaging for further investigations. Particularly, longitudinal studies with multiple behavioral paradigms translatable between preclinical and clinical models would provide data to elucidate the time course and underlying neurobiology of emotion processing dysfunction in AD.
Collapse
Affiliation(s)
- Shefali Chaudhary
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Simon Zhornitsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Herta H. Chao
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA,VA Connecticut Healthcare System, West Haven, CT, USA
| | - Christopher H. van Dyck
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA,Wu Tsai Institute, Yale University, New Haven, CT, USA
| |
Collapse
|
21
|
Bortz J, Klatt KC, Wallace TC. Perspective: Estrogen and the Risk of Cognitive Decline: A Missing Choline(rgic) Link? Adv Nutr 2021; 13:S2161-8313(22)00068-0. [PMID: 34849527 PMCID: PMC8970832 DOI: 10.1093/advances/nmab145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Factors that influence the risk of neurocognitive decline and Alzheimer's disease (AD) may provide insight into therapies for both disease treatment and prevention. While age is the most striking risk factor for AD, it is notable that the prevalence of AD is higher in women, representing two-thirds of cases. To explore potential underlying biological underpinnings of this observation, the intent of this article is to explore the interplay between cognitive aging and sex hormones, the cholinergic system, and novel hypotheses related to the essential nutrient, choline. Mechanistic evidence points toward estrogen's neuroprotective effects being strongly dependent on its interactions with the cholinergic system, a modulator of attentional functioning, learning, and memory. Estrogen has been shown to attenuate anticholinergic-induced impairments in verbal memory and normalize patterns of frontal and occipital cortex activation, resulting in a more "young adult" phenotype. However, similar to estrogen replacement's effect in cardiovascular diseases, its putative protective effects may be restricted to early postmenopausal women only, supportive of the "critical window hypothesis." Estrogen's impact on the cholinergic system may act both locally in the brain but also through peripheral tissues. Estrogen is critical for inducing endogenous choline synthesis via the phosphatidylethanolamine N-methyltransferase (PEMT) pathway of phosphatidylcholine (PC) synthesis. PEMT is dramatically induced in response to estrogen, producing not only a PC molecule and source of choline for the brain but also a key source of the long-chain omega-3 fatty acid, DHA. Herein, we highlight novel hypotheses related to hormone replacement therapy and nutrient metabolism aimed at directing future preclinical and clinical investigation.
Collapse
|
22
|
Emodin derivatives with multi-factor anti-AD activities: AChE inhibitor, anti-oxidant and metal chelator. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Cui W, Fu W, Lin Y, Zhang T. Application of Nanomaterials in Neurodegenerative Diseases. Curr Stem Cell Res Ther 2021; 16:83-94. [PMID: 32213159 DOI: 10.2174/1574888x15666200326093410] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/07/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease are very harmful brain lesions. Due to the difficulty in obtaining therapeutic drugs, the best treatment for neurodegenerative diseases is often not available. In addition, the bloodbrain barrier can effectively prevent the transfer of cells, particles and macromolecules (such as drugs) in the brain, resulting in the failure of the traditional drug delivery system to provide adequate cellular structure repair and connection modes, which are crucial for the functional recovery of neurodegenerative diseases. Nanomaterials are designed to carry drugs across the blood-brain barrier for targets. Nanotechnology uses engineering materials or equipment to interact with biological systems at the molecular level to induce physiological responses through stimulation, response and target site interactions, while minimizing the side effects, thus revolutionizing the treatment and diagnosis of neurodegenerative diseases. Some magnetic nanomaterials play a role as imaging agents or nanoprobes for Magnetic Resonance Imaging to assist in the diagnosis of neurodegenerative diseases. Although the current research on nanomaterials is not as useful as expected in clinical applications, it achieves a major breakthrough and guides the future development direction of nanotechnology in the application of neurodegenerative diseases. This review briefly discusses the application and advantages of nanomaterials in neurodegenerative diseases. Data for this review were identified by searches of PubMed, and references from relevant articles published in English between 2015 and 2019 using the search terms "nanomaterials", "neurodegenerative diseases" and "blood-brain barrier".
Collapse
Affiliation(s)
- Weitong Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Fu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Gámiz F, Gallo M. A Systematic Review of the Dietary Choline Impact on Cognition from a Psychobiological Approach: Insights from Animal Studies. Nutrients 2021; 13:nu13061966. [PMID: 34201092 PMCID: PMC8229126 DOI: 10.3390/nu13061966] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
The influence of dietary choline availability on cognition is currently being suggested by animal and human studies which have focused mainly on the early developmental stages. The aim of this review is to systematically search through the available rodent (rats and mice) research published during the last two decades that has assessed the effect of dietary choline interventions on cognition and related attentional and emotional processes for the entire life span. The review has been conducted according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement guidelines covering peer-reviewed studies included in PubMed and Scopus databases. After excluding duplicates and applying the inclusion/exclusion criteria we have reviewed a total of 44 articles published in 25 journals with the contribution of 146 authors. The results are analyzed based on the timing and duration of the dietary intervention and the behavioral tests applied, amongst other variables. Overall, the available results provide compelling support for the relevance of dietary choline in cognition. The beneficial effects of choline supplementation is more evident in recognition rather than in spatial memory tasks when assessing nonpathological samples whilst these effects extend to other relational memory tasks in neuropathological models. However, the limited number of studies that have evaluated other cognitive functions suggest a wider range of potential effects. More research is needed to draw conclusions about the critical variables and the nature of the impact on specific cognitive processes. The results are discussed on the terms of the theoretical framework underlying the relationship between the brain systems and cognition.
Collapse
|
25
|
Burgaletto C, Di Benedetto G, Munafò A, Bernardini R, Cantarella G. Beneficial Effects of Choline Alphoscerate on Amyloid-β Neurotoxicity in an In vitro Model of Alzheimer's Disease. Curr Alzheimer Res 2021; 18:298-309. [PMID: 34102970 DOI: 10.2174/1567205018666210608093658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/17/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of neurodegenerative disorder characterized by cognitive impairment, which represents an urgent public health concern. Given the worldwide impact of AD, there is a compelling need for effective therapies to slow down or halt this disorder. OBJECTIVE Choline alphoscerate (α-GPC) represents a potentially effective cholinergic neurotrans- mission enhancing agent with an interesting clinical profile in cognitive dysfunctions improve- ment, although only scanty data are available about the mechanisms underlying such beneficial ef- fects. METHOD The SH-SY5Y neuronal cell line, differentiated for 1 week with 10 μm of all-trans-reti- noic acid (RA), to achieve a switch towards a cholinergic phenotype, was used as an in vitro model of AD. SH-SY5Y cells were pre-treated for 1h with α-GPC (100nM) and treated for 72 h with Aβ25-35 (10μM). RESULTS α-GPC was able to antagonize Aβ25-35 mediated neurotoxicity and attenuate the Aβ-in- duced phosphorylation of the Tau protein. Moreover, α-GPC exerted its beneficial effects by em- ploying the NGF/TrkA system, knocked down in AD and, consequently, by sustaining the expres- sion level of synaptic vesicle proteins, such as synaptophysin. CONCLUSION Taken together, our data suggest that α-GPC can have a role in neuroprotection in the course of toxic challenges with Aβ. Thus, a deeper understanding of the mechanism underlying its beneficial effect, could provide new insights into potential future pharmacological applications of its functional cholinergic enhancement, with the aim to mitigate AD and could represent the basis for innovative therapy.Recent Advances in Anti-Infective Drug Discovery.
Collapse
Affiliation(s)
- Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Antonio Munafò
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| |
Collapse
|
26
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
27
|
Yang M, Xuan Z, Wang Q, Yan S, Zhou D, Naman CB, Zhang J, He S, Yan X, Cui W. Fucoxanthin has potential for therapeutic efficacy in neurodegenerative disorders by acting on multiple targets. Nutr Neurosci 2021; 25:2167-2180. [PMID: 33993853 DOI: 10.1080/1028415x.2021.1926140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Fucoxanthin, one of the most abundant carotenoids from edible brown seaweeds, for years has been used as a bioactive dietary supplement and functional food ingredient. Recently, fucoxanthin was reported to penetrate the blood-brain barrier, and was superior to other carotenoids to exert anti-neurodegenerative disorder effects via acting on multiple targets, including amyloid protein aggregation, oxidative stress, neuroinflammation, neuronal loss, neurotransmission dysregulation and gut microbiota disorder. However, the concentration of fucoxanthin required for in vivo neuroprotective effects is somewhat high, and the poor bioavailability of this molecule might prevent its clinical use. As such, new strategies have been introduced to overcome these obstacles, and may help to develop fucoxanthin as a novel lead for neurodegenerative disorders. Moreover, it has been shown that some metabolites of fucoxanthin may produce potent in vivo neuroprotective effects. Altogether, these studies suggest the possibility for future development of fucoxanthin as a one-compound-multiple-target or pro-drug type pharmaceutical or nutraceutical treatment for neurodegenerative disorders.Trial registration: ClinicalTrials.gov identifier: NCT03625284.Trial registration: ClinicalTrials.gov identifier: NCT02875392.Trial registration: ClinicalTrials.gov identifier: NCT03613740.Trial registration: ClinicalTrials.gov identifier: NCT04761406.
Collapse
Affiliation(s)
- Mengxiang Yang
- Ningbo Kangning Hospital, Ningbo, People's Republic of China.,Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| | - Zhenquan Xuan
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| | - Qiyao Wang
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| | - Sicheng Yan
- Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| | - Dongsheng Zhou
- Ningbo Kangning Hospital, Ningbo, People's Republic of China
| | - C Benjamin Naman
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, People's Republic of China
| | - Jinrong Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, People's Republic of China
| | - Shan He
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, People's Republic of China
| | - Xiaojun Yan
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, People's Republic of China.,Laboratory of Seafood Processing, Innovative and Application Institute, Zhejiang Ocean University, Zhoushan, People's Republic of China
| | - Wei Cui
- Ningbo Kangning Hospital, Ningbo, People's Republic of China.,Translational Medicine Center of Pain, Emotion and Cognition, Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, People's Republic of China
| |
Collapse
|
28
|
Crews FT, Fisher R, Deason C, Vetreno RP. Loss of Basal Forebrain Cholinergic Neurons Following Adolescent Binge Ethanol Exposure: Recovery With the Cholinesterase Inhibitor Galantamine. Front Behav Neurosci 2021; 15:652494. [PMID: 33716687 PMCID: PMC7953159 DOI: 10.3389/fnbeh.2021.652494] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Binge drinking and alcohol abuse are common during adolescence and cause both cognitive deficits and lasting cholinergic pathology in the adult basal forebrain. Acetylcholine is anti-inflammatory and studies using the preclinical adolescent intermittent ethanol (AIE; 5.0 g/kg, i.g., 2 day on/2 day off from postnatal day [P]25 to P54) model of human adolescent binge drinking report decreased basal forebrain cholinergic neurons (BFCNs) and induction of proinflammatory genes that persist long into adulthood. Recent studies link AIE-induced neuroimmune activation to cholinergic pathology, but the underlying mechanisms contributing to the persistent loss of BFCNs are unknown. We report that treatment with the cholinesterase inhibitor galantamine (4.0 mg/kg, i.p.) administered during AIE (i.e., P25-P54) or following the conclusion of AIE (i.e., P57-P72) recovered the persistent loss of cholinergic neuron phenotype markers (i.e., ChAT, TrkA, and p75NTR) and somal shrinkage of residual ChAT + neurons known to persist in AIE-exposed adults. Galantamine treatment also recovered the AIE-increased expression of the proinflammatory receptors TLR4 and RAGE, the endogenous TLR4/RAGE agonist HMGB1, and the transcription activation marker pNF-κB p65. Interestingly, we find BFCNs express TLR4 and RAGE, and that AIE treatment increased pNF-κB p65 expression in adult ChAT + IR neurons, consistent with intracellular HMGB1-TLR4/RAGE signaling within BFCNs. AIE increased epigenetic transcription silencing markers (i.e., H3K9me2 and H3K9me3) in the adult basal forebrain and H3K9me2 occupancy at cholinergic phenotype gene promoters (i.e., ChAT and TrkA). The finding of no AIE-induced changes in total basal forebrain NeuN + neurons with galantamine reversal of AIE-induced ChAT + neuron loss, TLR4/RAGE-pNF-κB p65 signals, and epigenetic transcription silencing markers suggests that AIE does not cause cell death, but rather the loss of the cholinergic phenotype. Together, these data suggest that AIE induces HMGB1-TLR4/RAGE-pNF-κB p65 signals, causing the loss of cholinergic phenotype (i.e., ChAT, TrkA, and p75NTR) through epigenetic histone transcription silencing that result in the loss of the BFCN phenotype that can be prevented and restored by galantamine.
Collapse
Affiliation(s)
- Fulton T. Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachael Fisher
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chloe Deason
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ryan P. Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
29
|
Toljan K, Homolak J. Circadian changes in Alzheimer's disease: Neurobiology, clinical problems, and therapeutic opportunities. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:285-300. [PMID: 34225969 DOI: 10.1016/b978-0-12-819975-6.00018-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The understanding of Alzheimer's disease (AD) pathophysiology is an active area of research, and the traditional focus on hippocampus, amyloid and tau protein, and memory impairment has been expanded with components like neuroinflammation, insulin resistance, and circadian rhythm alterations. The bidirectional vicious cycle of neuroinflammation and neurodegeneration on a molecular level may cause functional deficits already long before the appearance of overt clinical symptoms. Located at the crossroads of metabolic, circadian, and hormonal signaling, the hypothalamus has been identified as another brain region affected by AD pathophysiology. Current findings on hypothalamic dysfunction open a broader horizon for studying AD pathogenesis and offer new opportunities for diagnosis and therapy. While treatments with cholinomimetics and memantine form a first line of pharmacological treatment, additional innovative research is pursued toward the development of antiinflammatory, growth factor, or antidiabetic types of medication. Following recent epidemiological data showing associations of AD incidence with modern societal and "life-style"-related risk factors, also nonpharmacological interventions, including sleep optimization, are being developed and some have been shown to be beneficial. Circadian aspects in AD are relevant from a pathophysiological standpoint, but they can also have an important role in pharmacologic and nonpharmacologic interventions, and appropriate timing of sleep, meals, and medication may boost therapeutic efficacy.
Collapse
Affiliation(s)
- Karlo Toljan
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States.
| | - Jan Homolak
- Department of Pharmacology, and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
30
|
Xu P, Ning J, Jiang Q, Li C, Yan J, Zhao L, Gao H, Zheng H. Region-specific metabolic characterization of the type 1 diabetic brain in mice with and without cognitive impairment. Neurochem Int 2020; 143:104941. [PMID: 33333211 DOI: 10.1016/j.neuint.2020.104941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/08/2020] [Accepted: 12/12/2020] [Indexed: 11/28/2022]
Abstract
Type 1 diabetes (T1D) has been reported to cause cognitive decline, but brain metabolic changes during this process are still far from being fully understood. Here, we found that streptozotocin (STZ)-induced T1D mice exhibited impaired learning and memory at 11 weeks after STZ treatment but not at 3 weeks. Therefore, we studied metabolic alterations in six different brain regions of T1D mice with and without cognitive decline, and attempted to identify key metabolic pathways related to diabetic cognitive dysfunction. The results demonstrate that lactate had already increased in all brain regions of T1D mice prior to cognitive decline, but a decreased TCA cycle was only observed in hippocampus, cortex and striatum of T1D mice with cognitive impairment. Reduced N-acetylaspartate and choline were found in all brain regions of T1D mice, irrespective of cognitive decline. In addition, disrupted neurotransmitter metabolism was noted to occur in T1D mice before cognitive deficit. Of note, we found that the level of uridine was significantly reduced in cerebellum, cortex, hypothalamus and midbrain of T1D mice when cognitive decline was presented. Therefore, brain region-specific metabolic alterations may comprise possible biomarkers for the early-diagnosis and monitoring of diabetic cognitive decline. Moreover, down-regulated TCA cycle and pyrimidine metabolism could be closely related to T1D-associated cognitive impairment.
Collapse
Affiliation(s)
- Pengtao Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jie Ning
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qiaoying Jiang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen Li
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junjie Yan
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
31
|
Catanesi M, d'Angelo M, Antonosante A, Castelli V, Alfonsetti M, Benedetti E, Desideri G, Ferri C, Cimini A. Neuroprotective potential of choline alfoscerate against β-amyloid injury: Involvement of neurotrophic signals. Cell Biol Int 2020; 44:1734-1744. [PMID: 32343461 DOI: 10.1002/cbin.11369] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/10/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease represents the most prevalent neurodegeneration worldwide, clinically characterized by cognitive and memory impairment. New therapeutic approaches are extremely important to counteract this disorder. This research is focused on the potential use of choline alfoscerate in preventing neuronal death using in vitro models of Alzheimer's disease, representing the early stage of the disease, treated before or after the insult with glycerylphosphorylcholine. On the light of the results collected, we can postulate that choline alfoscerate, by the activation of the neurotrophin survival pathway, was able to counteract the detrimental effect of β-amyloid in both in vitro models, reducing apoptotic cell death and preserving the neuronal morphology.
Collapse
Affiliation(s)
- Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Margherita Alfonsetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giovambattista Desideri
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Claudio Ferri
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Choline as a prevention for Alzheimer's disease. Aging (Albany NY) 2020; 12:2026-2027. [PMID: 32039834 PMCID: PMC7041773 DOI: 10.18632/aging.102849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/06/2020] [Indexed: 11/25/2022]
|
33
|
Alzheimer's Disease Pharmacotherapy in Relation to Cholinergic System Involvement. Biomolecules 2019; 10:biom10010040. [PMID: 31888102 PMCID: PMC7022522 DOI: 10.3390/biom10010040] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease, a major and increasing global health challenge, is an irreversible, progressive form of dementia, associated with an ongoing decline of brain functioning. The etiology of this disease is not completely understood, and no safe and effective anti-Alzheimer’s disease drug to prevent, stop, or reverse its evolution is currently available. Current pharmacotherapy concentrated on drugs that aimed to improve the cerebral acetylcholine levels by facilitating cholinergic neurotransmission through inhibiting cholinesterase. These compounds, recognized as cholinesterase inhibitors, offer a viable target across key sign domains of Alzheimer’s disease, but have a modest influence on improving the progression of this condition. In this paper, we sought to highlight the current understanding of the cholinergic system involvement in Alzheimer’s disease progression in relation to the recent status of the available cholinesterase inhibitors as effective therapeutics.
Collapse
|