1
|
Patil S, Ayubcha C, Teichner E, Subtirelu R, Cho JH, Ghonim M, Ghonim M, Werner TJ, Høilund-Carlsen PF, Alavi A, Newberg AB. Clinical Applications of PET Imaging in Alzheimer's Disease. PET Clin 2025; 20:89-100. [PMID: 39547733 DOI: 10.1016/j.cpet.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) involves a complex pathophysiology of neurodegeneration that leads to severe cognitive deficiencies. Understanding the molecular alterations that underlie this disease is fundamental to clinical management and therapeutic innovation. Functional imaging with positron emission tomography (PET) enables a visualization of these impaired pathways, such as cerebral hypometabolism, amyloid and tau accumulation, and neurotransmitter dysfunction. This review discusses the clinical applications of PET in AD and mild cognitive impairment, focusing on relevant original research studies for disease diagnosis, progression, and treatment response.
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric Teichner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Mohanad Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | - Andrew B Newberg
- Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Sims SL, Frazier HN, Case SL, Lin RL, Trosper JN, Vekaria HJ, Sullivan PG, Thibault O. Variable bioenergetic sensitivity of neurons and astrocytes to insulin and extracellular glucose. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:33. [PMID: 39524535 PMCID: PMC11549053 DOI: 10.1038/s44324-024-00037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Energy flow within cellular elements of the brain is a well-orchestrated, tightly regulated process, however, details underlying these functions at the single-cell level are still poorly understood. Studying hypometabolism in aging and neurodegenerative diseases may benefit from experimentation on unicellular bioenergetics. Here, we examined energy status in neurons and astrocytes using mixed hippocampal cultures and PercevalHR, an ATP:ADP nanosensor. We assessed exposures of several compounds including KCl, glutamate, FCCP, insulin, and glucose. A mitochondrial stress test was performed, and PercevalHR's fluorescence was corrected for pH using pHrodo. Results demonstrate that PercevalHR can reliably report on the energetic status of two cell types that communicate in a mixed-culture setting. While KCl, glutamate, and FCCP showed clear changes in PercevalHR fluorescence, insulin and glucose responses were found to be more subtle and sensitive to extracellular glucose. These results may highlight mechanisms that mediate insulin sensitivity in the brain.
Collapse
Affiliation(s)
- Sophiya L. Sims
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hilaree N. Frazier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| | - Sami L. Case
- Department of Biomedical Sciences, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - James N. Trosper
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| |
Collapse
|
3
|
Ye Y, Fu C, Li Y, Sun J, Li X, Chai S, Li S, Hou M, Cai H, Wang Z, Wu M. Alternate-day fasting improves cognitive and brain energy deficits by promoting ketone metabolism in the 3xTg mouse model of Alzheimer's disease. Exp Neurol 2024; 381:114920. [PMID: 39142368 DOI: 10.1016/j.expneurol.2024.114920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/10/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is characterized by disorders in brain energy. The lack of sufficient energy for nerve function leads to cognitive dysfunction and massive neuronal loss in AD. Ketone bodies are an alternative to glucose as a source of energy in the brain, and alternate-day fasting (ADF) promotes the production of the ketone body β-hydroxybutyric acid (βOHB). In this study, 7-month-old male WT mice and 3xTg mice underwent dietary control for 20 weeks. We found that ADF increased circulating βOHB concentrations in 3xTg mice, improved cognitive function, reduced anxiety-like behaviors, improved hippocampal synaptic plasticity, and reduced neuronal loss, Aβ oligomers and tau hyperphosphorylation. In addition, ADF improved mitochondrial bioenergetic function by promoting brain ketone metabolism and rescued brain energy deficits in 3xTg mice. A safety evaluation showed that ADF improved exercise endurance and liver and kidney function in 3xTg mice without negatively affecting muscle motor and heart functions. This study provides a theoretical basis and strong support for the application of ADF as a non-drug strategy for preventing and treating brain energy defects in the early stage of AD.
Collapse
Affiliation(s)
- Yucai Ye
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Chaojing Fu
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Yan Li
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan 030001, China; Department of Toxicology, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Junli Sun
- School of Anesthesiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xinru Li
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Shifan Chai
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China
| | - Shuo Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Meng Hou
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, China
| | - Hongyan Cai
- Department of Microbiology and Immunology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Zhaojun Wang
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China.
| | - Meina Wu
- Department of Physiology, School of Basic Medicine, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Taiyuan 030001, China.
| |
Collapse
|
4
|
Dhar KS, Townsend B, Montgomery AP, Danon JJ, Pagan JK, Kassiou M. Enhancing CNS mitophagy: drug development and disease-relevant models. Trends Pharmacol Sci 2024; 45:982-996. [PMID: 39419743 DOI: 10.1016/j.tips.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Mitophagy, the selective degradation of mitochondria, is impaired in many neurodegenerative diseases (NDs), resulting in an accumulation of dysfunctional mitochondria and neuronal damage. Although enhancing mitophagy shows promise as a therapeutic strategy, the clinical significance of mitophagy activators remains uncertain due to limited understanding and poor representation of mitophagy in the central nervous system (CNS). This review explores recent insights into which mitophagy pathways to target and the extent of modulation necessary to be therapeutic towards NDs. We also highlight the complexities of mitophagy in the CNS, highlighting the need for disease-relevant models. Last, we outline crucial aspects of in vitro models to consider during drug discovery, aiming to bridge the gap between preclinical research and clinical applications in treating NDs through mitophagy modulation.
Collapse
Affiliation(s)
- Krishayant S Dhar
- School of Chemistry, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Brendan Townsend
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Andrew P Montgomery
- School of Chemistry, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan J Danon
- School of Chemistry, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Julia K Pagan
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Michael Kassiou
- School of Chemistry, Faculty of Science, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
5
|
Yang X, Chen YH, Liu L, Gu Z, You Y, Hao JR, Sun N, Gao C. Regulation of glycolysis-derived L-lactate production in astrocytes rescues the memory deficits and Aβ burden in early Alzheimer's disease models. Pharmacol Res 2024; 208:107357. [PMID: 39159732 DOI: 10.1016/j.phrs.2024.107357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/17/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024]
Abstract
Aberrant energy metabolism in the brain is a common pathological feature in the preclinical Alzheimer's Disease (AD). Recent studies have reported the early elevations of glycolysis-involved enzymes in AD brain and cerebrospinal fluid according to a large-scale proteomic analysis. It's well-known that astrocytes exhibit strong glycolytic metabolic ability and play a key role in the regulation of brain homeostasis. However, its relationship with glycolytic changes and cognitive deficits in early AD patients is unclear. Here, we investigated the mechanisms by which astrocyte glycolysis is involved in early AD and its potential as a therapeutic target. Our results suggest that Aβ-activated microglia can induce glycolytic-enhanced astrocytes in vitro, and that these processes are dependent on the activation of the AKT-mTOR-HIF-1α pathway. In early AD models, the increase in L-lactate produced by enhanced glycolysis of astrocytes leads to spatial cognitive impairment by disrupting synaptic plasticity and accelerating Aβ aggregation. Furthermore, we find rapamycin, the mTOR inhibitor, can rescue the impaired spatial memory and Aβ burden by inhibiting the glycolysis-derived L-lactate in the early AD models. In conclusion, we highlight that astrocytic glycolysis plays a critical role in the early onset of AD and that the modulation of glycolysis-derived L-lactate by rapamycin provides a new strategy for the treatment of AD.
Collapse
Affiliation(s)
- Xiu Yang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yuan-Hao Chen
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Le Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Zheng Gu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yue You
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jing-Ru Hao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Can Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
6
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:6844-6859. [PMID: 39171353 PMCID: PMC11485302 DOI: 10.1002/alz.14144] [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: 02/05/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Reduced brain energy metabolism, mammalian target of rapamycin (mTOR) dysregulation, and extracellular amyloid beta (Aβ) oligomer (xcAβO) buildup are some well-known Alzheimer's disease (AD) features; how they promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit nutrient-induced mitochondrial activity (NiMA) in cultured neurons. We now report NiMA disruption in vivo. METHODS Brain energy metabolism and oxygen consumption were recorded in heterozygous amyloid precursor protein knock-in (APPSAA) mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. RESULTS NiMA is inhibited in APPSAA mice before other defects are detected in these Aβ-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. Glycogen synthase kinase 3 (GSK3β) signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APPSAA mice. DISCUSSION NiMA disruption in vivo occurs before plaques, neuroinflammation, and cognitive decline in APPSAA mice, and may represent an early stage in human AD. HIGHLIGHTS Amyloid beta blocks communication between lysosomes and mitochondria in vivo. Nutrient-induced mitochondrial activity (NiMA) is disrupted long before the appearance of Alzheimer's disease (AD) histopathology in heterozygous amyloid precursor protein knock-in (APPSAA/+) mice. NiMA is disrupted long before learning and memory deficits in APPSAA/+ mice. Pharmacological interventions can rescue AD-related NiMA disruption in vivo.
Collapse
Affiliation(s)
- Andrés Norambuena
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Vijay Kumar Sagar
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Zhuoying Wang
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Prakash Raut
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Ziang Feng
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Horst Wallrabe
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Evelyn Pardo
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Taylor Kim
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Shagufta Rehman Alam
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Song Hu
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Ammasi Periasamy
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- W.M. Keck Center for Cellular ImagingUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - George S. Bloom
- Department of BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
7
|
Ameen AO, Nielsen SW, Kjær MW, Andersen JV, Westi EW, Freude KK, Aldana BI. Metabolic preferences of astrocytes: Functional metabolic mapping reveals butyrate outcompetes acetate. J Cereb Blood Flow Metab 2024:271678X241270457. [PMID: 39340267 PMCID: PMC11563520 DOI: 10.1177/0271678x241270457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/11/2024] [Accepted: 07/02/2024] [Indexed: 09/30/2024]
Abstract
Disruptions to the gut-brain-axis have been linked to neurodegenerative disorders. Of these disruptions, reductions in the levels of short-chain fatty acids (SCFAs), like butyrate, have been observed in mouse models of Alzheimer's disease (AD). Butyrate supplementation in mice has shown promise in reducing neuroinflammation, amyloid-β accumulation, and enhancing memory. However, the underlying mechanisms remain unclear. To address this, we investigated the impact of butyrate on energy metabolism in mouse brain slices, primary cultures of astrocytes and neurons and in-vivo by dynamic isotope labelling with [U-13C]butyrate and [1,2-13C]acetate to map metabolism via mass spectrometry. Metabolic competition assays in cerebral cortical slices revealed no competition between butyrate and the ketone body, β-hydroxybutyrate, but competition with acetate. Astrocytes favoured butyrate metabolism compared to neurons, suggesting that the astrocytic compartment is the primary site of butyrate metabolism. In-vivo metabolism investigated in the 5xFAD mouse, an AD pathology model, showed no difference in 13C-labelling of TCA cycle metabolites between wild-type and 5xFAD brains, but butyrate metabolism remained elevated compared to acetate in both groups, indicating sustained uptake and metabolism in 5xFAD mice. Overall, these findings highlight the role of astrocytes in butyrate metabolism and the potential use of butyrate as an alternative brain fuel source.
Collapse
Affiliation(s)
- Aishat O Ameen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sebastian W Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin W Kjær
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil W Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine K Freude
- Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Moawad MHED, Serag I, Alkhawaldeh IM, Abbas A, Sharaf A, Alsalah S, Sadeq MA, Shalaby MMM, Hefnawy MT, Abouzid M, Meshref M. Exploring the Mechanisms and Therapeutic Approaches of Mitochondrial Dysfunction in Alzheimer's Disease: An Educational Literature Review. Mol Neurobiol 2024:10.1007/s12035-024-04468-y. [PMID: 39254911 DOI: 10.1007/s12035-024-04468-y] [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: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Alzheimer's disease (AD) presents a significant challenge to global health. It is characterized by progressive cognitive deterioration and increased rates of morbidity and mortality among older adults. Among the various pathophysiologies of AD, mitochondrial dysfunction, encompassing conditions such as increased reactive oxygen production, dysregulated calcium homeostasis, and impaired mitochondrial dynamics, plays a pivotal role. This review comprehensively investigates the mechanisms of mitochondrial dysfunction in AD, focusing on aspects such as glucose metabolism impairment, mitochondrial bioenergetics, calcium signaling, protein tau and amyloid-beta-associated synapse dysfunction, mitophagy, aging, inflammation, mitochondrial DNA, mitochondria-localized microRNAs, genetics, hormones, and the electron transport chain and Krebs cycle. While lecanemab is the only FDA-approved medication to treat AD, we explore various therapeutic modalities for mitigating mitochondrial dysfunction in AD, including antioxidant drugs, antidiabetic agents, acetylcholinesterase inhibitors (FDA-approved to manage symptoms), nutritional supplements, natural products, phenylpropanoids, vaccines, exercise, and other potential treatments.
Collapse
Affiliation(s)
- Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Abdulrahman Sharaf
- Department of Clinical Pharmacy, Salmaniya Medical Complex, Government Hospital, Manama, Bahrain
| | - Sumaya Alsalah
- Ministry of Health, Primary Care, Governmental Health Centers, Manama, Bahrain
| | | | | | | | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
9
|
Yang T, Duan H, Li Y, Xu N, Wang Z, Li Z, Chen Y, Du Y, Zhang M, Yan J, Sun C, Wang G, Li W, Li X, Ma F, Huang G. β-hydroxybutyrate and mitochondria mediate the association between medium-chain fatty acids, DHA and mild cognitive impairment: a nested case-control study. Nutr Neurosci 2024:1-10. [PMID: 39225171 DOI: 10.1080/1028415x.2024.2398364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
BACKGROUND Medium-chain fatty acids (MCFAs) and docosahexaenoic acid (DHA) could affect the occurrence of mild cognitive impairment (MCI). β-hydroxybutyrate (BHB), mitochondrial DNA copy number (mtDNAcn) and mitochondrial DNA (mtDNA) deletions might be their potential mechanisms. This study aimed to explore the relationship between MCFAs, DHA and MCI, and potential mechanisms. METHODS This study used data from Tianjin Elderly Nutrition and Cognition (TENC) cohort study, 120 individuals were identified with new onset MCI during follow-up, 120 individuals without MCI were selected by 1:1 matching sex, age, and education levels as the control group from TENC. Conditional logistic regression analysis and mediation effect analysis were used to explore their relationship. RESULTS Higher serum octanoic acid levels (OR: 0.633, 95% CI: 0.520, 0.769), higher serum DHA levels (OR: 0.962, 95% CI: 0.942, 0.981), and more mtDNAcn (OR: 0.436, 95% CI: 0.240, 0.794) were associated with lower MCI risk, while more mtDNA deletions was associated with higher MCI risk (OR: 8.833, 95% CI: 3.909, 19.960). Mediation analysis suggested that BHB and mtDNAcn, in series, have mediation roles in the association between octanoic acid and MCI risk, and mtDNA deletions have mediation roles in the association between DHA and MCI risk. CONCLUSION Higher serum octanoic acid and DHA levels were associated with lower MCI risk. Octanoic acid could affect the incidence of MCI through BHB, then mitochondria function, or through mitochondria function, or directly. Serum DHA level could affect the incidence of MCI through mitochondria function, or directly.
Collapse
Affiliation(s)
- Tong Yang
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
| | - Huilian Duan
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yuan Li
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Ning Xu
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zehao Wang
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhenshu Li
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yongjie Chen
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
| | - Yue Du
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Social Medicine and Health Management, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Meilin Zhang
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Jing Yan
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Social Medicine and Health Management, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Changqing Sun
- Neurosurgical Department of Baodi Clinical College of Tianjin Medical University, Tianjin, People's Republic of China
| | - Guangshun Wang
- Department of Tumor, Baodi Clinical College of Tianjin Medical University, Tianjin, People's Republic of China
| | - Wen Li
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
| | - Xin Li
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Fei Ma
- Department of Epidemiology & Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
| | - Guowei Huang
- Key Laboratory of Prevention and Control of Major Diseases in the Population, Ministry of Education, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, People's Republic of China
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin, People's Republic of China
| |
Collapse
|
10
|
Zhu Q, Long Q, Wei C, Chen J, Nong L, Qin J, Huang Z, Zheng Y, Li S. Lactate dehydrogenase-1 may play a key role in the brain energy disturbance caused by cryptococcal meningitis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024:S1684-1182(24)00153-1. [PMID: 39214781 DOI: 10.1016/j.jmii.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Cryptococcal meningitis (CM) may affect the conversion of lactate to pyruvate in the brain, resulting in abnormal levels of adenosine triphosphate (ATP) throughout the brain. Lactate conversion to pyruvate is mainly caused by lactic dehydrogenase 1 (LDH1), which is composed of four LDHB subunits. However, the underlying mechanism of LDH1 in CM remains unclear. METHODS Cerebrospinal fluid (CSF) from 17 patients was collected, including eight patients with non-infectious diseases of the central nervous system and nine patients with CM. Based on clinical data and laboratory reports, data regarding intracranial pressure, CSF white cell counts, lactate dehydrogenase (LDH), adenosine deaminase, glucose, protein, and chloridion were collected. Meanwhile, LDH1, LDH5, lactate, pyruvate, and ATP levels were detected in CSF. Whereafter, the levels of lactate, pyruvate, ATP, and the amplitude and frequency of action potentials in the neurons with low expression of LDHB were explored. RESULTS Intracranial pressure and white cell count in CSF were significantly increased in patients with CM. In patients with CM, the LDH1, pyruvate, and ATP levels in the CSF were significantly decreased, and the levels of lactate were found to be increased. Furthermore, pyruvate and ATP levels were decreased, while lactate was increased in the neurons with low expression of LDHB. The amplitude and frequency of APs in the neurons with low expression of LDHB were significantly decreased. CONCLUSION Reduced levels of LDH1 in the brain of patients with CM may lead to increased lactate levels, decreased pyruvate and ATP levels, and negatively affect neuronal activity.
Collapse
Affiliation(s)
- Qingdong Zhu
- Department of Tuberculosis, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Qian Long
- Key Laboratory of Infectious Diseases of Nanning Municipal Health Commission, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Cailing Wei
- Infectious Disease Laboratory, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Jieling Chen
- Infectious Disease Laboratory, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Lanwei Nong
- Infectious Disease Laboratory, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Jianglong Qin
- Department of Infectious Disease, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Zhizhong Huang
- Department of Liver, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China
| | - Yanqing Zheng
- Infectious Disease Laboratory, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China.
| | - Sijun Li
- Infectious Disease Laboratory, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China; Department of Internal Medicine, Guangxi AlDs Clinical Treatment Center (Nanning)/The Fourth People's Hospital of Nanning, Nanning, Guangxi, China.
| |
Collapse
|
11
|
Mei J, Li Y, Niu L, Liang R, Tang M, Cai Q, Xu J, Zhang D, Yin X, Liu X, Shen Y, Liu J, Xu M, Xia P, Ling J, Wu Y, Liang J, Zhang J, Yu P. SGLT2 inhibitors: a novel therapy for cognitive impairment via multifaceted effects on the nervous system. Transl Neurodegener 2024; 13:41. [PMID: 39123214 PMCID: PMC11312905 DOI: 10.1186/s40035-024-00431-y] [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: 03/14/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.
Collapse
Affiliation(s)
- Jiaqi Mei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Yi Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Liyan Niu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Huan Kui College of Nanchang University, Nanchang, China
| | - Ruikai Liang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyue Tang
- Queen Mary College of Nanchang University, Nanchang, China
| | - Qi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingdong Xu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xiao Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
12
|
Zhong X, Gong S, Meng L, Yao W, Du K, Jiao L, Ma G, Liang J, Wei B, Jin X, Tong J, Dong J, Liu M, Gao M, Jia H, Jiang W, Yu Z, Wang Y, Sun X, Wei M, Liu M. Cordycepin Modulates Microglial M2 Polarization Coupled with Mitochondrial Metabolic Reprogramming by Targeting HKII and PDK2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304687. [PMID: 38889331 PMCID: PMC11336950 DOI: 10.1002/advs.202304687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 05/11/2024] [Indexed: 06/20/2024]
Abstract
The microenvironment mediated by the microglia (MG) M1/M2 phenotypic switch plays a decisive role in the neuronal fate and cognitive function of Alzheimer's disease (AD). However, the impact of metabolic reprogramming on microglial polarization and its underlying mechanism remains elusive. This study reveals that cordycepin improved cognitive function and memory in APP/PS1 mice, as well as attenuated neuronal damage by triggering MG-M2 polarization and metabolic reprogramming characterized by increased OXPHOS and glycolysis, rather than directly protecting neurons. Simultaneously, cordycepin partially alleviates mitochondrial damage in microglia induced by inhibitors of OXPHOS and glycolysis, further promoting MG-M2 transformation and increasing neuronal survival. Through confirmation of cordycepin distribution in the microglial mitochondria via mitochondrial isolation followed by HPLC-MS/MS techniques, HKII and PDK2 are further identified as potential targets of cordycepin. By investigating the effects of HKII and PDK2 inhibitors, the mechanism through which cordycepin targeted HKII to elevate ECAR levels in the glycolysis pathway while targeting PDK2 to enhance OCR levels in PDH-mediated OXPHOS pathway, thereby inducing MG-M2 polarization, promoting neuronal survival and exerting an anti-AD role is elucidated.
Collapse
Affiliation(s)
- Xin Zhong
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Shiqiang Gong
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
- Liaoning Medical Diagnosis and Treatment CenterShenyangLiaoning11067China
| | | | - Weifan Yao
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
- Liaoning Medical Diagnosis and Treatment CenterShenyangLiaoning11067China
| | - Ke Du
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Linchi Jiao
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Guowei Ma
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Jingwei Liang
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Binbin Wei
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Xin Jin
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Junhui Tong
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Jianru Dong
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Mengyu Liu
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Menglin Gao
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Huachao Jia
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| | - Wenjuan Jiang
- The First Affiliated Hospital of China Medical UniversityShenyangLiaoning110002China
| | - Zhihua Yu
- The Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoning110165China
| | - Yanzhe Wang
- The First Affiliated Hospital of China Medical UniversityShenyangLiaoning110002China
| | - Xiaohong Sun
- Science Experiment CenterChina Medical UniversityShenyangLiaoning110122China
| | - Minjie Wei
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
- Liaoning Medical Diagnosis and Treatment CenterShenyangLiaoning11067China
| | - Mingyan Liu
- School of PharmacyChina Medical UniversityShenyangLiaoning110122China
| |
Collapse
|
13
|
Leclerc M, Tremblay C, Bourassa P, Schneider JA, Bennett DA, Calon F. Lower GLUT1 and unchanged MCT1 in Alzheimer's disease cerebrovasculature. J Cereb Blood Flow Metab 2024; 44:1417-1432. [PMID: 38441044 PMCID: PMC11342728 DOI: 10.1177/0271678x241237484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 03/06/2024]
Abstract
The brain is a highly demanding organ, utilizing mainly glucose but also ketone bodies as sources of energy. Glucose transporter-1 (GLUT1) and monocarboxylates transporter-1 (MCT1) respectively transport glucose and ketone bodies across the blood-brain barrier. While reduced glucose uptake by the brain is one of the earliest signs of Alzheimer's disease (AD), no change in the uptake of ketone bodies has been evidenced yet. To probe for changes in GLUT1 and MCT1, we performed Western immunoblotting in microvessel extracts from the parietal cortex of 60 participants of the Religious Orders Study. Participants clinically diagnosed with AD had lower cerebrovascular levels of GLUT1, whereas MCT1 remained unchanged. GLUT1 reduction was associated with lower cognitive scores. No such association was found for MCT1. GLUT1 was inversely correlated with neuritic plaques and cerebrovascular β-secretase-derived fragment levels. No other significant associations were found between both transporters, markers of Aβ and tau pathologies, sex, age at death or apolipoprotein-ε4 genotype. These results suggest that, while a deficit of GLUT1 may underlie the reduced transport of glucose to the brain in AD, no such impairment occurs for MCT1. This study thus supports the exploration of ketone bodies as an alternative energy source for the aging brain.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Philippe Bourassa
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec – Université Laval, Québec, Canada
| |
Collapse
|
14
|
Rong L, Peng Y, Shen Q, Chen K, Fang B, Li W. Effects of ketogenic diet on cognitive function of patients with Alzheimer's disease: a systematic review and meta-analysis. J Nutr Health Aging 2024; 28:100306. [PMID: 38943982 DOI: 10.1016/j.jnha.2024.100306] [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: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND Ketogenic diets (KD) have shown remarkable effects in many disease areas. It has been demonstrated in numerous animal experiments that KD is effective in the treatment of Alzheimer's disease (AD). But the clinical effect of treating AD is uncertain. OBJECTIVE To systematically review the impact of KD on cognitive function in AD. METHODS We conducted a search of three international databases-PubMed, Cochrane Library, and Embase-to retrieve RCTs on the KD intervention for AD from the inception of the databases through October 2023. Two reviewers searched and screened the literature, extracted and checked relevant data independently, and assessed the risk of bias of the included studies. The meta-analysis was carried out utilizing RevMan 5.3 software. RESULTS A total of 10 RCTS involving 691 patients with AD were included. There were 357 participants in the intervention group and 334 participants in the control group. The duration of the KD intervention ranged from a minimum of 3 months to a maximum of 15 months. Meta-analysis results showed that KD could effectively improve the mental state of the elderly (NM scale) [MD = 7.56, 95%CI (3.02, 12.10), P = 0.001], MMSE [MD = 1.25, 95%CI (0.46, 2.04), P = 0.002], and ADAS-Cog [MD = -3.43, 95%CI (-5.98, -0.88), P = 0.008]. The elevation of ketone body (β-hydroxybutyric) [MD = 118.84, 95%CI (15.20, 222.48), P = 0.02] may also lead to the elevation of triglyceride [MD = 0.19, 95%CI (0.03, 0.35), P = 0.02] and low density lipoprotein [MD = 0.31, 95%CI (0.04, 0.58), P = 0.02]. CONCLUSION Research conducted has indicated that the KD can enhance the mental state and cognitive function of those with AD, albeit potentially leading to an elevation in blood lipid levels. In summary, the good intervention effect and safety of KD are worthy of promotion and application in clinical treatment of AD.
Collapse
Affiliation(s)
- Liyang Rong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China; Sanya Hospital of Traditional Chinese Medicine, Sanya, China
| | - Yating Peng
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Keying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bangjiang Fang
- Department of Emergency, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
15
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2024; 0:revneuro-2024-0046. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
16
|
Oliai SF, Shippy DC, Ulland TK. Mitigation of CXCL10 secretion by metabolic disorder drugs in microglial-mediated neuroinflammation. J Neuroimmunol 2024; 391:578364. [PMID: 38718558 PMCID: PMC11165694 DOI: 10.1016/j.jneuroim.2024.578364] [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: 02/13/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Metabolic disorders are associated with several neurodegenerative diseases. We previously identified C-X-C motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10), as a major contributor to the type I interferon response in microglial-mediated neuroinflammation. Therefore, we hypothesized FDA-approved metabolic disorder drugs that attenuate CXCL10 secretion may be repurposed as a treatment for neurodegenerative diseases. Screening, dose curves, and cytotoxicity assays in LPS-stimulated microglia yielded treprostinil (hypertension), pitavastatin (hyperlipidemia), and eplerenone (hypertension) as candidates that significantly reduced CXCL10 secretion (in addition to other pro-inflammatory mediators) without impacting cell viability. Altogether, these data suggest metabolic disorder drugs that attenuate CXCL10 as potential treatments for neurodegenerative disease through mitigating microglial-mediated neuroinflammation.
Collapse
Affiliation(s)
- Sophia F Oliai
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Daniel C Shippy
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Tyler K Ulland
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
17
|
Lemche E, Killick R, Mitchell J, Caton PW, Choudhary P, Howard JK. Molecular mechanisms linking type 2 diabetes mellitus and late-onset Alzheimer's disease: A systematic review and qualitative meta-analysis. Neurobiol Dis 2024; 196:106485. [PMID: 38643861 DOI: 10.1016/j.nbd.2024.106485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/23/2024] Open
Abstract
Research evidence indicating common metabolic mechanisms through which type 2 diabetes mellitus (T2DM) increases risk of late-onset Alzheimer's dementia (LOAD) has accumulated over recent decades. The aim of this systematic review is to provide a comprehensive review of common mechanisms, which have hitherto been discussed in separate perspectives, and to assemble and evaluate candidate loci and epigenetic modifications contributing to polygenic risk linkages between T2DM and LOAD. For the systematic review on pathophysiological mechanisms, both human and animal studies up to December 2023 are included. For the qualitative meta-analysis of genomic bases, human association studies were examined; for epigenetic mechanisms, data from human studies and animal models were accepted. Papers describing pathophysiological studies were identified in databases, and further literature gathered from cited work. For genomic and epigenomic studies, literature mining was conducted by formalised search codes using Boolean operators in search engines, and augmented by GeneRif citations in Entrez Gene, and other sources (WikiGenes, etc.). For the systematic review of pathophysiological mechanisms, 923 publications were evaluated, and 138 gene loci extracted for testing candidate risk linkages. 3 57 publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight insulin signalling, inflammation and inflammasome pathways, proteolysis, gluconeogenesis and glycolysis, glycosylation, lipoprotein metabolism and oxidation, cell cycle regulation or survival, autophagic-lysosomal pathways, and energy. Documented findings suggest interplay between brain insulin resistance, neuroinflammation, insult compensatory mechanisms, and peripheral metabolic dysregulation in T2DM and LOAD linkage. The results allow for more streamlined longitudinal studies of T2DM-LOAD risk linkages.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom.
| | - Richard Killick
- Section of Old Age Psychiatry, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, United Kingdom
| | - Jackie Mitchell
- Department of Basic and Clinical Neurosciences, Maurice Wohl CIinical Neurosciences Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 125 Coldharbour Lane, London SE5 9NU, United Kingdom
| | - Paul W Caton
- Diabetes Research Group, School of Life Course Sciences, King's College London, Hodgkin Building, Guy's Campus, London SE1 1UL, United Kingdom
| | - Pratik Choudhary
- Diabetes Research Group, Weston Education Centre, King's College London, 10 Cutcombe Road, London SE5 9RJ, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, Hodgkin Building, Guy's Campus, King's College London, Great Maze Pond, London SE1 1UL, United Kingdom
| |
Collapse
|
18
|
Qin J, Nong L, Zhu Q, Huang Z, Wu F, Li S. A Retrospective Analysis of Central and Peripheral Metabolic Characteristics in Patients with Cryptococcal Meningitis. Neurol Ther 2024; 13:763-784. [PMID: 38643256 PMCID: PMC11136911 DOI: 10.1007/s40120-024-00610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/19/2024] [Indexed: 04/22/2024] Open
Abstract
INTRODUCTION Most current treatment strategies and investigations on cryptococcal meningitis (CM) focus primarily on the central nervous system (CNS), often overlooking the complex interplay between the CNS and the peripheral system. This study aims to explore the characteristics of central and peripheral metabolism in patients with CM. METHODS Patients diagnosed with CM as per the hospital records of the Fourth People's Hospital of Nanning were retrospectively analyzed. Patients were divided into two groups, non-structural damage of the brain (NSDB) and structural damage of the brain (SDB), according to the presence of brain lesions as detected with imaging. Based on the presence of enlarged cerebral ventricles, the cases in the SDB group were classified into non-ventriculomegaly (NVM) and ventriculomegaly (VM). Various parameters of cerebrospinal fluid (CSF) and peripheral blood (PB) were analyzed. RESULTS A significant correlation was detected between CSF and PB parameters. The levels of CSF-adenosine dehydrogenase (ADA), CSF-protein, CSF-glucose, and CSF-chloride ions were significantly correlated with the levels of PB-aminotransferase, PB-bilirubin, PB-creatinine (Cr), PB-urea nitrogen, PB-electrolyte, PB-protein, and PB-lipid. Compared with NSDB, the levels of CSF-glucose were significantly decreased in the SDB group, while the levels of CSF-lactate dehydrogenase (LDH) and CSF-protein were significantly increased in the SDB group. In the SDB group, the levels of PB-potassium, PB-hemoglobin(Hb), and PB-albumin were significantly decreased in the patients with VM, while the level of PB-urea nitrogen was significantly increased in these patients. CONCLUSION Metabolic and structural alterations in the brain may be associated with peripheral metabolic changes.
Collapse
Affiliation(s)
- Jianglong Qin
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Lanwei Nong
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Qingdong Zhu
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Zhizhong Huang
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Fengyao Wu
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Sijun Li
- The Fourth People's Hospital of Nanning, 1 Chang-Gang-Two-Li Road, Nanning, Guangxi, 530023, People's Republic of China.
| |
Collapse
|
19
|
Fong V, Kanuri B, Traubert O, Lui M, Patel SB. Behavioral and Metabolic Effects of ABCG4 KO in the APP swe,Ind (J9) Mouse Model of Alzheimer's Disease. J Mol Neurosci 2024; 74:49. [PMID: 38668787 PMCID: PMC11052713 DOI: 10.1007/s12031-024-02214-6] [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: 06/02/2023] [Accepted: 03/21/2024] [Indexed: 04/29/2024]
Abstract
The pathogenesis of Alzheimer's disease (AD) is complex and involves an imbalance between production and clearance of amyloid-ß peptides (Aß), resulting in accumulation of Aß in senile plaques. Hypercholesterolemia is a major risk factor for developing AD, with cholesterol shown to accumulate in senile plaques and increase production of Aß. ABCG4 is a member of the ATP-binding cassette transporters predominantly expressed in the CNS and has been suggested to play a role in cholesterol and Aß efflux from the brain. In this study, we bred Abcg4 knockout (KO) with the APPSwe,Ind (J9) mouse model of AD to test the hypothesis that loss of Abcg4 would exacerbate the AD phenotype. Unexpectedly, no differences were observed in novel object recognition (NOR) and novel object placement (NOP) behavioral tests, or on histologic examinations of brain tissues for senile plaque numbers. Furthermore, clearance of radiolabeled Aß from the brains did not differ between Abcg4 KO and control mice. Metabolic testing by indirect calorimetry, glucose tolerance test (GTT), and insulin tolerance test (ITT) were also mostly similar between groups with only a few mild metabolic differences noted. Overall, these data suggest that the loss of ABCG4 did not exacerbate the AD phenotype.
Collapse
Affiliation(s)
- Vincent Fong
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Owen Traubert
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Min Lui
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Shailendra B Patel
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
20
|
Buchholz A, Deme P, Betz JF, Brandt J, Haughey N, Cervenka MC. A randomized feasibility trial of the modified Atkins diet in older adults with mild cognitive impairment due to Alzheimer's disease. Front Endocrinol (Lausanne) 2024; 15:1182519. [PMID: 38505743 PMCID: PMC10949529 DOI: 10.3389/fendo.2024.1182519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024] Open
Abstract
Background Alzheimer's disease (AD) is increasing in prevalence, but effective treatments for its cognitive impairment remain severely limited. This study investigates the impact of ketone body production through dietary manipulation on memory in persons with mild cognitive impairment due to early AD and explores potential mechanisms of action. Methods We conducted a 12-week, parallel-group, controlled feasibility trial of a ketogenic diet, the modified Atkins diet (MAD), compared to a control diet in patients with cognitive impairments attributed to AD. We administered neuropsychological assessments, including memory tests, and collected blood samples at baseline and after 12 weeks of intervention. We performed untargeted lipidomic and targeted metabolomic analyses on plasma samples to detect changes over time. Results A total of 839 individuals were screened to yield 38 randomized participants, with 20 assigned to receive MAD and 18 assigned to receive a control diet. Due to attrition, only 13 in the MAD arm and nine in the control arm were assessed for the primary endpoint, with two participants meeting ketosis levels used to define MAD adherence criteria. The average change from baseline in the Memory Composite Score was 1.37 (95% CI: -0.87, 4.90) points higher in the MAD group compared to the control group. The effect size of the intervention on baseline MAD change was moderate (Cohen's D = 0.57, 95% CI: -0.67, 1.33). In the 15 participants (nine MAD, six control) assessed for lipidomic and metabolomic-lipidomics and metabolomics, 13 metabolites and 10 lipids showed significant changes from baseline to 12 weeks, including triacylglycerols (TAGs, 50:5, 52:5, and 52:6), sphingomyelins (SM, 44:3, 46:0, 46:3, and 48:1), acetoacetate, fatty acylcarnitines, glycerol-3-phosphate, and hydroxy fatty acids. Conclusions Attrition was greatest between baseline and week 6. All participants retained at week 6 completed the study. Despite low rates of adherence by criteria defined a priori, lipidomic and metabolomic analyses indicate significant changes from baseline in circulating lipids and metabolites between MAD and control participants at 12-week postrandomization, and MAD participants showed greater, albeit nonsignificant, improvement in memory.
Collapse
Affiliation(s)
- Alison Buchholz
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pragney Deme
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joshua F. Betz
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jason Brandt
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Norman Haughey
- Department of Psychiatry & Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mackenzie C. Cervenka
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Norambuena A, Sagar VK, Wang Z, Raut P, Feng Z, Wallrabe H, Pardo E, Kim T, Alam SR, Hu S, Periasamy A, Bloom GS. Disrupted mitochondrial response to nutrients is a presymptomatic event in the cortex of the APP SAA knock-in mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578668. [PMID: 38352486 PMCID: PMC10862844 DOI: 10.1101/2024.02.02.578668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Introduction Reduced brain energy metabolism, mTOR dysregulation, and extracellular amyloid-β oligomer (xcAβO) buildup characterize AD; how they collectively promote neurodegeneration is poorly understood. We previously reported that xcAβOs inhibit N utrient-induced M itochondrial A ctivity (NiMA) in cultured neurons. We now report NiMA disruption in vivo . Methods Brain energy metabolism and oxygen consumption were recorded in APP SAA/+ mice using two-photon fluorescence lifetime imaging and multiparametric photoacoustic microscopy. Results NiMA is inhibited in APP SAA/+ mice before other defects are detected in these amyloid-β-producing animals that do not overexpress APP or contain foreign DNA inserts into genomic DNA. GSK3β signals through mTORC1 to regulate NiMA independently of mitochondrial biogenesis. Inhibition of GSK3β with lithium or TWS119 stimulates NiMA in cultured human neurons, and mitochondrial activity and oxygen consumption in APP SAA mice. Conclusion NiMA disruption in vivo occurs before histopathological changes and cognitive decline in APP SAA mice, and may represent an early stage in human AD.
Collapse
|
22
|
Wheeler KV, Irimia A, Braskie MN. Using Neuroimaging to Study Cerebral Amyloid Angiopathy and Its Relationship to Alzheimer's Disease. J Alzheimers Dis 2024; 97:1479-1502. [PMID: 38306032 DOI: 10.3233/jad-230553] [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: 02/03/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β aggregation in the media and adventitia of the leptomeningeal and cortical blood vessels. CAA is one of the strongest vascular contributors to Alzheimer's disease (AD). It frequently co-occurs in AD patients, but the relationship between CAA and AD is incompletely understood. CAA may drive AD risk through damage to the neurovascular unit and accelerate parenchymal amyloid and tau deposition. Conversely, early AD may also drive CAA through cerebrovascular remodeling that impairs blood vessels from clearing amyloid-β. Sole reliance on autopsy examination to study CAA limits researchers' ability to investigate CAA's natural disease course and the effect of CAA on cognitive decline. Neuroimaging allows for in vivo assessment of brain function and structure and can be leveraged to investigate CAA staging and explore its associations with AD. In this review, we will discuss neuroimaging modalities that can be used to investigate markers associated with CAA that may impact AD vulnerability including hemorrhages and microbleeds, blood-brain barrier permeability disruption, reduced cerebral blood flow, amyloid and tau accumulation, white matter tract disruption, reduced cerebrovascular reactivity, and lowered brain glucose metabolism. We present possible areas for research inquiry to advance biomarker discovery and improve diagnostics.
Collapse
Affiliation(s)
- Koral V Wheeler
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Corwin D. Denney Research Center, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| |
Collapse
|
23
|
Ye YC, Chai SF, Li XR, Wu MN, Cai HY, Wang ZJ. Intermittent fasting and Alzheimer's disease-Targeting ketone bodies as a potential strategy for brain energy rescue. Metab Brain Dis 2024; 39:129-146. [PMID: 37823968 DOI: 10.1007/s11011-023-01288-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023]
Abstract
Alzheimer's disease (AD) lacks effective clinical treatments. As the disease progresses, the cerebral glucose hypometabolism that appears in the preclinical phase of AD gradually worsens, leading to increasingly severe brain energy disorders. This review analyzes the brain energy deficit in AD and its etiology, brain energy rescue strategies based on ketone intervention, the effects and mechanisms of IF, the differences in efficacy between IF and ketogenic diet and the duality of IF. The evidence suggests that brain energy deficits lead to the development and progression of AD pathology. IF, which improves brain energy impairments by promoting ketone metabolism, thus has good therapeutic potential for AD.
Collapse
Affiliation(s)
- Yu- Cai Ye
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Shi-Fan Chai
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xin-Ru Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
24
|
Jang J, Kim SR, Lee JE, Lee S, Son HJ, Choe W, Yoon KS, Kim SS, Yeo EJ, Kang I. Molecular Mechanisms of Neuroprotection by Ketone Bodies and Ketogenic Diet in Cerebral Ischemia and Neurodegenerative Diseases. Int J Mol Sci 2023; 25:124. [PMID: 38203294 PMCID: PMC10779133 DOI: 10.3390/ijms25010124] [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: 11/20/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Ketone bodies (KBs), such as acetoacetate and β-hydroxybutyrate, serve as crucial alternative energy sources during glucose deficiency. KBs, generated through ketogenesis in the liver, are metabolized into acetyl-CoA in extrahepatic tissues, entering the tricarboxylic acid cycle and electron transport chain for ATP production. Reduced glucose metabolism and mitochondrial dysfunction correlate with increased neuronal death and brain damage during cerebral ischemia and neurodegeneration. Both KBs and the ketogenic diet (KD) demonstrate neuroprotective effects by orchestrating various cellular processes through metabolic and signaling functions. They enhance mitochondrial function, mitigate oxidative stress and apoptosis, and regulate epigenetic and post-translational modifications of histones and non-histone proteins. Additionally, KBs and KD contribute to reducing neuroinflammation and modulating autophagy, neurotransmission systems, and gut microbiome. This review aims to explore the current understanding of the molecular mechanisms underpinning the neuroprotective effects of KBs and KD against brain damage in cerebral ischemia and neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Jiwon Jang
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Su Rim Kim
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jo Eun Lee
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seoyeon Lee
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyeong Jig Son
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eui-Ju Yeo
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Insug Kang
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (J.J.); (S.R.K.); (J.E.L.); (S.L.); (H.J.S.); (W.C.); (K.-S.Y.); (S.S.K.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
25
|
de Veij Mestdagh CF, Smit AB, Henning RH, van Kesteren RE. Mitochondrial Targeting against Alzheimer's Disease: Lessons from Hibernation. Cells 2023; 13:12. [PMID: 38201215 PMCID: PMC10778235 DOI: 10.3390/cells13010012] [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: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide and yet remains without effective therapy. Amongst the many proposed causes of AD, the mitochondrial cascade hypothesis is gaining attention. Accumulating evidence shows that mitochondrial dysfunction is a driving force behind synaptic dysfunction and cognitive decline in AD patients. However, therapies targeting the mitochondria in AD have proven unsuccessful so far, and out-of-the-box options, such as hibernation-derived mitochondrial mechanisms, may provide valuable new insights. Hibernators uniquely and rapidly alternate between suppression and re-activation of the mitochondria while maintaining a sufficient energy supply and without acquiring ROS damage. Here, we briefly give an overview of mitochondrial dysfunction in AD, how it affects synaptic function, and why mitochondrial targeting in AD has remained unsuccessful so far. We then discuss mitochondria in hibernation and daily torpor in mice, covering current advancements in hibernation-derived mitochondrial targeting strategies. We conclude with new ideas on how hibernation-derived dual mitochondrial targeting of both the ATP and ROS pathways may boost mitochondrial health and induce local synaptic protein translation to increase synaptic function and plasticity. Further exploration of these mechanisms may provide more effective treatment options for AD in the future.
Collapse
Affiliation(s)
- Christina F. de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
- Alzheimer Center Amsterdam, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Ronald E. van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
| |
Collapse
|
26
|
Hernandez AR, Barrett ME, Lubke KN, Maurer AP, Burke SN. A long-term ketogenic diet in young and aged rats has dissociable effects on prelimbic cortex and CA3 ensemble activity. Front Aging Neurosci 2023; 15:1274624. [PMID: 38155737 PMCID: PMC10753023 DOI: 10.3389/fnagi.2023.1274624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction Age-related cognitive decline has been linked to distinct patterns of cellular dysfunction in the prelimbic cortex (PL) and the CA3 subregion of the hippocampus. Because higher cognitive functions require both structures, selectively targeting a neurobiological change in one region, at the expense of the other, is not likely to restore normal behavior in older animals. One change with age that both the PL and CA3 share, however, is a reduced ability to utilize glucose, which can produce aberrant neural activity patterns. Methods The current study used a ketogenic diet (KD) intervention, which reduces the brain's reliance on glucose, and has been shown to improve cognition, as a metabolic treatment for restoring neural ensemble dynamics in aged rats. Expression of the immediate-early genes Arc and Homer1a were used to quantify the neural ensembles that were active in the home cage prior to behavior, during a working memory/biconditional association task, and a continuous spatial alternation task. Results Aged rats on the control diet had increased activity in CA3 and less ensemble overlap in PL between different task conditions than did the young animals. In the PL, the KD was associated with increased activation of neurons in the superficial cortical layers, establishing a clear link between dietary macronutrient content and frontal cortical activity. The KD did not lead to any significant changes in CA3 activity. Discussion These observations suggest that the availability of ketone bodies may permit the engagement of compensatory mechanisms in the frontal cortices that produce better cognitive outcomes.
Collapse
Affiliation(s)
- Abbi R. Hernandez
- Division of Gerontology, Geriatrics, and Palliative Care, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Maya E. Barrett
- Department of Psychology, The University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Katelyn N. Lubke
- Department of Neuroscience, McKnight Brain Institute, and Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, United States
| | - Andrew P. Maurer
- Department of Neuroscience, McKnight Brain Institute, and Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, United States
| | - Sara N. Burke
- Department of Neuroscience, McKnight Brain Institute, and Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, United States
| |
Collapse
|
27
|
Hao L, Wang L, Ju M, Feng W, Guo Z, Sun X, Xiao R. 27-Hydroxycholesterol impairs learning and memory ability via decreasing brain glucose uptake mediated by the gut microbiota. Biomed Pharmacother 2023; 168:115649. [PMID: 37806088 DOI: 10.1016/j.biopha.2023.115649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
Brain glucose hypometabolism is a significant manifestation of Alzheimer's disease (AD). 27-hydroxycholesterol (27-OHC) and the gut microbiota have been recognized as factors possibly influencing the pathogenesis of AD. This study aimed to investigate the link between 27-OHC, the gut microbiota, and brain glucose uptake in AD. Here, 6-month-old male C57BL/6 J mice were treated with sterile water or antibiotic cocktails, with or without 27-OHC and/or 27-OHC synthetic enzyme CYP27A1 inhibitor anastrozole (ANS). The gut microbiota, brain glucose uptake levels, and memory ability were measured. We observed that 27-OHC altered microbiota composition, damaged brain tissue structures, decreased the 2-deoxy-2-[18 F] fluorodeoxyglucose (18F-FDG) uptake value, downregulated the gene expression of glucose transporter type 4 (GLUT4), reduced the colocalization of GLUT1/glial fibrillary acidic protein (GFAP) in the hippocampus, and impaired spatial memory. ANS reversed the effects of 27-OHC. The antibiotic-treated mice did not exhibit similar results after 27-OHC treatment. This study reveals a potential molecular mechanism wherein 27-OHC-induced memory impairment might be linked to reduced brain glucose uptake, mediated by the gut microbiota.
Collapse
Affiliation(s)
- Ling Hao
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Lijing Wang
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Mengwei Ju
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Wenjing Feng
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Zhiting Guo
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Xuejing Sun
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China
| | - Rong Xiao
- School of Public Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing 100069, China.
| |
Collapse
|
28
|
Zilberter Y, Tabuena DR, Zilberter M. NOX-induced oxidative stress is a primary trigger of major neurodegenerative disorders. Prog Neurobiol 2023; 231:102539. [PMID: 37838279 DOI: 10.1016/j.pneurobio.2023.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Neurodegenerative diseases (NDDs) causing cognitive impairment and dementia are difficult to treat due to the lack of understanding of primary initiating factors. Meanwhile, major sporadic NDDs share many risk factors and exhibit similar pathologies in their early stages, indicating the existence of common initiation pathways. Glucose hypometabolism associated with oxidative stress is one such primary, early and shared pathology, and a likely major cause of detrimental disease-associated cascades; targeting this common pathology may therefore be an effective preventative strategy for most sporadic NDDs. However, its exact cause and trigger remain unclear. Recent research suggests that early oxidative stress caused by NADPH oxidase (NOX) activation is a shared initiating mechanism among major sporadic NDDs and could prove to be the long-sought ubiquitous NDD trigger. We focus on two major NDDs - Alzheimer's disease (AD) and Parkinson's disease (PD), as well as on acquired epilepsy which is an increasingly recognized comorbidity in NDDs. We also discuss available data suggesting the relevance of the proposed mechanisms to other NDDs. We delve into the commonalities among these NDDs in neuroinflammation and NOX involvement to identify potential therapeutic targets and gain a deeper understanding of the underlying causes of NDDs.
Collapse
Affiliation(s)
- Yuri Zilberter
- Aix-Marseille Université, INSERM UMR1106, Institut de Neurosciences des Systèmes, Marseille, France
| | - Dennis R Tabuena
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
| |
Collapse
|
29
|
Zhou Z, Ryan J, Tonkin AM, Zoungas S, Lacaze P, Wolfe R, Orchard SG, Murray AM, McNeil JJ, Yu C, Watts GF, Hussain SM, Beilin LJ, Ernst ME, Stocks N, Woods RL, Zhu C, Reid CM, Shah RC, Chong TTJ, Sood A, Sheets KM, Nelson MR. Association Between Triglycerides and Risk of Dementia in Community-Dwelling Older Adults: A Prospective Cohort Study. Neurology 2023; 101:e2288-e2299. [PMID: 37879942 PMCID: PMC10727221 DOI: 10.1212/wnl.0000000000207923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND AND OBJECTIVES It has been suggested that higher triglyceride levels were associated with a lower risk of Alzheimer disease. This study aimed to examine the association of triglycerides with dementia and cognition change in community-dwelling older adults. METHODS This prospective longitudinal study used data from the Aspirin in Reducing Events in the Elderly (ASPREE) randomized trial of adults aged 65 years or older without dementia or previous cardiovascular events at enrollment. The main outcome was incident dementia. Other outcomes included changes in composite cognition and domain-specific cognition (global cognition, memory, language and executive function, and psychomotor speed). The association between baseline triglycerides and dementia risk was estimated using Cox proportional hazard models adjusting for relevant risk factors. Linear mixed models were used to investigate cognitive change. The analysis was repeated in a subcohort of participants with available APOE-ε4 genetic data with additional adjustment for APOE-ε4 carrier status and an external cohort (UK Biobank) with similar selection criteria applied. RESULTS This study included 18,294 ASPREE participants and 68,200 UK Biobank participants (mean age: 75.1 and 66.9 years; female: 56.3% and 52.7%; median [interquartile range] triglyceride: 106 [80-142] mg/dL and 139 [101-193] mg/dL), with dementia recorded in 823 and 2,778 individuals over a median follow-up of 6.4 and 12.5 years, respectively. Higher triglyceride levels were associated with lower dementia risk in the entire ASPREE cohort (hazard ratio [HR] with doubling of triglyceride: 0.82, 95% CI 0.72-0.94). Findings were similar in the subcohort of participants with APOE-ε4 genetic data (n = 13,976) and in the UK Biobank cohort (HR was 0.82 and 0.83, respectively, all p ≤ 0.01). Higher triglycerides were also associated with slower decline in composite cognition and memory over time (p ≤ 0.05). DISCUSSION Older adults with higher triglyceride levels within the normal to high-normal range had a lower dementia risk and slower cognitive decline over time compared with individuals with lower triglyceride levels. Higher triglyceride levels may be reflective of better overall health and/or lifestyle behaviors that would protect against dementia development. Future studies are warranted to investigate whether specific components within the total circulating pool of plasma triglycerides may promote better cognitive function, with the hope of informing the development of new preventive strategies.
Collapse
Affiliation(s)
- Zhen Zhou
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia.
| | - Joanne Ryan
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Andrew M Tonkin
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Sophia Zoungas
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Paul Lacaze
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Rory Wolfe
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Suzanne G Orchard
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Anne M Murray
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - John J McNeil
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Chenglong Yu
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Gerald F Watts
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Sultana Monira Hussain
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Lawrence J Beilin
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Michael E Ernst
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Nigel Stocks
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Robyn L Woods
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Chao Zhu
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Christopher M Reid
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Raj C Shah
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Trevor T-J Chong
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Ajay Sood
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Kerry M Sheets
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| | - Mark R Nelson
- From the School of Public Health and Preventive Medicine (Z.Z., J.R., A.M.T., S.Z., P.L., R.W., S.G.O., J.J.M., C.Y., S.M.H., R.L.W.), Central Clinical School (C.Z.), and Turner Institute for Brain & Mental Health (T.T.-J.C.), Monash University, Melbourne, Victoria, Australia; Berman Center for Outcomes and Clinical Research (A.M.M.), Hennepin Healthcare Research Institute, Division of Geriatrics, Department of Medicine Hennepin HealthCare, Minneapolis, MN; School of Medicine (G.F.W., L.J.B.), University of Western Australia, Perth; Department of Pharmacy Practice and Science (M.E.E.), College of Pharmacy, The University of Iowa, Iowa City; Discipline of General Practice (N.S.), University of Adelaide, South Australia; School of Population Health (C.M.R.), Curtin University, Perth, Western Australia; Department of Family and Preventive Medicine and Rush Alzheimer's Disease Center (R.C.S., A.S.), Rush University Medical Center, Chicago, IL; Division of Geriatric Medicine (K.M.S.), Department of Medicine, Hennepin Healthcare, Minneapolis, MN; and Menzies Institute for Medical Research (M.R.N.), University of Tasmania, Hobart, Australia
| |
Collapse
|
30
|
Ehret F, Pelz MS, Senko AN, Soto KEG, Liu H, Kempermann G. Presymptomatic Reduction of Individuality in the App NL-F Knockin Model of Alzheimer's Disease. Biol Psychiatry 2023; 94:721-731. [PMID: 37076091 DOI: 10.1016/j.biopsych.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND One-third of the risk for Alzheimer's disease is explained by environment and lifestyle, but Alzheimer's disease pathology might also affect lifestyle and thereby impair the individual potential for health behavior and prevention. METHODS We examined in mice how the AppNL-F/NL-F (NL-F) knockin mutation affects the presymptomatic response to environmental enrichment (ENR) as an experimental paradigm addressing nongenetic factors. We assessed the emergence of interindividual phenotypic variation under the condition that both the genetic background and the shared environment were held constant, thereby isolating the contribution of individual behavior (nonshared environment). RESULTS After 4 months of ENR, the mean and variability of plasma ApoE were increased in NL-F mice, suggesting a presymptomatic variation in pathogenic processes. Roaming entropy as a measure of behavioral activity was continuously assessed with radiofrequency identification (RFID) technology and revealed reduced habituation and variance in NL-F mice compared with control animals, which do not carry a Beyreuther/Iberian mutation. Intraindividual variation decreased, while behavioral stability was reduced in NL-F mice. Seven months after discontinuation of ENR, we found no difference in plaque size and number, but ENR increased variance in hippocampal plaque counts in NL-F mice. A reactive increase in adult hippocampal neurogenesis in NL-F mice, known from other models, was normalized by ENR. CONCLUSIONS Our data suggest that while NL-F has early effects on individual behavioral patterns in response to ENR, there are lasting effects on cellular plasticity even after the discontinuation of ENR. Hence, early behavior matters for maintaining individual behavioral trajectories and brain plasticity even under maximally constrained conditions.
Collapse
Affiliation(s)
- Fanny Ehret
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany; Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Meike S Pelz
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Anna N Senko
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Karla E G Soto
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Hang Liu
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases Dresden, Dresden, Germany; Center for Regenerative Therapies Technical University Dresden, Dresden, Germany.
| |
Collapse
|
31
|
Lee H, Cho S, Kim MJ, Park YJ, Cho E, Jo YS, Kim YS, Lee JY, Thoudam T, Woo SH, Lee SI, Jeon J, Lee YS, Suh BC, Yoon JH, Go Y, Lee IK, Seo J. ApoE4-dependent lysosomal cholesterol accumulation impairs mitochondrial homeostasis and oxidative phosphorylation in human astrocytes. Cell Rep 2023; 42:113183. [PMID: 37777962 DOI: 10.1016/j.celrep.2023.113183] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 08/07/2023] [Accepted: 09/13/2023] [Indexed: 10/03/2023] Open
Abstract
Recent developments in genome sequencing have expanded the knowledge of genetic factors associated with late-onset Alzheimer's disease (AD). Among them, genetic variant ε4 of the APOE gene (APOE4) confers the greatest disease risk. Dysregulated glucose metabolism is an early pathological feature of AD. Using isogenic ApoE3 and ApoE4 astrocytes derived from human induced pluripotent stem cells, we find that ApoE4 increases glycolytic activity but impairs mitochondrial respiration in astrocytes. Ultrastructural and autophagy flux analyses show that ApoE4-induced cholesterol accumulation impairs lysosome-dependent removal of damaged mitochondria. Acute treatment with cholesterol-depleting agents restores autophagic activity, mitochondrial dynamics, and associated proteomes, and extended treatment rescues mitochondrial respiration in ApoE4 astrocytes. Taken together, our study provides a direct link between ApoE4-induced lysosomal cholesterol accumulation and abnormal oxidative phosphorylation.
Collapse
Affiliation(s)
- Hyein Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Sukhee Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Mi-Jin Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Yeo Jin Park
- Korean Medicine Life Science, University of Science and Technology, Daejeon 34054, South Korea; Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, South Korea
| | - Eunji Cho
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Yeon Suk Jo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea; Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Yong-Seok Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Jung Yi Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Seung-Hwa Woo
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Se-In Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Juyeong Jeon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Younghoon Go
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, South Korea.
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41944, South Korea; Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu 41944, South Korea.
| | - Jinsoo Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea.
| |
Collapse
|
32
|
Si B, Liu K, Huang G, Chen M, Yang J, Wu X, Li N, Tang W, Zhao S, Zheng N, Zhang Y, Wang J. Relationship between rumen bacterial community and milk fat in dairy cows. Front Microbiol 2023; 14:1247348. [PMID: 37886063 PMCID: PMC10598608 DOI: 10.3389/fmicb.2023.1247348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
Introduction Milk fat is the most variable nutrient in milk, and recent studies have shown that rumen bacteria are closely related to milk fat. However, there is limited research on the relationship between rumen bacteria and milk fatty. Fatty acids (FAs) are an important component of milk fat and are associated with various potential benefits and risks to human health. Methods In this experiment, forty-five healthy Holstein dairy cows with alike physiological and productive conditions were selected from medium-sized dairy farms and raised under the same feeding and management conditions. The experimental period was two weeks. During the experiment, raw milk and rumen fluid were collected, and milk components were determined. In this study, 8 high milk fat percentage (HF) dairy cows and 8 low milk fat percentage (LF) dairy cows were selected for analysis. Results Results showed that the milk fat percentage in HF group was significantly greater than that of the dairy cows in the LF group. 16S rRNA gene sequencing showed that the rumen bacterial abundance of HF dairy cows was significantly higher than that in LF dairy cows; at the genus level, the bacterial abundances of Prevotellaceae_UCG-001, Candidatus_Saccharimonas, Prevotellaceae_UCG-003, Ruminococcus_1, Lachnospiraceae_XPB1014_group, Lachnospiraceae_AC2044_group, probable_genus_10 and U29-B03 in HF group were significantly higher than those in the LF group. Spearman rank correlation analysis indicated that milk fat percentage was positively related to Prevotellaceae_UCG-001, Candidatus_Saccharimonas, Prevotellaceae_UCG-003, Ruminococcus_1, Lachnospiraceae_XPB1014_group, Lachnospiraceae_AC2044_group, probable_genus_10 and U29-B03. Furthermore, Prevotellaceae_UCG-001 was positively related to C14:0 iso, C15:0 iso, C18:0, Ruminococcus_1 with C18:1 t9, Lachnospiraceae_AC2044_group with C18:1 t9 and C18:1 t11, U29-B03 with C15:0 iso. Discussion To sum up, rumen bacteria in dairy cows are related to the variation of milk fat, and some rumen bacteria have potential effects on the deposition of certain fatty acids in raw milk.
Collapse
Affiliation(s)
- Boxue Si
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kaizhen Liu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Guoxin Huang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meiqing Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiyong Yang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xufang Wu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ning Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenhao Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shengguo Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yangdong Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Quality & Safety Control for Milk and Dairy Products of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
33
|
Cuenoud B, Croteau E, St-Pierre V, Richard G, Fortier M, Vandenberghe C, Carpentier AC, Cunnane SC. Cardiorenal ketone metabolism: a positron emission tomography study in healthy humans. Front Physiol 2023; 14:1280191. [PMID: 37869718 PMCID: PMC10587428 DOI: 10.3389/fphys.2023.1280191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023] Open
Abstract
Ketones are alternative energy substrates for the heart and kidney but no studies have investigated their metabolism simultaneously in both organs in humans. The present double tracer positron emission tomography (PET) study evaluated the organ distribution and basal kinetic rates of the radiolabeled ketone, 11C-acetoacetate (11C-AcAc), in the heart and kidney compared to 11C-acetate (11C-Ac), which is a well-validated metabolic radiotracer. Both tracers were highly metabolized by the left ventricle and the renal cortex. In the heart, kinetic rates were similar for both tracers. But in the renal cortex, uptake of 11C-Ac was higher compared to 11C-AcAc, while the reverse was observed for the clearance. Interestingly, infusion of 11C-AcAc led to a significantly delayed release of radioactivity in the renal medulla and pelvis, a phenomenon not observed with 11C-Ac. This suggests an equilibrium of 11C-AcAc with the other ketone, 11C-D-beta-hydroxybutyrate, and a different clearance profile. Overall, this suggests that in the kidney, the absorption and metabolism of 11C-AcAc is different compared to 11C-Ac. This dual tracer PET protocol provides the opportunity to explore the relative importance of ketone metabolism in cardiac and renal diseases, and to improve our mechanistic understanding of new metabolic interventions targeting these two organs.
Collapse
Affiliation(s)
- Bernard Cuenoud
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
- Centre D’imagerie Moléculaire de Sherbrooke, Sherbrooke, Canada
- Centre de Recherche Du CHUS, Sherbrooke, Canada
- Nestlé Health Science, Lausanne, Switzerland
| | - Etienne Croteau
- Centre D’imagerie Moléculaire de Sherbrooke, Sherbrooke, Canada
- Centre de Recherche Du CHUS, Sherbrooke, Canada
| | | | - Gabriel Richard
- Centre D’imagerie Moléculaire de Sherbrooke, Sherbrooke, Canada
- Centre de Recherche Du CHUS, Sherbrooke, Canada
| | - Mélanie Fortier
- Centre de Recherche sur le Vieillissement, Sherbrooke, Canada
| | | | - André C. Carpentier
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
- Centre de Recherche Du CHUS, Sherbrooke, Canada
| | - Stephen C. Cunnane
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
- Centre de Recherche sur le Vieillissement, Sherbrooke, Canada
| |
Collapse
|
34
|
Ashton JS, Roberts JW, Wakefield CJ, MacLaren DPM, Marwood S, Malone JJ. Medium chain triglycerides with a C8:C10 ratio of 30:70 enhances cognitive performance and mitigates the cognitive decline associated with prolonged exercise in young and healthy adults. Physiol Behav 2023; 269:114284. [PMID: 37394051 DOI: 10.1016/j.physbeh.2023.114284] [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: 10/19/2022] [Revised: 06/09/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION Prolonged exercise has been linked to a decline in cognitive function due to a variety of factors, such as a drop in oxygen in the prefrontal cortex and an increase in stress hormones and neurotransmitters. Medium chain triglycerides (MCTs) may possibly offset this decline as they provide energy for the brain via both direct and indirect pathways, alongside promoting chronic physiological adaptations within the brain. METHODS Participants were divided into two groups; MCT (n = 9) and Placebo (n = 10). The MCT gels contained 6 g of MCT with a C8:C10 ratio of 30:70, whereas the placebo gels contained carbohydrates of similar calorific value to the MCT gels. Participants visited the laboratory on three occasions (familiarisation/fitness test, pre-supplementation, post-supplementation), during which they performed a battery of cognitive tasks assessing domains such as processing speed, working memory, selective attention, decision making and coordination, before and after a prolonged bout of exercise (60 mins at 90% gas exchange threshold (GET). A 2-week supplementation period between visits 2 and 3 involved the ingestion of 2 gels per day. RESULTS Exercise resulted in detriments in most cognitive tasks pre-supplementation for both groups, and post-supplementation for the Placebo group (main effect ps< 0.05). Post-supplementation, the effect of exercise was mediated in the MCT group for all cognitive tasks (main effect ps< 0.05), except for the Digit and Spatial Span Backwards test phases (main effect ps> 0.05). Furthermore, MCT supplementation enhanced before-exercise cognitive performance and in some measures, such as working memory, this was maintained after-exercise (interaction effect ps> 0.05). CONCLUSIONS Chronic MCT supplementation enhanced before-exercise cognitive performance and offset the cognitive decline caused by a prolonged bout of exercise. In some cases, improvements in before-exercise cognitive performance were maintained after-exercise.
Collapse
Affiliation(s)
- Jake S Ashton
- School of Health and Sport Sciences, Liverpool Hope University, Liverpool, UK.
| | - James W Roberts
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | | | - Don P M MacLaren
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - Simon Marwood
- School of Health and Sport Sciences, Liverpool Hope University, Liverpool, UK
| | - James J Malone
- School of Health and Sport Sciences, Liverpool Hope University, Liverpool, UK
| |
Collapse
|
35
|
Li X, Young AJ, Pereira-Rufino LS, Shi Z, Byanyima J, Vesslee S, Reddy R, Pond T, Elliott M, Reddy R, Doot RK, van der Veen JW, Kranzler HR, Reddy Nanga RP, Dubroff JG, Wiers CE. Pharmacokinetic effects of a single-dose nutritional ketone ester supplement on brain ketone and glucose metabolism in alcohol use disorder - a pilot study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.25.23296090. [PMID: 37808798 PMCID: PMC10557835 DOI: 10.1101/2023.09.25.23296090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Introduction Acute alcohol intake decreases brain glucose metabolism and increases brain uptake of acetate, a metabolite of alcohol. Individuals with alcohol use disorder (AUD) show elevated brain acetate metabolism at the expense of glucose, a shift in energy utilization that persists beyond acute intoxication. We recently reported that nutritional ketosis and administration of ketone bodies as an alternative energy source to glucose reduce alcohol withdrawal severity and alcohol craving in AUD. However, the regional effects of nutritional ketosis on brain ketone (beta-hydroxybutyrate [BHB]) and glucose metabolism have not been studied in AUD. Methods Five participants with AUD underwent two magnetic resonance imaging (MRI) sessions and 4 participants with AUD underwent two positron emission tomography (PET) sessions with 18 F-fluorodeoxyglucose. All participants completed one session without KE intervention and one session during which they consumed 395 mg/kg (R) -3-hydroxybutyl (R) -3-hydroxybutyrate Ketone Ester (KE) intervention (TdeltaS Global Inc.) before the scan. The order of the sessions was randomized. For the PET cohort, blood glucose and ketone levels were assessed and voxel-wise maps of the cerebral metabolic rate of glucose (CMRglc) were computed at each session. For the MRI cohort, brain anterior cingulate BHB levels were assessed using magnetic resonance spectroscopy. Results A single dose of KE elevated blood BHB and anterior cingulate BHB levels compared to baseline. Moreover, blood glucose levels were lower with KE than baseline, and whole-brain CMRglc decreased by 17%. The largest KE-induced CMRglc reductions were in the frontal, occipital, cortex, and anterior cingulate cortices. Conclusion These findings provide preliminary evidence that KE administration elevates ketone and reduces brain glucose metabolism in humans, consistent with a shift from glucose to ketones as a brain energy source. Average reductions in CMRglc of 17% are similar to global average reductions documented with administration of 0.25-0.5 g/kg of alcohol. Documenting the clinical and neurometabolic effects of nutritional ketosis will yield fundamental knowledge as to its potential beneficial effects as a treatment for AUD and its underlying neural mechanisms.
Collapse
|
36
|
Feng S, Liu Y, Zhou Y, Shu Z, Cheng Z, Brenner C, Feng P. Mechanistic insights into the role of herpes simplex virus 1 in Alzheimer's disease. Front Aging Neurosci 2023; 15:1245904. [PMID: 37744399 PMCID: PMC10512732 DOI: 10.3389/fnagi.2023.1245904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Alzheimer's Disease (AD) is an aging-associated neurodegenerative disorder, threatening millions of people worldwide. The onset and progression of AD can be accelerated by environmental risk factors, such as bacterial and viral infections. Human herpesviruses are ubiquitous infectious agents that underpin numerous inflammatory disorders including neurodegenerative diseases. Published studies concerning human herpesviruses in AD imply an active role HSV-1 in the pathogenesis of AD. This review will summarize the current understanding of HSV-1 infection in AD and highlight some barriers to advance this emerging field.
Collapse
Affiliation(s)
- Shu Feng
- Department of Diabetes and Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, United States
| | - Yongzhen Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Yu Zhou
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Zhenfeng Shu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Zhuxi Cheng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
- International Department, Beijing Bayi School, Beijing, China
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, United States
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
37
|
Dunn E, Zhang B, Sahota VK, Augustin H. Potential benefits of medium chain fatty acids in aging and neurodegenerative disease. Front Aging Neurosci 2023; 15:1230467. [PMID: 37680538 PMCID: PMC10481710 DOI: 10.3389/fnagi.2023.1230467] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Neurodegenerative diseases are a large class of neurological disorders characterized by progressive dysfunction and death of neurones. Examples include Alzheimer's disease, Parkinson's disease, frontotemporal dementia, and amyotrophic lateral sclerosis. Aging is the primary risk factor for neurodegeneration; individuals over 65 are more likely to suffer from a neurodegenerative disease, with prevalence increasing with age. As the population ages, the social and economic burden caused by these diseases will increase. Therefore, new therapies that address both aging and neurodegeneration are imperative. Ketogenic diets (KDs) are low carbohydrate, high-fat diets developed initially as an alternative treatment for epilepsy. The classic ketogenic diet provides energy via long-chain fatty acids (LCFAs); naturally occurring medium chain fatty acids (MCFAs), on the other hand, are the main components of the medium-chain triglyceride (MCT) ketogenic diet. MCT-based diets are more efficient at generating the ketone bodies that are used as a secondary energy source for neurones and astrocytes. However, ketone levels alone do not closely correlate with improved clinical symptoms. Recent findings suggest an alternative mode of action for the MCFAs, e.g., via improving mitochondrial biogenesis and glutamate receptor inhibition. MCFAs have been linked to the treatment of both aging and neurodegenerative disease via their effects on metabolism. Through action on multiple disease-related pathways, MCFAs are emerging as compounds with notable potential to promote healthy aging and ameliorate neurodegeneration. MCFAs have been shown to stimulate autophagy and restore mitochondrial function, which are found to be disrupted in aging and neurodegeneration. This review aims to provide insight into the metabolic benefits of MCFAs in neurodegenerative disease and healthy aging. We will discuss the use of MCFAs to combat dysregulation of autophagy and mitochondrial function in the context of "normal" aging, Parkinson's disease, amyotrophic lateral sclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | - Hrvoje Augustin
- Department of Biological Sciences, Centre for Biomedical Sciences, Royal Holloway University of London, Egham, United Kingdom
| |
Collapse
|
38
|
Lima-Filho RAS, Benedet AL, De Bastiani MA, Povala G, Cozachenco D, Ferreira ST, De Felice FG, Rosa-Neto P, Zimmer ER, Lourenco MV. Association of the fibronectin type III domain-containing protein 5 rs1746661 single nucleotide polymorphism with reduced brain glucose metabolism in elderly humans. Brain Commun 2023; 5:fcad216. [PMID: 37601408 PMCID: PMC10438215 DOI: 10.1093/braincomms/fcad216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/23/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
Fibronectin type III domain-containing protein 5 (FNDC5) and its derived hormone, irisin, have been associated with metabolic control in humans, with described FNDC5 single nucleotide polymorphisms being linked to obesity and metabolic syndrome. Decreased brain FNDC5/irisin has been reported in subjects with dementia due to Alzheimer's disease. Since impaired brain glucose metabolism develops in ageing and is prominent in Alzheimer's disease, here, we examined associations of a single nucleotide polymorphism in the FNDC5 gene (rs1746661) with brain glucose metabolism and amyloid-β deposition in a cohort of 240 cognitively unimpaired and 485 cognitively impaired elderly individuals from the Alzheimer's Disease Neuroimaging Initiative. In cognitively unimpaired elderly individuals harbouring the FNDC5 rs1746661(T) allele, we observed a regional reduction in low glucose metabolism in memory-linked brain regions and increased brain amyloid-β PET load. No differences in cognition or levels of cerebrospinal fluid amyloid-β42, phosphorylated tau and total tau were observed between FNDC5 rs1746661(T) allele carriers and non-carriers. Our results indicate that a genetic variant of FNDC5 is associated with low brain glucose metabolism in elderly individuals and suggest that FNDC5 may participate in the regulation of brain metabolism in brain regions vulnerable to Alzheimer's disease pathophysiology. Understanding the associations between genetic variants in metabolism-linked genes and metabolic brain signatures may contribute to elucidating genetic modulators of brain metabolism in humans.
Collapse
Affiliation(s)
- Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Andréa L Benedet
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, 413 45, Sweden
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Guilherme Povala
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ 22281-100, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ 22281-100, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health University Institute, Departments of Neurology and Neurosurgery, Psychiatry, and Pharmacology, McGill University, Montreal, QC H4H 1R3, Canada
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | | |
Collapse
|
39
|
Castro CB, Dias CB, Hillebrandt H, Sohrabi HR, Chatterjee P, Shah TM, Fuller SJ, Garg ML, Martins RN. Medium-chain fatty acids for the prevention or treatment of Alzheimer's disease: a systematic review and meta-analysis. Nutr Rev 2023; 81:1144-1162. [PMID: 36633304 DOI: 10.1093/nutrit/nuac104] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
CONTEXT In preclinical Alzheimer's disease (AD), the brain gradually becomes insulin resistant. As a result, brain glucose utilization is compromised, causing a cellular energy deficit that leads to the accumulation of free radicals, which increases inflammation and damages neurons. When glucose utilization is impaired, ketone bodies offer an alternative energy source. Ketone bodies are synthesized from fats, obtained from either the diet or adipose tissue. Dietary medium-chain fatty acids (MCFAs), which are preferentially metabolized into ketone bodies, have the potential to supply the insulin-resistant brain with energy. OBJECTIVE This systematic review and meta-analysis aims to review the effect of MCFA supplements on circulating ketone bodies and cognition in individuals with subjective cognitive decline, mild cognitive impairment, and AD. DATA SOURCES A comprehensive search of electronic databases was performed on August 12, 2019, to retrieve all publications meeting the inclusion criteria. Alerts were then set to identify any publications after the search date up until January 31, 2021. DATA EXTRACTION Data were extracted by 2 authors and assessed by a third. In total, 410 publications were identified, of which 16 (n = 17 studies) met the inclusion criteria. DATA ANALYSIS All studies assessing change in levels of blood ketone bodies due to MCFA supplementation (n = 12) reported a significant increase. Cognition outcomes (measured in 13 studies), however, varied, ranging from no improvement (n = 4 studies) to improvement (n = 8 studies) or improvement only in apolipoprotein E allele 4 (APOE ε4) noncarriers (n = 2 studies). One study reported an increase in regional cerebral blood flow in APOE ε4 noncarriers and another reported an increase in energy metabolism in the brain. CONCLUSION MCFA supplementation increases circulating ketone body levels, resulting in increased brain energy metabolism. Further research is required to determine whether this MCFA-mediated increase in brain energy metabolism improves cognition. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration number CRD42019146967.
Collapse
Affiliation(s)
- Carolina B Castro
- Murdoch University Centre for Healthy Ageing, Murdoch University, Perth, Western Australia, Australia
- Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
| | - Cintia B Dias
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Heidi Hillebrandt
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Hamid R Sohrabi
- Murdoch University Centre for Healthy Ageing, Murdoch University, Perth, Western Australia, Australia
- Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
| | - Pratishtha Chatterjee
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Tejal M Shah
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
- Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
| | - Stephanie J Fuller
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Manohar L Garg
- Nutraceuticals Research Program, Faculty of Health and Medicine, University of Newcastle, Newcastle, New South Wales, Australia
| | - Ralph N Martins
- Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
- Australian Alzheimer's Research Foundation, Perth, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowen University, Perth, Western Australia, Australia
| |
Collapse
|
40
|
Robison LS, Gannon OJ, Salinero AE, Abi-Ghanem C, Kelly RD, Riccio DA, Mansour FM, Zuloaga KL. Sex differences in metabolic phenotype and hypothalamic inflammation in the 3xTg-AD mouse model of Alzheimer's disease. Biol Sex Differ 2023; 14:51. [PMID: 37559092 PMCID: PMC10410820 DOI: 10.1186/s13293-023-00536-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is notably associated with cognitive decline resulting from impaired function of hippocampal and cortical areas; however, several other domains and corresponding brain regions are affected. One such brain region is the hypothalamus, shown to atrophy and develop amyloid and tau pathology in AD patients. The hypothalamus controls several functions necessary for survival, including energy and glucose homeostasis. Changes in appetite and body weight are common in AD, often seen several years prior to the onset of cognitive symptoms. Therefore, altered metabolic processes may serve as a biomarker for AD, as well as a target for treatment, considering they are likely both a result of pathological changes and contributor to disease progression. Previously, we reported sexually dimorphic metabolic disturbances in ~ 7-month-old 3xTg-AD mice, accompanied by differences in systemic and hypothalamic inflammation. METHODS In the current study, we investigated metabolic outcomes and hypothalamic inflammation in 3xTg-AD males and females at 3, 6, 9, and 12 months of age to determine when these sex differences emerge. RESULTS In agreement with our previous study, AD males displayed less weight gain and adiposity, as well as reduced blood glucose levels following a glucose challenge, compared to females. These trends were apparent by 6-9 months of age, coinciding with increased expression of inflammatory markers (Iba1, GFAP, TNF-α, and IL-1β) in the hypothalamus of AD males. CONCLUSIONS These findings provide additional evidence for sex-dependent effects of AD pathology on energy and glucose homeostasis, which may be linked to hypothalamic inflammation.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL, 33328, USA.
| | - Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Richard D Kelly
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - David A Riccio
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Febronia M Mansour
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
41
|
Madhavan SS, Roa Diaz S, Peralta S, Nomura M, King CD, Lin A, Bhaumik D, Shah S, Blade T, Gray W, Chamoli M, Eap B, Panda O, Diaz D, Garcia TY, Stubbs BJ, Lithgow GJ, Schilling B, Verdin E, Chaudhuri AR, Newman JC. β-hydroxybutyrate is a metabolic regulator of proteostasis in the aged and Alzheimer disease brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547547. [PMID: 37461525 PMCID: PMC10349929 DOI: 10.1101/2023.07.03.547547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Loss of proteostasis is a hallmark of aging and Alzheimer disease (AD). Here, we identify β-hydroxybutyrate (βHB), a ketone body, as a regulator of protein solubility in the aging brain. βHB is a small molecule metabolite which primarily provides an oxidative substrate for ATP during hypoglycemic conditions, and also regulates other cellular processes through covalent and noncovalent protein interactions. We demonstrate βHB-induced protein insolubility across in vitro, ex vivo, and in vivo mouse systems. This activity is shared by select structurally similar metabolites, is not dependent on covalent protein modification, pH, or solute load, and is observable in mouse brain in vivo after delivery of a ketone ester. Furthermore, this phenotype is selective for pathological proteins such as amyloid-β, and exogenous βHB ameliorates pathology in nematode models of amyloid-β aggregation toxicity. We have generated a comprehensive atlas of the βHB-induced protein insolublome ex vivo and in vivo using mass spectrometry proteomics, and have identified common protein domains within βHB target sequences. Finally, we show enrichment of neurodegeneration-related proteins among βHB targets and the clearance of these targets from mouse brain, likely via βHB-induced autophagy. Overall, these data indicate a new metabolically regulated mechanism of proteostasis relevant to aging and AD.
Collapse
Affiliation(s)
- S S Madhavan
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Geriatrics, University of California San Francisco, San Francisco, CA, USA
| | - S Roa Diaz
- Buck Institute for Research on Aging, Novato, CA, USA
- Department of Geriatrics, University of California San Francisco, San Francisco, CA, USA
| | - S Peralta
- Buck Institute for Research on Aging, Novato, CA, USA
| | - M Nomura
- Buck Institute for Research on Aging, Novato, CA, USA
| | - C D King
- Buck Institute for Research on Aging, Novato, CA, USA
| | - A Lin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - D Bhaumik
- Buck Institute for Research on Aging, Novato, CA, USA
| | - S Shah
- Buck Institute for Research on Aging, Novato, CA, USA
| | - T Blade
- Buck Institute for Research on Aging, Novato, CA, USA
| | - W Gray
- Buck Institute for Research on Aging, Novato, CA, USA
| | - M Chamoli
- Buck Institute for Research on Aging, Novato, CA, USA
| | - B Eap
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - O Panda
- Buck Institute for Research on Aging, Novato, CA, USA
| | - D Diaz
- Buck Institute for Research on Aging, Novato, CA, USA
| | - T Y Garcia
- Buck Institute for Research on Aging, Novato, CA, USA
- Department of Geriatrics, University of California San Francisco, San Francisco, CA, USA
| | - B J Stubbs
- Buck Institute for Research on Aging, Novato, CA, USA
| | - G J Lithgow
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - B Schilling
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - E Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - A R Chaudhuri
- Buck Institute for Research on Aging, Novato, CA, USA
| | - J C Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Geriatrics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
42
|
Juby AG, Cunnane SC, Mager DR. Refueling the post COVID-19 brain: potential role of ketogenic medium chain triglyceride supplementation: an hypothesis. Front Nutr 2023; 10:1126534. [PMID: 37415915 PMCID: PMC10320593 DOI: 10.3389/fnut.2023.1126534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 04/25/2023] [Indexed: 07/08/2023] Open
Abstract
COVID-19 infection causes cognitive changes in the acute phase, but also after apparent recovery. Over fifty post (long)-COVID symptoms are described, including cognitive dysfunction ("brain fog") precluding return to pre-COVID level of function, with rates twice as high in females. Additionally, the predominant demographic affected by these symptoms is younger and still in the workforce. Lack of ability to work, even for six months, has significant socio-economic consequences. This cognitive dysfunction is associated with impaired cerebral glucose metabolism, assessed using 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET), showing brain regions that are abnormal compared to age and sex matched controls. In other cognitive conditions such as Alzheimer's disease (AD), typical patterns of cerebral glucose hypometabolism, frontal hypometabolism and cerebellar hypermetabolism are common. Similar FDG-PET changes have also been observed in post-COVID-19, raising the possibility of a similar etiology. Ketone bodies (B-hydroxybutyrate, acetoacetate and acetone) are produced endogenously with very low carbohydrate intake or fasting. They improve brain energy metabolism in the face of cerebral glucose hypometabolism in other conditions [mild cognitive impairment (MCI) and AD]. Long-term low carbohydrate intake or prolonged fasting is not usually feasible. Medium chain triglyceride (MCT) is an exogenous route to nutritional ketosis. Research has supported their efficacy in managing intractable seizures, and cognitive impairment in MCI and AD. We hypothesize that cerebral glucose hypometabolism associated with post COVID-19 infection can be mitigated with MCT supplementation, with the prediction that cognitive function would also improve. Although there is some suggestion that post COVID-19 cognitive symptoms may diminish over time, in many individuals this may take more than six months. If MCT supplementation is able to speed the cognitive recovery, this will impact importantly on quality of life. MCT is readily available and, compared to pharmaceutical interventions, is cost-effective. Research shows general tolerability with dose titration. MCT is a component of enteral and parenteral nutrition supplements, including in pediatrics, so has a long record of safety in vulnerable populations. It is not associated with weight gain or adverse changes in lipid profiles. This hypothesis serves to encourage the development of clinical trials evaluating the impact of MCT supplementation on the duration and severity of post COVID-19 cognitive symptoms.
Collapse
Affiliation(s)
- Angela G. Juby
- Division of Geriatrics, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Stephen C. Cunnane
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Diana R. Mager
- Agriculture Food and Nutrition Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
43
|
Fong V, Kanuri B, Traubert O, Lui M, Patel SB. Behavioral and metabolic and effects of ABCG4 KO in the APPswe,Ind (J9) mouse model of Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-3014093. [PMID: 37333297 PMCID: PMC10275060 DOI: 10.21203/rs.3.rs-3014093/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The pathogenesis of Alzheimer's disease (AD) is complex and involves an imbalance between production and clearance of amyloid-ß peptides (Aß), resulting in accumulation of Aß in senile plaques. Hypercholesterolemia is a major risk factor for developing AD, with cholesterol shown to accumulate in senile plaques and increase production of Aß. ABCG4 is a member of the ATP-binding cassette transporters predominantly expressed in the CNS, and has been suggested to play a role in cholesterol and Aß efflux from the brain. In this study, we bred Abcg4 knockout (KO) with the APPSwe,Ind (J9) mouse model of AD to test the hypothesis that loss of Abcg4 would exacerbate the AD phenotype. Unexpectedly, no differences were observed in Novel object recognition (NOR) and Novel object placement (NOP) behavioral tests, or on histologic examinations of brain tissues for senile plaque numbers. Furthermore, clearance of radiolabeled Aß from the brains did not differ between Abcg4 KO and control mice. Metabolic testing by indirect calorimetry, glucose tolerance test (GTT) and insulin tolerance test (ITT), were also mostly similar between groups with only a few mild metabolic differences noted. Overall these data suggest that the loss of ABCG4 did not exacerbate the AD phenotype.
Collapse
Affiliation(s)
- Vincent Fong
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Owen Traubert
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Min Lui
- Department of Pathology & Laboratory Medicine, University of Cincinnati
| | - Shailendra B Patel
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| |
Collapse
|
44
|
Sun L, Ye KX, Wong HLK, Wang L, Lim SL, Chao YX, Zhang C, Yap KZ, Feng L. The Effects of Medium Chain Triglyceride for Alzheimer's Disease Related Cognitive Impairment: A Systematic Review and Meta-Analysis. J Alzheimers Dis 2023:JAD230406. [PMID: 37248908 PMCID: PMC10357178 DOI: 10.3233/jad-230406] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
BACKGROUND The current lack of effective drug therapies for Alzheimer's disease (AD) has prompted researchers to seek alternative nutritional therapies, such as medium chain triglycerides (MCTs). However, results are inconclusive. OBJECTIVE This systematic review and meta-analysis aims to summarize current evidence on the effect of MCT on cognitive function in patients with mild cognitive impairment (MCI) or AD. METHODS A systematic search was conducted up until December 16, 2022, to identify human interventions reporting the effects of MCT on cognitive functioning of MCI or AD patients. 995 non-duplicated publications were identified, of which nine (n = 10 studies) met the inclusion criteria. RESULTS Meta-analysis showed cognitive improvements in general (SMD = 0.64; 95% CI [0.05, 1.24]), but not in memory, language, and attention domains after oral MCT administration, compared to placebo. The effect of MCT was greater among APOEɛ4 (-) subjects than APOEɛ4 (+) subjects (SMD = 1.87; 95% CI [0.35, 3.40]). CONCLUSION This review provides some evidence that treatment with MCT could improve general cognitive function in APOEɛ4 (-) cognitive impaired patients. Better characterized clinical studies are warranted before making a definitive conclusion on the use of MCT for MCI and AD management.
Collapse
Affiliation(s)
- Lina Sun
- School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Kaisy Xinhong Ye
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Lingyan Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China
| | - Su Lin Lim
- Department of Dietetics, National University Hospital, Singapore, Singapore
| | - Yin Xia Chao
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Can Zhang
- Mass General Institute for Neurodegenerative, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kai Zhen Yap
- Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
45
|
Westi EW, Andersen JV, Aldana BI. Using stable isotope tracing to unravel the metabolic components of neurodegeneration: Focus on neuron-glia metabolic interactions. Neurobiol Dis 2023; 182:106145. [PMID: 37150307 DOI: 10.1016/j.nbd.2023.106145] [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: 02/23/2023] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023] Open
Abstract
Disrupted brain metabolism is a critical component of several neurodegenerative diseases. Energy metabolism of both neurons and astrocytes is closely connected to neurotransmitter recycling via the glutamate/GABA-glutamine cycle. Neurons and astrocytes hereby work in close metabolic collaboration which is essential to sustain neurotransmission. Elucidating the mechanistic involvement of altered brain metabolism in disease progression has been aided by the advance of techniques to monitor cellular metabolism, in particular by mapping metabolism of substrates containing stable isotopes, a technique known as isotope tracing. Here we review key aspects of isotope tracing including advantages, drawbacks and applications to different cerebral preparations. In addition, we narrate how isotope tracing has facilitated the discovery of central metabolic features in neurodegeneration with a focus on the metabolic cooperation between neurons and astrocytes.
Collapse
Affiliation(s)
- Emil W Westi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
46
|
Kumar P, Osahon OW, Sekhar RV. GlyNAC (Glycine and N-Acetylcysteine) Supplementation in Old Mice Improves Brain Glutathione Deficiency, Oxidative Stress, Glucose Uptake, Mitochondrial Dysfunction, Genomic Damage, Inflammation and Neurotrophic Factors to Reverse Age-Associated Cognitive Decline: Implications for Improving Brain Health in Aging. Antioxidants (Basel) 2023; 12:antiox12051042. [PMID: 37237908 DOI: 10.3390/antiox12051042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cognitive decline frequently occurs with increasing age, but mechanisms contributing to age-associated cognitive decline (ACD) are not well understood and solutions are lacking. Understanding and reversing mechanisms contributing to ACD are important because increased age is identified as the single most important risk factor for dementia. We reported earlier that ACD in older humans is associated with glutathione (GSH) deficiency, oxidative stress (OxS), mitochondrial dysfunction, glucose dysmetabolism and inflammation, and that supplementing GlyNAC (glycine and N-acetylcysteine) improved these defects. To test whether these defects occur in the brain in association with ACD, and could be improved/reversed with GlyNAC supplementation, we studied young (20-week) and old (90-week) C57BL/6J mice. Old mice received either regular or GlyNAC supplemented diets for 8 weeks, while young mice received the regular diet. Cognition and brain outcomes (GSH, OxS, mitochondrial energetics, autophagy/mitophagy, glucose transporters, inflammation, genomic damage and neurotrophic factors) were measured. Compared to young mice, the old-control mice had significant cognitive impairment and multiple brain defects. GlyNAC supplementation improved/corrected the brain defects and reversed ACD. This study finds that naturally-occurring ACD is associated with multiple abnormalities in the brain, and provides proof-of-concept that GlyNAC supplementation corrects these defects and improves cognitive function in aging.
Collapse
Affiliation(s)
- Premranjan Kumar
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ob W Osahon
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rajagopal V Sekhar
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
47
|
Zhou D, Sun Y, Qian Z, Wang Z, Zhang D, Li Z, Zhao J, Dong C, Li W, Huang G. Long-term dietary folic acid supplementation attenuated aging-induced hippocampus atrophy and promoted glucose uptake in 25-month-old rats with cognitive decline. J Nutr Biochem 2023; 117:109328. [PMID: 36958416 DOI: 10.1016/j.jnutbio.2023.109328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/18/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023]
Abstract
The brain has high energy demand making it sensitive to changes in energy fuel supply. Aging shrinks brain volume, decreases glucose uptake availability of the brain, and finally, causes cognitive dysfunction. Folic acid supplementation delayed cognitive decline and neurodegeneration. However, whether folic acid affects brain energy metabolism and structural changes is unclear. The study aimed to determine if long-term dietary folic acid supplementation could alleviate age-related cognitive decline by attenuating hippocampus atrophy and promoting brain glucose uptake in Sprague-Dawley (SD) rats. According to folic acid levels in diet, three-month-old male SD rats were randomly divided into four intervention groups for 22 months in equal numbers: folic acid-deficient diet (FA-D) group, folic acid-normal diet (FA-N) group, low folic acid-supplemented diet (FA-L) group, and high folic acid-supplemented diet (FA-H) group. The results showed that serum folate concentrations decreased and serum homocysteine (Hcy) concentrations increased with age, and dietary folic acid supplementation increased serum folate concentrations and decreased Hcy concentrations at 11, 18, and 22 months of intervention. Dietary folic acid supplementation attenuated aging-induced hippocampus atrophy, which was showed by higher fractional anisotropy and lower mean diffusivity in the hippocampus, increased brain 18F-Fluorodeoxyglucose (18F-FDG) uptake, then stimulated neuronal survival, and alleviated age-related cognitive decline in SD rats. In conclusion, long-term dietary folic acid supplementation alleviated age-related cognitive decline by attenuating hippocampus atrophy and promoting brain glucose uptake in SD rats.
Collapse
Affiliation(s)
- Dezheng Zhou
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Yue Sun
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Zhiyong Qian
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Zehao Wang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Dalong Zhang
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Zhenshu Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Jing Zhao
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Cuixia Dong
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Wen Li
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China.
| | - Guowei Huang
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin 300070, China.
| |
Collapse
|
48
|
Deng S, Yi P, Xu M, Yi Q, Feng J. Dysfunctional gene splicing in glucose metabolism may contribute to Alzheimer's disease. Chin Med J (Engl) 2023; 136:666-675. [PMID: 35830275 PMCID: PMC10129079 DOI: 10.1097/cm9.0000000000002214] [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: 12/14/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT The glucose metabolism is crucial for sustained brain activity as it provides energy and is a carbon source for multiple biomacromolecules; glucose metabolism decreases dramatically in Alzheimer's disease (AD) and may be a fundamental cause for its development. Recent studies reveal that the alternative splicing events of certain genes effectively regulate several processes in glucose metabolism including insulin receptor, insulin-degrading enzyme, pyruvate kinase M, receptor for advanced glycation endproducts, and others, thereby, influencing glucose uptake, glycolysis, and advanced glycation end-products-mediated signaling pathways. Indeed, the discovery of aberrant alternative splicing that changes the proteomic diversity and protein activity in glucose metabolism has been pivotal in our understanding of AD development. In this review, we summarize the alternative splicing events of the glucose metabolism-related genes in AD pathology and highlight the crucial regulatory roles of splicing factors in the alternative splicing process. We also discuss the emerging therapeutic approaches for targeting splicing factors for AD treatment.
Collapse
Affiliation(s)
- Shengfeng Deng
- Laboratory of Anesthesiology, Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Peng Yi
- Laboratory of Anesthesiology, Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Mingliang Xu
- Laboratory of Anesthesiology, Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qian Yi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jianguo Feng
- Laboratory of Anesthesiology, Department of Anesthesiology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Affiliated Xinhui Hospital, Southern Medical University (People's Hospital of Xinhui District), Jiangmen, Guangdong 529100, China
| |
Collapse
|
49
|
Somin S, Kulasiri D, Samarasinghe S. Alleviating the unwanted effects of oxidative stress on Aβ clearance: a review of related concepts and strategies for the development of computational modelling. Transl Neurodegener 2023; 12:11. [PMID: 36907887 PMCID: PMC10009979 DOI: 10.1186/s40035-023-00344-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Treatment for Alzheimer's disease (AD) can be more effective in the early stages. Although we do not completely understand the aetiology of the early stages of AD, potential pathological factors (amyloid beta [Aβ] and tau) and other co-factors have been identified as causes of AD, which may indicate some of the mechanism at work in the early stages of AD. Today, one of the primary techniques used to help delay or prevent AD in the early stages involves alleviating the unwanted effects of oxidative stress on Aβ clearance. 4-Hydroxynonenal (HNE), a product of lipid peroxidation caused by oxidative stress, plays a key role in the adduction of the degrading proteases. This HNE employs a mechanism which decreases catalytic activity. This process ultimately impairs Aβ clearance. The degradation of HNE-modified proteins helps to alleviate the unwanted effects of oxidative stress. Having a clear understanding of the mechanisms associated with the degradation of the HNE-modified proteins is essential for the development of strategies and for alleviating the unwanted effects of oxidative stress. The strategies which could be employed to decrease the effects of oxidative stress include enhancing antioxidant activity, as well as the use of nanozymes and/or specific inhibitors. One area which shows promise in reducing oxidative stress is protein design. However, more research is needed to improve the effectiveness and accuracy of this technique. This paper discusses the interplay of potential pathological factors and AD. In particular, it focuses on the effect of oxidative stress on the expression of the Aβ-degrading proteases through adduction of the degrading proteases caused by HNE. The paper also elucidates other strategies that can be used to alleviate the unwanted effects of oxidative stress on Aβ clearance. To improve the effectiveness and accuracy of protein design, we explain the application of quantum mechanical/molecular mechanical approach.
Collapse
Affiliation(s)
- Sarawoot Somin
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand.,Department of Wine, Food and Molecular Biosciences, Lincoln University, Christchurch, 7647, New Zealand
| | - Don Kulasiri
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand. .,Department of Wine, Food and Molecular Biosciences, Lincoln University, Christchurch, 7647, New Zealand.
| | - Sandhya Samarasinghe
- Centre for Advanced Computational Solutions (C-fACS), Lincoln University, Christchurch, 7647, New Zealand
| |
Collapse
|
50
|
Sun Y, Zhang H, Zhang X, Wang W, Chen Y, Cai Z, Wang Q, Wang J, Shi Y. Promotion of astrocyte-neuron glutamate-glutamine shuttle by SCFA contributes to the alleviation of Alzheimer's disease. Redox Biol 2023; 62:102690. [PMID: 37018970 PMCID: PMC10122027 DOI: 10.1016/j.redox.2023.102690] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/29/2023] Open
Abstract
The brain is particularly susceptible to oxidative damage which is a key feature of several neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease. The shuttling of glutathione (GSH) precursors from astrocytes to neurons has been shown to be instrumental for the neuroprotective activity. Here, we revealed that short chain fatty acids (SCFA), which have been related to AD and PD, could promote glutamate-glutamine shuttle to potentially resist oxidative damage in neurons at cellular level. Furthermore, we performed nine-month-long dietary SCFA supplementations in APPswe/PS1dE9 (APP/PS1) mice, and showed that it reshaped the homeostasis of microbiota and alleviated the cognitive impairment by reducing Aβ deposition and tau hyperphosphorylation. Single-cell RNA sequencing analysis of the hippocampus revealed SCFA can enhance astrocyte-neuron communication including glutamate-glutamine shuttle, mainly by acting on astrocyte in vivo. Collectively, our findings indicate that long-term dietary SCFA supplementations at early aging stage can regulate the neuroenergetics to alleviate AD, providing a promising direction for the development of new AD drug.
Collapse
|