1
|
Madhavan SS, Roa Diaz S, Peralta S, Nomura M, King CD, Ceyhan KE, Lin A, Bhaumik D, Foulger AC, Shah S, Blade T, Gray W, Chamoli M, Eap B, Panda O, Diaz D, Garcia TY, Stubbs BJ, Ulrich SM, 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. Cell Chem Biol 2025; 32:174-191.e8. [PMID: 39626664 PMCID: PMC11741930 DOI: 10.1016/j.chembiol.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/23/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024]
Abstract
Loss of proteostasis is a hallmark of aging and Alzheimer disease (AD). We identify β-hydroxybutyrate (βHB), a ketone body, as a regulator of protein solubility. βHB primarily provides ATP substrate during periods of reduced glucose availability, and regulates other cellular processes through protein interactions. We demonstrate βHB-induced protein insolubility 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. This mechanism is selective for pathological proteins such as amyloid-β, and exogenous βHB ameliorates pathology in nematode models of amyloid-β aggregation toxicity. We generate libraries of the βHB-induced protein insolublome using mass spectrometry proteomics, and identify common protein domains and upstream regulators. We show enrichment of neurodegeneration-related proteins among βHB targets and the clearance of these targets from mouse brain. These data indicate a metabolically regulated mechanism of proteostasis relevant to aging and AD.
Collapse
Affiliation(s)
- Sidharth S Madhavan
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Division of Geriatrics, University of California, San Francisco, San Francisco, CA 94118, USA
| | - Stephanie Roa Diaz
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Division of Geriatrics, University of California, San Francisco, San Francisco, CA 94118, USA
| | - Sawyer Peralta
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | - Kaya E Ceyhan
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Anwen Lin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Dipa Bhaumik
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Anna C Foulger
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Samah Shah
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Thanh Blade
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Wyatt Gray
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Manish Chamoli
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Brenda Eap
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Oishika Panda
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Diego Diaz
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Thelma Y Garcia
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Division of Geriatrics, University of California, San Francisco, San Francisco, CA 94118, USA
| | | | - Scott M Ulrich
- Department of Chemistry, Ithaca College, Ithaca, NY 14850, USA
| | - Gordon J Lithgow
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | | | - John C Newman
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; Division of Geriatrics, University of California, San Francisco, San Francisco, CA 94118, USA.
| |
Collapse
|
2
|
Lathika Rajendrakumar A, Arbeev KG, Bagley O, Duan M, Yashin AI, Ukraintseva S. APOE4 and infectious diseases jointly contribute to brain glucose hypometabolism, a biomarker of Alzheimer's pathology: New findings from the ADNI. PLoS One 2025; 20:e0316808. [PMID: 39774485 PMCID: PMC11706463 DOI: 10.1371/journal.pone.0316808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Impaired brain glucose metabolism is a preclinical feature of neurodegenerative diseases such as Alzheimer's disease (AD). Infections may promote AD-related pathology. Therefore, we investigated the interplay between infections and APOE4, a strong genetic risk factor for AD. METHODS We analyzed data on 1,509 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database using multivariate linear regression models. The outcomes were rank-normalized hypometabolic convergence index (HCI), statistical regions of interest (SROI) for AD, and mild cognitive impairment (MCI). Marginal mean estimates for infections, stratified by APOE4 carrier status, were then computed. RESULTS Prior infections were associated with greater HCI [β = 0.15, 95% CI: 0.03, 0.27, p = 0.01]. The combined effects of infections and APOE4 carriers on HCI levels were significantly greater than either variable alone. Among APOE4 carriers, the estimated marginal mean was 0.62, rising to 0.77, with infections (p<0.001), indicating an interaction effect. Carriers with multiple infections showed greater hypometabolism (higher HCI), with an estimate of 0.44 (p = 0.01) compared to 0.11 (p = 0.08) for those with a single infection, revealing a dose-response relationship. The estimates for the association of infections with SROI AD and SROI MCI were β = -0.01 (p = 0.02) and β = -0.01 (p = 0.04), respectively. CONCLUSION Our findings suggest that infections and APOE4 jointly contribute to brain glucose hypometabolism and AD pathology, supporting a "multi-hit" mechanism in AD development.
Collapse
Affiliation(s)
- Aravind Lathika Rajendrakumar
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | | |
Collapse
|
3
|
Mares J, Kumar G, Sharma A, Emrani S, McIntire LB, Guo J, Menon V, Nuriel T. APOE ε4-associated heterogeneity of neuroimaging biomarkers across the Alzheimer's disease continuum. Alzheimers Dement 2025; 21:e14392. [PMID: 39575672 PMCID: PMC11775459 DOI: 10.1002/alz.14392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025]
Abstract
INTRODUCTION While the role of apolipoprotein E (APOE) ε4 in Alzheimer's disease (AD) susceptibility has been studied extensively, much less is known about the differences in disease presentation in APOE ε4 carriers versus non-carriers. METHODS To help elucidate these differences, we performed a broad analysis comparing the regional levels of six different neuroimaging biomarkers in the brains of APOE ε4 carriers versus non-carriers who participated in the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS We observed significant APOE ε4-associated heterogeneity in regional amyloid beta deposition, tau accumulation, glucose uptake, brain volume, cerebral blood flow, and white matter hyperintensities within each AD diagnostic group. We also observed important APOE ε4-associated differences in cognitively unimpaired individuals who converted to mild cognitive impairment/AD versus those who did not convert. DISCUSSION This observed heterogeneity in neuroimaging biomarkers between APOE ε4 carriers versus non-carriers may have important implications regarding the prevention, diagnosis, and treatment of AD in different subpopulations. HIGHLIGHTS An extensive study was performed on the apolipoprotein E (APOE) ε4-associated heterogeneity in neuroimaging biomarkers from the Alzheimer's Disease Neuroimaging Initiative. Robust APOE ε4-associated increases in amyloid beta (Aβ) deposition throughout the brain, in every diagnostic group, were observed. APOE ε4-associated increases in tau pathology, decreases in glucose uptake, and increases in brain atrophy, which expand in regional scope and magnitude with disease progression, were observed. Significant sex- and age-related differences in APOE ε4-associated neuroimaging biomarker heterogeneity, with overall increases in pathological presentation in female APOE ε4 carriers, were observed. Regional differences in Aβ deposition, tau accumulation, glucose uptake, ventricle size, and white matter hyperintensities were observed in cognitively normal participants who converted to mild cognitive impairment/Alzheimer's disease, which may hold potential predictive value.
Collapse
Affiliation(s)
- Jason Mares
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Gautam Kumar
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Department of NeurobiologyUniversity of MarylandBaltimoreMarylandUSA
| | - Anurag Sharma
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Sheina Emrani
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura Beth McIntire
- Lipidomics and Biomarker Discovery LabBrain Health Imaging InstituteDepartment of RadiologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Jia Guo
- Department of PsychiatryColumbia UniversityNew YorkNew YorkUSA
- Zuckerman InstituteColumbia UniversityNew YorkNew YorkUSA
| | - Vilas Menon
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
4
|
Day IL, Tamboline M, Lipshutz GS, Xu S. Recent developments in translational imaging of in vivo gene therapy outcomes. Mol Ther 2024:S1525-0016(24)00849-9. [PMID: 39741403 DOI: 10.1016/j.ymthe.2024.12.049] [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: 07/31/2024] [Revised: 11/18/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Gene therapy achieves therapeutic benefits by delivering genetic materials, packaged within a delivery vehicle, to target cells with defective genes. This approach has shown promise in treating various conditions, including cancer, metabolic disorders, and tissue-degenerative diseases. Over the past 5 years, molecular imaging has increasingly supported gene therapy development in both preclinical and clinical studies. High-quality images from positron emission tomography (PET), single-photon emission computed tomography (SPECT), magnetic resonance imaging (MRI), and computed tomography (CT) enable quantitative and reliable monitoring of gene therapy. Most reported studies have applied imaging biomarkers to non-invasively evaluate the outcomes of gene therapy. This review aims to inform researchers in molecular imaging and gene therapy about the integration of these two disciplines. We highlight recent developments in using imaging biomarkers to monitor the outcome of in vivo gene therapy, where the therapeutic delivery vehicle is administered systemically. In addition, we discuss prospects for further incorporating imaging biomarkers to support the development and application of gene therapy.
Collapse
Affiliation(s)
- Isabel L Day
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mikayla Tamboline
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gerald S Lipshutz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Semel Institute for Neuroscience, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shili Xu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Crump Institute for Molecular Imaging, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
5
|
Johnson CN, Lysaker CR, McCoin CS, Evans MR, Thyfault JP, Wilkins HM, Morris JK, Geiger PC. Skeletal muscle proteome differs between young APOE3 and APOE4 targeted replacement mice in a sex-dependent manner. Front Aging Neurosci 2024; 16:1486762. [PMID: 39634654 PMCID: PMC11615480 DOI: 10.3389/fnagi.2024.1486762] [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: 08/26/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD), yet it's unclear how this allele mediates risk. APOE4 carriers experience reduced mobility and faster decline in muscle strength, suggesting skeletal muscle involvement. Mitochondria are critical for muscle function and although we have reported defects in muscle mitochondrial respiration during early cognitive decline, APOE4-mediated effects on muscle mitochondria are unknown. Methods Here, we sought to determine the impact of APOE4 on skeletal muscle bioenergetics using young, male and female APOE3 (control) and APOE4 targeted replacement mice (n = 8 per genotype/sex combination). We examined the proteome, mitochondrial respiration, fiber size, and fiber-type distribution in skeletal muscle. Results We found that APOE4 alters mitochondrial pathway expression in young mouse muscle in a sex-dependent manner without affecting respiration and fiber size or composition relative to APOE3. In both sexes, the expression of mitochondrial pathways involved in electron transport, ATP synthesis, and heat production by uncoupling proteins and mitochondrial dysfunction significantly differed between APOE4 and APOE3 muscle. For pathways with predicted direction of activation, electron transport and oxidative phosphorylation were upregulated while mitochondrial dysfunction and sirtuin signaling were downregulated in female APOE4 vs. APOE3 muscle. In males, sulfur amino acid metabolism was upregulated in APOE4 vs. APOE3 muscle. Discussion This work highlights early involvement of skeletal muscle in a mouse model of APOE4-linked AD, which may contribute to AD pathogenesis or serve as a biomarker for brain health.
Collapse
Affiliation(s)
- Chelsea N. Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, MO, United States
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, MO, United States
| | - Colin S. McCoin
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, MO, United States
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, MO, United States
| | - Mara R. Evans
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, MO, United States
| | - John P. Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, MO, United States
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, MO, United States
| | - Heather M. Wilkins
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, MO, United States
| | - Jill K. Morris
- University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Fairway, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, MO, United States
| | - Paige C. Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, MO, United States
- University of Kansas Diabetes Institute, University of Kansas Medical Center, Kansas City, MO, United States
| |
Collapse
|
6
|
Menendez-Gonzalez M. Therapeutic Challenges Derived from the Interaction Among Apolipoprotein E, Cholesterol, and Amyloid in Alzheimer's Disease. Int J Mol Sci 2024; 25:12029. [PMID: 39596098 PMCID: PMC11593474 DOI: 10.3390/ijms252212029] [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: 10/19/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The isoform E4 of the Apolipoprotein E (ApoE) represents one of the strongest genetic risk factors for late-onset Alzheimer's disease (AD). ApoE has key roles in cholesterol transport and amyloid-β (Aβ) metabolism, which are both central to AD pathogenesis. The E4 isoform has been implicated in reduced cholesterol homeostasis, increased Aβ aggregation, and heightened tau phosphorylation, contributing to amyloid plaques and neurodegeneration. This manuscript examines the complex interactions among ApoE isoforms, cholesterol metabolism, and amyloid pathology. Moreover, the therapeutic challenges associated with lipid-lowering agents (e.g., statins, PCSK9 inhibitors), anti-amyloid immunotherapies, and anticoagulants are described, focusing on ApoE4 carriers. Decision-making challenges are discussed by analyzing the pros and cons of these therapies.
Collapse
Affiliation(s)
- Manuel Menendez-Gonzalez
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universidad de Oviedo, ES-33006 Oviedo, Spain;
- Servicio de Neurología, Hospital Universitario Central de Asturias, ES-33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), ES-33011 Oviedo, Spain
| |
Collapse
|
7
|
Jagust WJ, Koeppe RA, Rabinovici GD, Villemagne VL, Harrison TM, Landau SM. The ADNI PET Core at 20. Alzheimers Dement 2024; 20:7340-7349. [PMID: 39108002 PMCID: PMC11485322 DOI: 10.1002/alz.14165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 10/18/2024]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) PET Core has evolved over time, beginning with positron emission tomography (PET) imaging of a subsample of participants with [18F]fluorodeoxyglucose (FDG)-PET, adding tracers for measurement of β-amyloid, followed by tau tracers. This review examines the evolution of the ADNI PET Core, the novel aspects of PET imaging in each stage of ADNI, and gives an accounting of PET images available in the ADNI database. The ADNI PET Core has been and continues to be a rich resource that provides quantitative PET data and preprocessed PET images to the scientific community, allowing interrogation of both basic and clinically relevant questions. By standardizing methods across different PET scanners and multiple PET tracers, the Core has demonstrated the feasibility of large-scale, multi-center PET studies. Data managed and disseminated by the PET Core has been critical to defining pathophysiological models of Alzheimer's disease (AD) and helped to drive methods used in modern therapeutic trials. HIGHLIGHTS: The ADNI PET Core began with FDG-PET and now includes three amyloid and three tau PET ligands. The PET Core has standardized acquisition and analysis of multitracer PET images. The ADNI PET Core helped to develop methods that have facilitated clinical trials in AD.
Collapse
Affiliation(s)
- William J. Jagust
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Robert A. Koeppe
- Department of RadiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Gil D. Rabinovici
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | | | - Susan M. Landau
- Department of NeuroscienceUniversity of CaliforniaBerkeleyCaliforniaUSA
| | | |
Collapse
|
8
|
Salmon E, Collette F, Bastin C. Cerebral glucose metabolism in Alzheimer's disease. Cortex 2024; 179:50-61. [PMID: 39141935 DOI: 10.1016/j.cortex.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/05/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
18F-fluoro-deoxy-glucose positron emission tomography (FDG-PET) is a useful paraclinical exam for the diagnosis of Alzheimer's disease (AD). In this narrative review, we report seminal studies in clinically probable AD that have shown the importance of posterior brain metabolic decrease and the paradoxical variability of the hippocampal metabolism. The FDG-PET pattern was a sensitive indicator of AD in pathologically confirmed cases and it was used for differential diagnosis of dementia conditions. In prodromal AD, the AD FDG-PET pattern was observed in converters and predicted conversion. Automated data analysis techniques provided variable accuracy according to the reported indices and machine learning methods showed variable reliability of results. FDG-PET could confirm AD clinical heterogeneity and image data driven analyses identified hypometabolic subtypes with variable involvement of the hippocampus, reminiscent if the paradoxical FDG uptake. In studies dedicated to clinical and metabolic correlations, episodic memory was related to metabolism in the default mode network (and Papez's circuit) in prodromal and mild AD stages, and specific cognitive processes were associated to precisely distributed brain metabolism. Cerebral metabolic correlates of anosognosia could also be related to current neuropsychological models. AD FDG-PET pattern was reported in preclinical AD stages and related to cognition or to conversion to mild cognitive impairment (MCI). Using other biomarkers, the AD FDG-PET pattern was confirmed in AD participants with positive PET-amyloid. Intriguing observations reported increased metabolism related to brain amyloid and/or tau deposition. Preserved glucose metabolism sometimes appear as a compensation, but it was frequently detrimental and the nature of such a preservation of glucose metabolism remains an open question. Limbic metabolic involvement was frequently related to non-AD biomarkers profile and clinical stability, and it was reported in non-AD pathologies, such as the limbic predominant age-related encephalopathy (LATE). FDG-PET abnormalities observed in the absence of classical AD proteinopathies can be useful to search for pathological mechanisms and differential diagnosis of AD.
Collapse
Affiliation(s)
- Eric Salmon
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| | - Fabienne Collette
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| | - Christine Bastin
- GIGA Research, CRC Human Imaging, University of Liege, Liege, Belgium.
| |
Collapse
|
9
|
Rajendrakumar AL, Arbeev KG, Bagley O, Duan M, Yashin AI, Ukraintseva S. APOE4 and Infectious Diseases Jointly Contribute to Brain Glucose Hypometabolism, a Biomarker of Alzheimer's Pathology: New Findings from the ADNI. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313582. [PMID: 39314962 PMCID: PMC11419198 DOI: 10.1101/2024.09.13.24313582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Impaired brain glucose metabolism is a preclinical feature of neurodegenerative diseases such as Alzheimer's disease (AD). Infections may promote AD-related pathology. Therefore, we investigated the interplay between infections and APOE4, a strong genetic risk factor for AD. Methods We analyzed data on 1,509 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database using multivariate linear regression models. The outcomes were rank-normalized hypometabolic convergence index (HCI), statistical regions of interest (SROI) for AD, and mild cognitive impairment (MCI). Marginal mean estimates for infections, stratified by APOE4 carrier status, were then computed. Results Prior infections were associated with greater HCI [β=0.15, 95% CI: 0.03, 0.27, p=0.01]. The combined effects of infections and APOE4 carriers on HCI levels were significantly greater than either variable alone. Among APOE4 carriers, the estimated marginal mean was 0.62, rising to 0.77, with infections (p<0.001), indicating an interaction effect. Carriers with multiple infections showed greater hypometabolism (higher HCI), with an estimate of 0.44 (p=0.01) compared to 0.11 (p=0.08) for those with a single infection, revealing a dose-response relationship. The estimates for the association of infections with SROI AD and SROI MCI were β=-0.01 (p=0.02) and β=-0.01 (p=0.04), respectively. Conclusion Our findings suggest that infections and APOE4 jointly contribute to brain glucose hypometabolism and AD pathology, supporting a "multi-hit" mechanism in AD development.
Collapse
Affiliation(s)
- Aravind Lathika Rajendrakumar
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| |
Collapse
|
10
|
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
|
11
|
Kloske CM, Belloy ME, Blue EE, Bowman GR, Carrillo MC, Chen X, Chiba‐Falek O, Davis AA, Paolo GD, Garretti F, Gate D, Golden LR, Heinecke JW, Herz J, Huang Y, Iadecola C, Johnson LA, Kanekiyo T, Karch CM, Khvorova A, Koppes‐den Hertog SJ, Lamb BT, Lawler PE, Guen YL, Litvinchuk A, Liu C, Mahinrad S, Marcora E, Marino C, Michaelson DM, Miller JJ, Morganti JM, Narayan PS, Naslavsky MS, Oosthoek M, Ramachandran KV, Ramakrishnan A, Raulin A, Robert A, Saleh RNM, Sexton C, Shah N, Shue F, Sible IJ, Soranno A, Strickland MR, TCW J, Thierry M, Tsai L, Tuckey RA, Ulrich JD, van der Kant R, Wang N, Wellington CL, Weninger SC, Yassine HN, Zhao N, Bu G, Goate AM, Holtzman DM. Advancements in APOE and dementia research: Highlights from the 2023 AAIC Advancements: APOE conference. Alzheimers Dement 2024; 20:6590-6605. [PMID: 39031528 PMCID: PMC11497726 DOI: 10.1002/alz.13877] [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: 02/02/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION The apolipoprotein E gene (APOE) is an established central player in the pathogenesis of Alzheimer's disease (AD), with distinct apoE isoforms exerting diverse effects. apoE influences not only amyloid-beta and tau pathologies but also lipid and energy metabolism, neuroinflammation, cerebral vascular health, and sex-dependent disease manifestations. Furthermore, ancestral background may significantly impact the link between APOE and AD, underscoring the need for more inclusive research. METHODS In 2023, the Alzheimer's Association convened multidisciplinary researchers at the "AAIC Advancements: APOE" conference to discuss various topics, including apoE isoforms and their roles in AD pathogenesis, progress in apoE-targeted therapeutic strategies, updates on disease models and interventions that modulate apoE expression and function. RESULTS This manuscript presents highlights from the conference and provides an overview of opportunities for further research in the field. DISCUSSION Understanding apoE's multifaceted roles in AD pathogenesis will help develop targeted interventions for AD and advance the field of AD precision medicine. HIGHLIGHTS APOE is a central player in the pathogenesis of Alzheimer's disease. APOE exerts a numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The AAIC Advancements: APOE conference encouraged discussions and collaborations on understanding the role of APOE.
Collapse
Affiliation(s)
| | - Michael E. Belloy
- Department of Neurology and Neurological SciencesStanford University, StanfordPalo AltoCaliforniaUSA
- NeuroGenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
| | - Elizabeth E. Blue
- Division of Medical GeneticsDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Institute for Public Health GeneticsUniversity of WashingtonSeattleWashingtonUSA
| | - Gregory R. Bowman
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Xiaoying Chen
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Ornit Chiba‐Falek
- Division of Translational Brain SciencesDepartment of NeurologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Albert A. Davis
- Department of Neurology Hope Center for Neurological Disorders Washington University School of MedicineSt. LouisMissouriUSA
| | | | - Francesca Garretti
- Ronald M. Loeb Center for Alzheimer's DiseaseNew YorkNew YorkUSA
- Department of Genetics & Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David Gate
- The Ken & Ruth Davee Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Lesley R. Golden
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Jay W. Heinecke
- Department of MedicineUniversity of Washington, UV MedicineSeattleWashingtonUSA
| | - Joachim Herz
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Yadong Huang
- Gladstone Institute of Neurological DiseaseGladstone InstitutesSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Lance A. Johnson
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Takahisa Kanekiyo
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | - Celeste M. Karch
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
| | - Anastasia Khvorova
- RNA Therapeutic InstituteUMass Chan Medical SchoolWorcesterMassachusettsUSA
| | - Sascha J. Koppes‐den Hertog
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute Indiana University School of MedicineIndianapolisIndianaUSA
| | - Paige E. Lawler
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
- The Tracy Family SILQ CenterWashington University School of MedicineIndianapolisIndianaUSA
| | - Yann Le Guen
- Department of Neurology and Neurological SciencesStanford UniversityPalo AltoCaliforniaUSA
- Institut du Cerveau–Paris Brain Institute–ICMParisFrance
| | - Alexandra Litvinchuk
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Chia‐Chen Liu
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | | | - Edoardo Marcora
- Department of Genetics and Genomic SciencesNash Family Department of NeuroscienceIcahn Genomics Institute; Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Justin J. Miller
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biochemistry and Molecular BiophysicsWashington University School of MedicineSt. LouisMissouriUSA
| | - Josh M. Morganti
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Priyanka S. Narayan
- Genetics and Biochemistry BranchNational Institute of Diabetes and Digestive and Kidney DiseasesNational Institute of Neurological Disorders and StrokeCenter for Alzheimer's and Related Dementias (CARD)National Institutes of HealthMarylandUSA
| | - Michel S. Naslavsky
- Human Genome and Stem‐cell Research CenterBiosciences InstituteUniversity of São PauloRua do MataoSão PauloBrazil
- Hospital Israelita Albert EinsteinAvenida Albert EinsteinSão PauloBrazil
| | - Marlies Oosthoek
- Neurochemistry LaboratoryDepartment of Laboratory MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Kapil V. Ramachandran
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeuroscienceColumbia University Vagelos College of Physicians and SurgeonsNew YorkUSA
| | - Abhirami Ramakrishnan
- The Ken & Ruth Davee Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | | | - Aiko Robert
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Rasha N. M. Saleh
- Norwich Medical SchoolUniversity of East Anglia, UK Clinical and Chemical PathologyNorfolkUK
- Faculty of MedicineAlexandria UniversityAlexandria GovernorateEgypt
| | | | | | | | | | - Andrea Soranno
- Washington University in Saint Louis, St. Louis, Missouri, USASt. LouisMissouriUSA
| | - Michael R. Strickland
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
| | - Julia TCW
- Department of PharmacologyPhysiology & BiophysicsChobanian and Avedisian School of MedicineBoston UniversityBostonMassachusettsUSA
- Bioinformatics ProgramFaculty of Computing & Data SciencesBoston UniversityBostonMassachusettsUSA
| | - Manon Thierry
- Center for Cognitive NeurologyDepartment of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Li‐Huei Tsai
- Picower Institute for Learning and MemoryDepartment of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ryan A. Tuckey
- Department of NeurologyCenter for Neurodegeneration and Experimental TherapeuticsMedical Scientist Training ProgramUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jason D. Ulrich
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Rik van der Kant
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Na Wang
- Mayo Clinic RochesterRochesterMinnesotaUSA
| | - Cheryl L. Wellington
- Djavad Mowafaghian Centre for Brain Health Department of Pathology and Laboratory Medicine International Collaboration on Repair Discoveries School of Biomedical Engineering University of British ColumbiaVancouverCanada
| | | | - Hussein N. Yassine
- Department of NeurologyKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Na Zhao
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | - Guojun Bu
- Division of Life ScienceHong Kong University of Science and TechnologyClear Water BayKowloonHong Kong
| | - Alison M. Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's diseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David M. Holtzman
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
12
|
Daly J, De Luca F, Berens SC, Field AP, Rusted JM, Bird CM. The effect of apolipoprotein E genotype on spatial processing in humans: A meta-analysis and systematic review. Cortex 2024; 177:268-284. [PMID: 38878339 DOI: 10.1016/j.cortex.2024.05.006] [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: 01/03/2024] [Revised: 04/04/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
The ε4 allele of the apolipoprotein E (APOE4) gene is an established risk factor for Alzheimer's disease but its impact on cognition in healthy adults across the lifespan is unclear. One cognitive domain that is affected early in the course of Alzheimer's disease is spatial cognition, yet the evidence for APOE-related changes in spatial cognition is mixed. In this meta-analysis we assessed the impact of carrying the APOE4 allele on five subdomains of spatial cognition across the lifespan. We included studies of healthy human participants where an APOE4-carrier group (heterozygous or homozygous) could be compared to a homozygous group of APOE3-carriers. We identified 156 studies in total from three databases (Pubmed, Scopus and Web of Science) as well as through searching cited literature and contacting authors for unpublished data. 122 studies involving 32,547 participants were included in a meta-analysis, and the remaining studies are included in a descriptive review. APOE4 carriers scored significantly lower than APOE3 carriers (θˆ = -.08 [-.14, -.02]) on tests of spatial long-term memory; this effect was very small and was not modulated by age. On other subdomains of spatial cognition (spatial construction, spatial working memory, spatial reasoning, navigation) there were no effects of genotype. Overall, our results demonstrate that the APOE4 allele exerts little influence on spatial cognitive abilities in healthy adults.
Collapse
Affiliation(s)
- Jessica Daly
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Flavia De Luca
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Sam C Berens
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Andy P Field
- School of Psychology, University of Sussex, United Kingdom
| | | | - Chris M Bird
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom.
| |
Collapse
|
13
|
Beshir SA, Hussain N, Menon VB, Al Haddad AHI, Al Zeer RAK, Elnour AA. Advancements and Challenges in Antiamyloid Therapy for Alzheimer's Disease: A Comprehensive Review. Int J Alzheimers Dis 2024; 2024:2052142. [PMID: 39081336 PMCID: PMC11288696 DOI: 10.1155/2024/2052142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/20/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder caused by the accumulation of amyloid-beta (Aβ) proteins and neurofibrillary tangles in the brain. There have been recent advancements in antiamyloid therapy for AD. This narrative review explores the recent advancements and challenges in antiamyloid therapy. In addition, a summary of evidence from antiamyloid therapy trials is presented with a focus on lecanemab. Lecanemab is the most recently approved monoclonal antibody that targets Aβ protofibrils for the treatment of patients with early AD and mild cognitive impairment (MCI). Lecanemab was the first drug shown to slow cognitive decline in patients with MCI or early onset AD dementia when administered as an infusion once every two weeks. In the Clarity AD trial, lecanemab was associated with infusion-site reactions (26.4%) and amyloid-related imaging abnormalities (12.6%). The clinical relevance and long-term side effects of lecanemab require further longitudinal observation. However, several challenges must be addressed before the drug can be routinely used in clinical practice. The drug's route of administration, need for imaging and genetic testing, affordability, accessibility, infrastructure, and potential for serious side effects are some of these challenges. Lecanemab's approval has fueled interest in the potential of other antiamyloid therapies, such as donanemab. Future research must focus on developing strategies to prevent AD; identify easy-to-use validated plasma-based assays; and discover newer user-friendly, and cost-effective drugs that target multiple pathways in AD pathology.
Collapse
Affiliation(s)
- Semira Abdi Beshir
- Department of Pharmacy PracticeDubai Pharmacy College for Girls, Dubai, UAE
| | - Nadia Hussain
- Department of Pharmaceutical SciencesCollege of PharmacyAl Ain University, Al Ain, UAE
- AAU Health and Biomedical Research CentreAl Ain University, Abu Dhabi, UAE
| | | | - Amal H. I. Al Haddad
- Chief Operations OfficeSheikh Shakhbout Medical City (SSMC)PureHealth, Abu Dhabi, UAE
| | | | - Asim Ahmed Elnour
- AAU Health and Biomedical Research CentreAl Ain University, Abu Dhabi, UAE
- College of PharmacyAl Ain UniversityAbu Dhabi Campus, Abu Dhabi, UAE
| |
Collapse
|
14
|
Liu Y, Kwok W, Yoon H, Ryu JC, Stevens P, Hawkinson TR, Shedlock CJ, Ribas RA, Medina T, Keohane SB, Scharre D, Bruschweiler-Li L, Bruschweiler R, Gaultier A, Obrietan K, Sun RC, Yoon SO. Imbalance in Glucose Metabolism Regulates the Transition of Microglia from Homeostasis to Disease-Associated Microglia Stage 1. J Neurosci 2024; 44:e1563232024. [PMID: 38565291 PMCID: PMC11097271 DOI: 10.1523/jneurosci.1563-23.2024] [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: 08/16/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
Microglia undergo two-stage activation in neurodegenerative diseases, known as disease-associated microglia (DAM). TREM2 mediates the DAM2 stage transition, but what regulates the first DAM1 stage transition is unknown. We report that glucose dyshomeostasis inhibits DAM1 activation and PKM2 plays a role. As in tumors, PKM2 was aberrantly elevated in both male and female human AD brains, but unlike in tumors, it is expressed as active tetramers, as well as among TREM2+ microglia surrounding plaques in 5XFAD male and female mice. snRNAseq analyses of microglia without Pkm2 in 5XFAD mice revealed significant increases in DAM1 markers in a distinct metabolic cluster, which is enriched in genes for glucose metabolism, DAM1, and AD risk. 5XFAD mice incidentally exhibited a significant reduction in amyloid pathology without microglial Pkm2 Surprisingly, microglia in 5XFAD without Pkm2 exhibited increases in glycolysis and spare respiratory capacity, which correlated with restoration of mitochondrial cristae alterations. In addition, in situ spatial metabolomics of plaque-bearing microglia revealed an increase in respiratory activity. These results together suggest that it is not only glycolytic but also respiratory inputs that are critical to the development of DAM signatures in 5XFAD mice.
Collapse
Affiliation(s)
- Yuxi Liu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio 43210
| | - Witty Kwok
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio 43210
| | - Hyojung Yoon
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Jae Cheon Ryu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio 43210
| | - Patrick Stevens
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio 43210
| | - Tara R Hawkinson
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Cameron J Shedlock
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Roberto A Ribas
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Terrymar Medina
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Shannon B Keohane
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Douglas Scharre
- Department of Neurology, The Ohio State University, Columbus, Ohio 43210
| | - Lei Bruschweiler-Li
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| | - Rafael Bruschweiler
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210
| | - Alban Gaultier
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, 22908
| | - Karl Obrietan
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida 32610
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, Florida, 32610
| | - Sung Ok Yoon
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
15
|
Liu X, Beck T, Dhana K, Tangney CC, Desai P, Krueger K, Evans DA, Rajan KB. Dietary fats and the APOE-e4 risk allele in relation to cognitive decline: a longitudinal investigation in a biracial population sample. J Nutr Health Aging 2024; 28:100211. [PMID: 38507884 PMCID: PMC11623058 DOI: 10.1016/j.jnha.2024.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND APOE-e4 is the strongest genetic risk factor for Alzheimer's disease. However, the influence of APOE-e4 on dietary fat intake and cognition has not been investigated. OBJECTIVE We aim to examine the association of types of dietary fat and their association to cognitive decline among those with and without the APOE-e4 allele. METHODS The study included 3,360 Chicago Health and Aging Project (CHAP) participants from four Southside Chicago communities. Global cognition was assessed using a composite score of episodic memory, perceptual speed, MMSE, and diet using a 144-item food frequency questionnaire. APOE genotype was assessed by the hME Sequenom mass-array platform. Longitudinal mixed-effect regression models were used to examine the association of dietary fat and the APOE-e4 allele with cognitive decline, adjusted for age, sex, education, smoking status, and calorie intake. RESULTS The present study involved 3,360 participants with a mean age of 74 at baseline, 62% African Americans, 63% females, and a mean follow-up of 7.8 years. Among participants with the APOE-e4 risk allele, higher intakes of total and saturated fat (SFA) were associated with a faster decline in global cognition. Among individuals with the APOE-e4 risk allele, a 5% increase in calories from SFA was associated with a 21% faster decline (β = -0.0197, P = 0.0038). In contrast, a higher intake of long-chain n-3 polyunsaturated fatty acids (LC-n3 PUFA) was associated with a slower rate of decline in global cognition among APOE-e4 carriers. Specifically, for every 1% energy increment from LC-n3 PUFA, the annual rate of global cognitive decline was slower by 0.024 standardized unit (SD 0.010, P = 0.023), about 30.4% slower annual cognitive decline. Higher SFA or other types of dietary fat were not associated with cognitive decline among APOE-e4 non-carriers. CONCLUSIONS Our study found a significant association between SFA and faster cognitive decline, LC-n3 PUFA and slower cognitive decline among those with the APOE-e4 allele. Our findings suggested that higher intake of SFA might contribute faster cognitive decline in combination with APOE-e4 whereas LC-n3 PUFA might compensate the adverse effects of APOE-e4. The interaction between intakes of different types of dietary fat and APOE-e4 on cognitive function warrants further research.
Collapse
Affiliation(s)
- Xiaoran Liu
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Todd Beck
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Klodian Dhana
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Christy C Tangney
- Department of Clinical Nutrition & Preventive Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Pankaja Desai
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kristin Krueger
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Denis A Evans
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kumar B Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, IL, USA; Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
16
|
Yang Q, Han X, Ye M, Jiang T, Wang B, Zhang Z, Li F. Association of genetically predicted 486 blood metabolites on the risk of Alzheimer's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1372605. [PMID: 38681667 PMCID: PMC11047179 DOI: 10.3389/fnagi.2024.1372605] [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: 01/18/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Background Studies have reported that metabolic disturbance exhibits in patients with Alzheimer's disease (AD). Still, the presence of definitive evidence concerning the genetic effect of metabolites on AD risk remains insufficient. A systematic exploration of the genetic association between blood metabolites and AD would contribute to the identification of new targets for AD screening and prevention. Methods We conducted an exploratory two-sample Mendelian randomization (MR) study aiming to preliminarily identify the potential metabolites involved in AD development. A genome-wide association study (GWAS) involving 7,824 participants provided information on 486 human blood metabolites. Outcome information was obtained from a large-scale GWAS meta-analysis of AD, encompassing 21,982 cases and 41,944 controls of Europeans. The primary two-sample MR analysis utilized the inverse variance weighted (IVW) model while supplementary analyses used Weighted median (WM), MR Egger, Simple mode, and Weighted mode, followed by sensitivity analyses such as the heterogeneity test, horizontal pleiotropy test, and leave-one-out analysis. For the further identification of metabolites, replication and meta-analysis with FinnGen data, steiger test, linkage disequilibrium score regression, confounding analysis, and were conducted for further evaluation. Multivariable MR was performed to assess the direct effect of metabolites on AD. Besides, an extra replication analysis with EADB data was conducted for final evaluation of the most promising findings. Results After rigorous genetic variant selection, IVW, complementary analysis, sensitivity analysis, replication and meta-analysis with the FinnGen data, five metabolites (epiandrosterone sulfate, X-12680, pyruvate, docosapentaenoate, and 1-stearoylglycerophosphocholine) were identified as being genetically associated with AD. MVMR analysis disclosed that genetically predicted these four known metabolites can directly influence AD independently of other metabolites. Only epiandrosterone sulfate and X-12680 remained suggestive significant associations with AD after replication analysis with the EADB data. Conclusion By integrating genomics with metabonomics, this study furnishes evidence substantiating the genetic association of epiandrosterone sulfate and X-12680 with AD. These findings hold significance for the screening, prevention, and treatment strategies for AD.
Collapse
Affiliation(s)
- Qiqi Yang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Xinyu Han
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Min Ye
- The First Clinical Medical School, Anhui University of Chinese Medicine, Hefei, China
| | - Tianxin Jiang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Baoguo Wang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Zhenfeng Zhang
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Fei Li
- Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
- Intelligent Manufacturing Institute, Hefei University of Technology, Hefei, China
| |
Collapse
|
17
|
Riviere M, Langbaum JB, Turner RS, Rinne JO, Sui Y, Cazorla P, Ricart J, Meneses K, Caputo A, Tariot PN, Reiman EM, Graf A. Effects of the active amyloid beta immunotherapy CAD106 on PET measurements of amyloid plaque deposition in cognitively unimpaired APOE ε4 homozygotes. Alzheimers Dement 2024; 20:1839-1850. [PMID: 38145469 PMCID: PMC10984441 DOI: 10.1002/alz.13532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION Alzheimer's Prevention Initiative Generation Study 1 evaluated amyloid beta (Aβ) active immunotherapy (vaccine) CAD106 and BACE-1 inhibitor umibecestat in cognitively unimpaired 60- to 75-year-old participants at genetic risk for Alzheimer's disease (AD). The study was reduced in size and terminated early. Results from the CAD106 cohort are presented. METHODS Sixty-five apolipoprotein E ε4 homozygotes with/without amyloid deposition received intramuscular CAD106 450 μg (n = 42) or placebo (n = 23) at baseline; Weeks 1, 7, 13; and quarterly; 51 of them had follow-up Aβ positron emission tomography (PET) scans at 18 to 24 months. RESULTS CAD106 induced measurable serum Aβ immunoglobulin G titers in 41/42 participants, slower rates of Aβ plaque accumulation (mean [standard deviation] annualized change from baseline in amyloid PET Centiloid: -0.91[5.65] for CAD106 versus 8.36 [6.68] for placebo; P < 0.001), and three amyloid-related imaging abnormality cases (one symptomatic). DISCUSSION Despite early termination, these findings support the potential value of conducting larger prevention trials of Aβ active immunotherapies in individuals at risk for AD. HIGHLIGHTS This was the first amyloid-lowering prevention trial in persons at genetic risk of late-onset Alzheimer's disease (AD). Active immunotherapy targeting amyloid (CAD106) was tested in this prevention trial. CAD106 significantly slowed down amyloid plaque deposition in apolipoprotein E homozygotes. CAD106 was generally safe and well tolerated, with only three amyloid-related imaging abnormality cases (one symptomatic). Such an approach deserves further evaluation in larger AD prevention trials.
Collapse
Affiliation(s)
| | | | - R. Scott Turner
- Department of NeurologyGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Juha O. Rinne
- Turku PET CentreUniversity of Turku and Turku University HospitalTurkuFinland
- Department of NeurologyCRST – Clinical Research Services TurkuTurkuFinland
| | - Yihan Sui
- Clinical Development, NeuroscienceNovartis PharmaceuticalsEast HanoverNew JerseyUSA
| | - Pilar Cazorla
- Clinical Development, NeuroscienceNovartis PharmaceuticalsEast HanoverNew JerseyUSA
| | - Javier Ricart
- Clinical Development, NeuroscienceNovartis Farmaceutica SABarcelonaSpain
| | - Kathleen Meneses
- Clinical Development, NeuroscienceNovartis PharmaceuticalsEast HanoverNew JerseyUSA
| | - Angelika Caputo
- Clinical Development, NeuroscienceNovartis Pharma AGBaselSwitzerland
| | | | | | - Ana Graf
- Clinical Development, NeuroscienceNovartis Pharma AGBaselSwitzerland
| |
Collapse
|
18
|
Mak E, Dounavi ME, Operto G, Ziukelis ET, Jones PS, Low A, Swann P, Newton C, Muniz Terrera G, Malhotra P, Koychev I, Falcon C, Mackay C, Lawlor B, Naci L, Wells K, Ritchie C, Ritchie K, Su L, Gispert JD, O’Brien JT. APOE ɛ4 exacerbates age-dependent deficits in cortical microstructure. Brain Commun 2024; 6:fcad351. [PMID: 38384997 PMCID: PMC10881196 DOI: 10.1093/braincomms/fcad351] [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: 05/30/2023] [Revised: 10/20/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024] Open
Abstract
The apolipoprotein E ɛ4 allele is the primary genetic risk factor for the sporadic type of Alzheimer's disease. However, the mechanisms by which apolipoprotein E ɛ4 are associated with neurodegeneration are still poorly understood. We applied the Neurite Orientation Dispersion Model to characterize the effects of apolipoprotein ɛ4 and its interactions with age and education on cortical microstructure in cognitively normal individuals. Data from 1954 participants were included from the PREVENT-Dementia and ALFA (ALzheimer and FAmilies) studies (mean age = 57, 1197 non-carriers and 757 apolipoprotein E ɛ4 carriers). Structural MRI datasets were processed with FreeSurfer v7.2. The Microstructure Diffusion Toolbox was used to derive Orientation Dispersion Index maps from diffusion MRI datasets. Primary analyses were focused on (i) the main effects of apolipoprotein E ɛ4, and (ii) the interactions of apolipoprotein E ɛ4 with age and education on lobar and vertex-wise Orientation Dispersion Index and implemented using Permutation Analysis of Linear Models. There were apolipoprotein E ɛ4 × age interactions in the temporo-parietal and frontal lobes, indicating steeper age-dependent Orientation Dispersion Index changes in apolipoprotein E ɛ4 carriers. Steeper age-related Orientation Dispersion Index declines were observed among apolipoprotein E ɛ4 carriers with lower years of education. We demonstrated that apolipoprotein E ɛ4 worsened age-related Orientation Dispersion Index decreases in brain regions typically associated with atrophy patterns of Alzheimer's disease. This finding also suggests that apolipoprotein E ɛ4 may hasten the onset age of dementia by accelerating age-dependent reductions in cortical Orientation Dispersion Index.
Collapse
Affiliation(s)
- Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Grégory Operto
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
| | - Elina T Ziukelis
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Peter Simon Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Audrey Low
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Peter Swann
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Coco Newton
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Paresh Malhotra
- Department of Brain Sciences, Imperial College, London W12 0NN, UK
| | - Ivan Koychev
- Department of Psychiatry, Oxford University, Oxford OX3 7JX, UK
| | - Carles Falcon
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Clare Mackay
- Department of Psychiatry, Oxford University, Oxford OX3 7JX, UK
| | - Brian Lawlor
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin D02 PX31, Ireland
| | - Lorina Naci
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin D02 PX31, Ireland
| | - Katie Wells
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Craig Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen Ritchie
- Institut National de la Santé et de la Recherche Médicale, U1061 Neuropsychiatrie, Montpellier 34093, France
- Faculty of Medicine, University of Montpellier, Montpellier 34093, France
| | - Li Su
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona 08005, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - John T O’Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
19
|
Cammisuli DM, Tuena C, Riva G, Repetto C, Axmacher N, Chandreswaran V, Isella V, Pomati S, Zago S, Difonzo T, Pavanello G, Prete LA, Stramba-Badiale M, Mauro A, Cattaldo S, Castelnuovo G. Exploring the Remediation of Behavioral Disturbances of Spatial Cognition in Community-Dwelling Senior Citizens with Mild Cognitive Impairment via Innovative Technological Apparatus (BDSC-MCI Project): Protocol for a Prospective, Multi-Center Observational Study. J Pers Med 2024; 14:192. [PMID: 38392625 PMCID: PMC10890288 DOI: 10.3390/jpm14020192] [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: 12/22/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Spatial navigation (SN) has been reported to be one of the first cognitive domains to be affected in Alzheimer's disease (AD), which occurs as a result of progressive neuropathology involving specific brain areas. Moreover, the epsilon 4 isoform of apolipoprotein-E (APOE-ε4) has been associated with both sporadic and familial late-onset AD, and patients with mild cognitive impairment (MCI) due to AD are more likely to progressively deteriorate. Spatial navigation performance will be examined on a sample of 76 community-dwelling senior citizens (25 healthy controls; 25 individuals with subjective cognitive decline (SCD); and 26 patients with MCI due to AD) via a virtual computer-based task (i.e., the AppleGame) and a naturalistic task (i.e., the Detour Navigation Test-modified version) for which a wearable device with sensors will be used for recording gait data and revealing physiological parameters that may be associated with spatial disorientation. We expect that patients with MCI due to AD and APOE-ε4 carriers will show altered SN performances compared to individuals with SCD and healthy controls in the experimental tasks, and that VR testing may predict ecological performance. Impaired SN performances in people at increased risk of developing AD may inform future cognitive rehabilitation protocols for counteracting spatial disorientation that may occur during elders' traveling to unfamiliar locations. The research protocol has been approved by the Ethics Committee of the Istituto Auxologico Italiano. Findings will be published in peer-reviewed medical journals and discussed in national and international congresses.
Collapse
Affiliation(s)
| | - Cosimo Tuena
- Applied Technology for Neuro-Psychology Lab, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy; (C.T.); (G.R.)
| | - Giuseppe Riva
- Applied Technology for Neuro-Psychology Lab, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy; (C.T.); (G.R.)
- Human Technology Lab, Catholic University, 20145 Milan, Italy
| | - Claudia Repetto
- Department of Psychology, Catholic University, 20123 Milan, Italy; (D.M.C.); (C.R.)
| | - Nikolai Axmacher
- Department of Neuropsychology, Faculty of Psychology, Institute of Cognitive Neuroscience, Ruhr University, 44801 Bochum, Germany (V.C.)
| | - Varnan Chandreswaran
- Department of Neuropsychology, Faculty of Psychology, Institute of Cognitive Neuroscience, Ruhr University, 44801 Bochum, Germany (V.C.)
| | - Valeria Isella
- Department of Neurology, School of Medicine, University of Milano-Bicocca, 20126 Milan, Italy;
- Milan Center for Neurosciences, 20133 Milan, Italy
| | - Simone Pomati
- Neurology Unit, Luigi Sacco University Hospital, 20157 Milan, Italy;
| | - Stefano Zago
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, 20122 Milan, Italy; (S.Z.); (T.D.)
| | - Teresa Difonzo
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, 20122 Milan, Italy; (S.Z.); (T.D.)
| | - Giada Pavanello
- School of Specialization in Clinical Psychology, Catholic University, 20123 Milan, Italy; (G.P.); (L.A.P.)
| | - Lorenzo Augusto Prete
- School of Specialization in Clinical Psychology, Catholic University, 20123 Milan, Italy; (G.P.); (L.A.P.)
| | - Marco Stramba-Badiale
- Department of Geriatrics and Cardiovascular Medicine, IRCCS Istituto Auxologico Italiano, 20145 Milan, Italy;
| | - Alessandro Mauro
- “Rita Levi Montalcini” Department of Neurosciences, University of Turin, 10126 Turin, Italy;
- Neurology and Neurorehabilitation Unit, IRCCS Istituto Auxologico Italiano, “San Giuseppe” Hospital, 33081 Piancavallo, Italy
| | - Stefania Cattaldo
- Clinic Neurobiology Laboratory, IRCCS Istituto Auxologico Italiano, “San Giuseppe” Hospital, 33081 Piancavallo, Italy;
| | - Gianluca Castelnuovo
- Department of Psychology, Catholic University, 20123 Milan, Italy; (D.M.C.); (C.R.)
- Clinical Psychology Research Laboratory, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
20
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. Mol Neurodegener 2024; 19:13. [PMID: 38282024 PMCID: PMC10823734 DOI: 10.1186/s13024-023-00690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/28/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. METHODS We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of techniques, including genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. RESULTS We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. CONCLUSION NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Zhen-Xian Niou
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Andrea Enriquez
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jacob LaMar
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
- Present address: Department of Biomedical Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Jui-Yen Huang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
| | - Karen Ling
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Paymaan Jafar-Nejad
- Neuroscience Drug Discovery, Ionis Pharmaceuticals, Inc., 2855, Gazelle Court, Carlsbad, CA, 92010, USA
| | - Jonathan Gilley
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Michael P Coleman
- Department of Clinical Neuroscience, Cambridge University, Cambridge, UK
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
21
|
Li XY, Yuan LX, Ding CC, Guo TF, Du WY, Jiang JH, Jessen F, Zang YF, Han Y. Convergent Multimodal Imaging Abnormalities in the Dorsal Precuneus in Subjective Cognitive Decline. J Alzheimers Dis 2024; 101:589-601. [PMID: 39213059 DOI: 10.3233/jad-231360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background A range of imaging modalities have reported Alzheimer's disease-related abnormalities in individuals experiencing subjective cognitive decline (SCD). However, there has been no consistent local abnormality identified across multiple neuroimaging modalities for SCD. Objective We aimed to investigate the convergent local alterations in amyloid-β (Aβ) deposition, glucose metabolism, and resting-state functional MRI (RS-fMRI) metrics in SCD. Methods Fifty SCD patients (66.4±5.7 years old, 19 men [38%]) and 15 normal controls (NC) (66.3±4.4 years old, 5 men [33.3%]) were scanned with both [18F]-florbetapir PET and [18F]-fluorodeoxyglucose PET, as well as simultaneous RS-fMRI from February 2018 to November 2018. Voxel-wise metrics were retrospectively analyzed, including Aβ deposition, glucose metabolism, amplitude of low frequency fluctuation (ALFF), regional homogeneity (ReHo), and degree centrality(DC). Results The SCD group showed increased Aβ deposition and glucose metabolism (p < 0.05, corrected), as well as decreased ALFF, ReHo, and DC (p < 0.05, uncorrected) in the left dorsal precuneus (dPCu). Furthermore, the dPCu illustrated negative resting-state functional connectivity with the default mode network. Regarding global Aβ deposition positivity, the Aβ deposition in the left dPCu showed a gradient change, i.e., Aβ positive SCD > Aβ negative SCD > Aβ negative NC. Additionally, both Aβ positive SCD and Aβ negative SCD showed increased glucose metabolism and decreased RS-fMRI metrics in the dPCu. Conclusions The dorsal precuneus, an area implicated in early AD, shows convergent neuroimaging alterations in SCD, and might be more related to other cognitive functions (e.g., unfocused attention) than episodic memory.
Collapse
Affiliation(s)
- Xuan-Yu Li
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing, China
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li-Xia Yuan
- Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, China
- Institutes of Psychological Sciences, Hangzhou Normal University, Hangzhou, China
| | - Chang-Chang Ding
- Department of Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Teng-Fei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Wen-Ying Du
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing, China
| | - Jie-Hui Jiang
- Department of Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Yu-Feng Zang
- Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, China
- Institutes of Psychological Sciences, Hangzhou Normal University, Hangzhou, China
| | - Ying Han
- Department of Neurology, XuanWu Hospital of Capital Medical University, Beijing, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- School of Biomedical Engineering, Hainan University, Haikou, China
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
- The Central Hospital of Karamay, Xinjiang, China
| |
Collapse
|
22
|
Ji X, Peng X, Tang H, Pan H, Wang W, Wu J, Chen J, Wei N. Alzheimer's disease phenotype based upon the carrier status of the apolipoprotein E ɛ4 allele. Brain Pathol 2024; 34:e13208. [PMID: 37646624 PMCID: PMC10711266 DOI: 10.1111/bpa.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
The apolipoprotein E ɛ4 allele (APOE4) is universally acknowledged as the most potent genetic risk factor for Alzheimer's disease (AD). APOE4 promotes the initiation and progression of AD. Although the underlying mechanisms are unclearly understood, differences in lipid-bound affinity among the three APOE isoforms may constitute the basis. The protein APOE4 isoform has a high affinity with triglycerides and cholesterol. A distinction in lipid metabolism extensively impacts neurons, microglia, and astrocytes. APOE4 carriers exhibit phenotypic differences from non-carriers in clinical examinations and respond differently to multiple treatments. Therefore, we hypothesized that phenotypic classification of AD patients according to the status of APOE4 carrier will help specify research and promote its use in diagnosing and treating AD. Recent reviews have mainly evaluated the differences between APOE4 allele carriers and non-carriers from gene to protein structures, clinical features, neuroimaging, pathology, the neural network, and the response to various treatments, and have provided the feasibility of phenotypic group classification based on APOE4 carrier status. This review will facilitate the application of APOE phenomics concept in clinical practice and promote further medical research on AD.
Collapse
Affiliation(s)
- Xiao‐Yu Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Xin‐Yuan Peng
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Hai‐Liang Tang
- Fudan University Huashan Hospital, Department of Neurosurgery, State Key Laboratory for Medical NeurobiologyInstitutes of Brain Science, Shanghai Medical College‐Fudan UniversityShanghaiChina
| | - Hui Pan
- Shantou Longhu People's HospitalShantouGuangdongChina
| | - Wei‐Tang Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Jie Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Jian Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Nai‐Li Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| |
Collapse
|
23
|
Panyard DJ, McKetney J, Deming YK, Morrow AR, Ennis GE, Jonaitis EM, Van Hulle CA, Yang C, Sung YJ, Ali M, Kollmorgen G, Suridjan I, Bayfield A, Bendlin BB, Zetterberg H, Blennow K, Cruchaga C, Carlsson CM, Johnson SC, Asthana S, Coon JJ, Engelman CD. Large-scale proteome and metabolome analysis of CSF implicates altered glucose and carbon metabolism and succinylcarnitine in Alzheimer's disease. Alzheimers Dement 2023; 19:5447-5470. [PMID: 37218097 PMCID: PMC10663389 DOI: 10.1002/alz.13130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/23/2023] [Accepted: 04/04/2023] [Indexed: 05/24/2023]
Abstract
INTRODUCTION A hallmark of Alzheimer's disease (AD) is the aggregation of proteins (amyloid beta [A] and hyperphosphorylated tau [T]) in the brain, making cerebrospinal fluid (CSF) proteins of particular interest. METHODS We conducted a CSF proteome-wide analysis among participants of varying AT pathology (n = 137 participants; 915 proteins) with nine CSF biomarkers of neurodegeneration and neuroinflammation. RESULTS We identified 61 proteins significantly associated with the AT category (P < 5.46 × 10-5 ) and 636 significant protein-biomarker associations (P < 6.07 × 10-6 ). Proteins from glucose and carbon metabolism pathways were enriched among amyloid- and tau-associated proteins, including malate dehydrogenase and aldolase A, whose associations with tau were replicated in an independent cohort (n = 717). CSF metabolomics identified and replicated an association of succinylcarnitine with phosphorylated tau and other biomarkers. DISCUSSION These results implicate glucose and carbon metabolic dysregulation and increased CSF succinylcarnitine levels with amyloid and tau pathology in AD. HIGHLIGHTS Cerebrospinal fluid (CSF) proteome enriched for extracellular, neuronal, immune, and protein processing. Glucose/carbon metabolic pathways enriched among amyloid/tau-associated proteins. Key glucose/carbon metabolism protein associations independently replicated. CSF proteome outperformed other omics data in predicting amyloid/tau positivity. CSF metabolomics identified and replicated a succinylcarnitine-phosphorylated tau association.
Collapse
Affiliation(s)
- Daniel J. Panyard
- Department of Population Health Sciences, University of Wisconsin-Madison; 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| | - Justin McKetney
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison; Madison, WI 53706, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison; Madison, WI 53506, United States of America
| | - Yuetiva K. Deming
- Department of Population Health Sciences, University of Wisconsin-Madison; 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
| | - Autumn R. Morrow
- Department of Population Health Sciences, University of Wisconsin-Madison; 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| | - Gilda E. Ennis
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
| | - Erin M. Jonaitis
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison; 610 Walnut Street, 9 Floor, Madison, WI 53726, United States of America
| | - Carol A. Van Hulle
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine; St Louis, MO 63110, United States of America
- NeuroGenomics and Informatics Center, Washington University School of Medicine; St Louis, MO 63110, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine; St Louis, MO 63110, United States of America
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine; St Louis, MO 63110, United States of America
- NeuroGenomics and Informatics Center, Washington University School of Medicine; St Louis, MO 63110, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine; St Louis, MO 63110, United States of America
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine; St Louis, MO 63110, United States of America
- NeuroGenomics and Informatics Center, Washington University School of Medicine; St Louis, MO 63110, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine; St Louis, MO 63110, United States of America
| | | | | | | | - Barbara B. Bendlin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison; 610 Walnut Street, 9 Floor, Madison, WI 53726, United States of America
- William S. Middleton Memorial Veterans Hospital; 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital; Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology; London, UK
- UK Dementia Research Institute at UCL; London, UK
- Hong Kong Center for Neurodegenerative Diseases; Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg; Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital; Mölndal, Sweden
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine; St Louis, MO 63110, United States of America
- NeuroGenomics and Informatics Center, Washington University School of Medicine; St Louis, MO 63110, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine; St Louis, MO 63110, United States of America
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison; 610 Walnut Street, 9 Floor, Madison, WI 53726, United States of America
- William S. Middleton Memorial Veterans Hospital; 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- Wisconsin Alzheimer’s Institute, University of Wisconsin-Madison; 610 Walnut Street, 9 Floor, Madison, WI 53726, United States of America
- William S. Middleton Memorial Veterans Hospital; 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin-Madison; 600 Highland Avenue, J5/1 Mezzanine, Madison, WI 53792, United States of America
- Department of Medicine, University of Wisconsin-Madison; 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705, United States of America
- William S. Middleton Memorial Veterans Hospital; 2500 Overlook Terrace, Madison, WI 53705, United States of America
| | - Joshua J. Coon
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison; Madison, WI 53706, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison; Madison, WI 53506, United States of America
- Morgridge Institute for Research; Madison, WI 53706, United States of America
- Department of Chemistry, University of Wisconsin-Madison; Madison, WI 53506, United States of America
| | - Corinne D. Engelman
- Department of Population Health Sciences, University of Wisconsin-Madison; 610 Walnut Street, 707 WARF Building, Madison, WI 53726, United States of America
| |
Collapse
|
24
|
Sudwarts A, Thinakaran G. Alzheimer's genes in microglia: a risk worth investigating. Mol Neurodegener 2023; 18:90. [PMID: 37986179 PMCID: PMC10662636 DOI: 10.1186/s13024-023-00679-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Despite expressing many key risk genes, the role of microglia in late-onset Alzheimer's disease pathophysiology is somewhat ambiguous, with various phenotypes reported to be either harmful or protective. Herein, we review some key findings from clinical and animal model investigations, discussing the role of microglial genetics in mediating perturbations from homeostasis. We note that impairment to protective phenotypes may include prolonged or insufficient microglial activation, resulting in dysregulated metabolomic (notably lipid-related) processes, compounded by age-related inflexibility in dynamic responses. Insufficiencies of mouse genetics and aggressive transgenic modelling imply severe limitations in applying current methodologies for aetiological investigations. Despite the shortcomings, widely used amyloidosis and tauopathy models of the disease have proven invaluable in dissecting microglial functional responses to AD pathophysiology. Some recent advances have brought modelling tools closer to human genetics, increasing the validity of both aetiological and translational endeavours.
Collapse
Affiliation(s)
- Ari Sudwarts
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| | - Gopal Thinakaran
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
25
|
Jia D, Wang F, Yu H. Systemic alterations of tricarboxylic acid cycle enzymes in Alzheimer's disease. Front Neurosci 2023; 17:1206688. [PMID: 37575300 PMCID: PMC10413568 DOI: 10.3389/fnins.2023.1206688] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Mitochondrial dysfunction, especially tricarboxylic acid (TCA) cycle arrest, is strongly associated with Alzheimer's disease (AD), however, its systemic alterations in the central and peripheral of AD patients are not well defined. Here, we performed an integrated analysis of AD brain and peripheral blood cells transcriptomics to reveal the expression levels of nine TCA cycle enzymes involving 35 genes. The results showed that TCA cycle related genes were consistently down-regulated in the AD brain, whereas 11 genes were increased and 16 genes were decreased in the peripheral system. Pearson analysis of the TCA cycle genes with Aβ, Tau and mini-mental state examination (MMSE) revealed several significant correlated genes, including pyruvate dehydrogenase complex subunit (PDHB), isocitrate dehydrogenase subunits (IDH3B, IDH3G), 2-oxoglutarate dehydrogenase complex subunit (DLD), succinyl-CoA synthetase subunit (SUCLA2), malate dehydrogenase subunit (MDH1). In addition, SUCLA2, MDH1, and PDHB were also uniformly down-regulated in peripheral blood cells, suggesting that they may be candidate biomarkers for the early diagnosis of AD. Taken together, TCA cycle enzymes were systemically altered in AD progression, PDHB, SUCLA2, and MDH1 may be potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Dongdong Jia
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Fangzhou Wang
- Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Haitao Yu
- The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
- Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
26
|
Yang S, Park JH, Lu HC. Axonal energy metabolism, and the effects in aging and neurodegenerative diseases. Mol Neurodegener 2023; 18:49. [PMID: 37475056 PMCID: PMC10357692 DOI: 10.1186/s13024-023-00634-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023] Open
Abstract
Human studies consistently identify bioenergetic maladaptations in brains upon aging and neurodegenerative disorders of aging (NDAs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and Amyotrophic lateral sclerosis. Glucose is the major brain fuel and glucose hypometabolism has been observed in brain regions vulnerable to aging and NDAs. Many neurodegenerative susceptible regions are in the topological central hub of the brain connectome, linked by densely interconnected long-range axons. Axons, key components of the connectome, have high metabolic needs to support neurotransmission and other essential activities. Long-range axons are particularly vulnerable to injury, neurotoxin exposure, protein stress, lysosomal dysfunction, etc. Axonopathy is often an early sign of neurodegeneration. Recent studies ascribe axonal maintenance failures to local bioenergetic dysregulation. With this review, we aim to stimulate research in exploring metabolically oriented neuroprotection strategies to enhance or normalize bioenergetics in NDA models. Here we start by summarizing evidence from human patients and animal models to reveal the correlation between glucose hypometabolism and connectomic disintegration upon aging/NDAs. To encourage mechanistic investigations on how axonal bioenergetic dysregulation occurs during aging/NDAs, we first review the current literature on axonal bioenergetics in distinct axonal subdomains: axon initial segments, myelinated axonal segments, and axonal arbors harboring pre-synaptic boutons. In each subdomain, we focus on the organization, activity-dependent regulation of the bioenergetic system, and external glial support. Second, we review the mechanisms regulating axonal nicotinamide adenine dinucleotide (NAD+) homeostasis, an essential molecule for energy metabolism processes, including NAD+ biosynthetic, recycling, and consuming pathways. Third, we highlight the innate metabolic vulnerability of the brain connectome and discuss its perturbation during aging and NDAs. As axonal bioenergetic deficits are developing into NDAs, especially in asymptomatic phase, they are likely exaggerated further by impaired NAD+ homeostasis, the high energetic cost of neural network hyperactivity, and glial pathology. Future research in interrogating the causal relationship between metabolic vulnerability, axonopathy, amyloid/tau pathology, and cognitive decline will provide fundamental knowledge for developing therapeutic interventions.
Collapse
Affiliation(s)
- Sen Yang
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Jung Hyun Park
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- The Linda and Jack Gill Center for Biomolecular Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA.
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
27
|
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
|
28
|
Lee EG, Leong L, Chen S, Tulloch J, Yu CE. APOE Locus-Associated Mitochondrial Function and Its Implication in Alzheimer's Disease and Aging. Int J Mol Sci 2023; 24:10440. [PMID: 37445616 PMCID: PMC10341489 DOI: 10.3390/ijms241310440] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The Apolipoprotein E (APOE) locus has garnered significant clinical interest because of its association with Alzheimer's disease (AD) and longevity. This genetic association appears across multiple genes in the APOE locus. Despite the apparent differences between AD and longevity, both conditions share a commonality of aging-related changes in mitochondrial function. This commonality is likely due to accumulative biological effects partly exerted by the APOE locus. In this study, we investigated changes in mitochondrial structure/function-related markers using oxidative stress-induced human cellular models and postmortem brains (PMBs) from individuals with AD and normal controls. Our results reveal a range of expressional alterations, either upregulated or downregulated, in these genes in response to oxidative stress. In contrast, we consistently observed an upregulation of multiple APOE locus genes in all cellular models and AD PMBs. Additionally, the effects of AD status on mitochondrial DNA copy number (mtDNA CN) varied depending on APOE genotype. Our findings imply a potential coregulation of APOE locus genes possibly occurring within the same topologically associating domain (TAD) of the 3D chromosome conformation. The coordinated expression of APOE locus genes could impact mitochondrial function, contributing to the development of AD or longevity. Our study underscores the significant role of the APOE locus in modulating mitochondrial function and provides valuable insights into the underlying mechanisms of AD and aging, emphasizing the importance of this locus in clinical research.
Collapse
Affiliation(s)
- Eun-Gyung Lee
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Lesley Leong
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Sunny Chen
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Jessica Tulloch
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Chang-En Yu
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Yang S, Niou ZX, Enriquez A, LaMar J, Huang JY, Ling K, Jafar-Nejad P, Gilley J, Coleman MP, Tennessen JM, Rangaraju V, Lu HC. NMNAT2 supports vesicular glycolysis via NAD homeostasis to fuel fast axonal transport. RESEARCH SQUARE 2023:rs.3.rs-2859584. [PMID: 37292715 PMCID: PMC10246254 DOI: 10.21203/rs.3.rs-2859584/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Bioenergetic maladaptations and axonopathy are often found in the early stages of neurodegeneration. Nicotinamide adenine dinucleotide (NAD), an essential cofactor for energy metabolism, is mainly synthesized by Nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2) in CNS neurons. NMNAT2 mRNA levels are reduced in the brains of Alzheimer's, Parkinson's, and Huntington's disease. Here we addressed whether NMNAT2 is required for axonal health of cortical glutamatergic neurons, whose long-projecting axons are often vulnerable in neurodegenerative conditions. We also tested if NMNAT2 maintains axonal health by ensuring axonal ATP levels for axonal transport, critical for axonal function. Methods We generated mouse and cultured neuron models to determine the impact of NMNAT2 loss from cortical glutamatergic neurons on axonal transport, energetic metabolism, and morphological integrity. In addition, we determined if exogenous NAD supplementation or inhibiting a NAD hydrolase, sterile alpha and TIR motif-containing protein 1 (SARM1), prevented axonal deficits caused by NMNAT2 loss. This study used a combination of genetics, molecular biology, immunohistochemistry, biochemistry, fluorescent time-lapse imaging, live imaging with optical sensors, and anti-sense oligos. Results We provide in vivo evidence that NMNAT2 in glutamatergic neurons is required for axonal survival. Using in vivo and in vitro studies, we demonstrate that NMNAT2 maintains the NAD-redox potential to provide "on-board" ATP via glycolysis to vesicular cargos in distal axons. Exogenous NAD+ supplementation to NMNAT2 KO neurons restores glycolysis and resumes fast axonal transport. Finally, we demonstrate both in vitro and in vivo that reducing the activity of SARM1, an NAD degradation enzyme, can reduce axonal transport deficits and suppress axon degeneration in NMNAT2 KO neurons. Conclusion NMNAT2 ensures axonal health by maintaining NAD redox potential in distal axons to ensure efficient vesicular glycolysis required for fast axonal transport.
Collapse
|
30
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
31
|
Guardiola M, Muntané G, Martínez I, Martorell L, Girona J, Ibarretxe D, Plana N, Bullido MJ, Vilella E, Ribalta J. Metabolic Overlap between Alzheimer's Disease and Metabolic Syndrome Identifies the PVRL2 Gene as a New Modulator of Diabetic Dyslipidemia. Int J Mol Sci 2023; 24:ijms24087415. [PMID: 37108578 PMCID: PMC10139078 DOI: 10.3390/ijms24087415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) share metabolic alterations such as abnormal insulin and lipid metabolism and have some common genetic factors such as APOE genotype. Taking this into account, we hypothesized that we could identify common genetic factors involved in the development of diabetes and cardiovascular diseases. METHODOLOGY We first genotyped 48 single nucleotide polymorphisms (SNPs) previously associated with AD in a cohort composed of 330 patients with cognitive impairment (CI) to assess their association with plasma lipids. Second, we conducted pleiotropy-informed conjunctional false discovery rate (FDR) analysis designed to identify shared variants between AD and plasma lipid levels. Finally, we used the SNPs to be found associated with lipid parameters and AD to search for associations with lipoprotein parameters in 281 patients with cardiometabolic risk. RESULTS Five SNPs were significantly associated with lower levels of cholesterol transported in remnant lipoprotein particles (RLPc) in subjects with CI; among these SNPs was the rs73572039 variant in PVRL2. Stratified QQ-plots were conducted on GWAS designed for AD and triglycerides (TG). The cross-trait analysis resulted in a total of 22 independent genomic loci associated with both AD and TG levels with a conjFDR < 0.05. Among these loci, two pleiotropic variants were located in PVRL2 (rs12978931 and rs11667640). The three SNPs in PVRL2 were significantly associated with RLPc, TG, and number of circulating VLDL and HDL particles in subjects with cardiometabolic risk. CONCLUSIONS We have identified three variants in PVRL2 that predispose individuals to AD that also influence the lipid profile that confers cardiovascular risk in T2DM subjects. PVRL2 is a potential new modulating factor of atherogenic dyslipidemia.
Collapse
Affiliation(s)
- Montse Guardiola
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gerard Muntané
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Hospital Universitari Institut Pere Mata, 43206 Reus, Spain
- Genètica i Ambient en Psiquiatria, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Iris Martínez
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Lourdes Martorell
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Hospital Universitari Institut Pere Mata, 43206 Reus, Spain
- Genètica i Ambient en Psiquiatria, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josefa Girona
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daiana Ibarretxe
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Núria Plana
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM-Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unitat de Medicina Vascular i Metabolisme, Servei de Medicina Interna, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - María J Bullido
- Centro de Biología Molecular "Severo Ochoa" (C.S.I.C.-U.A.M.), Universidad Autónoma de Madrid, 28049 Madrid, Spain
- CIBERNED, Center for Networked Biomedical Research on Neurodegenerative Diseases, Carlos III Institute of Health, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz, IdiPAZ (Hospital Universitario La Paz-Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Elisabet Vilella
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Hospital Universitari Institut Pere Mata, 43206 Reus, Spain
- Genètica i Ambient en Psiquiatria, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Centro de Investigación Biomédica en Red en Salud Mental, CIBERSAM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Josep Ribalta
- Unitat de Recerca en Lípids i Arteriosclerosi, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, 43201 Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili-CERCA, 43204 Reus, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM-Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
32
|
Malek-Ahmadi M, Su Y, Ghisays V, Luo J, Devadas V, Chen Y, Lee W, Protas H, Chen K, Zetterberg H, Blennow K, Caselli RJ, Reiman EM. Plasma NfL is associated with the APOE ε4 allele, brain imaging measurements of neurodegeneration, and lower recall memory scores in cognitively unimpaired late-middle-aged and older adults. Alzheimers Res Ther 2023; 15:74. [PMID: 37038190 PMCID: PMC10084600 DOI: 10.1186/s13195-023-01221-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Plasma neurofilament light (NfL) is an indicator of neurodegeneration and/or neuroaxonal injury in persons with Alzheimer's disease (AD) and a wide range of other neurological disorders. Here, we characterized and compared plasma NfL concentrations in cognitively unimpaired (CU) late-middle-aged and older adults with two, one, or no copies of the APOE ε4 allele, the major genetic risk factor for AD. We then assessed plasma NfL associations with brain imaging measurements of AD-related neurodegeneration (hippocampal atrophy and a hypometabolic convergence index [HCI]), brain imaging measurements of amyloid-β plaque burden, tau tangle burden and white matter hyperintensity volume (WMHV), and delayed and total recall memory scores. METHODS Plasma NfL concentrations were measured in 543 CU 69 ± 9 year-old participants in the Arizona APOE Cohort Study, including 66 APOE ε4 homozygotes (HM), 165 heterozygotes (HT), and 312 non-carriers (NC). Robust regression models were used to characterize plasma NfL associations with APOE ε4 allelic dose before and after adjustment for age, sex, and education. They were also used to characterize plasma NfL associations with MRI-based hippocampal volume and WMHV measurements, an FDG PET-based HCI, mean cortical PiB PET measurements of amyloid-β plaque burden and meta-region-of-interest (meta-ROI) flortaucipir PET measurements of tau tangle burden, and Auditory Verbal Learning Test (AVLT) Delayed and Total Recall Memory scores. RESULTS After the adjustments noted above, plasma NfL levels were significantly greater in APOE ε4 homozygotes and heterozygotes than non-carriers and significantly associated with smaller hippocampal volumes (r = - 0.43), greater tangle burden in the entorhinal cortex and inferior temporal lobes (r = 0.49, r = 0.52, respectively), and lower delayed (r = - 0.27), and total (r = - 0.27) recall memory scores (p < 0.001). NfL levels were not significantly associated with PET measurements of amyloid-β plaque or total tangle burden. CONCLUSIONS Plasma NfL concentrations are associated with the APOE ε4 allele, brain imaging biomarkers of neurodegeneration, and less good recall memory in CU late-middle-aged and older adults, supporting its value as an indicator of neurodegeneration in the preclinical study of AD.
Collapse
Affiliation(s)
| | - Yi Su
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Valentina Ghisays
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Ji Luo
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Vivek Devadas
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Yinghua Chen
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Wendy Lee
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Hillary Protas
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Kewei Chen
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | - Eric M Reiman
- Banner Alzheimer's Institute, 901 E. Willetta St., Phoenix, AZ, 85006, USA
- Translation Genomics Research Institute, Phoenix, AZ, USA
- University of Arizona, Phoenix, AZ, USA
- Arizona State University, Tempe, AZ, USA
| |
Collapse
|
33
|
Sung JH, Ou Y, Barger SW. Amyloid β-Peptide Effects on Glucose Regulation Are Dependent on Apolipoprotein E Genotype. eNeuro 2023; 10:ENEURO.0376-22.2023. [PMID: 37163733 PMCID: PMC10135078 DOI: 10.1523/eneuro.0376-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 05/12/2023] Open
Abstract
The apolipoprotein E gene (APOE) confers the greatest genetic risk factor for Alzheimer's disease (AD), wherein the ε4 allele confers an elevated risk compared with the ε3 allele. Biological mechanisms that differ across these alleles have been explored in mouse models wherein the murine Apoe gene has undergone targeted replacement with sequences encoding human ApoE3 or ApoE4 (ApoE-TR mice). Such models have indicated that the two variants of ApoE produce differential effects on energy metabolism, including metabolic syndrome. However, glucose regulation has not been compared in ApoE-TR mice with and without amyloid β-peptide (Aβ) accumulation. We crossed ApoE3-TR and ApoE4-TR mice with a transgenic line that accumulates human Aβ1-42 In male ApoE3-TR mice, introduction of Aβ caused aberrations in glucose tolerance and in membrane translocation of astrocytic glucose transporter 1 (GLUT1). Phosphorylation of Tau at AD-relevant sites was correlated with glucose intolerance. These effects appeared independent of insulin dysregulation and were not observed in females. In ApoE4-TR mice, the addition of Aβ had no significant effects because of a trend toward perturbation of the baseline values.
Collapse
Affiliation(s)
- Jin Hee Sung
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Yang Ou
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205
| |
Collapse
|
34
|
Mahley RW. Apolipoprotein E4 targets mitochondria and the mitochondria-associated membrane complex in neuropathology, including Alzheimer's disease. Curr Opin Neurobiol 2023; 79:102684. [PMID: 36753858 DOI: 10.1016/j.conb.2023.102684] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 02/08/2023]
Abstract
Apolipoprotein (apo) E4 sets the stage for neuropathology in Alzheimer's disease (AD) by causing mitochondrial dysfunction and altering mitochondria-associated membranes. Contact and apposition of mitochondrial-endoplasmic reticulum membranes are enhanced in brain cells in AD and associated with increases in tethering and spacing proteins that modulate many cellular processes. Contact site protein levels are higher in apoE4 cells. In apoE4 neurons, the NAD+/NADH ratio is lowered, reactive oxygen species are increased, and NAD/NADH pathway components and redox proteins are decreased. Oxidative phosphorylation is impaired and reserve ATP generation capacity is lacking. ApoE4 neurons have ∼50% fewer respiratory complex subunits (e.g., ATP synthase) and may increase translocase levels of the outer and inner mitochondrial membranes to facilitate delivery of nucleus-encoded complex subunits. Respiratory complex assembly relies on mitochondrial cristae organizing system subunits that are altered in apoE4 cells, and apoE4 increases mitochondrial proteases that control respiratory subunit composition for complex assembly.
Collapse
Affiliation(s)
- Robert W Mahley
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158, USA; Departments of Pathology and Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
35
|
Li W, Li R, Yan S, Zhao Z, Shan Y, Qi Z, Lu J. Effect of APOE ε4 genotype on amyloid-β, glucose metabolism, and gray matter volume in cognitively normal individuals and amnestic mild cognitive impairment. Eur J Neurol 2023; 30:587-596. [PMID: 36448771 PMCID: PMC10107141 DOI: 10.1111/ene.15656] [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: 10/09/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND PURPOSE The presence of apolipoprotein E ε4 (APOE ε4) is associated with an increased risk of developing Alzheimer disease (AD). The aim of this study was to assess the effects of APOE ε4 on amyloid-β (Aβ) pathology, glucose metabolism, and gray matter (GM) volume and their longitudinal changes in healthy control (HC) and amnestic mild cognitive impairment (aMCI). METHODS We included 50 HCs and 109 aMCI patients from the Alzheimer's Disease Neuroimaging Initiative phase 2/GO based on availability of baseline T1-weighted magnetic resonance imaging, 18 F-florbetapir positron emission tomography (PET), and 18 F-fluorodeoxyglucose (FDG) PET. Of these, 35 HCs and 67 aMCI patients who underwent 24-month scans were included for follow-up study. RESULTS Voxelwise analysis revealed that APOE ε4 carriers exhibited greater baseline Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. However, there was no significant difference between APOE ε4 noncarriers and APOE ε4 carriers in terms of 18 F-FDG PET standardized uptake value ratio and GM volume. Region of interest-based analysis showed statistically significant greater Aβ deposition in APOE ε4 carriers than APOE ε4 noncarriers only in aMCI patients. Furthermore, APOE ε4 carriers generally exhibited a greater magnitude and spatial extent of longitudinal changes in Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. CONCLUSIONS Our findings suggest a differential effect of APOE ε4 on Aβ pathology, glucose metabolism, and GM volume. Studying APOE ε4-related brain changes with neuroimaging biomarkers in preclinical AD offers an opportunity to further our understanding of the pathophysiology of AD at an early stage.
Collapse
Affiliation(s)
- Weihua Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhilian Zhao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | |
Collapse
|
36
|
Li X, Kaur Y, Wilhelm O, Reuter M, Montag C, Sommer W, Zhou C, Hildebrandt A. Resting-state brain signal complexity discriminates young healthy APOE e4 carriers from non-e4 carriers. Eur J Neurosci 2023; 57:854-866. [PMID: 36656069 DOI: 10.1111/ejn.15915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
It is well established that the e4 allele of the APOE gene is associated with impaired brain functionality and cognitive decline in humans at elder age. However, it is controversial whether and how the APOE e4 allele is associated with superior brain function among young healthy individuals, thus indicates a case of antagonistic pleiotropy of APOE e4 allele. Signal complexity is a critical aspect of brain activity that has been associated with brain function. In this study, the multiscale entropy (MSE) of resting-state EEG signals among a sample of young healthy adults (N = 260) as an indicator of brain signal complexity was investigated. It was of interest whether MSE differs across APOE genotype groups while age and education level were controlled for and whether the APOE genotype effect on MSE interacts with MSE time scale, as well as EEG recording condition. Results of linear mixed models indicate overall larger MSE in APOE e4 carriers. This genotype-dependent difference is larger at high as compared with low time scales. The interaction effect between APOE genotype and recording condition indicates increased between-state MSE change in young healthy APOE e4 carriers as compared with non-carriers. Because higher complexity is commonly taken to be associated with better cognitive functioning, the present results complement previous findings and therefore point to a pleiotropic spectrum of the APOE gene polymorphism.
Collapse
Affiliation(s)
- Xiaojing Li
- Chinese Academy of Disability Data Science, Nanjing Normal University of Special Education, Nanjing, China.,Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong.,Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Yadwinder Kaur
- Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | | | - Martin Reuter
- Centre for Economics and Neuroscience, University of Bonn, Bonn, Germany.,Department of Psychology, University of Bonn, Bonn, Germany
| | - Christian Montag
- Department of Psychology, Ulm University, Ulm, Germany.,The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Werner Sommer
- Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong.,Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Psychology, Zhejiang Normal University, Jinhua, China
| | - Changsong Zhou
- Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong
| | - Andrea Hildebrandt
- Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| |
Collapse
|
37
|
Swerdlow RH. The Alzheimer's Disease Mitochondrial Cascade Hypothesis: A Current Overview. J Alzheimers Dis 2023; 92:751-768. [PMID: 36806512 DOI: 10.3233/jad-221286] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Viable Alzheimer's disease (AD) hypotheses must account for its age-dependence; commonality; association with amyloid precursor protein, tau, and apolipoprotein E biology; connection with vascular, inflammation, and insulin signaling changes; and systemic features. Mitochondria and parameters influenced by mitochondria could link these diverse characteristics. Mitochondrial biology can initiate changes in pathways tied to AD and mediate the dysfunction that produces the clinical phenotype. For these reasons, conceptualizing a mitochondrial cascade hypothesis is a straightforward process and data accumulating over decades argue the validity of its principles. Alternative AD hypotheses may yet account for its mitochondria-related phenomena, but absent this happening a primary mitochondrial cascade hypothesis will continue to evolve and attract interest.
Collapse
Affiliation(s)
- Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, USA.,Departments of Neurology, Molecular and Integrative Physiology, and Biochemistry and Molecular Biology, University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
38
|
Zhang X, Wu L, Swerdlow RH, Zhao L. Opposing Effects of ApoE2 and ApoE4 on Glycolytic Metabolism in Neuronal Aging Supports a Warburg Neuroprotective Cascade against Alzheimer's Disease. Cells 2023; 12:410. [PMID: 36766752 PMCID: PMC9914046 DOI: 10.3390/cells12030410] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Apolipoprotein E4 (ApoE4) is the most recognized genetic risk factor for late-onset Alzheimer's disease (LOAD), whereas ApoE2 reduces the risk for LOAD. The underlying mechanisms are unclear but may include effects on brain energy metabolism. Here, we used neuro-2a (N2a) cells that stably express human ApoE isoforms (N2a-hApoE), differentiated N2a-hApoE neuronal cells, and humanized ApoE knock-in mouse models to investigate relationships among ApoE isoforms, glycolytic metabolism, and neuronal health and aging. ApoE2-expressing cells retained robust hexokinase (HK) expression and glycolytic activity, whereas these endpoints progressively declined with aging in ApoE4-expressing cells. These divergent ApoE2 and ApoE4 effects on glycolysis directly correlated with markers of cellular wellness. Moreover, ApoE4-expressing cells upregulated phosphofructokinase and pyruvate kinase with the apparent intent of compensating for the HK-dependent glycolysis reduction. The introduction of ApoE2 increased HK levels and glycolysis flux in ApoE4 cells. PI3K/Akt signaling was distinctively regulated by ApoE isoforms but was only partially responsible for the ApoE-mediated effects on HK. Collectively, our findings indicate that human ApoE isoforms differentially modulate neuronal glycolysis through HK regulation, with ApoE2 upregulating and ApoE4 downregulating, which markedly impacts neuronal health during aging. These findings lend compelling support to the emerging inverse-Warburg theory of AD and highlight a therapeutic opportunity for bolstering brain glycolytic resilience to prevent and treat AD.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Long Wu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
39
|
Huguenard CJC, Cseresznye A, Darcey T, Nkiliza A, Evans JE, Hazen SL, Mullan M, Crawford F, Abdullah L. Age and APOE affect L-carnitine system metabolites in the brain in the APOE-TR model. Front Aging Neurosci 2023; 14:1059017. [PMID: 36688151 PMCID: PMC9853982 DOI: 10.3389/fnagi.2022.1059017] [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: 09/30/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
With age the apolipoprotein E (APOE) E4 allele (involved in lipid homeostasis) is associated with perturbation of bioenergetics pathways in Alzheimer's disease (AD). We therefore hypothesized that in aging mice APOE genotype would affect the L-carnitine system (central to lipid bioenergetics), in the brain and in the periphery. Using liquid chromatography-mass spectrometry, levels of L-carnitine and associated metabolites: γ-butyrobetaine (GBB), crotonobetaine, as well as acylcarnitines, were evaluated at 10-, 25-, and 50-weeks, in the brain and the periphery, in a targeted replacement mouse model of human APOE (APOE-TR). Aged APOE-TR mice were also orally administered 125 mg/kg of L-carnitine daily for 7 days followed by evaluation of brain, liver, and plasma L-carnitine system metabolites. Compared to E4-TR, an age-dependent increase among E2- and E3-TR mice was detected for medium- and long-chain acylcarnitines (MCA and LCA, respectively) within the cerebrovasculature and brain parenchyma. While following L-carnitine oral challenge, E4-TR mice had higher increases in the L-carnitine metabolites, GBB and crotonobetaine in the brain and a reduction of plasma to brain total acylcarnitine ratios compared to other genotypes. These studies suggest that with aging, the presence of the E4 allele may contribute to alterations in the L-carnitine bioenergetic system and to the generation of L-carnitine metabolites that could have detrimental effects on the vascular system. Collectively the E4 allele and aging may therefore contribute to AD pathogenesis through aging-related lipid bioenergetics as well as cerebrovascular dysfunctions.
Collapse
Affiliation(s)
- Claire J. C. Huguenard
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes, United Kingdom
| | - Adam Cseresznye
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
| | - Teresa Darcey
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
| | - Aurore Nkiliza
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
- James A. Haley VA Hospital, Tampa, FL, United States
| | - James E. Evans
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Michael Mullan
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes, United Kingdom
| | - Fiona Crawford
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes, United Kingdom
- James A. Haley VA Hospital, Tampa, FL, United States
| | - Laila Abdullah
- Department of Metabolomics, Roskamp Institute, Sarasota, FL, United States
- School of Life, Health and Chemical Sciences, Open University, Milton Keynes, United Kingdom
- James A. Haley VA Hospital, Tampa, FL, United States
| |
Collapse
|
40
|
Smith GS, Protas H, Kuwabara H, Savonenko A, Nassery N, Gould NF, Kraut M, Avramopoulos D, Holt D, Dannals RF, Nandi A, Su Y, Reiman EM, Chen K. Molecular imaging of the association between serotonin degeneration and beta-amyloid deposition in mild cognitive impairment. Neuroimage Clin 2023; 37:103322. [PMID: 36680976 PMCID: PMC9869478 DOI: 10.1016/j.nicl.2023.103322] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
BACKGROUND Degeneration of the serotonin system has been observed in Alzheimer's disease (AD) and in mild cognitive impairment (MCI). In transgenic amyloid mouse models, serotonin degeneration is detected prior to widespread cortical beta-amyloid (Aβ) deposition, also suggesting that serotonin degeneration may be observed in preclinical AD. METHODS The differences in the distribution of serotonin degeneration (reflected by the loss of the serotonin transporter, 5-HTT) relative to Aβ deposition was measured with positron emission tomography in a group of individuals with MCI and a group of healthy older adults. A multi-modal partial least squares (mmPLS) algorithm was applied to identify the spatial covariance pattern between 5-HTT availability and Aβ deposition. RESULTS Forty-five individuals with MCI and 35 healthy older adults were studied, 22 and 27 of whom were included in the analyses who were "amyloid positive" and "amyloid negative", respectively. A pattern of lower cortical, subcortical and limbic 5-HTT availability and higher cortical Aβ deposition distinguished the MCI from the healthy older control participants. Greater expression of this pattern was correlated with greater deficits in memory and executive function in the MCI group, not in the control group. CONCLUSION A spatial covariance pattern of lower 5-HTT availability and Aβ deposition was observed to a greater extent in an MCI group relative to a control group and was associated with cognitive impairment in the MCI group. The results support the application of mmPLS to understand the neurochemical changes associated with Aβ deposition in the course of preclinical AD.
Collapse
Affiliation(s)
- Gwenn S Smith
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | - Hiroto Kuwabara
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alena Savonenko
- Department of Pathology (Neuropathology), Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Najlla Nassery
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neda F Gould
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Kraut
- Division of Neuroradiology, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitri Avramopoulos
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel Holt
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Dannals
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ayon Nandi
- Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| |
Collapse
|
41
|
Chhimpa N, Singh N, Puri N, Kayath HP. The Novel Role of Mitochondrial Citrate Synthase and Citrate in the Pathophysiology of Alzheimer's Disease. J Alzheimers Dis 2023; 94:S453-S472. [PMID: 37393492 PMCID: PMC10473122 DOI: 10.3233/jad-220514] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Citrate synthase is a key mitochondrial enzyme that utilizes acetyl-CoA and oxaloacetate to form citrate in the mitochondrial membrane, which participates in energy production in the TCA cycle and linked to the electron transport chain. Citrate transports through a citrate malate pump and synthesizes acetyl-CoA and acetylcholine (ACh) in neuronal cytoplasm. In a mature brain, acetyl-CoA is mainly utilized for ACh synthesis and is responsible for memory and cognition. Studies have shown low citrate synthase in different regions of brain in Alzheimer's disease (AD) patients, which reduces mitochondrial citrate, cellular bioenergetics, neurocytoplasmic citrate, acetyl-CoA, and ACh synthesis. Reduced citrate mediated low energy favors amyloid-β (Aβ) aggregation. Citrate inhibits Aβ25-35 and Aβ1-40 aggregation in vitro. Hence, citrate can be a better therapeutic option for AD by improving cellular energy and ACh synthesis, and inhibiting Aβ aggregation, which prevents tau hyperphosphorylation and glycogen synthase kinase-3 beta. Therefore, we need clinical studies if citrate reverses Aβ deposition by balancing mitochondrial energy pathway and neurocytoplasmic ACh production. Furthermore, in AD's silent phase pathophysiology, when neuronal cells are highly active, they shift ATP utilization from oxidative phosphorylation to glycolysis and prevent excessive generation of hydrogen peroxide and reactive oxygen species (oxidative stress) as neuroprotective action, which upregulates glucose transporter-3 (GLUT3) and pyruvate dehydrogenase kinase-3 (PDK3). PDK3 inhibits pyruvate dehydrogenase, which decreases mitochondrial-acetyl-CoA, citrate, and cellular bioenergetics, and decreases neurocytoplasmic citrate, acetyl-CoA, and ACh formation, thus initiating AD pathophysiology. Therefore, GLUT3 and PDK3 can be biomarkers for silent phase of AD.
Collapse
Affiliation(s)
- Neeraj Chhimpa
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
- Department of Pharmacology, Meharishi Markandeshwar College of Medical Science & Research, Ambala, India
| | - Neha Singh
- Department of Pharmacology, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Nikkita Puri
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | | |
Collapse
|
42
|
He DL, Fan YG, Wang ZY. Energy Crisis Links to Autophagy and Ferroptosis in Alzheimer's Disease: Current Evidence and Future Avenues. Curr Neuropharmacol 2023; 21:67-86. [PMID: 35980072 PMCID: PMC10193753 DOI: 10.2174/1570159x20666220817140737] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/14/2022] [Accepted: 08/11/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases worldwide. The occult nature of the onset and the uncertainty of the etiology largely impede the development of therapeutic strategies for AD. Previous studies revealed that the disorder of energy metabolism in the brains of AD patients appears far earlier than the typical pathological features of AD, suggesting a tight association between energy crisis and the onset of AD. Energy crisis in the brain is known to be induced by the reductions in glucose uptake and utilization, which may be ascribed to the diminished expressions of cerebral glucose transporters (GLUTs), insulin resistance, mitochondrial dysfunctions, and lactate dysmetabolism. Notably, the energy sensors such as peroxisome proliferators-activated receptor (PPAR), transcription factor EB (TFEB), and AMP-activated protein kinase (AMPK) were shown to be the critical regulators of autophagy, which play important roles in regulating beta-amyloid (Aβ) metabolism, tau phosphorylation, neuroinflammation, iron dynamics, as well as ferroptosis. In this study, we summarized the current knowledge on the molecular mechanisms involved in the energy dysmetabolism of AD and discussed the interplays existing between energy crisis, autophagy, and ferroptosis. In addition, we highlighted the potential network in which autophagy may serve as a bridge between energy crisis and ferroptosis in the progression of AD. A deeper understanding of the relationship between energy dysmetabolism and AD may provide new insight into developing strategies for treating AD; meanwhile, the energy crisis in the progression of AD should gain more attention.
Collapse
Affiliation(s)
- Da-Long He
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| | - Zhan-You Wang
- Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang, 110122, China
- Key Laboratory of Medical Cell Biology of Ministry of Education, Health Sciences Institute of China Medical University, Shenyang, 110122, China
| |
Collapse
|
43
|
Weber CM, Moiz B, Zic SM, Alpízar Vargas V, Li A, Clyne AM. Induced pluripotent stem cell-derived cells model brain microvascular endothelial cell glucose metabolism. Fluids Barriers CNS 2022; 19:98. [PMID: 36494870 PMCID: PMC9733016 DOI: 10.1186/s12987-022-00395-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Glucose transport from the blood into the brain is tightly regulated by brain microvascular endothelial cells (BMEC), which also use glucose as their primary energy source. To study how BMEC glucose transport contributes to cerebral glucose hypometabolism in diseases such as Alzheimer's disease, it is essential to understand how these cells metabolize glucose. Human primary BMEC (hpBMEC) can be used for BMEC metabolism studies; however, they have poor barrier function and may not recapitulate in vivo BMEC function. iPSC-derived BMEC-like cells (hiBMEC) are readily available and have good barrier function but may have an underlying epithelial signature. In this study, we examined differences between hpBMEC and hiBMEC glucose metabolism using a combination of dynamic metabolic measurements, metabolic mass spectrometry, RNA sequencing, and Western blots. hiBMEC had decreased glycolytic flux relative to hpBMEC, and the overall metabolomes and metabolic enzyme levels were different between the two cell types. However, hpBMEC and hiBMEC had similar glucose metabolism, including nearly identical glucose labeled fractions of glycolytic and TCA cycle metabolites. Treatment with astrocyte conditioned media and high glucose increased glycolysis in both hpBMEC and hiBMEC, though hpBMEC decreased glycolysis in response to fluvastatin while hiBMEC did not. Together, these results suggest that hiBMEC can be used to model cerebral vascular glucose metabolism, which expands their use beyond barrier models.
Collapse
Affiliation(s)
| | - Bilal Moiz
- University of Maryland, College Park, MD, 20742, USA
| | - Sophia M Zic
- University of Maryland, College Park, MD, 20742, USA
| | | | - Andrew Li
- University of Maryland, College Park, MD, 20742, USA
| | | |
Collapse
|
44
|
Savignac C, Villeneuve S, Badhwar A, Saltoun K, Shafighi K, Zajner C, Sharma V, Gagliano Taliun SA, Farhan S, Poirier J, Bzdok D. APOE alleles are associated with sex-specific structural differences in brain regions affected in Alzheimer's disease and related dementia. PLoS Biol 2022; 20:e3001863. [PMID: 36512526 PMCID: PMC9747055 DOI: 10.1371/journal.pbio.3001863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 09/30/2022] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease is marked by intracellular tau aggregates in the medial temporal lobe (MTL) and extracellular amyloid aggregates in the default network (DN). Here, we examined codependent structural variations between the MTL's most vulnerable structure, the hippocampus (HC), and the DN at subregion resolution in individuals with Alzheimer's disease and related dementia (ADRD). By leveraging the power of the approximately 40,000 participants of the UK Biobank cohort, we assessed impacts from the protective APOE ɛ2 and the deleterious APOE ɛ4 Alzheimer's disease alleles on these structural relationships. We demonstrate ɛ2 and ɛ4 genotype effects on the inter-individual expression of HC-DN co-variation structural patterns at the population level. Across these HC-DN signatures, recurrent deviations in the CA1, CA2/3, molecular layer, fornix's fimbria, and their cortical partners related to ADRD risk. Analyses of the rich phenotypic profiles in the UK Biobank cohort further revealed male-specific HC-DN associations with air pollution and female-specific associations with cardiovascular traits. We also showed that APOE ɛ2/2 interacts preferentially with HC-DN co-variation patterns in estimating social lifestyle in males and physical activity in females. Our structural, genetic, and phenotypic analyses in this large epidemiological cohort reinvigorate the often-neglected interplay between APOE ɛ2 dosage and sex and link APOE alleles to inter-individual brain structural differences indicative of ADRD familial risk.
Collapse
Affiliation(s)
- Chloé Savignac
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Centre (BIC), MNI, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Centre for Studies in the Prevention of Alzheimer’s Disease, Douglas Mental Health Institute, McGill University, Montreal, Quebec, Canada
| | - AmanPreet Badhwar
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Centre de recherche de l’Institut universitaire de gériatrie de Montréal (CRIUGM), Montreal, Quebec, Canada
| | - Karin Saltoun
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kimia Shafighi
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Chris Zajner
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Vaibhav Sharma
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sarah A. Gagliano Taliun
- Department of Neurosciences & Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Montréal, Quebec, Canada
| | - Sali Farhan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Judes Poirier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Centre for Studies in the Prevention of Alzheimer’s Disease, Douglas Mental Health Institute, McGill University, Montreal, Quebec, Canada
| | - Danilo Bzdok
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- McConnell Brain Imaging Centre (BIC), MNI, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- School of Computer Science, McGill University, Montreal, Quebec, Canada
- Mila—Quebec Artificial Intelligence Institute, Montreal, Quebec, Canada
| |
Collapse
|
45
|
Suchy-Dicey A, Howard B, Longstreth WT, Reiman EM, Buchwald D. APOE genotype, hippocampus, and cognitive markers of Alzheimer's disease in American Indians: Data from the Strong Heart Study. Alzheimers Dement 2022; 18:2518-2526. [PMID: 35142437 PMCID: PMC9363523 DOI: 10.1002/alz.12573] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND The apolipoprotein E (APOE) ε4 allele confers higher risk of neurodegeneration and Alzheimer's disease (AD), but differs by race/ethnicity. We examined this association in American Indians. METHODS The Strong Heart Study is a population-based cohort of American Indians who were 64 to 95 years of age in 2010 to 2013. APOE ε4 status, brain imaging, and neuropsychological testing was collected in N = 811 individuals. Summary statistics, graphics, and generalized linear regressions-adjusted for sociodemographics, clinical features, and intracranial volume with bootstrap variance estimator-compared APOE ε4 carriers with non-carriers. RESULTS APOE ε4 carriers comprised 22% of the population (0.7% homozygotes). Participants were mean 73 years, 67% female, and 54% had some college education. The majority were obese (>50%), hypertensive (>80%), and diabetic (>50%). Neither imaging findings nor multidomain cognitive testing showed any substantive differences between APOE ε4 carriers and non-carriers. CONCLUSION We found no evidence of neurodegenerative risk from APOE ε4 in American Indians. Additional studies are needed to examine potential protective features.
Collapse
Affiliation(s)
- Astrid Suchy-Dicey
- Washington State University Elson S Floyd College of Medicine, Seattle, Washington, USA
| | - Barbara Howard
- MedStar Health Research Institute, Phoenix, Arizona, USA
| | - W T Longstreth
- University of Washington Neurology and Epidemiology Departments, Seattle, Washington, USA
| | | | - Dedra Buchwald
- Washington State University Elson S Floyd College of Medicine, Seattle, Washington, USA
| |
Collapse
|
46
|
Li J, Zhang Y, Lu T, Liang R, Wu Z, Liu M, Qin L, Chen H, Yan X, Deng S, Zheng J, Liu Q. Identification of diagnostic genes for both Alzheimer's disease and Metabolic syndrome by the machine learning algorithm. Front Immunol 2022; 13:1037318. [PMID: 36405716 PMCID: PMC9667080 DOI: 10.3389/fimmu.2022.1037318] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/23/2022] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Alzheimer's disease is the most common neurodegenerative disease worldwide. Metabolic syndrome is the most common metabolic and endocrine disease in the elderly. Some studies have suggested a possible association between MetS and AD, but few studied genes that have a co-diagnostic role in both diseases. METHODS The microarray data of AD (GSE63060 and GSE63061 were merged after the batch effect was removed) and MetS (GSE98895) in the GEO database were downloaded. The WGCNA was used to identify the co-expression modules related to AD and MetS. RF and LASSO were used to identify the candidate genes. Machine learning XGBoost improves the diagnostic effect of hub gene in AD and MetS. The CIBERSORT algorithm was performed to assess immune cell infiltration MetS and AD samples and to investigate the relationship between biomarkers and infiltrating immune cells. The peripheral blood mononuclear cells (PBMCs) single-cell RNA (scRNA) sequencing data from patients with AD and normal individuals were visualized with the Seurat standard flow dimension reduction clustering the metabolic pathway activity changes each cell with ssGSEA. RESULTS The brown module was identified as the significant module with AD and MetS. GO analysis of shared genes showed that intracellular transport and establishment of localization in cell and organelle organization were enriched in the pathophysiology of AD and MetS. By using RF and Lasso learning methods, we finally obtained eight diagnostic genes, namely ARHGAP4, SNRPG, UQCRB, PSMA3, DPM1, MED6, RPL36AL and RPS27A. Their AUC were all greater than 0.7. Higher immune cell infiltrations expressions were found in the two diseases and were positively linked to the characteristic genes. The scRNA-seq datasets finally obtained seven cell clusters. Seven major cell types including CD8 T cell, monocytes, T cells, NK cell, B cells, dendritic cells and macrophages were clustered according to immune cell markers. The ssGSEA revealed that immune-related gene (SNRPG) was significantly regulated in the glycolysis-metabolic pathway. CONCLUSION We identified genes with common diagnostic effects on both MetS and AD, and found genes involved in multiple metabolic pathways associated with various immune cells.
Collapse
Affiliation(s)
- Jinwei Li
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yang Zhang
- General Surgery, The First Affiliated Hospital of Dali University, Dali, China
| | - Tanli Lu
- Department of Neurology, The Tenth Affiliated Hospital of Guangxi Medical University, Qinzhou, China
| | - Rui Liang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Zhikang Wu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Meimei Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Linyao Qin
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Hongmou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xianlei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Shan Deng
- Department of Neurology, The Fourth Affliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Jiemin Zheng
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|
47
|
Steele OG, Stuart AC, Minkley L, Shaw K, Bonnar O, Anderle S, Penn AC, Rusted J, Serpell L, Hall C, King S. A multi-hit hypothesis for an APOE4-dependent pathophysiological state. Eur J Neurosci 2022; 56:5476-5515. [PMID: 35510513 PMCID: PMC9796338 DOI: 10.1111/ejn.15685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 01/01/2023]
Abstract
The APOE gene encoding the Apolipoprotein E protein is the single most significant genetic risk factor for late-onset Alzheimer's disease. The APOE4 genotype confers a significantly increased risk relative to the other two common genotypes APOE3 and APOE2. Intriguingly, APOE4 has been associated with neuropathological and cognitive deficits in the absence of Alzheimer's disease-related amyloid or tau pathology. Here, we review the extensive literature surrounding the impact of APOE genotype on central nervous system dysfunction, focussing on preclinical model systems and comparison of APOE3 and APOE4, given the low global prevalence of APOE2. A multi-hit hypothesis is proposed to explain how APOE4 shifts cerebral physiology towards pathophysiology through interconnected hits. These hits include the following: neurodegeneration, neurovascular dysfunction, neuroinflammation, oxidative stress, endosomal trafficking impairments, lipid and cellular metabolism disruption, impaired calcium homeostasis and altered transcriptional regulation. The hits, individually and in combination, leave the APOE4 brain in a vulnerable state where further cumulative insults will exacerbate degeneration and lead to cognitive deficits in the absence of Alzheimer's disease pathology and also a state in which such pathology may more easily take hold. We conclude that current evidence supports an APOE4 multi-hit hypothesis, which contributes to an APOE4 pathophysiological state. We highlight key areas where further study is required to elucidate the complex interplay between these individual mechanisms and downstream consequences, helping to frame the current landscape of existing APOE-centric literature.
Collapse
Affiliation(s)
| | | | - Lucy Minkley
- School of Life SciencesUniversity of SussexBrightonUK
| | - Kira Shaw
- School of Life SciencesUniversity of SussexBrightonUK
| | - Orla Bonnar
- School of Life SciencesUniversity of SussexBrightonUK
| | | | | | | | | | | | - Sarah King
- School of PsychologyUniversity of SussexBrightonUK
| |
Collapse
|
48
|
Nazarian A, Loiko E, Yassine HN, Finch CE, Kulminski AM. APOE alleles modulate associations of plasma metabolites with variants from multiple genes on chromosome 19q13.3. Front Aging Neurosci 2022; 14:1023493. [PMID: 36389057 PMCID: PMC9650319 DOI: 10.3389/fnagi.2022.1023493] [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: 08/19/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
The APOE ε2, ε3, and ε4 alleles differentially impact various complex diseases and traits. We examined whether these alleles modulated associations of 94 single-nucleotide polymorphisms (SNPs) harbored by 26 genes in 19q13.3 region with 217 plasma metabolites using Framingham Heart Study data. The analyses were performed in the E2 (ε2ε2 or ε2ε3 genotype), E3 (ε3ε3 genotype), and E4 (ε3ε4 or ε4ε4 genotype) groups separately. We identified 31, 17, and 22 polymorphism-metabolite associations in the E2, E3, and E4 groups, respectively, at a false discovery rate P FDR < 0.05. These entailed 51 and 19 associations with 20 lipid and 12 polar analytes. Contrasting the effect sizes between the analyzed groups showed 20 associations with group-specific effects at Bonferroni-adjusted P < 7.14E-04. Three associations with glutamic acid or dimethylglycine had significantly larger effects in the E2 than E3 group and 12 associations with triacylglycerol 56:5, lysophosphatidylethanolamines 16:0, 18:0, 20:4, or phosphatidylcholine 38:6 had significantly larger effects in the E2 than E4 group. Two associations with isocitrate or propionate and three associations with phosphatidylcholines 32:0, 32:1, or 34:0 had significantly larger effects in the E4 than E3 group. Nine of 70 SNP-metabolite associations identified in either E2, E3, or E4 groups attained P FDR < 0.05 in the pooled sample of these groups. However, none of them were among the 20 group-specific associations. Consistent with the evolutionary history of the APOE alleles, plasma metabolites showed higher APOE-cluster-related variations in the E4 than E2 and E3 groups. Pathway enrichment mainly highlighted lipids and amino acids metabolism and citrate cycle, which can be differentially impacted by the APOE alleles. These novel findings expand insights into the genetic heterogeneity of plasma metabolites and highlight the importance of the APOE-allele-stratified genetic analyses of the APOE-related diseases and traits.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| | - Hussein N. Yassine
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Caleb E. Finch
- Andrus Gerontology Center, University of Southern California, Los Angeles, CA, United States
| | - Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, United States
| |
Collapse
|
49
|
Kim H, Kim S, Cho B, Shin J, Kim J. APOE ε4-dependent effects on the early amyloid pathology in induced neurons of patients with Alzheimer's disease. Transl Neurodegener 2022; 11:45. [PMID: 36284363 PMCID: PMC9594913 DOI: 10.1186/s40035-022-00319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/11/2022] [Indexed: 12/04/2022] Open
Abstract
Background The ε4 allele of apolipoprotein E (APOE ε4) is the strongest known genetic risk factor for late-onset Alzheimer’s disease (AD), associated with amyloid pathogenesis. However, it is not clear how APOE ε4 accelerates amyloid-beta (Aβ) deposition during the seeding stage of amyloid development in AD patient neurons. Methods AD patient induced neurons (iNs) with an APOE ε4 inducible system were prepared from skin fibroblasts of AD patients. Transcriptome analysis was performed using RNA isolated from the AD patient iNs expressing APOE ε4 at amyloid-seeding and amyloid-aggregation stages. Knockdown of IGFBP3 was applied in the iNs to investigate the role of IGFBP3 in the APOE ε4-mediated amyloidosis. Results We optimized amyloid seeding stage in the iNs of AD patients that transiently expressed APOE ε4. Remarkably, we demonstrated that Aβ pathology was aggravated by the induction of APOE ε4 gene expression at the amyloid early-seeding stage in the iNs of AD patients. Moreover, transcriptome analysis in the early-seeding stage revealed that IGFBP3 was functionally important in the molecular pathology of APOE ε4-associated AD. Conclusions Our findings suggest that the presence of APOE ε4 at the early Aβ-seeding stage in patient iNs is critical for aggravation of sporadic AD pathology. These results provide insights into the importance of APOE ε4 expression for the progression and pathogenesis of sporadic AD. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00319-9.
Collapse
Affiliation(s)
- Hongwon Kim
- grid.255168.d0000 0001 0671 5021Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea ,grid.255168.d0000 0001 0671 5021Laboratory of Stem Cells & Gene Editing, Department of Chemistry, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea
| | - Siyoung Kim
- grid.255168.d0000 0001 0671 5021Laboratory of Stem Cells & Gene Editing, Department of Chemistry, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea
| | - Byounggook Cho
- grid.255168.d0000 0001 0671 5021Laboratory of Stem Cells & Gene Editing, Department of Chemistry, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea
| | - Jaein Shin
- grid.255168.d0000 0001 0671 5021Laboratory of Stem Cells & Gene Editing, Department of Chemistry, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea
| | - Jongpil Kim
- grid.255168.d0000 0001 0671 5021Department of Biomedical Engineering, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea ,grid.255168.d0000 0001 0671 5021Laboratory of Stem Cells & Gene Editing, Department of Chemistry, Dongguk University, Pildong-ro 1-gil 30, Jung-Gu, Seoul, 04620 Republic of Korea
| |
Collapse
|
50
|
Liu C, Zhu N, Sun H, Zhang J, Feng X, Gjerswold-Selleck S, Sikka D, Zhu X, Liu X, Nuriel T, Wei HJ, Wu CC, Vaughan JT, Laine AF, Provenzano FA, Small SA, Guo J. Deep learning of MRI contrast enhancement for mapping cerebral blood volume from single-modal non-contrast scans of aging and Alzheimer's disease brains. Front Aging Neurosci 2022; 14:923673. [PMID: 36034139 PMCID: PMC9407020 DOI: 10.3389/fnagi.2022.923673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
While MRI contrast agents such as those based on Gadolinium are needed for high-resolution mapping of brain metabolism, these contrast agents require intravenous administration, and there are rising concerns over their safety and invasiveness. Furthermore, non-contrast MRI scans are more commonly performed than those with contrast agents and are readily available for analysis in public databases such as the Alzheimer's Disease Neuroimaging Initiative (ADNI). In this article, we hypothesize that a deep learning model, trained using quantitative steady-state contrast-enhanced structural MRI datasets, in mice and humans, can generate contrast-equivalent information from a single non-contrast MRI scan. The model was first trained, optimized, and validated in mice, and was then transferred and adapted to humans. We observe that the model can substitute for Gadolinium-based contrast agents in approximating cerebral blood volume, a quantitative representation of brain activity, at sub-millimeter granularity. Furthermore, we validate the use of our deep-learned prediction maps to identify functional abnormalities in the aging brain using locally obtained MRI scans, and in the brain of patients with Alzheimer's disease using publicly available MRI scans from ADNI. Since it is derived from a commonly-acquired MRI protocol, this framework has the potential for broad clinical utility and can also be applied retrospectively to research scans across a host of neurological/functional diseases.
Collapse
Affiliation(s)
- Chen Liu
- Department of Electrical Engineering, Columbia University, New York, NY, United States
| | - Nanyan Zhu
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Haoran Sun
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Junhao Zhang
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Xinyang Feng
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | | | - Dipika Sikka
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Xuemin Zhu
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Xueqing Liu
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Tal Nuriel
- Department of Radiation Oncology, Columbia University, New York, NY, United States
| | - Hong-Jian Wei
- Department of Radiation Oncology, Columbia University, New York, NY, United States
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University, New York, NY, United States
| | - J. Thomas Vaughan
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Andrew F. Laine
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | | | - Scott A. Small
- Department of Neurology, Columbia University, New York, NY, United States
- Department of Psychiatry, Columbia University, New York, NY, United States
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, United States
| | - Jia Guo
- Department of Psychiatry, Columbia University, New York, NY, United States
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- *Correspondence: Jia Guo
| |
Collapse
|