1
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4-associated phenotypes with the longevity-promoting intervention 17α-estradiol in middle-aged male mice. COMMUNICATIONS MEDICINE 2025; 5:222. [PMID: 40494894 PMCID: PMC12152157 DOI: 10.1038/s43856-025-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/30/2025] [Indexed: 06/18/2025] Open
Abstract
BACKGROUND The apolipoprotein ε4 allele (APOE4) is associated with decreased longevity and increased vulnerability to age-related declines and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4-associated aging phenotypes. Here, we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. METHODS Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared body-wide for indices of middle-aged phenotypes. RESULTS Across peripheral and neural measures, APOE4 associates with poorer outcomes. Notably, 17αE2 treatment generally improves outcomes in a genotype-dependent manner, favoring APOE4 mice, including reductions in body weight, plasma leptin, hepatic steatosis, learning and memory, and oxidative damage in the brain. Plasma lipidomics and microglial transcriptomics show reductions in genotype-specific differences with 17αE2 treatment. CONCLUSIONS These findings demonstrate that APOE4 promotes systemic and neural aging phenotypes linked to AD and that 17αE2-mediated healthspan actions show a positive APOE4 bias. Collectively, the findings suggest that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with the APOE4 genotype.
Collapse
Affiliation(s)
- Cassandra J McGill
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wenjie Qian
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jose Godoy Lugo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Sara Namvari
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Olivia S White
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
- Molecular and Computational Biology Department, USC Dornsife College of Letters, Arts and Sciences, Los Angeles, CA, USA.
- Cancer Biology Department, USC Keck School of Medicine, Los Angeles, CA, USA.
- USC Norris Comprehensive Cancer Center, Epigenetics and Gene Regulation, Los Angeles, CA, USA.
- USC Stem Cell Initiative, Los Angeles, CA, USA.
| | - Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Yu K, Zhou X, Li B, Sun J, Yang T, Li W, Wang N, Gu X, Cui S, Cao R. Astrocytic spermidine insufficiency contributes to enhanced pain sensitivity associated with ApoE4. J Headache Pain 2025; 26:116. [PMID: 40375206 DOI: 10.1186/s10194-025-02054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025] Open
Abstract
Neuropathic pain is a chronic condition with limited effective treatments, closely associated with astrocytes and their role in central sensitization. Apolipoprotein E (ApoE), predominantly expressed in astrocytes in central nervous system, exists in three ApoE isoforms (ApoE2, ApoE3, and ApoE4) in humans, with ApoE4 linked to increased susceptibility to neurological diseases. However, the relationship between ApoE4 and neuropathic pain, as well as underlying mechanisms, remains poorly understood. Here, we demonstrated that mice expressing human ApoE4 (ApoE4-TR) displayed increased pain sensitivity following spared nerve injury (SNI) compared to ApoE3-TR mice. This increased sensitivity was also observed in mice with astrocyte-specific expression of ApoE4, achieved through Cre-mediated recombination. Metabolomic profiling revealed reduced spermidine levels in the spinal dorsal horn of ApoE4-TR mice relative to ApoE3-TR mice. Daily gavage administration of spermidine alleviated mechanical pain to a comparable level in ApoE3-TR and ApoE4-TR mice, as assessed by von Frey test. However, lower dose of spermidine effectively alleviated neuropathic pain in ApoE3-TR mice but showed reduced efficacy in ApoE4-TR mice, likely due to limited spermidine retention in ApoE4 astrocytes, as demonstrated in vitro. Transcriptomic analysis identified Nos2 as a critical gene upregulated in ApoE4-TR mice. Mechanistically, spermidine suppressed Nos2 expression by inhibiting the NF-κB pathway in astrocytes, thereby alleviating neuropathic pain. These findings highlight an enhanced pain sensitivity associated with ApoE4 and suggest spermidine as a potential therapeutic strategy, emphasizing a tailored dosage approach for ApoE4 carriers.
Collapse
Affiliation(s)
- Kaiming Yu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Xiongyao Zhou
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Baolong Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Jialu Sun
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Tuo Yang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Ningning Wang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| | - Shusen Cui
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, China.
| |
Collapse
|
3
|
Ellis D, Watanabe K, Wilmanski T, Lustgarten MS, Korat AVA, Glusman G, Hadlock J, Fiehn O, Sebastiani P, Price ND, Hood L, Magis AT, Evans SJ, Pflieger L, Lovejoy JC, Gibbons SM, Funk CC, Baloni P, Rappaport N. APOE genotype and biological age impact inter-omic associations related to bioenergetics. Aging (Albany NY) 2025; 17:206243. [PMID: 40323280 DOI: 10.18632/aging.206243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Apolipoprotein E (APOE) modifies human aging; specifically, the ε2 and ε4 alleles are among the strongest genetic predictors of longevity and Alzheimer's disease (AD) risk, respectively. However, detailed mechanisms for their influence on aging remain unclear. In the present study, we analyzed multi-omic association patterns across APOE genotypes, sex, and biological age (BA) axes in 2,229 community dwelling individuals. Our analysis, supported by validation in an independent cohort, identified diacylglycerols as the top APOE-associated plasma metabolites. However, despite the known opposing aging effects of the allele variants, both ε2- and ε4-carriers showed higher diacylglycerols compared to ε3-homozygotes. 'Omics association patterns of ε2-carriers and increased biological age were also counter-intuitively similar, displaying significantly increased associations between insulin resistance markers and energy-generating pathway metabolites. These results demonstrate the context-dependence of the influence of APOE, with ε2 potentially strengthening insulin resistance-like pathways in the decades prior to imparting its longevity benefits. Additionally, they provide an atlas of APOE-related 'omic associations and support the involvement of bioenergetic pathways in mediating the impact of APOE on aging.
Collapse
Affiliation(s)
- Dylan Ellis
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Kengo Watanabe
- Institute for Systems Biology, Seattle, WA 98109, USA
- Present address: Department of Medical Artificial Intelligence and Data Science, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | | | - Michael S Lustgarten
- Metabolism and Basic Biology of Aging, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | - Andres V Ardisson Korat
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | | | - Jennifer Hadlock
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA 95616, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Nathan D Price
- Thorne HealthTech, New York, NY 10019, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Phenome Health, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Simon J Evans
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Lance Pflieger
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jennifer C Lovejoy
- Institute for Systems Biology, Seattle, WA 98109, USA
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- eScience Institute, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Priyanka Baloni
- Institute for Systems Biology, Seattle, WA 98109, USA
- Present address: School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Noa Rappaport
- Institute for Systems Biology, Seattle, WA 98109, USA
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
4
|
Kalkman HO, Smigielski L. Ceramides may Play a Central Role in the Pathogenesis of Alzheimer's Disease: a Review of Evidence and Horizons for Discovery. Mol Neurobiol 2025:10.1007/s12035-025-04989-0. [PMID: 40295359 DOI: 10.1007/s12035-025-04989-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
While several hypotheses have been proposed to explain the underlying mechanisms of Alzheimer's disease, none have been entirely satisfactory. Both genetic and non-genetic risk factors, such as infections, metabolic disorders and psychological stress, contribute to this debilitating disease. Multiple lines of evidence indicate that ceramides may be central to the pathogenesis of Alzheimer's disease. Tumor necrosis factor-α, saturated fatty acids and cortisol elevate the brain levels of ceramides, while genetic risk factors, such as mutations in APP, presenilin, TREM2 and APOE ε4, also elevate ceramide synthesis. Importantly, ceramides displace sphingomyelin and cholesterol from lipid raft-like membrane patches that connect the endoplasmic reticulum and mitochondria, disturbing mitochondrial oxidative phosphorylation and energy production. As a consequence, the flattening of lipid rafts alters the function of γ-secretase, leading to increased production of Aβ42. Moreover, ceramides inhibit the insulin-signaling cascade via at least three mechanisms, resulting in the activation of glycogen synthase kinase-3 β. Activation of this kinase has multiple consequences, as it further deteriorates insulin resistance, promotes the transcription of BACE1, causes hyperphosphorylation of tau and inhibits the transcription factor Nrf2. Functional Nrf2 prevents apoptosis, mediates anti-inflammatory activity and improves blood-brain barrier function. Thus, various seemingly unrelated Alzheimer's disease risk factors converge on ceramide production, whereas the elevated levels of ceramides give rise to the well-known pathological features of Alzheimer's disease. Understanding and targeting these mechanisms may provide a promising foundation for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Hans O Kalkman
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Lukasz Smigielski
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Bhattarai P, Yilmaz E, Cakir EÖ, Korkmaz HY, Lee AJ, Ma Y, Celikkaya H, Cosacak MI, Haage V, Wang X, Nelson N, Lin W, Zhang Y, Nuriel T, Jülich D, Iş Ö, Holley SA, de Jager P, Fisher E, Tubbesing K, Teich AF, Bertucci T, Temple S, Ertekin-Taner N, Vardarajan BN, Mayeux R, Kizil C. APOE- ε4-induced Fibronectin at the blood-brain barrier is a conserved pathological mediator of disrupted astrocyte-endothelia interaction in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634732. [PMID: 39975303 PMCID: PMC11838230 DOI: 10.1101/2025.01.24.634732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Blood-brain barrier (BBB) dysfunction is a key feature of Alzheimer's disease (AD), particularly in individuals carrying the APOE-ε4 allele. This dysfunction worsens neuroinflammation and hinders the removal of toxic proteins, such as amyloid-beta (Aβ42), from the brain. In post-mortem brain tissues and in animal models, we previously reported that fibronectin accumulates at the BBB predominantly in APOE-ε4 carriers. Furthermore, we found a loss-of-function variant in the fibronectin 1 ( FN1 ) gene significantly reduces aggregated fibronectin levels and decreases AD risk among APOE-ε4 carriers. Yet, the molecular mechanisms downstream of fibronectin at the BBB remain unclear. The extracellular matrix (ECM) plays a crucial role in maintaining BBB homeostasis and orchestrating the interactions between BBB cell types, including endothelia and astrocytes. Understanding the mechanisms affecting the ECM and BBB cell types will be critical for developing effective therapies against AD, especially among APOE-ε4 carriers. Here, we demonstrate that APOE-ε4 , Aβ42, and inflammation drive the induction of FN1 expression in several models including zebrafish, mice, iPSC-derived human 3D astrocyte and 3D cerebrovascular cell cultures, and in human brains. Fibronectin accumulation disrupts astroglial-endothelial interactions and the signalling cascade between vascular endothelial growth factor (VEGF), heparin-binding epidermal growth factor (HBEGF) and Insulin-like growth factor 1 (IGF1). This accumulation of fibronectin in APOE-ε4- associated AD potentiates BBB dysfunction, which strongly implicates reducing fibronectin deposition as a potential therapeutic target for AD. Graphical abstract Accessibility text This image illustrates the effects of different APOE isoforms (ApoE-ε3 and ApoE-ε4) on blood-brain barrier (BBB) integrity, focusing on the molecular interactions between astrocytes and endothelial cells. This figure emphasizes the detrimental effects of ApoE-ε4 on BBB integrity via fibronectin accumulation and altered signaling pathways. The top section provides a schematic overview of the blood-brain barrier, highlighting astrocytes, endothelial cells, and their interface. The left panel represents the ApoE-ε3 condition: Normal fibronectin (FN1) levels support healthy interactions between astrocytes and endothelial cells. Growth factors, including VEGFA, HBEGF, and IGF1, maintain BBB integrity through their respective receptors (VEGFR and EGFR). Green arrows indicate activation of these signaling pathways. The right panel depicts the ApoE-ε4 condition: Elevated fibronectin (FN1) disrupts astrocyte-endothelium interactions. FN1 binds integrins and activates focal adhesion kinase (FAK), inhibiting VEGFA, which is required for endothelial HBEGF that in turn activates IGF1 signaling. Red symbols indicate inhibition of HBEGF, VEGFA, and IGF1 pathways, leading to BBB dysfunction. Highlights APOE-ε4 drives fibronectin deposition in Alzheimer's, disrupting astrocyte-endothelia interactions. APOE-ε4 and fibronectin co-localize, forming aggregates at blood-brain barrier (BBB). Fibronectin alters the signaling between VEGF, IGF1, and HBEGF impairing BBB function. Reducing fibronectin restores BBB integrity and offsets APOE-ε4 pathology.
Collapse
|
6
|
Guo JL, Braun D, Fitzgerald GA, Hsieh YT, Rougé L, Litvinchuk A, Steffek M, Propson NE, Heffner CM, Discenza C, Han SJ, Rana A, Skuja LL, Lin BQ, Sun EW, Davis SS, Balasundar S, Becerra I, Dugas JC, Ha C, Hsiao-Nakamoto J, Huang F, Jain S, Kung JE, Liau NPD, Mahon CS, Nguyen HN, Nguyen N, Samaddar M, Shi Y, Tatarakis D, Tian Y, Zhu Y, Suh JH, Sandmann T, Calvert MEK, Arguello A, Kane LA, Lewcock JW, Holtzman DM, Koth CM, Di Paolo G. Decreased lipidated ApoE-receptor interactions confer protection against pathogenicity of ApoE and its lipid cargoes in lysosomes. Cell 2025; 188:187-206.e26. [PMID: 39532095 PMCID: PMC11724755 DOI: 10.1016/j.cell.2024.10.027] [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: 01/05/2024] [Revised: 07/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
While apolipoprotein E (APOE) is the strongest genetic modifier for late-onset Alzheimer's disease (LOAD), the molecular mechanisms underlying isoform-dependent risk and the relevance of ApoE-associated lipids remain elusive. Here, we report that impaired low-density lipoprotein (LDL) receptor (LDLR) binding of lipidated ApoE2 (lipApoE2) avoids LDLR recycling defects observed with lipApoE3/E4 and decreases the uptake of cholesteryl esters (CEs), which are lipids linked to neurodegeneration. In human neurons, the addition of ApoE carrying polyunsaturated fatty acids (PUFAs)-CE revealed an allelic series (ApoE4 > ApoE3 > ApoE2) associated with lipofuscinosis, an age-related lysosomal pathology resulting from lipid peroxidation. Lipofuscin increased lysosomal accumulation of tau fibrils and was elevated in the APOE4 mouse brain with exacerbation by tau pathology. Intrahippocampal injection of PUFA-CE-lipApoE4 was sufficient to induce lipofuscinosis in wild-type mice. Finally, the protective Christchurch mutation also reduced LDLR binding and phenocopied ApoE2. Collectively, our data strongly suggest decreased lipApoE-LDLR interactions minimize LOAD risk by reducing the deleterious effects of endolysosomal targeting of ApoE and associated pathogenic lipids.
Collapse
Affiliation(s)
- Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA, USA.
| | - Dylan Braun
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Lionel Rougé
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Alexandra Litvinchuk
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Micah Steffek
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Suk Ji Han
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Anil Rana
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Lukas L Skuja
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Bi Qi Lin
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | - Jason C Dugas
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Connie Ha
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Fen Huang
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Shourya Jain
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | - Nathan Nguyen
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Yajuan Shi
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Yuxi Tian
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Yuda Zhu
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Lesley A Kane
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
7
|
Breeze B, Connell E, Wileman T, Muller M, Vauzour D, Pontifex MG. Menopause and Alzheimer's disease susceptibility: Exploring the potential mechanisms. Brain Res 2024; 1844:149170. [PMID: 39163895 DOI: 10.1016/j.brainres.2024.149170] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Alzheimer's Disease (AD), responsible for 62% of all dementia cases, is a progressive neurodegenerative condition that leads to cognitive dysfunction. The prevalence of AD is consistently higher in women suggesting they are disproportionately affected by this disease. Despite this, our understanding of this female AD vulnerability remains limited. Menopause has been identified as a potential contributing factor to AD in women, with earlier menopause onset associated with greater AD risk. However, the underlying mechanisms responsible for this increased risk are not fully understood. This review examines the potential role of menopause in the development of Alzheimer's Disease providing a mechanistic overview of the available literature from hormones to pathology. While literature is now emerging that indicates a role of hormonal shifts, gut dysbiosis, lipid dysregulation and inflammation, more research is needed to fully elucidate the mechanisms involved.
Collapse
Affiliation(s)
- Bernadette Breeze
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Emily Connell
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Tom Wileman
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom; Quadram Institute Biosciences, Norwich NR4 7UQ, United Kingdom
| | - Michael Muller
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - David Vauzour
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Matthew G Pontifex
- Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom.
| |
Collapse
|
8
|
Bulfon D, Breithofer J, Grabner GF, Fawzy N, Pirchheim A, Wolinski H, Kolb D, Hartig L, Tischitz M, Zitta C, Bramerdorfer G, Lass A, Taschler U, Kratky D, Greimel P, Zimmermann R. Functionally overlapping intra- and extralysosomal pathways promote bis(monoacylglycero)phosphate synthesis in mammalian cells. Nat Commun 2024; 15:9937. [PMID: 39548099 PMCID: PMC11568333 DOI: 10.1038/s41467-024-54213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
Bis(monoacylglycero)phosphate (BMP) is a major phospholipid constituent of intralumenal membranes in late endosomes/lysosomes, where it regulates the degradation and sorting of lipid cargo. Recent observations suggest that the Batten disease-associated protein CLN5 functions as lysosomal BMP synthase. Here, we show that transacylation reactions catalyzed by cytosolic and secreted enzymes enhance BMP synthesis independently of CLN5. The transacylases identified in this study are capable of acylating the precursor lipid phosphatidylglycerol (PG), generating acyl-PG, which is subsequently hydrolyzed to BMP. Extracellularly, acyl-PG and BMP are generated by endothelial lipase in cooperation with other serum enzymes of the pancreatic lipase family. The intracellular acylation of PG is catalyzed by several members of the cytosolic phospholipase A2 group IV (PLA2G4) family. Overexpression of secreted or cytosolic transacylases was sufficient to correct BMP deficiency in HEK293 cells lacking CLN5. Collectively, our observations suggest that functionally overlapping pathways promote BMP synthesis in mammalian cells.
Collapse
Grants
- Funding: this work was supported by SFB Lipid hydrolysis (10.55776/F73, D.K., R.Z.), 10.55776/P28533 (R.Z.), 10.55776/P35532 (R.Z.), the doctoral program doc-fund “Molecular Metabolism” 10.55776/DOC50 funded by the Austrian Science Fund FWF, Field of Excellence BioHealth – University of Graz, Graz, Austria, Province of Styria, City of Graz, BioTechMed-Graz, and NAWI Graz, and the Glycolipidologue Program of RIKEN (P.G.). For open access purposes, the authors have applied a CC BY public copyright license to any author accepted manuscript version arising from this submission
Collapse
Affiliation(s)
- Dominik Bulfon
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Nermeen Fawzy
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructure Analysis, Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Lennart Hartig
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martin Tischitz
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clara Zitta
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, Center for Brain Science, RIKEN, Wako, Saitama, Japan
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
9
|
Ellis D, Watanabe K, Wilmanski T, Lustgarten MS, Korat AVA, Glusman G, Hadlock JJ, Fiehn O, Sebastiani P, Price ND, Hood L, Magis AT, Evans SJ, Pflieger L, Lovejoy JC, Gibbons SM, Funk CC, Baloni P, Rappaport N. APOE Genotype and Biological Age Impact Inter-Omic Associations Related to Bioenergetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618322. [PMID: 39605362 PMCID: PMC11601402 DOI: 10.1101/2024.10.17.618322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Apolipoprotein E ( APOE ) modifies human aging; specifically, the ε2 and ε4 alleles are among the strongest genetic predictors of longevity and Alzheimer's disease (AD) risk, respectively. However, detailed mechanisms for their influence on aging remain unclear. Herein, we analyzed inter-omic, context-dependent association patterns across APOE genotypes, sex, and health axes in 2,229 community-dwelling individuals to test APOE genotypes for variation in metabolites and metabolite-associations tied to a previously-validated metric of biological aging (BA) based on blood biomarkers. Our analysis, supported by validation in an independent cohort, identified top APOE -associated plasma metabolites as diacylglycerols, which were increased in ε2-carriers and trended higher in ε4-carriers compared to ε3-homozygotes, despite the known opposing aging effects of the allele variants. 'Omics association patterns of ε2-carriers and increased biological age were also counter-intuitively similar, displaying increased associations between insulin resistance markers and energy-generating pathway metabolites. These results provide an atlas of APOE -related 'omic associations and support the involvement of bioenergetic pathways in mediating the impact of APOE on aging.
Collapse
|
10
|
Lindner K, Gavin AC. Isoform- and cell-state-specific APOE homeostasis and function. Neural Regen Res 2024; 19:2456-2466. [PMID: 38526282 PMCID: PMC11090418 DOI: 10.4103/nrr.nrr-d-23-01470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 12/26/2023] [Indexed: 03/26/2024] Open
Abstract
Apolipoprotein E is the major lipid transporter in the brain and an important player in neuron-astrocyte metabolic coupling. It ensures the survival of neurons under stressful conditions and hyperactivity by nourishing and detoxifying them. Apolipoprotein E polymorphism, combined with environmental stresses and/or age-related alterations, influences the risk of developing late-onset Alzheimer's disease. In this review, we discuss our current knowledge of how apolipoprotein E homeostasis, i.e. its synthesis, secretion, degradation, and lipidation, is affected in Alzheimer's disease.
Collapse
Affiliation(s)
- Karina Lindner
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
11
|
Moraes RCM, Roth JR, Mao H, Crawley SR, Xu BP, Watson JC, Melkani GC. Apolipoprotein E Induces Lipid Accumulation Through Dgat2 That Is Prevented with Time-Restricted Feeding in Drosophila. Genes (Basel) 2024; 15:1376. [PMID: 39596576 PMCID: PMC11594465 DOI: 10.3390/genes15111376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Apolipoprotein E (ApoE) is the leading genetic risk factor for late-onset Alzheimer's disease (AD), which is the leading cause of dementia worldwide. Most people have two ApoE-ε3 (ApoE3) alleles, while ApoE-ε2 (ApoE2) is protective from AD, and ApoE-ε4 (ApoE4) confers AD risk. How these alleles modulate AD risk is not clearly defined, and ApoE's role in lipid metabolism is also not fully known. Lipid droplets increase in AD. However, how ApoE contributes to lipid accumulation in the brain remains unknown. Methods: Here, we use Drosophila to study the effects of ApoE alleles on lipid accumulation in the brain and muscle in a cell-autonomous and non-cell-autonomous manner. Results: We report that pan-neuronal expression of each ApoE allele induces lipid accumulation specifically in the brain, but not in the muscle. However, this was not the case when expressed with muscle-specific drivers. ApoE2- and ApoE3-induced lipid accumulation is dependent on the expression of Dgat2, a key regulator of triacylglycerol production, while ApoE4 still induces lipid accumulation even with knock-down of Dgat2. Additionally, we find that implementation of time-restricted feeding (TRF), a dietary intervention in which food access only occurs in the active period (day), prevents ApoE-induced lipid accumulation in the brain of flies and modulates lipid metabolism genes. Conclusions: Altogether, our results demonstrate that ApoE induces lipid accumulation in the brain, that ApoE4 is unique in causing lipid accumulation independent of Dgat2, and that TRF prevents ApoE-induced lipid accumulation. These results support the idea that lipid metabolism is critical in AD, and that TRF could be a promising therapeutic approach to prevent ApoE-associated dysfunction in lipid metabolism.
Collapse
Affiliation(s)
- Ruan C. M. Moraes
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jonathan R. Roth
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Mao
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Savannah R. Crawley
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Brittney P. Xu
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John C. Watson
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Girish C. Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- UAB Nathan Shock Center, 1300 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
Christensen A, McGill CJ, Qian W, Pike CJ. Effects of obesogenic diet and 17β-estradiol in female mice with APOE 3/3, 3/4, and 4/4 genotypes. Front Aging Neurosci 2024; 16:1415072. [PMID: 39347015 PMCID: PMC11427389 DOI: 10.3389/fnagi.2024.1415072] [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: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The main genetic risk factor for Alzheimer's disease (AD) is the apolipoprotein E ε4 allele (APOE4). AD risk associated with APOE4 disproportionately affects women. Furthermore, human and rodent studies indicate that the cognitive deficits associated with APOE4 are greater in females. One modifiable AD risk factor is obesity during middle age. Given that approximately two-thirds of US adults are overweight, it is important to understand how obesity affects AD risk, how it interacts with APOE4, and the extent to which its detrimental effects can be mitigated with therapeutics. One intervention study for women is estrogen-based hormone therapy, which can exert numerous health benefits when administered in early middle age. No experimental studies have examined the interactions among APOE4, obesity, and hormone therapy in aging females. To begin to explore these issues, we considered how obesity outcomes are affected by treatment with estradiol at the onset of middle age in female mice with human APOE3 and APOE4. Furthermore, to explore how gene dosage affects outcomes, we compared mice homozygous for APOE3 (3/3) and homozygous (4/4) or hemizygous (3/4) for APOE4. Mice were examined over a 4-month period that spans the transition into reproductive senescence, a normal age-related change that models many aspects of human perimenopause. Beginning at 5 months of age, mice were maintained on a control diet (10% fat) or high-fat diet (HFD; 60% fat). After 8 weeks, by which time obesity was present in all HFD groups, mice were implanted with an estradiol or vehicle capsule that was maintained for the final 8 weeks. Animals were assessed on a range of metabolic and neural measures. Overall, APOE4 was associated with poorer metabolic function and cognitive performance. However, an obesogenic diet induced relatively greater impairments in metabolic function and cognitive performance in APOE3/3 mice. Estradiol treatment improved metabolic and cognitive outcomes across all HFD groups, with APOE4/4 generally exhibiting the greatest benefit. APOE3/4 mice were intermediate to the homozygous genotypes on many measures but also exhibited unique profiles. Together, these findings highlight the importance of the APOE genotype as a modulator of the risks associated with obesity and the beneficial outcomes of estradiol.
Collapse
Affiliation(s)
| | | | | | - Christian J. Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
13
|
Shan X, Zhao Z, Lai P, Liu Y, Li B, Ke Y, Jiang H, Zhou Y, Li W, Wang Q, Qin P, Xue Y, Zhang Z, Wei C, Ma B, Liu W, Luo C, Lu X, Lin J, Shu L, Jie Y, Xian X, Delcassian D, Ge Y, Miao L. RNA nanotherapeutics with fibrosis overexpression and retention for MASH treatment. Nat Commun 2024; 15:7263. [PMID: 39191801 DOI: 10.1038/s41467-024-51571-8] [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: 01/11/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) poses challenges for targeted delivery and retention of therapeutic proteins due to excess extracellular matrix (ECM). Here we present a new approach to treat MASH, termed "Fibrosis overexpression and retention (FORT)". In this strategy, we design (1) retinoid-derivative lipid nanoparticle (LNP) to enable enhanced mRNA overexpression in fibrotic regions, and (2) mRNA modifications which facilitate anchoring of therapeutic proteins in ECM. LNPs containing carboxyl-retinoids, rather than alcohol- or ester-retinoids, effectively deliver mRNA with over 10-fold enhancement of protein expression in fibrotic livers. The carboxyl-retinoid rearrangement on the LNP surface improves protein binding and membrane fusion. Therapeutic proteins are then engineered with an endogenous collagen-binding domain. These fusion proteins exhibit increased retention in fibrotic lesions and reduced systemic toxicity. In vivo, fibrosis-targeting LNPs encoding fusion proteins demonstrate superior therapeutic efficacy in three clinically relevant male-animal MASH models. This approach holds promise in fibrotic diseases unsuited for protein injection.
Collapse
Affiliation(s)
- Xinzhu Shan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhiqiang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Pingping Lai
- Institute of Cardiovascular Sciences and State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuxiu Liu
- Chinese Institute for Brain Research, Beijing, China
| | - Buyao Li
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yubin Ke
- China Spallation Neutron Source, Institute of High Energy Physics, Chinese Academy of Science, Dongguan, China
| | - Hanqiu Jiang
- China Spallation Neutron Source, Institute of High Energy Physics, Chinese Academy of Science, Dongguan, China
| | - Yilong Zhou
- Department of Surgery, Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pengxia Qin
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yizhe Xue
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zihan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Chenlong Wei
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Bin Ma
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wei Liu
- Keymed Biosciences (Chengdu) Limited, Chengdu, Sichuan, China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueguang Lu
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Jiaqi Lin
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Li Shu
- Interdisplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yin Jie
- Chinese Institute for Brain Research, Beijing, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences and State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing, China
| | | | - Yifan Ge
- Interdisplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China.
- Peking University-Yunnan Baiyao International Medical Research Center, Beijing, China.
| |
Collapse
|
14
|
Schepers J, Löser T, Behl C. Lipids and α-Synuclein: adding further variables to the equation. Front Mol Biosci 2024; 11:1455817. [PMID: 39188788 PMCID: PMC11345258 DOI: 10.3389/fmolb.2024.1455817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Aggregation of alpha-Synuclein (αSyn) has been connected to several neurodegenerative diseases, such as Parkinson's disease (PD), dementia with Lewy Bodies (DLB), and multiple system atrophy (MSA), that are collected under the umbrella term synucleinopathies. The membrane binding abilities of αSyn to negatively charged phospholipids have been well described and are connected to putative physiological functions of αSyn. Consequently, αSyn-related neurodegeneration has been increasingly connected to changes in lipid metabolism and membrane lipid composition. Indeed, αSyn aggregation has been shown to be triggered by the presence of membranes in vitro, and some genetic risk factors for PD and DLB are associated with genes coding for proteins directly involved in lipid metabolism. At the same time, αSyn aggregation itself can cause alterations of cellular lipid composition and brain samples of patients also show altered lipid compositions. Thus, it is likely that there is a reciprocal influence between cellular lipid composition and αSyn aggregation, which can be further affected by environmental or genetic factors and ageing. Little is known about lipid changes during physiological ageing and regional differences of the lipid composition of the aged brain. In this review, we aim to summarise our current understanding of lipid changes in connection to αSyn and discuss open questions that need to be answered to further our knowledge of αSyn related neurodegeneration.
Collapse
Affiliation(s)
| | | | - Christian Behl
- The Autophagy Lab, Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
15
|
McGill CJ, Christensen A, Qian W, Thorwald MA, Lugo JG, Namvari S, White OS, Finch CE, Benayoun BA, Pike CJ. Protection against APOE4 -associated aging phenotypes with the longevity-promoting intervention 17α-estradiol in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584678. [PMID: 38559059 PMCID: PMC10980056 DOI: 10.1101/2024.03.12.584678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The apolipoprotein ε4 allele ( APOE4 ) is associated with decreased longevity, increased vulnerability to age-related declines, and disorders across multiple systems. Interventions that promote healthspan and lifespan represent a promising strategy to attenuate the development of APOE4 -associated aging phenotypes. Here we studied the ability of the longevity-promoting intervention 17α-estradiol (17αE2) to protect against age-related impairments in APOE4 versus the predominant APOE3 genotype using early middle-aged mice with knock-in of human APOE alleles. Beginning at age 10 months, male APOE3 or APOE4 mice were treated for 20 weeks with 17αE2 or vehicle then compared for indices of aging phenotypes body-wide. Across peripheral and neural measures, APOE4 was associated with poorer outcomes. Notably, 17αE2 treatment improved outcomes in a genotype-dependent manner favoring APOE4 mice. These data demonstrate a positive APOE4 bias in 17αE2-mediated healthspan actions, suggesting that longevity-promoting interventions may be useful in mitigating deleterious age-related risks associated with APOE4 genotype.
Collapse
|
16
|
Litvinchuk A, Suh JH, Guo JL, Lin K, Davis SS, Bien-Ly N, Tycksen E, Tabor GT, Remolina Serrano J, Manis M, Bao X, Lee C, Bosch M, Perez EJ, Yuede CM, Cashikar AG, Ulrich JD, Di Paolo G, Holtzman DM. Amelioration of Tau and ApoE4-linked glial lipid accumulation and neurodegeneration with an LXR agonist. Neuron 2024; 112:384-403.e8. [PMID: 37995685 PMCID: PMC10922706 DOI: 10.1016/j.neuron.2023.10.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Litvinchuk
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Karin Lin
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Nga Bien-Ly
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, St. Louis, MO 63110, USA
| | - G Travis Tabor
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Choonghee Lee
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Megan Bosch
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Enmanuel J Perez
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Carla M Yuede
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Anil G Cashikar
- Department of Psychiatry, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
17
|
Almeida FC, Patra K, Giannisis A, Niesnerova A, Nandakumar R, Ellis E, Oliveira TG, Nielsen HM. APOE genotype dictates lipidomic signatures in primary human hepatocytes. J Lipid Res 2024; 65:100498. [PMID: 38216055 PMCID: PMC10875595 DOI: 10.1016/j.jlr.2024.100498] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/27/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024] Open
Abstract
Apolipoprotein E (APOE) genetic variants are most notably known for their divergent impact on the risk of developing Alzheimer's disease. While APOE genotype has been consistently shown to modulate lipid metabolism in a variety of cellular contexts, the effect of APOE alleles on the lipidome in hepatocytes is unknown. In this study, we investigated the contribution of APOE alleles to lipidomic profiles of donor-derived primary human hepatocytes from 77 subjects. Lipidomic data obtained by liquid chromatography-mass spectrometry were analyzed across ε2/ε3, ε3/ε3, and ε3/ε4 genotypes to reveal how APOE modulates lipid relative levels over age and between groups. Hepatic APOE concentration, measured by ELISA, was assessed for correlation with lipid abundance in subjects grouped as per APOE genotype and sex. APOE genotype-specific differential lipidomic signatures associated with age for multiple lipid classes but did not differ between sexes. Compared to ε2/ε3, ε3/ε4 hepatocytes had higher abundance of acylcarnitines (AC) and acylphosphatidylglycerol (AcylPG) as a class, as well as higher medium and long-chain ACs, AcylPG, phosphatidylglycerol (PG), bis(monoacylglycerol)phosphate (BMP), monoacylglycerol (MG) and diacylglycerol (DG) species. The ε3/ε4 hepatocytes also exhibited a higher abundance of medium and long-chain ACs compared to the ε3/ε3 hepatocytes. Only in the ε3/ε4 hepatocytes, APOE concentration was lower and showed a negative correlation with BMP levels, specifically in females. APOE genotype dictates a differential lipidome in primary human hepatocytes. The lipids involved suggest mitochondrial dysfunction with accompanying alterations in neutral lipid storage, reflective of a general disturbance of free fatty acid metabolism in human hepatocytes with the ε4 allele.
Collapse
Affiliation(s)
- Francisco C Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Neuroradiology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Kalicharan Patra
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Anezka Niesnerova
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Renu Nandakumar
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, USA
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology, (CLINTEC), Division of Transplantation surgery, Karolinska Institutet and ME Transplantation, Karolinska University Hospital, Huddinge, Sweden
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, Braga, Portugal.
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
18
|
Ferré-González L, Balaguer Á, Roca M, Ftara A, Lloret A, Cháfer-Pericás C. Brain areas lipidomics in female transgenic mouse model of Alzheimer's disease. Sci Rep 2024; 14:870. [PMID: 38195731 PMCID: PMC10776612 DOI: 10.1038/s41598-024-51463-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/05/2024] [Indexed: 01/11/2024] Open
Abstract
Lipids are the major component of the brain with important structural and functional properties. Lipid disruption could play a relevant role in Alzheimer's disease (AD). Some brain lipidomic studies showed significant differences compared to controls, but few studies have focused on different brain areas related to AD. Furthermore, AD is more prevalent in females, but there is a lack of studies focusing on this sex. This work aims to perform a lipidomic study in selected brain areas (cerebellum, amygdala, hippocampus, entire cortex) from wild-type (WT, n = 10) and APPswe/PS1dE9 transgenic (TG, n = 10) female mice of 5 months of age, as a model of early AD, to identify alterations in lipid composition. A lipidomic mass spectrometry-based method was optimized and applied to brain tissue. As result, some lipids showed statistically significant differences between mice groups in cerebellum (n = 68), amygdala (n = 49), hippocampus (n = 48), and the cortex (n = 22). In addition, some lipids (n = 15) from the glycerolipid, phospholipid, and sphingolipid families were statistically significant in several brain areas simultaneously between WT and TG. A selection of lipid variables was made to develop a multivariate approach to assess their discriminant potential, showing high diagnostic indexes, especially in cerebellum and amygdala (sensitivity 70-100%, sensibility 80-100%).
Collapse
Affiliation(s)
- Laura Ferré-González
- Alzheimer's Disease Research Group, Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Ángel Balaguer
- Faculty of Mathematics, University of Valencia, Valencia, Spain
| | - Marta Roca
- Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | | | - Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer's Disease Research Group, Health Research Institute La Fe, Avda de Fernando Abril Martorell, 106, 46026, Valencia, Spain.
| |
Collapse
|
19
|
Vardarajan B, Kalia V, Reyes-Dumeyer D, Dubey S, Nandakumar R, Lee A, Lantigua R, Medrano M, Rivera D, Honig L, Mayeux R, Miller G. Lysophosphatidylcholines are associated with P-tau181 levels in early stages of Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-3346076. [PMID: 38260644 PMCID: PMC10802729 DOI: 10.21203/rs.3.rs-3346076/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background We profiled circulating plasma metabolites to identify systemic biochemical changes in clinical and biomarker-assisted diagnosis of Alzheimer's disease (AD). Methods We used an untargeted approach with liquid chromatography coupled to high-resolution mass spectrometry to measure small molecule plasma metabolites from 150 clinically diagnosed AD patients and 567 age-matched healthy elderly of Caribbean Hispanic ancestry. Plasma biomarkers of AD were measured including P-tau181, Aβ40, Aβ42, total-tau, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP). Association of individual and co-abundant modules of metabolites were tested with clinical diagnosis of AD, as well as biologically-defined AD pathological process based on P-tau181 and other biomarker levels. Results Over 6000 metabolomic features were measured with high accuracy. First principal component (PC) of lysophosphatidylcholines (lysoPC) that bind to or interact with docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and arachidonic acid (AHA) was associated with decreased risk of AD (OR = 0.91 [0.89-0.96], p = 2e-04). Association was restricted to individuals without an APOE ε4 allele (OR = 0.89 [0.84-0.94], p = 8.7e-05). Among individuals carrying at least one APOE ε4 allele, PC4 of lysoPCs moderately increased risk of AD (OR = 1.37 [1.16-1.6], p = 1e-04). Essential amino acids including tyrosine metabolism pathways were enriched among metabolites associated with P-tau181 levels and heparan and keratan sulfate degradation pathways were associated with Aβ42/Aβ40 ratio. Conclusions Unbiased metabolic profiling can identify critical metabolites and pathways associated with β-amyloid and phosphotau pathology. We also observed an APOE-ε4 dependent association of lysoPCs with AD and biologically based diagnostic criteria may aid in the identification of unique pathogenic mechanisms.
Collapse
Affiliation(s)
| | - Vrinda Kalia
- Columbia University Mailman School of Public Health
| | | | | | | | - Annie Lee
- Center for Translational & Computational Neuroimmunology
| | | | | | | | | | | | | |
Collapse
|
20
|
Lozupone M, Panza F. Impact of apolipoprotein E isoforms on sporadic Alzheimer's disease: beyond the role of amyloid beta. Neural Regen Res 2024; 19:80-83. [PMID: 37488848 PMCID: PMC10479857 DOI: 10.4103/1673-5374.375316] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 04/16/2023] [Indexed: 07/26/2023] Open
Abstract
The impact of apolipoprotein E (ApoE) isoforms on sporadic Alzheimer's disease has long been studied; however, the influences of apolipoprotein E gene (APOE) on healthy and pathological human brains are not fully understood. ApoE exists as three common isoforms (ApoE2, ApoE3, and ApoE4), which differ in two amino acid residues. Traditionally, ApoE binds cholesterol and phospholipids and ApoE isoforms display different affinities for their receptors, lipids transport and distribution in the brain and periphery. The role of ApoE in the human depends on ApoE isoforms, brain regions, aging, and neural injury. APOE ε4 is the strongest genetic risk factor for sporadic Alzheimer's disease, considering its role in influencing amyloid-beta metabolism. The exact mechanisms by which APOE gene variants may increase or decrease Alzheimer's disease risk are not fully understood, but APOE was also known to affect directly and indirectly tau-mediated neurodegeneration, lipids metabolism, neurovascular unit, and microglial function. Consistent with the biological function of ApoE, ApoE4 isoform significantly altered signaling pathways associated with cholesterol homeostasis, transport, and myelination. Also, the rare protective APOE variants confirm that ApoE plays an important role in Alzheimer's disease pathogenesis. The objectives of the present mini-review were to describe classical and new roles of various ApoE isoforms in Alzheimer's disease pathophysiology beyond the deposition of amyloid-beta and to establish a functional link between APOE, brain function, and memory, from a molecular to a clinical level. APOE genotype also exerted a heterogeneous effect on clinical Alzheimer's disease phenotype and its outcomes. Not only in learning and memory but also in neuropsychiatric symptoms that occur in a premorbid condition. Clarifying the relationships between Alzheimer's disease-related pathology with neuropsychiatric symptoms, particularly suicidal ideation in Alzheimer's disease patients, may be useful for elucidating also the underlying pathophysiological process and its prognosis. Also, the effects of anti-amyloid-beta drugs, recently approved for the treatment of Alzheimer's disease, could be influenced by the APOE genotype.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology “Saverio de Bellis”, Research Hospital, Castellana Grotte, Bari, Italy
| |
Collapse
|
21
|
Medoh UN, Hims A, Chen JY, Ghoochani A, Nyame K, Dong W, Abu-Remaileh M. The Batten disease gene product CLN5 is the lysosomal bis(monoacylglycero)phosphate synthase. Science 2023; 381:1182-1189. [PMID: 37708259 DOI: 10.1126/science.adg9288] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023]
Abstract
Lysosomes critically rely on bis(monoacylglycero)phosphate (BMP) to stimulate lipid catabolism, cholesterol homeostasis, and lysosomal function. Alterations in BMP levels in monogenic and complex neurodegeneration suggest an essential function in human health. However, the site and mechanism responsible for BMP synthesis have been subject to debate for decades. Here, we report that the Batten disease gene product CLN5 is the elusive BMP synthase (BMPS). BMPS-deficient cells exhibited a massive accumulation of the BMP synthesis precursor lysophosphatidylglycerol (LPG), depletion of BMP species, and dysfunctional lipid metabolism. Mechanistically, we found that BMPS mediated synthesis through an energy-independent base exchange reaction between two LPG molecules with increased activity on BMP-laden vesicles. Our study elucidates BMP biosynthesis and reveals an anabolic function of late endosomes/lysosomes.
Collapse
Affiliation(s)
- Uche N Medoh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andy Hims
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Julie Y Chen
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Ali Ghoochani
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Guardia-Escote L, Biosca-Brull J, Cabré M, Blanco J, Mladenova-Koleva M, Basaure P, Pérez-Fernández C, Sánchez-Santed F, Domingo JL, Colomina MT. Developmental brain lipidomics is influenced by postnatal chlorpyrifos exposure and APOE genetic background in mice. Arch Toxicol 2023; 97:2463-2475. [PMID: 37439814 PMCID: PMC10404178 DOI: 10.1007/s00204-023-03555-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Lipids are a major component of the brain, and are involved in structural and neurodevelopmental processes such as neurogenesis, synaptogenesis and signaling. Apolipoprotein E (apoE) is the main lipoprotein involved in lipid transport in the brain. The apoE isoforms can determine vulnerability to the toxic effects of the pesticide chlorpyrifos (CPF), which can interfere with normal neurodevelopment. We aimed to study the effects of postnatal exposure to CPF and of the APOE genotype on the lipid composition of the brain at early ages. For it, we used apoE3 and apoE4 targeted-replacement (TR) male mice, as well as wild-type C57BL/6. The mice were orally exposed to 1 mg/kg/day of CPF on postnatal days 10-15 and, four hours after the treatment, we obtained samples to assess the cerebral lipid composition. Differences between APOE genotypes were found in the cerebral lipid profile in the postnatal period. ApoE4-TR mice exhibited higher lipid concentrations compared to the other groups in most of the cases. CPF exposure led to a decrease in cholesteryl ester and triglyceride concentrations, while modulating the levels of phosphatidylcholine species based on the apoE isoform. Specifically, CPF treatment decreased the concentration of some species of this lipid (PC30:0, PC31:0, PC32:2, PC36:5, PC40:4 and PC40:5) in C57BL/6 mice exposed to CPF, increased (PC31:0 and PC37:6) in apoE3-TR exposed mice while exposed apoE4-TR mice remained unaltered. These results provide further insights into the lipid composition of the brain at early ages, and how it can be modulated by environmental and genetic factors.
Collapse
Affiliation(s)
- Laia Guardia-Escote
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
| | - Judit Biosca-Brull
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
| | - Maria Cabré
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Tarragona, Spain
| | - Jordi Blanco
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
- Department of Basic Medical Sciences, Universitat Rovira i Virgili, Reus, Spain
| | - Mikaela Mladenova-Koleva
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
| | - Pia Basaure
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain
| | | | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almería University, Almería, Spain
| | - José L Domingo
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain
| | - Maria Teresa Colomina
- Research Group in Neurobehavior and Health (NEUROLAB), Universitat Rovira i Virgili, Tarragona, Spain.
- Department of Psychology and Research Center for Behavior Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.
- Laboratory of Toxicology and Environmental Health (TECNATOX), Universitat Rovira i Virgili, Reus, Spain.
| |
Collapse
|
23
|
Kalia V, Reyes-Dumeyer D, Dubey S, Nandakumar R, Lee AJ, Lantigua R, Medrano M, Rivera D, Honig LS, Mayeux R, Miller GW, Vardarajan BN. Lysophosphatidylcholines are associated with P-tau181 levels in early stages of Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.24.23294581. [PMID: 37662203 PMCID: PMC10473810 DOI: 10.1101/2023.08.24.23294581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background We investigated systemic biochemical changes in Alzheimer's disease (AD) by investigating the relationship between circulating plasma metabolites and both clinical and biomarker-assisted diagnosis of AD. Methods We used an untargeted approach with liquid chromatography coupled to high-resolution mass spectrometry to measure exogenous and endogenous small molecule metabolites in plasma from 150 individuals clinically diagnosed with AD and 567 age-matched elderly without dementia of Caribbean Hispanic ancestry. Plasma biomarkers of AD were also measured including P-tau181, Aβ40, Aβ42, total tau, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP). Association of individual and co-expressed modules of metabolites were tested with the clinical diagnosis of AD, as well as biologically-defined AD pathological process based on P-tau181 and other biomarker levels. Results Over 4000 metabolomic features were measured with high accuracy. First principal component (PC) of lysophosphatidylcholines (lysoPC) that bind to or interact with docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and arachidonic acid (AHA) was associated with decreased risk of AD (OR=0.91 [0.89-0.96], p=2e-04). Restricted to individuals without an APOE ε4 allele (OR=0.89 [0.84-0.94], p= 8.7e-05), the association remained. Among individuals carrying at least one APOE ε4 allele, PC4 of lysoPCs moderately increased risk of AD (OR=1.37 [1.16-1.6], p=1e-04). Essential amino acids including tyrosine metabolism pathways were enriched among metabolites associated with P-tau181 levels and heparan and keratan sulfate degradation pathways were associated with Aβ42/Aβ40 ratio reflecting different pathways enriched in early and middle stages of disease. Conclusions Our findings indicate that unbiased metabolic profiling can identify critical metabolites and pathways associated with β-amyloid and phosphotau pathology. We also observed an APOE ε4 dependent association of lysoPCs with AD and that biologically-based diagnostic criteria may aid in the identification of unique pathogenic mechanisms.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
| | - Saurabh Dubey
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Renu Nandakumar
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
| | - Rafael Lantigua
- Department of Medicine, College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital. 630 West 168 Street, New York, NY 10032
| | - Martin Medrano
- School of Medicine, Pontificia Universidad Católica Madre y Maestra, Santiago, Dominican Republic
| | - Diones Rivera
- Department of Neurosurgery, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
- Department of Epidemiology, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
- Department of Epidemiology, Mailman School of Public Health, Columbia University. 722 West 168 Street, New York, NY 10032
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University. 630 West 168 Street, New York, NY 10032
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital. 710 West 168 Street, New York, NY 10032
| |
Collapse
|
24
|
Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab 2023; 34:430-445. [PMID: 37357100 PMCID: PMC10365028 DOI: 10.1016/j.tem.2023.05.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/27/2023]
Abstract
Dysregulation of lipid metabolism has emerged as a central component of many neurodegenerative diseases. Variants of the lipid transport protein, apolipoprotein E (APOE), modulate risk and resilience in several neurodegenerative diseases including late-onset Alzheimer's disease (LOAD). Allelic variants of the gene, APOE, alter the lipid metabolism of cells and tissues and have been broadly associated with several other cellular and systemic phenotypes. Targeting APOE-associated metabolic pathways may offer opportunities to alter disease-related phenotypes and consequently, attenuate disease risk and impart resilience to multiple neurodegenerative diseases. We review the molecular, cellular, and tissue-level alterations to lipid metabolism that arise from different APOE isoforms. These changes in lipid metabolism could help to elucidate disease mechanisms and tune neurodegenerative disease risk and resilience.
Collapse
Affiliation(s)
- Linda G Yang
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zachary M March
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Roxan A Stephenson
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Priyanka S Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.; National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias (CARD), National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
25
|
Zhang L, Xia Y, Gui Y. Neuronal ApoE4 in Alzheimer's disease and potential therapeutic targets. Front Aging Neurosci 2023; 15:1199434. [PMID: 37333457 PMCID: PMC10272394 DOI: 10.3389/fnagi.2023.1199434] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
The most prevalent genetic risk factor for Alzheimer's disease (AD) is Apolipoprotein E (ApoE), a gene located on chromosome 19 that encodes three alleles (e2, e3, and e4) that give rise to the ApoE subtypes E2, E3, and E4, respectively. E2 and E4 have been linked to increased plasma triglyceride concentrations and are known to play a critical role in lipoprotein metabolism. The prominent pathological features of AD mainly include senile plaques formed by amyloid β (Aβ42) aggregation and neuronal fibrous tangles (NFTs), and the deposited plaques are mainly composed of Aβ hyperphosphorylation and truncated head. In the central nervous system, the ApoE protein is primarily derived from astrocytes, but ApoE is also produced when neurons are stressed or affected by certain stress, injury, and aging conditions. ApoE4 in neurons induces Aβ and tau protein pathologies, leading to neuroinflammation and neuronal damage, impairing learning and memory functions. However, how neuronal ApoE4 mediates AD pathology remains unclear. Recent studies have shown that neuronal ApoE4 may lead to greater neurotoxicity, which increases the risk of AD development. This review focuses on the pathophysiology of neuronal ApoE4 and explains how neuronal ApoE4 mediates Aβ deposition, pathological mechanisms of tau protein hyperphosphorylation, and potential therapeutic targets.
Collapse
|
26
|
Lee S, Devanney NA, Golden LR, Smith CT, Schwartz JL, Walsh AE, Clarke HA, Goulding DS, Allenger EJ, Morillo-Segovia G, Friday CM, Gorman AA, Hawkinson TR, MacLean SM, Williams HC, Sun RC, Morganti JM, Johnson LA. APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge. Cell Rep 2023; 42:112196. [PMID: 36871219 PMCID: PMC10117631 DOI: 10.1016/j.celrep.2023.112196] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/29/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The E4 allele of Apolipoprotein E (APOE) is associated with both metabolic dysfunction and a heightened pro-inflammatory response: two findings that may be intrinsically linked through the concept of immunometabolism. Here, we combined bulk, single-cell, and spatial transcriptomics with cell-specific and spatially resolved metabolic analyses in mice expressing human APOE to systematically address the role of APOE across age, neuroinflammation, and AD pathology. RNA sequencing (RNA-seq) highlighted immunometabolic changes across the APOE4 glial transcriptome, specifically in subsets of metabolically distinct microglia enriched in the E4 brain during aging or following an inflammatory challenge. E4 microglia display increased Hif1α expression and a disrupted tricarboxylic acid (TCA) cycle and are inherently pro-glycolytic, while spatial transcriptomics and mass spectrometry imaging highlight an E4-specific response to amyloid that is characterized by widespread alterations in lipid metabolism. Taken together, our findings emphasize a central role for APOE in regulating microglial immunometabolism and provide valuable, interactive resources for discovery and validation research.
Collapse
Affiliation(s)
- Sangderk Lee
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Nicholas A Devanney
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Cathryn T Smith
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - James L Schwartz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Harrison A Clarke
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Danielle S Goulding
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | | | | | - Cassi M Friday
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Amy A Gorman
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Tara R Hawkinson
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Steven M MacLean
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ramon C Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Josh M Morganti
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
27
|
Ferré-González L, Lloret A, Cháfer-Pericás C. Systematic review of brain and blood lipidomics in Alzheimer's disease mouse models. Prog Lipid Res 2023; 90:101223. [PMID: 36871907 DOI: 10.1016/j.plipres.2023.101223] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Alzheimer's disease (AD) diagnosis is based on invasive and expensive biomarkers. Regarding AD pathophysiological mechanisms, there is evidence of a link between AD and aberrant lipid homeostasis. Alterations in lipid composition have been observed in blood and brain samples, and transgenic mouse models represent a promising approach. Nevertheless, there is great variability among studies in mice for the determination of different types of lipids in targeted and untargeted methods. It could be explained by the different variables (model, age, sex, analytical technique), and experimental conditions used. The aim of this work is to review the studies on lipid alteration in brain tissue and blood samples from AD mouse models, focusing on different experimental parameters. As result, great disparity has been observed among the reviewed studies. Brain studies showed an increase in gangliosides, sphingomyelins, lysophospholipids and monounsaturated fatty acids and a decrease in sulfatides. In contrast, blood studies showed an increase in phosphoglycerides, sterols, diacylglycerols, triacylglycerols and polyunsaturated fatty acids, and a decrease in phospholipids, lysophospholipids and monounsaturated fatty acids. Thus, lipids are closely related to AD, and a consensus on lipidomics studies could be used as a diagnostic tool and providing insight into the mechanisms involved in AD.
Collapse
Affiliation(s)
- Laura Ferré-González
- Alzheimer's Disease Research Group, Health Research Institute La Fe, Valencia, Spain
| | - Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain.
| | | |
Collapse
|
28
|
Sebastiani P, Song Z, Ellis D, Tian Q, Schwaiger-Haber M, Stancliffe E, Lustgarten MS, Funk CC, Baloni P, Yao CH, Joshi S, Marron MM, Gurinovich A, Li M, Leshchyk A, Xiang Q, Andersen SL, Feitosa MF, Ukraintseva S, Soerensen M, Fiehn O, Ordovas JM, Haigis M, Monti S, Barzilai N, Milman S, Ferrucci L, Rappaport N, Patti GJ, Perls TT. A metabolomic signature of the APOE2 allele. GeroScience 2023; 45:415-426. [PMID: 35997888 PMCID: PMC9886693 DOI: 10.1007/s11357-022-00646-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/15/2022] [Indexed: 02/03/2023] Open
Abstract
With the goal of identifying metabolites that significantly correlate with the protective e2 allele of the apolipoprotein E (APOE) gene, we established a consortium of five studies of healthy aging and extreme human longevity with 3545 participants. This consortium includes the New England Centenarian Study, the Baltimore Longitudinal Study of Aging, the Arivale study, the Longevity Genes Project/LonGenity studies, and the Long Life Family Study. We analyzed the association between APOE genotype groups E2 (e2e2 and e2e3 genotypes, N = 544), E3 (e3e3 genotypes, N = 2299), and E4 (e3e4 and e4e4 genotypes, N = 702) with metabolite profiles in the five studies and used fixed effect meta-analysis to aggregate the results. Our meta-analysis identified a signature of 19 metabolites that are significantly associated with the E2 genotype group at FDR < 10%. The group includes 10 glycerolipids and 4 glycerophospholipids that were all higher in E2 carriers compared to E3, with fold change ranging from 1.08 to 1.25. The organic acid 6-hydroxyindole sulfate, previously linked to changes in gut microbiome that were reflective of healthy aging and longevity, was also higher in E2 carriers compared to E3 carriers. Three sterol lipids and one sphingolipid species were significantly lower in carriers of the E2 genotype group. For some of these metabolites, the effect of the E2 genotype opposed the age effect. No metabolites reached a statistically significant association with the E4 group. This work confirms and expands previous results connecting the APOE gene to lipid regulation and suggests new links between the e2 allele, lipid metabolism, aging, and the gut-brain axis.
Collapse
Affiliation(s)
- Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA.
| | - Zeyuan Song
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Dylan Ellis
- Institute for Systems Biology, Seattle, WA, USA
| | - Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute On Aging, Baltimore, MD, USA
| | - Michaela Schwaiger-Haber
- Department of Chemistry, Department of Medicine, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, USA
| | - Ethan Stancliffe
- Department of Chemistry, Department of Medicine, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, USA
| | - Michael S Lustgarten
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center On Aging at Tufts University, Boston, MA, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, USA
| | | | - Cong-Hui Yao
- Department of Cell Biology at Harvard Medical School, Boston, MA, USA
| | - Shakchhi Joshi
- Department of Cell Biology at Harvard Medical School, Boston, MA, USA
| | - Megan M Marron
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA
| | - Mengze Li
- Bioinformatics Program, Boston University, Boston, MA, USA
| | | | - Qingyan Xiang
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Stacy L Andersen
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St Louis, MI, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research, Duke University, Durham, NC, USA
| | - Mette Soerensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA
| | - Jose M Ordovas
- Nutrition and Genomics Team, Jean Mayer USDA Human Nutrition Research Center On Aging and Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MB, USA
| | - Marcia Haigis
- Department of Cell Biology at Harvard Medical School, Boston, MA, USA
| | - Stefano Monti
- Bioinformatics Program, Boston University, Boston, MA, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sofiya Milman
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute On Aging, Baltimore, MD, USA
| | | | - Gary J Patti
- Department of Chemistry, Department of Medicine, Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, USA
| | - Thomas T Perls
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Leikin-Frenkel A, Schnaider Beeri M, Cooper I. How Alpha Linolenic Acid May Sustain Blood-Brain Barrier Integrity and Boost Brain Resilience against Alzheimer's Disease. Nutrients 2022; 14:nu14235091. [PMID: 36501121 PMCID: PMC9737216 DOI: 10.3390/nu14235091] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Cognitive decline, the primary clinical phenotype of Alzheimer's disease (AD), is currently attributed mainly to amyloid and tau protein deposits. However, a growing body of evidence is converging on brain lipids, and blood-brain barrier (BBB) dysfunction, as crucial players involved in AD development. The critical role of lipids metabolism in the brain and its vascular barrier, and its constant modifications particularly throughout AD development, warrants investigation of brain lipid metabolism as a high value therapeutic target. Yet, there is limited knowledge on the biochemical and structural roles of lipids in BBB functionality in AD. Within this framework, we hypothesize that the ApoE4 genotype, strongly linked to AD risk and progression, may be related to altered fatty acids composition in the BBB. Interestingly, alpha linolenic acid (ALA), the precursor of the majoritarian brain component docosahexaenoic acid (DHA), emerges as a potential novel brain savior, acting via BBB functional improvements, and this may be primarily relevant to ApoE4 carriers.
Collapse
Affiliation(s)
- Alicia Leikin-Frenkel
- Bert Strassburger Lipid Center, Sheba Medical Center, Tel-Hashomer 52621, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- School of Psychology, The Reichman University (IDC), Herzliya 4610101, Israel
- Correspondence: ; Tel.: +972-3-5303693
| |
Collapse
|
30
|
Toledano-Díaz A, Álvarez MI, Toledano A. The relationships between neuroglial alterations and neuronal changes in Alzheimer's disease, and the related controversies I: Gliopathogenesis and glioprotection. J Cent Nerv Syst Dis 2022; 14:11795735221128703. [PMID: 36238130 PMCID: PMC9551335 DOI: 10.1177/11795735221128703] [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] [Indexed: 12/05/2022] Open
Abstract
Since Alois Alzheimer described the pathology of Alzheimer's disease in 1907, an increasing number of studies have attempted to discover its causes and possible ways to treat it. For decades, research has focused on neuronal degeneration and the disruption to the neural circuits that occurs during disease progression, undervaluing in some extent the alterations to glial cells even though these alterations were described in the very first studies of this disease. In recent years, it has been recognized that different families of neuroglia are not merely support cells for neurons but rather key and active elements in the physiology and pathology of the nervous system. Alterations to different types of neuroglia (especially astroglia and microglia but also mature oligodendroglia and oligodendroglial progenitors) have been identified in the initial neuropathological changes that lead to dementia, suggesting that they may represent therapeutic targets to prevent neurodegeneration. In this review, based on our own studies and on the relevant scientific literature, we argue that a careful and in-depth study of glial cells will be fundamental to understanding the origin and progression of Alzheimer's disease. In addition, we analyze the main issues regarding the neuroprotective and neurotoxic role of neuroglial changes, reactions and/or involutions in both humans with Alzheimer's disease and in experimental models of this condition.
Collapse
|
31
|
Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, Gouras GK, Deierborg T. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener 2022; 17:62. [PMID: 36153580 PMCID: PMC9509584 DOI: 10.1186/s13024-022-00566-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
ApoE is the major lipid and cholesterol carrier in the CNS. There are three major human polymorphisms, apoE2, apoE3, and apoE4, and the genetic expression of APOE4 is one of the most influential risk factors for the development of late-onset Alzheimer's disease (AD). Neuroinflammation has become the third hallmark of AD, together with Amyloid-β plaques and neurofibrillary tangles of hyperphosphorylated aggregated tau protein. This review aims to broadly and extensively describe the differential aspects concerning apoE. Starting from the evolution of apoE to how APOE's single-nucleotide polymorphisms affect its structure, function, and involvement during health and disease. This review reflects on how APOE's polymorphisms impact critical aspects of AD pathology, such as the neuroinflammatory response, particularly the effect of APOE on astrocytic and microglial function and microglial dynamics, synaptic function, amyloid-β load, tau pathology, autophagy, and cell-cell communication. We discuss influential factors affecting AD pathology combined with the APOE genotype, such as sex, age, diet, physical exercise, current therapies and clinical trials in the AD field. The impact of the APOE genotype in other neurodegenerative diseases characterized by overt inflammation, e.g., alpha- synucleinopathies and Parkinson's disease, traumatic brain injury, stroke, amyotrophic lateral sclerosis, and multiple sclerosis, is also addressed. Therefore, this review gathers the most relevant findings related to the APOE genotype up to date and its implications on AD and CNS pathologies to provide a deeper understanding of the knowledge in the APOE field.
Collapse
Affiliation(s)
- Rosalía Fernández-Calle
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Javier Frontiñán-Rubio
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Juan García-Revilla
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Martina Svensson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Isak Martinson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luís Venero
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Henrietta M. Nielsen
- Department of Biochemistry and Biophysics at, Stockholm University, Stockholm, Sweden
| | - Gunnar K. Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|