995 results on '"APOE4"'
Search Results
2. APOE4, Alzheimer’s and periodontal disease: A scoping review
- Author
-
Arévalo-Caro, Catalina, Arce Retana, Marianela, Losada Amaya, Sergio, Arboleda, Humberto, Gallart-Palau, Xavier, and Serra, Aida
- Published
- 2025
- Full Text
- View/download PDF
3. Suppressing APOE4-induced neural pathologies by targeting the VHL-HIF axis.
- Author
-
Jiang, Wei, Cao, Yiming, Xue, Yue, Ji, Yichun, Winer, Benjamin, Chandra, Rashmi, Zhang, Xingyuan, Zhang, Mengqi, Singhal, Neel, Pierce, Jonathan, Chen, Song, and Ma, Dengke
- Subjects
APOE4 ,VHL–HIF axis ,mitochondrial dysfunction ,neurodegeneration ,oxidative stress ,Animals ,Von Hippel-Lindau Tumor Suppressor Protein ,Caenorhabditis elegans ,Apolipoprotein E4 ,Humans ,Mice ,Caenorhabditis elegans Proteins ,Oxidative Stress ,Alzheimer Disease ,Mitochondria ,Mice ,Transgenic ,Transcription Factors - Abstract
The ε4 variant of human apolipoprotein E (APOE4) is a key genetic risk factor for neurodegeneration in Alzheimers disease and elevated all-cause mortality in humans. Understanding the factors and mechanisms that can mitigate the harmful effects of APOE4 has significant implications. In this study, we find that inactivating the VHL-1 (Von Hippel-Lindau) protein can suppress mortality, neural and behavioral pathologies caused by transgenic human APOE4 in Caenorhabditis elegans. The protective effects of VHL-1 deletion are recapitulated by stabilized HIF-1 (hypoxia-inducible factor), a transcription factor degraded by VHL-1. HIF-1 activates a genetic program that safeguards against mitochondrial dysfunction, oxidative stress, proteostasis imbalance, and endolysosomal rupture-critical cellular events linked to neural pathologies and mortality. Furthermore, genetic inhibition of Vhl reduces cerebral vascular injury and synaptic lesions in APOE4 mice, suggesting an evolutionarily conserved mechanism. Thus, we identify the VHL-HIF axis as a potent modulator of APOE4-induced neural pathologies and propose that targeting this pathway in nonproliferative tissues may curb cellular damage, protect against neurodegeneration, and reduce tissue injuries and mortality.
- Published
- 2025
4. Uncovering dark mass in population proteomics: Pan-analysis of single amino acid polymorphism relevant to cognition and aging
- Author
-
Gao, Xiaojing, Yin, Yuanyuan, Chen, Yiqian, Lu, Ling, Zhao, Jian, Lin, Xu, Wu, Jiarui, Li, Qingrun, and Zeng, Rong
- Published
- 2025
- Full Text
- View/download PDF
5. Optimized prime editing of the Alzheimer’s disease-associated APOE4 mutation
- Author
-
Rottner, Antje K., Lundin, Anders, Li, Songyuan, Firth, Mike, Maresca, Marcello, and Sienski, Grzegorz
- Published
- 2025
- Full Text
- View/download PDF
6. Endothelial progenitor cells and cerebral small vessel disease in APOE4 carriers
- Author
-
Kapoor, Arunima, Dutt, Shubir, Nguyen, Amy, Lohman, Trevor, Gaubert, Aimée, Alitin, John Paul M., Sible, Isabel J, Marshall, Anisa, Shenasa, Fatemah, Engstrom, Allison C, Bradford, David Robert, Rodgers, Kathleen, and Nation, Daniel A
- Published
- 2025
- Full Text
- View/download PDF
7. Precuneus Activity during Retrieval Is Positively Associated with Amyloid Burden in Cognitively Normal Older APOE4 Carriers.
- Author
-
Fischer, Larissa, Molloy, Eóin N., Binette, Alexa Pichet, Vockert, Niklas, Marquardt, Jonas, Pilar, Andrea Pacha, Kreissl, Michael C., Remz, Jordana, Tremblay-Mercier, Jennifer, Poirier, Judes, Rajah, Maria Natasha, Villeneuve, Sylvia, and Maass, Anne
- Abstract
The precuneus is a site of early amyloid-beta (Aß) accumulation. Previous cross-sectional studies reported increased precuneus fMRI activity in older adults with mild cognitive deficits or elevated Aß. However, longitudinal studies in early Alzheimer's disease (AD) are lacking and the relationship to the Apolipoprotein-E (APOE) genotype is unclear. Investigating the PREVENT-AD dataset, we assessed how baseline and longitudinal precuneus activity during successful memory retrieval relates to future Aß and tau burden and change in memory performance. We further studied the moderation by APOE4 genotype. We included 165 older adults (age, 62.8 ± 4.4 years; 113 female; 66 APOE4 carriers) who were cognitively normal at baseline with a family history of AD. All participants performed task-fMRI at baseline and underwent 18F-flortaucipir-PET and 18F-NAV4694-Aß-PET on average 5 years later. We found that higher baseline activity and greater longitudinal increase in precuneus activity were associated with higher Aß burden in APOE4 carriers but not noncarriers. We observed no effects of precuneus activity on tau burden. Finally, APOE4 noncarriers with low baseline precuneus activity exhibited better longitudinal performance in an independent memory test compared with (1) noncarriers with higher baseline activity and (2) APOE4 carriers. Our findings suggest that higher task-related precuneus activity duringmemory retrieval at baseline and over time are associated with greater Aß burden in cognitively normal APOE4 carriers. Our results further indicate that the absence of "hyperactivation" and the absence of the APOE4 allele is related with better future cognitive outcomes in cognitively normal older adults at risk for AD. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
8. Selenium and Episodic Memory: The Moderating Role of Apolipoprotein E ε4.
- Author
-
Kim, Shin Gyeom, Keum, Musung, Choe, Young Min, Suh, Guk-Hee, Lee, Boung Chul, Kim, Hyun Soo, Lee, Jun Hyung, Hwang, Jaeuk, Yi, Dahyun, and Kim, Jee Wook
- Abstract
Background: Selenium (Se), a vital trace element, plays a neuroprotective role by mitigating oxidative stress through selenoproteins and regulating metal balance. The apolipoprotein E ε4 allele (APOE4), a significant genetic risk factor for Alzheimer's disease (AD), has been linked to reduced Se levels and weakened antioxidant capacity. This research explores the association between serum Se concentrations and cognitive performance, with an emphasis on how APOE4 status influences this relationship. Methods: This study included 196 older adults from community and memory clinic settings, who underwent assessments for episodic memory, global cognition, and non-memory functions using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological battery, with serum selenium levels analyzed via inductively coupled plasma–mass spectrometry (ICP-MS) and APOE genotyping conducted to determine allele status. Results: Higher serum Se levels were associated with better episodic memory score (EMS) (B = 0.065, 95% CI = 0.020–0.110, p = 0.005) and CERAD total score (TS) (B = 0.119, 95% CI = 0.046–0.193, p = 0.002). However, the interaction between Se and APOE4 status significantly affected EMS (B = −0.074, 95% CI = −0.109 to −0.039, p < 0.001), with significant benefits observed in APOE4-negative participants. Conclusions: This study highlights the genotype-specific impact of Se on cognitive health, emphasizing the need for personalized nutritional interventions targeting Se levels, particularly for APOE4-negative individuals. Future research should further elucidate the mechanisms of Se's effects and assess its therapeutic potential in aging populations. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
9. Lower activity of cholesteryl ester transfer protein (CETP) and the risk of dementia: a Mendelian randomization analysis.
- Author
-
Schmidt, Amand F., Davidson, Michael H., Ditmarsch, Marc, Kastelein, John J., and Finan, Chris
- Subjects
- *
CHOLESTERYL ester transfer protein , *LEWY body dementia , *HDL cholesterol , *LDL cholesterol , *PARKINSON'S disease - Abstract
Background: Elevated concentrations of low-density lipoprotein cholesterol (LDL-C) are linked to dementia risk, and conversely, increased plasma concentrations of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein-A1 (Apo-A1) associate with decreased dementia risk. Inhibition of cholesteryl ester transfer protein (CETP) meaningfully affects the concentrations of these blood lipids and may therefore provide an opportunity to treat dementia. Methods: Drug target Mendelian randomization (MR) was employed to anticipate the on-target effects of lower CETP concentration (μg/mL) on plasma lipids, cardiovascular disease outcomes, autopsy confirmed Lewy body dementia (LBD), as well as Parkinson's dementia. Results: MR analysis of lower CETP concentration recapitulated the blood lipid effects observed in clinical trials of CETP-inhibitors, as well as protective effects on coronary heart disease (odds ratio (OR) 0.92, 95% confidence interval (CI) 0.89; 0.96), heart failure, abdominal aortic aneurysm, any stroke, ischemic stroke, and small vessel stroke (0.90, 95%CI 0.85; 0.96). Consideration of dementia related traits indicated that lower CETP concentrations were associated higher total brain volume (0.04 per standard deviation, 95%CI 0.02; 0.06), lower risk of LBD (OR 0.81, 95%CI 0.74; 0.89) and Parkinson's dementia risk (OR 0.26, 95%CI 0.14; 0.48). APOE4 stratified analyses suggested the LBD effect was most pronounced in APOE-ε4 + participants (OR 0.61 95%CI 0.51; 0.73), compared to APOE-ε4- (OR 0.89 95%CI 0.79; 1.01); interaction p-value 5.81 × 10− 4. Conclusions: These results suggest that inhibition of CETP may be a viable strategy to treat dementia, with a more pronounced effect expected in APOE-ε4 carriers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Proteome profiling of cerebrospinal fluid using machine learning shows a unique protein signature associated with APOE4 genotype.
- Author
-
Shvetcov, Artur, Thomson, Shannon, Cho, Ann‐Na, Wilkins, Heather M., Reed, Joanne H., Swerdlow, Russell H., Brown, David A., and Finney, Caitlin A.
- Subjects
- *
SUPERVISED learning , *ALZHEIMER'S disease , *MILD cognitive impairment , *CEREBROSPINAL fluid , *APOLIPOPROTEIN E4 , *CEREBROSPINAL fluid examination - Abstract
Proteome changes associated with APOE4 variant carriage that are independent of Alzheimer's disease (AD) pathology and diagnosis are unknown. This study investigated APOE4 proteome changes in people with AD, mild cognitive impairment, and no impairment. Clinical, APOE genotype, and cerebrospinal fluid (CSF) proteome and AD biomarker data was sourced from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Proteome profiling was done using supervised machine learning. We found an APOE4‐specific proteome signature that was independent of cognitive diagnosis and AD pathological biomarkers, and increased the risk of progression to cognitive impairment. Proteins were enriched in brain regions including the caudate and cortex and cells including endothelial cells, oligodendrocytes, and astrocytes. Enriched peripheral immune cells included T cells, macrophages, and B cells. APOE4 carriers have a unique CSF proteome signature associated with a strong brain and peripheral immune and inflammatory phenotype that likely underlies APOE4 carriers' vulnerability to cognitive decline and AD as they age. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Skeletal muscle proteome differs between young APOE3 and APOE4 targeted replacement mice in a sex-dependent manner.
- Author
-
Johnson, Chelsea N., Lysaker, Colton R., McCoin, Colin S., Evans, Mara R., Thyfault, John P., Wilkins, Heather M., Morris, Jill K., and Geiger, Paige C.
- Subjects
ALZHEIMER'S disease risk factors ,MUSCLE analysis ,GENETICS of Alzheimer's disease ,ALZHEIMER'S disease ,SKELETAL muscle ,MITOCHONDRIA ,RESEARCH funding ,PHOSPHORYLATION ,SEX distribution ,BODY composition ,CELLULAR signal transduction ,FLUORESCENT antibody technique ,MICE ,GENETIC risk score ,APOLIPOPROTEINS ,PROTEOMICS ,ANIMAL experimentation ,MASS spectrometry ,ANALYSIS of variance ,SULFUR amino acids ,COMPARATIVE studies ,DATA analysis software ,ELECTRON transport ,GENOTYPES ,ALLELES - Abstract
Introduction: Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD), yet it's unclear how this allele mediates risk. APOE4 carriers experience reduced mobility and faster decline in muscle strength, suggesting skeletal muscle involvement. Mitochondria are critical for muscle function and although we have reported defects in muscle mitochondrial respiration during early cognitive decline, APOE4 -mediated effects on muscle mitochondria are unknown. Methods: Here, we sought to determine the impact of APOE4 on skeletal muscle bioenergetics using young, male and female APOE3 (control) and APOE4 targeted replacement mice (n = 8 per genotype/sex combination). We examined the proteome, mitochondrial respiration, fiber size, and fiber-type distribution in skeletal muscle. Results: We found that APOE4 alters mitochondrial pathway expression in young mouse muscle in a sex-dependent manner without affecting respiration and fiber size or composition relative to APOE3. In both sexes, the expression of mitochondrial pathways involved in electron transport, ATP synthesis, and heat production by uncoupling proteins and mitochondrial dysfunction significantly differed between APOE4 and APOE3 muscle. For pathways with predicted direction of activation, electron transport and oxidative phosphorylation were upregulated while mitochondrial dysfunction and sirtuin signaling were downregulated in female APOE4 vs. APOE3 muscle. In males, sulfur amino acid metabolism was upregulated in APOE4 vs. APOE3 muscle. Discussion: This work highlights early involvement of skeletal muscle in a mouse model of APOE4 -linked AD, which may contribute to AD pathogenesis or serve as a biomarker for brain health. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Microglial APOE4 promotes neuron degeneration in Alzheimer's disease through inhibition of lipid droplet autophagy
- Author
-
Meng Mao, Xiwen Ma, Xiaochuan Wang, and Jianping Ye
- Subjects
APOE4 ,Lipid droplets ,ACSL1 ,Alzheimer's disease (AD) ,Microglia ,Therapeutics. Pharmacology ,RM1-950 - Published
- 2025
- Full Text
- View/download PDF
13. Microglial APOE4: more is less and less is more.
- Author
-
Eskandari-Sedighi, Ghazaleh and Blurton-Jones, Mathew
- Subjects
APOE3 ,APOE4 ,Alzheimer’s disease ,Apolipoprotein E ,Lgals3 ,Microglia ,TGFβ ,Humans ,Animals ,Mice ,Apolipoprotein E4 ,Microglia ,Apolipoprotein E3 ,Alzheimer Disease ,Apolipoproteins E ,Mice ,Transgenic - Abstract
Apolipoprotein E (APOE) is the single greatest genetic risk factor for late onset Alzheimers disease (AD). Yet, the cell-specific effects of APOE on microglia function have remained unclear. Fortunately, two comprehensive new studies published in the latest issue of Nature Immunology have employed complementary gain-of-function and loss-of-function approaches to provide critical new insight into the impact of microglial APOE on AD pathogenesis.
- Published
- 2023
14. Associations between sex and lifestyle activities with cognitive reserve in mid-life adults with genetic risk for Alzheimer’s disease
- Author
-
Qing Qi, Feng Deng, Rebecca Sammon, Karen Ritchie, Graciela Muniz-Terrera, Ivan Koychev, Paresh Malhotra, Siobhan Hutchinson, David Robinson, John T. O’Brien, Craig W. Ritchie, Brian Lawlor, and Lorina Naci
- Subjects
Alzheimer’s disease ,Sex differences ,Cognitive reserve ,APOE4 ,Modifiable ,Mid-life ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Females have a higher age-adjusted incidence of Alzheimer’s Disease (AD) than males, even when accounting for longer lifespan and, therefore, stand to benefit the most from dementia prevention efforts. As exposure to many modifiable risk factors for dementia begins in mid-life, interventions must be implemented from middle-age. Building cognitive reserve, particularly through stimulating avocational activities and occupational attainment presents a crucial, underexplored, dementia prevention approach for mid-life. It is currently unknown, however, whether modifiable lifestyle factors can protect against AD processes, from mid-life, differentially for females and males who carry inherited risk for late-life dementia. To address this gap, this study investigated the impact of biological sex and APOE4 carrier status on the relationship between stimulating activities, occupational attainment, and cognition in mid-life. Methods We leveraged the PREVENT–Dementia program, the world’s largest study investigating the origins and early diagnosis of dementia in mid-life at-risk individuals (N = 700; 40–59 years). Cognitive performance was measured using the Cognito Battery and the Visual Short Term Memory Binding task. Mid-life specific reserve contributors were assessed via the Lifetime of Experiences Questionnaire. Results Females had significantly better episodic and relational memory (p
- Published
- 2024
- Full Text
- View/download PDF
15. TRPV1 alleviates APOE4-dependent microglial antigen presentation and T cell infiltration in Alzheimer's disease
- Author
-
Jia Lu, Kexin Wu, Xudong Sha, Jiayuan Lin, Hongzhuan Chen, and Zhihua Yu
- Subjects
APOE4 ,Cholesterol ,TRPV1 ,Microglia ,Tau ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Persistent innate and adaptive immune responses in the brain contribute to the progression of Alzheimer’s disease (AD). APOE4, the most important genetic risk factor for sporadic AD, encodes apolipoprotein E4, which by itself is a potent modulator of immune response. However, little is known about the immune hub that governs the crosstalk between the nervous and the adaptive immune systems. Transient receptor potential vanilloid type 1 (TRPV1) channel is a ligand-gated, nonselective cation channel with Ca2+ permeability, which has been proposed as a neuroprotective target in AD. Methods Using Ca2+-sensitive dyes, dynamic changes of Ca2+ in microglia were measured, including exogenous Ca2+ uptake and endoplasmic reticulum Ca2+ release. The mRFP-GFP-tagged LC3 plasmid was expressed in microglia to characterize the role of TRPV1 in the autophagic flux. Transcriptomic analyses and flow cytometry were performed to investigate the effects of APOE4 on brain microglia and T cells from APOE-targeted replacement mice with microglia-specific TRPV1 gene deficiency. Results Both APOE4 microglia derived from induced pluripotent stem cells of AD patients and APOE4-related tauopathy mouse model showed significantly increased cholesterol biosynthesis and accumulation compared to their APOE3 counterparts. Further, cholesterol dysregulation was associated with persistent activation of microglia and elevation of major histocompatibility complex II-dependent antigen presentation in microglia, subsequently accompanied by T cell infiltration. In addition, TRPV1-mediated transient Ca2+ influx mitigated cholesterol biosynthesis in microglia by suppressing the transcriptional activation of sterol regulatory element-binding protein 2, promoted autophagic activity and reduced lysosomal cholesterol accumulation, which were sufficient to resolve excessive immune response and neurodegeneration in APOE4-related tauopathy mouse model. Moreover, microglia-specific deficiency of TRPV1 gene accelerated glial inflammation, T cell response and associated neurodegeneration in an APOE4-related tauopathy mouse model. Conclusions The findings provide new perspectives for the treatment of APOE4-dependent neurodegeneration including AD.
- Published
- 2024
- Full Text
- View/download PDF
16. Associated risk and resilience factors of Alzheimer's disease in women with early bilateral oophorectomy: Data from the UK Biobank.
- Author
-
Calvo, Noelia, McFall, G Peggy, Ramana, Shreeyaa, Galper, Michelle, Fuller-Thomson, Esme, Dixon, Roger A, and Einstein, Gillian
- Subjects
- *
ALZHEIMER'S disease , *BODY mass index , *APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E , *HORMONE therapy - Abstract
Background: Bilateral oophorectomy (BO) confers immediate estradiol loss. We examined prevalence and predictors of Alzheimer's disease (AD) in women with early BO comparing their odds ratios of AD to those of women with spontaneous menopause (SM). Methods: A cohort from UK Biobank (n = 34,603) included women aged 60 + at baseline with and without AD who had early BO or SM. AD was determined based on AD related ICD-10 or ICD-9 code. We used logistic regression to model the association of menopause type with AD. Model predictors included age, education, age at menopause, hormone therapy (HT), APOE4, body mass index (BMI), cancer history, and smoking history. Results: Those with early BO had four times the odds of developing AD (OR = 4.12, 95% CI [2.02, 8.44]) compared to those with SM. APOE4 (OR = 4.29, 95% CI [2.43, 7.56]), and older age (OR = 1.16, 95% CI [1.05, 1.28]) were associated with increased odds of AD in the BO group. Greater years of education were associated with reduced odds of AD for both BO (OR = 0.91, 95% CI [0.85, 0.98]), and SM (OR = 0.95, 95% CI [0.90, 0.99]), while ever use of HT was associated with decreased odds of AD only for the BO group (OR = 0.43, 95% CI [0.23, 0.82]). Conclusions: Women with early BO, particularly with an APOE4 allele, are at high risk of AD. Women with early BO who use HT and those with increased education have lower odds of developing AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Associations between sex and lifestyle activities with cognitive reserve in mid-life adults with genetic risk for Alzheimer's disease.
- Author
-
Qi, Qing, Deng, Feng, Sammon, Rebecca, Ritchie, Karen, Muniz-Terrera, Graciela, Koychev, Ivan, Malhotra, Paresh, Hutchinson, Siobhan, Robinson, David, O'Brien, John T., Ritchie, Craig W., Lawlor, Brian, and Naci, Lorina
- Subjects
DISEASE risk factors ,SHORT-term memory ,OCCUPATIONAL prestige ,SEX (Biology) ,ALZHEIMER'S disease ,EPISODIC memory - Abstract
Background: Females have a higher age-adjusted incidence of Alzheimer's Disease (AD) than males, even when accounting for longer lifespan and, therefore, stand to benefit the most from dementia prevention efforts. As exposure to many modifiable risk factors for dementia begins in mid-life, interventions must be implemented from middle-age. Building cognitive reserve, particularly through stimulating avocational activities and occupational attainment presents a crucial, underexplored, dementia prevention approach for mid-life. It is currently unknown, however, whether modifiable lifestyle factors can protect against AD processes, from mid-life, differentially for females and males who carry inherited risk for late-life dementia. To address this gap, this study investigated the impact of biological sex and APOE4 carrier status on the relationship between stimulating activities, occupational attainment, and cognition in mid-life. Methods: We leveraged the PREVENT–Dementia program, the world's largest study investigating the origins and early diagnosis of dementia in mid-life at-risk individuals (N = 700; 40–59 years). Cognitive performance was measured using the Cognito Battery and the Visual Short Term Memory Binding task. Mid-life specific reserve contributors were assessed via the Lifetime of Experiences Questionnaire. Results: Females had significantly better episodic and relational memory (p < 0.001), and lower occupational attainment than males (p < 0.001). Engagement in stimulating activities was positively associated with episodic and relational memory, regardless of sex and APOE4 status (β = 0.05, CI 0.03–0.07, p < 0.001). APOE4 carriers showed significant sex differences in the association between occupational attainment and episodic and relational memory (β = 0.38, CI 0.12–0.63, p = 0.003). APOE4 carrier females with higher occupational attainment showed better cognition (β = 0.16, CI -0.002–0.32, p = 0.053), whereas APOE4 carrier males showed the opposite effect (β = -0.20, CI -0.40 – -0.001, p = 0.049). Conclusion: Our findings suggest that occupational attainment in mid-life contributes to cognitive reserve against inherited risk of dementia in females, but not males. They highlight the need for high precision approaches that consider biological sex and APOE4 carrier status to inform Alzheimer's disease prevention strategies and clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells.
- Author
-
Kaltschmidt, Barbara, Czaniera, Nele Johanne, Schulten, Wiebke, and Kaltschmidt, Christian
- Subjects
- *
TRANSCRIPTION factors , *DOUBLE-strand DNA breaks , *ALZHEIMER'S disease , *TUMOR necrosis factors , *NEUROGLIA , *DNA repair - Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Suppression of CNS APOE4 Expression by miRNAs Delivered by the S2 AAVrh.10 Capsid-Modified AAV Vector.
- Author
-
Karan, Kalpita R., Andrzejewski, Slawomir, Stiles, Katie M., Hackett, Neil R., and Crystal, Ronald G.
- Subjects
- *
GENE expression , *PEPTIDES , *APOLIPOPROTEIN E4 , *GENOME editing , *ALZHEIMER'S disease - Abstract
The homozygous Apolipoprotein E (APOE4) genotype is the major risk factor for the development of early Alzheimer's disease. Genome engineering studies in mouse models of human APOE4-dependent pathology have established that reduction of APOE4 expression can rescue the phenotype. We hypothesized that APOE4 could be suppressed in the CNS of APOE4 homozygotes using adeno-associated virus (AAV) expression of microRNAs (miRNA) designed to hybridize to APOE mRNA. We screened nine different miRNAs targeting APOE following transfection in HEK293T and Huh7 cells. Optimal APOE suppression was obtained with mir2A (targeting coding region nt330-351) and mirN4 (3′ untranslated region nt1142-1162). miRNA expression cassettes were designed with two copies of each of these two miRNAs co-expressed with a mCherry transgene. To optimize delivery of these miRNAs, an engineered AAVrh.10 variant was identified from a screen of multiple peptide insertions into capsid loop IV and substitutions in loop VIII. This led to identifying the AAV.S2 capsid with enhanced transduction of both neurons and glia and enhanced distribution in the brain. The engineered capsid was used to deliver the APOE miRNA suppression cassette to the hippocampus of TRE4 mice (human APOE4 knock-in replacement of the murine apoE locus). Two weeks after intra-hippocampus administration, regional expression of miRNA at the injection site was quantified at the mRNA level relative to an endogenous reference. The AAV.S2 capsid provided 2.31 ± 0.37-fold higher expression of miRNA over that provided by AAVrh.10 (p < 0.05). In the targeted region, a single intra-hippocampus AAV.S2 administration suppressed hippocampal APOE4 mRNA levels by 76.5 ± 3.9% compared with 41.3 ± 3.3% with the same cassette delivered by the wildtype AAVrh.10 capsid (p < 0.0001). We conclude that an expression cassette with two different miRNAs targeting APOE4 delivered by the AAV.S2 capsid will generate highly significant suppression of APOE4 in the CNS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. TRPV1 alleviates APOE4-dependent microglial antigen presentation and T cell infiltration in Alzheimer's disease.
- Author
-
Lu, Jia, Wu, Kexin, Sha, Xudong, Lin, Jiayuan, Chen, Hongzhuan, and Yu, Zhihua
- Subjects
STEROL regulatory element-binding proteins ,IMMUNOMODULATORS ,TRPV cation channels ,INDUCED pluripotent stem cells ,APOLIPOPROTEIN E4 ,ALZHEIMER'S disease - Abstract
Background: Persistent innate and adaptive immune responses in the brain contribute to the progression of Alzheimer's disease (AD). APOE4, the most important genetic risk factor for sporadic AD, encodes apolipoprotein E4, which by itself is a potent modulator of immune response. However, little is known about the immune hub that governs the crosstalk between the nervous and the adaptive immune systems. Transient receptor potential vanilloid type 1 (TRPV1) channel is a ligand-gated, nonselective cation channel with Ca
2+ permeability, which has been proposed as a neuroprotective target in AD. Methods: Using Ca2+ -sensitive dyes, dynamic changes of Ca2+ in microglia were measured, including exogenous Ca2+ uptake and endoplasmic reticulum Ca2+ release. The mRFP-GFP-tagged LC3 plasmid was expressed in microglia to characterize the role of TRPV1 in the autophagic flux. Transcriptomic analyses and flow cytometry were performed to investigate the effects of APOE4 on brain microglia and T cells from APOE-targeted replacement mice with microglia-specific TRPV1 gene deficiency. Results: Both APOE4 microglia derived from induced pluripotent stem cells of AD patients and APOE4-related tauopathy mouse model showed significantly increased cholesterol biosynthesis and accumulation compared to their APOE3 counterparts. Further, cholesterol dysregulation was associated with persistent activation of microglia and elevation of major histocompatibility complex II-dependent antigen presentation in microglia, subsequently accompanied by T cell infiltration. In addition, TRPV1-mediated transient Ca2+ influx mitigated cholesterol biosynthesis in microglia by suppressing the transcriptional activation of sterol regulatory element-binding protein 2, promoted autophagic activity and reduced lysosomal cholesterol accumulation, which were sufficient to resolve excessive immune response and neurodegeneration in APOE4-related tauopathy mouse model. Moreover, microglia-specific deficiency of TRPV1 gene accelerated glial inflammation, T cell response and associated neurodegeneration in an APOE4-related tauopathy mouse model. Conclusions: The findings provide new perspectives for the treatment of APOE4-dependent neurodegeneration including AD. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
21. mTOR inhibition enhances synaptic and mitochondrial function in Alzheimer's disease in an APOE genotype-dependent manner.
- Author
-
Sanganahalli, Basavaraju G, Mihailovic, Jelena M, Vekaria, Hemendra J, Coman, Daniel, Yackzan, Andrew T, Flemister, Abeoseh, Aware, Chetan, Wenger, Kathryn, Hubbard, W Brad, Sullivan, Patrick G, Hyder, Fahmeed, and Lin, Ai-Ling
- Abstract
Apolipoprotein ε4 (APOE4) carriers develop brain metabolic dysfunctions decades before the onset of Alzheimer's disease (AD). A goal of the study is to identify if rapamycin, an inhibitor for the mammalian target of rapamycin (mTOR) inhibitor, would enhance synaptic and mitochondrial function in asymptomatic mice with human APOE4 gene (E4FAD) before they showed metabolic deficits. A second goal is to determine whether there may be genetic-dependent responses to rapamycin when compared to mice with human APOE3 alleles (E3FAD), a neutral AD genetic risk factor. We fed asymptomatic E4FAD and E3FAD mice with control or rapamycin diets for 16 weeks from starting from 3 months of age. Neuronal mitochondrial oxidative metabolism and excitatory neurotransmission rates were measured using in vivo 1H-[13C] proton-observed carbon-edited magnetic resonance spectroscopy, and isolated mitochondrial bioenergetic measurements using Seahorse. We found that rapamycin enhanced neuronal mitochondrial function, glutamate-glutamine cycling, and TCA cycle rates in the asymptomatic E4FAD mice. In contrast, rapamycin enhances glycolysis, non-neuronal activities, and inhibitory neurotransmission of the E3FAD mice. These findings indicate that rapamycin might be able to mitigate the risk for AD by enhancing brain metabolic functions for cognitively intact APOE4 carriers, and the responses to rapamycin are varied by APOE genotypes. Consideration of precision medicine may be needed for future rapamycin therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Readdressing the Localization of Apolipoprotein E (APOE) in Mitochondria-Associated Endoplasmic Reticulum (ER) Membranes (MAMs): An Investigation of the Hepatic Protein–Protein Interactions of APOE with the Mitochondrial Proteins Lon Protease (LONP1), Mitochondrial Import Receptor Subunit TOM40 (TOMM40) and Voltage-Dependent Anion-Selective Channel 1 (VDAC1)
- Author
-
Rueter, Johanna, Rimbach, Gerald, Bilke, Stephanie, Tholey, Andreas, and Huebbe, Patricia
- Subjects
- *
CELL receptors , *UNFOLDED protein response , *MITOCHONDRIAL proteins , *CELL physiology , *LIPOPROTEIN receptors , *APOLIPOPROTEIN E - Abstract
As a component of circulating lipoproteins, APOE binds to cell surface receptors mediating lipoprotein metabolism and cholesterol transport. A growing body of evidence, including the identification of a broad variety of cellular proteins interacting with APOE, suggests additional independent functions. Investigating cellular localization and protein–protein interactions in cultured human hepatocytes, we aimed to contribute to the elucidation of hitherto unnoted cellular functions of APOE. We observed a strong accumulation of APOE in MAMs, equally evident for the two major isoforms APOE3 and APOE4. Using mass spectrometry proteome analyses, novel and previously noted APOE interactors were identified, including the mitochondrial proteins TOMM40, LONP1 and VDAC1. All three interactors were present in MAM fractions, which we think initially facilitates interactions with APOE. LONP1 is a protease with chaperone activity, which migrated to MAMs in response to ER stress, displaying a reinforced interaction with APOE. We therefore hypothesize that APOE may help in the unfolded protein response (UPR) by acting as a co-chaperone in cooperation with LONP1 at the interface of mitochondria and ER membranes. The interaction of APOE with the integral proteins TOMM40 and VDAC1 may point to the formation of bridging complexes connecting mitochondria with other organelles. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Andrographolide Improves ApoE4-Mediated Blood–Brain Barrier Injury by Alleviating Inflammation.
- Author
-
Zhou, Xuebin, Li, Jinhua, Quan, Shengli, Zhang, Xinyue, Gu, Lili, Hu, Min, Huang, Wenhai, and Li, Qin
- Abstract
The pathological and physiological studies of Alzheimer's disease (AD) have been in-depth, and apolipoprotein E4 (ApoE4) has been proven to be highly correlated with AD, and clinical and experimental data show that ApoE4 can cause blood–brain barrier (BBB) injury, and the change of BBB permeability is an important factor affecting the development of AD. Andrographolide (Andro), as the active component of the natural plant Andrographis paniculata, has been proven to have anti-inflammatory and antioxidant effects, which have potential neuroprotective effects. To verify the protective effect of Andro on BBB in a short term, our research group used atorvastatin (Atorva)-mediated zebrafish brain injury model and the ApoE4-mediated cell co-culture model of BBB injury to explore the protective effects and mechanisms of Andro on BBB injury. Studies have shown that Andro can inhibit the activation of CypA/NF-κB/MMP-9 signaling pathway and has achieved the effect of antagonizing the inhibition of ApoE4 on intercellular tight junction proteins (occludin, claudin-5, and ZO-1). At the same time, Andro can inhibit the secretion of cell adhesion molecules (VCAM-1 and ICAM-1) in cells, thereby delaying the occurrence and progression of neuroinflammation and playing a protective role in BBB. In conclusion, Andro is a potent natural product which can protect the blood–brain barrier. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. In 2024, the amyloid-cascade-hypothesis still remains a working hypothesis, no less but certainly no more.
- Author
-
Behl, Christian
- Subjects
THERAPEUTIC use of monoclonal antibodies ,BIOLOGICAL models ,ALZHEIMER'S disease ,AUTOPHAGY ,BRAIN ,NEURODEGENERATION ,MONOCLONAL antibodies ,HYPOTHESIS ,AGING ,AMYLOID beta-protein precursor ,DISEASE progression - Abstract
The amyloid-cascade-hypothesis of the pathogenesis of Alzheimer's disease (AD) was introduced 32 years ago, in 1992. From early on, this clear and straight forward hypothesis received a lot of attention, but also a lot of substantial criticism. Foremost, there have always been massive doubts that a complex age-associated disorder of the most intricate organ of the human body, the brain, can be explained by a linear, one-dimensional cause-and-effect model. The amyloid-cascade defines the generation, aggregation, and deposition of the amyloid beta peptide as the central pathogenic mechanism in AD, as the ultimate trigger of the disease, and, consequently, as the key pharmacological target. Certainly, the original 1992 version of this hypothesis has been refined by various means, and the 'formulating fathers' followed up with a few reappraisals and partly very open reflections in 2002, 2006, 2009, and 2016. However, up until today, for the supporters of this hypothesis, the central and initial steps of the cascade are believed to be driven by amyloid beta--even if now displayed somewhat more elaborate. In light of the recently published clinical results achieved with anti-amyloid antibodies, the controversy in the field about (1) the clinical meaningfulness of this approach, (2) the significance of clearance of the amyloid beta peptide, and last but not least (3) the relevance of the amyloid-cascade-hypothesis is gaining momentum. This review addresses the interesting manifestation of the amyloid-cascade-hypothesis as well as its ups and downs over the decades. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Hormone replacement therapy, menopausal age and lifestyle variables are associated with better cognitive performance at follow-up but not cognition over time in older-adult women irrespective of APOE4 carrier status and co-morbidities
- Author
-
Tamlyn J. Watermeyer, Sarah Gregory, Emmi Leetham, Chinedu T. Udeh-Momoh, and Graciela Muniz-Terrera
- Subjects
hormone replacement therapy (HRT) ,menopausal age ,cognition ,APOE4 ,cognitive aging ,lifestyle factors ,Medicine - Abstract
IntroductionThe impact of Hormone Replacement Therapy (HRT) on cognitive function in postmenopausal women remains a topic of considerable debate. Although estrogen's neuroprotective effects suggest potential cognitive benefits, empirical findings are mixed.MethodsThis study uses data from the Cognitive Function and Ageing Study Wales (CFAS Wales) cohort to explore the relationships between HRT use, age at menopause, APOE4 carrier status, lifestyle factors, comorbidities, and cognitive outcomes in older adult women. Two regression models were employed: one analyzing cognitive performance at follow-up and another examining changes in cognitive scores over time.ResultsResults indicate that while age, education, HRT use, age at menopause, alcohol consumption, and diet were associated with cognitive function at a single later time point, only age remained a significant predictor when modeling cognition over time.DiscussionThese findings suggest that while HRT, menopausal age and lifestyle factors may support cognitive stability, they do not necessarily predict cognitive decline in post-menopausal older women. A major limitation of the current work is the lack of detail regarding HRT use, such as formulation, timing and duration; caveats that future studies should address. The study underscores the need for longer follow-up periods, consideration of other female-specific risk factors, and more comprehensive lifestyle and health assessments to clarify the complex interplay between HRT use, reproductive history, lifestyle, comorbidities and cognitive aging in women.
- Published
- 2025
- Full Text
- View/download PDF
26. Loss of Endothelial APOE4 Dysregulates Neural Function In Vivo
- Author
-
Felecia M. Marottoli, Deebika Balu, Eden Flores‐Barrera, Emilce Artur de la Villarmois, Hui Zhang, Rohan Chaudhary, Ruju Talati, Kuei Y. Tseng, and Leon M. Tai
- Subjects
ApoE4 ,behavior ,brain endothelial cells ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background We recently found that loss of endothelial cell APOE3 disrupts neurovascular and synaptic function. However, whether endothelial APOE4 is detrimental or protective for neural function under physiological conditions is unknown. Therefore, the goal of this study was to determine the role of endothelial cell APOE4 in regulating brain function in vivo. Methods and Results We developed APOE4fl/fl/Cdh5(PAC)‐CreERT2+/− and APOE4fl/fl/Cdh5(PAC)‐CreERT2−/− (control) mice. Knockdown of endothelial cell APOE4 was induced at ≈4 to 5 weeks of age. Experiments were conducted at 9 months of age to evaluate neurovascular and neuronal function via biochemistry, immunohistochemistry, behavior tests, and electrophysiology. Endothelial cell APOE4 knockdown resulted in higher neurovascular permeability, lower claudin‐5 vessel coverage, impaired trace fear memory extinction, and disruption of cortical excitatory‐inhibitory balance of synaptic activity. Conclusions Our data support the novel concept that endothelial cell APOE4 is protective for brain function when other cell types express APOE4.
- Published
- 2024
- Full Text
- View/download PDF
27. Editorial: APOE4 -associated heterogeneity in the pathogenesis of Alzheimer's disease.
- Author
-
Torres, Eileen Ruth Samson, Rebeck, G. William, and Nuriel, Tal
- Subjects
SERIAL publications ,ALZHEIMER'S disease ,LIPIDS ,DIETARY fats ,ESTROGEN ,APOLIPOPROTEINS ,GENETICS ,DISEASE complications - Published
- 2024
- Full Text
- View/download PDF
28. Protein intake and episodic memory: the moderating role of the apolipoprotein E ε4 status
- Author
-
Musung Keum, Boung Chul Lee, Young Min Choe, Guk-Hee Suh, Shin Gyeom Kim, Hyun Soo Kim, Jaeuk Hwang, Dahyun Yi, and Jee Wook Kim
- Subjects
Protein intake ,Cognition ,APOE4 ,Episodic memory ,Alzheimer’s disease ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background This study investigated the correlation between protein intake and Alzheimer’s disease (AD)-related cognitive decline, particularly in episodic memory, among older adults without dementia. Furthermore, we assessed the moderating effect of apolipoprotein ε4 (APOE4) on this association and analyzed its influence on other cognitive functions beyond memory. Methods The study involved 196 participants who underwent assessments for protein intake, cognitive performance, APOE4 genotyping, and nutritional biomarkers. Protein intake was categorized into low, medium, and high based on the consumption of dairy, legumes, eggs, meat, and fish. Results High protein intake was significantly associated with better episodic memory and overall cognition. Moreover, a significant interaction was found between high protein intake and APOE4, indicating that APOE4 moderates the association between high protein intake level and episodic memory. Sensitivity analysis confirmed these results among participants with stable food intake. Conclusions Our study results demonstrated that high protein intake is associated with better episodic memory among older adults without dementia. Furthermore, the findings highlight the significant role of APOE4 status in moderating the relationship between protein consumption and episodic memory. These results suggest that dietary interventions focusing on protein intake could be beneficial for cognitive health, particularly in individuals with a genetic predisposition to AD.
- Published
- 2024
- Full Text
- View/download PDF
29. Protein intake and episodic memory: the moderating role of the apolipoprotein E ε4 status.
- Author
-
Keum, Musung, Lee, Boung Chul, Choe, Young Min, Suh, Guk-Hee, Kim, Shin Gyeom, Kim, Hyun Soo, Hwang, Jaeuk, Yi, Dahyun, and Kim, Jee Wook
- Subjects
ALZHEIMER'S disease ,APOLIPOPROTEIN E ,COGNITIVE ability ,APOLIPOPROTEIN E4 ,OLDER people ,EPISODIC memory - Abstract
Background: This study investigated the correlation between protein intake and Alzheimer's disease (AD)-related cognitive decline, particularly in episodic memory, among older adults without dementia. Furthermore, we assessed the moderating effect of apolipoprotein ε4 (APOE4) on this association and analyzed its influence on other cognitive functions beyond memory. Methods: The study involved 196 participants who underwent assessments for protein intake, cognitive performance, APOE4 genotyping, and nutritional biomarkers. Protein intake was categorized into low, medium, and high based on the consumption of dairy, legumes, eggs, meat, and fish. Results: High protein intake was significantly associated with better episodic memory and overall cognition. Moreover, a significant interaction was found between high protein intake and APOE4, indicating that APOE4 moderates the association between high protein intake level and episodic memory. Sensitivity analysis confirmed these results among participants with stable food intake. Conclusions: Our study results demonstrated that high protein intake is associated with better episodic memory among older adults without dementia. Furthermore, the findings highlight the significant role of APOE4 status in moderating the relationship between protein consumption and episodic memory. These results suggest that dietary interventions focusing on protein intake could be beneficial for cognitive health, particularly in individuals with a genetic predisposition to AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Texture‐based morphometry in relation to apolipoprotein ε4 genotype, ageing and sex in a midlife population.
- Author
-
Dounavi, Maria‐Eleni, Mak, Elijah, Operto, Gregory, Muniz‐Terrera, Graciela, Bridgeman, Katie, Koychev, Ivan, Malhotra, Paresh, Naci, Lorina, Lawlor, Brian, Su, Li, Falcon, Carles, Ritchie, Karen, Ritchie, Craig W., Gispert, Juan Domingo, and O'Brien, John T.
- Subjects
- *
OLDER people , *ALZHEIMER'S disease , *APOLIPOPROTEIN E4 , *CEREBRAL cortical thinning , *DISEASE risk factors - Abstract
Brain atrophy and cortical thinning are typically observed in people with Alzheimer's disease (AD) and, to a lesser extent, in those with mild cognitive impairment. In asymptomatic middle‐aged apolipoprotein ε4 (ΑPOE4) carriers, who are at higher risk of future AD, study reports are discordant with limited evidence of brain structural differences between carriers and non‐carriers of the ε4 allele. Alternative imaging markers with higher sensitivity at the presymptomatic stage, ideally quantified using typically acquired structural MRI scans, would thus be of great benefit for the detection of early disease, disease monitoring and subject stratification. In the present cross‐sectional study, we investigated textural properties of T1‐weighted 3T MRI scans in relation to APOE4 genotype, age and sex. We pooled together data from the PREVENT‐Dementia and ALFA studies focused on midlife healthy populations with dementia risk factors (analysable cohort: 1585 participants; mean age 56.2 ± 7.4 years). Voxel‐based and texture (examined features: contrast, entropy, energy, homogeneity) based morphometry was used to identify areas of volumetric and textural differences between APOE4 carriers and non‐carriers. Textural maps were generated and were subsequently harmonised using voxel‐wise COMBAT. For all analyses, APOE4, sex, age and years of education were used as model predictors. Interactions between APOE4 and age were further examined. There were no group differences in regional brain volume or texture based on APOE4 carriership or when age × APOE4 interactions were examined. Older people tended to have a less homogeneous textural profile in grey and white matter and a more homogeneous profile in the ventricles. A more heterogeneous textural profile was observed for females in areas such as the ventricles, frontal and parietal lobes and for males in the brainstem, cerebellum, precuneus and cingulate. Overall, we have shown the absence of volumetric and textural differences between APOE4 carriers and non‐carriers at midlife and have established associations of textural features with ageing and sex. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Effects of APOE4 on omega-3 brain metabolism across the lifespan.
- Author
-
Ebright, Brandon, Duro, Marlon V., Chen, Kai, Louie, Stan, and Yassine, Hussein N.
- Subjects
- *
DOCOSAHEXAENOIC acid , *UNSATURATED fatty acids , *ALZHEIMER'S disease , *POSITRON emission tomography , *FATTY acid oxidation , *APOLIPOPROTEIN E , *APOLIPOPROTEIN E4 - Abstract
Inconsistencies between Alzheimer's disease (AD) clinical trials and prevention studies using omega-3 (n-3) polyunsaturated fatty acid (PUFA) supplementation can be largely attributed to differences in age, environmental factors, genetic factors, baseline n-3/n-6 intake, and disease stage. Changes in docosahexaenoic acid (DHA) brain uptake throughout AD progression are influenced by the apolipoprotein E ε4 (APOE4) allele and lifestyle factors, such as DHA intake or exercise, and can be monitored by DHA positron emission tomography (PET) brain imaging. APOE4 carriers are more susceptible to blood–brain barrier dysfunction, oxidative stress, neuroinflammation, and fatty acid oxidation with aging compared to noncarriers. We hypothesize that increasing n-3 PUFA intake provides APOE4 carriers with the highest potential for protection against AD dementia when implemented early in life, many years before the onset of cognitive decline. During the AD dementia phase, alternative strategies targeting neuroinflammation and PUFA metabolism may offer potential benefits. Omega-3 (n-3) polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA), have important roles in human nutrition and brain health by promoting neuronal functions, maintaining inflammatory homeostasis, and providing structural integrity. As Alzheimer's disease (AD) pathology progresses, DHA metabolism in the brain becomes dysregulated, the timing and extent of which may be influenced by the apolipoprotein E ε4 (APOE4) allele. Here, we discuss how maintaining adequate DHA intake early in life may slow the progression to AD dementia in cognitively normal individuals with APOE4 , how recent advances in DHA brain imaging could offer insights leading to more personalized preventive strategies, and how alternative strategies targeting PUFA metabolism pathways may be more effective in mitigating disease progression in patients with existing AD dementia. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Associations of endogenous estrogens, plasma Alzheimer's disease biomarkers, and APOE4 carrier status on regional brain volumes in postmenopausal women.
- Author
-
Wugalter, Katrina A., Schroeder, Rachel A., Thurston, Rebecca C., Minjie Wu, Aizenstein, Howard J., Cohen, Ann D., Kamboh, M. Ilyas, Karikari, Thomas K., Derby, Carol A., and Maki, Pauline M.
- Subjects
ALZHEIMER'S disease risk factors ,RISK assessment ,SEX hormones ,LIQUID chromatography-mass spectrometry ,T-test (Statistics) ,RESEARCH funding ,BRAIN ,MULTIPLE regression analysis ,POSTMENOPAUSE ,ESTROGEN ,GENETIC carriers ,DESCRIPTIVE statistics ,CHI-squared test ,LONGITUDINAL method ,AMYLOID ,NEURORADIOLOGY ,DATA analysis software ,BIOMARKERS ,MOLECULAR diagnosis ,GENOTYPES ,MIDDLE age - Abstract
Background: Women carrying the APOE4 allele are at greater risk of developing Alzheimer's disease (AD) from ages 65-75 years compared to men. To better understand the elevated risk conferred by APOE4 carrier status among midlife women, we investigated the separate and interactive associations of endogenous estrogens, plasma AD biomarkers, and APOE4 carrier status on regional brain volumes in a sample of late midlife postmenopausal women. Methods: Participants were enrolled in MsBrain, a cohort study of postmenopausal women (n = 171, mean age = 59.4 years, mean MoCA score = 26.9; race = 83.2% white, APOE4 carriers = 40). Serum estrone (E1) and estradiol (E2) levels were assessed using liquid chromatography-tandem mass spectrometry. APOE genotype was determined using TaqMan SNP genotyping assays. Plasma AD biomarkers were measured using single molecule array technology. Cortical volume was measured and segmented by FreeSurfer software using individual T1w MPRAGE images. Multiple linear regression models were conducted to determine whether separate and interactive associations between endogenous estrogen levels, plasma AD biomarkers (Aβ42/Aβ40, Aβ42/p-tau181), and APOE4 carrier status predict regional brain volume (21 regions per hemisphere, selected a priori); and, whether significant interactive associations between estrogens and AD biomarkers on brain volume differed by APOE4 carrier status. Results: There was no main effect of APOE4 carrier status on regional brain volumes, endogenous estrogen levels, or plasma AD biomarkers. Estrogens did not associate with regional brain volumes, except for positive associations with left caudal middle frontal gyrus and fusiform volumes. The interactive association of estrogens and APOE4 carrier status on brain volume was not significant for any region. The interactive association of estrogens and plasma AD biomarkers predicted brain volume of several regions. Higher E1 and E2 were more strongly associated with greater regional brain volumes among women with a poorer AD biomarker profile (lower Aβ42/40, lower Aβ42/p-tau181 ratios). In APOE4- stratified analyses, these interactions were driven by non-APOE4 carriers. Conclusion: We demonstrate that the brain volumes of postmenopausal women with poorer AD biomarker profiles benefit most from higher endogenous estrogen levels. These findings are driven by non-APOE4 carriers, suggesting that APOE4 carriers may be insensitive to the favorable effects of estrogens on brain volume in the postmenopause. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. In vivo validation of late‐onset Alzheimer's disease genetic risk factors.
- Author
-
Sasner, Michael, Preuss, Christoph, Pandey, Ravi S., Uyar, Asli, Garceau, Dylan, Kotredes, Kevin P., Williams, Harriet, Oblak, Adrian L., Lin, Peter Bor‐Chian, Perkins, Bridget, Soni, Disha, Ingraham, Cindy, Lee‐Gosselin, Audrey, Lamb, Bruce T., Howell, Gareth R., and Carter, Gregory W.
- Abstract
INTRODUCTION: Genome‐wide association studies have identified over 70 genetic loci associated with late‐onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS: Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD‐sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS: We created new models for 11 coding and loss‐of‐function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION: These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. Highlights: A novel approach to validate genetic risk factors for late‐onset AD (LOAD) is presented.LOAD risk variants were knocked in to conserved mouse loci.Variant effects were assayed by transcriptional analysis.Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease.This approach should generate more translationally relevant animal models. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Divergent Roles of APOAI and APOM in the Identification of Alcohol Use Disorder and Their Association With Inflammation and Cognitive Decline: A Pilot Study.
- Author
-
Escudero, Berta, López-Valencia, Leticia, Horcajadas, Francisco Arias, and Orio, Laura
- Subjects
ALCOHOLISM ,APOLIPOPROTEINS ,COGNITION disorders ,LOGISTIC regression analysis ,APOLIPOPROTEIN B - Abstract
Background Alcohol use disorder (AUD) courses with inflammation and cognitive decline. Apolipoproteins have emerged as novel target compounds related to inflammatory processes and cognition. Methods A cross-sectional study was performed on abstinent AUD patients with at least 1 month of abstinence (n = 33; 72.7% men) and healthy controls (n = 34; 47.1% men). A battery of plasma apolipoproteins (APOAI, APOAII, APOB, APOCII, APOE, APOJ, and APOM), plasma inflammatory markers (LPS, LBP), and their influence on cognition and presence of the disorder were investigated. Results Higher levels of plasma APOAI, APOB, APOE, and APOJ, as well as the proinflammatory LPS, were observed in the AUD group, irrespective of sex, whereas APOM levels were lower vs controls. Hierarchical logistic regression analyses, adjusting for covariates (age, sex, education), associated APOM with the absence of cognitive impairment in AUD and identified APOAI and APOM as strong predictors of the presence or absence of the disorder, respectively. APOAI and APOM did not correlate with alcohol abuse variables or liver status markers, but they showed an opposite profile in their associations with LPS (positive for APOAI; negative for APOM) and cognition (negative for APOAI; positive for APOM) in the entire sample. Conclusions The HDL constituents APOAI and APOM were differentially regulated in the plasma of AUD patients compared with controls, playing divergent roles in the disorder identification and associations with inflammation and cognitive decline. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Could Alcohol-Related Cognitive Decline Be the Result of Iron-Induced Neuroinflammation?
- Author
-
Wilcockson, Thomas D. W. and Roy, Sankanika
- Subjects
- *
COGNITION disorders , *NEUROINFLAMMATION , *ENCEPHALITIS , *NEURAL circuitry , *REACTIVE oxygen species - Abstract
Excessive and prolonged alcohol use can have long-term severe neurological consequences. The mechanisms involved may be complicated; however, new evidence seems to indicate the involvement of iron accumulation and neuroinflammation. Prolonged alcohol consumption has been linked to the accumulation of iron in specific regions of the brain. Evidence suggests that excess iron in the brain can trigger microglia activation in response. This activation leads to the release of pro-inflammatory cytokines and reactive oxygen species, which can cause damage to neurons and surrounding brain tissue. Additionally, iron-induced oxidative stress and inflammation can disrupt the blood–brain barrier, allowing immune cells from the periphery to infiltrate the brain. This infiltration can lead to further neuroinflammatory responses. Inflammation in the brain subsequently disrupts neuronal networks, impairs synaptic plasticity, and accelerates neuronal cell death. Consequently, cognitive functions such as memory, attention, and decision-making are compromised. Additionally, chronic neuroinflammation can hasten the development and progression of neurodegenerative diseases, further exacerbating cognitive impairment. Therefore, alcohol could act as a trigger for iron-induced neuroinflammation and cognitive decline. Overall, the mechanisms at play here seem to strongly link alcohol with cognitive decline, with neuroinflammation resulting from alcohol-induced iron accumulation playing a pivotal role. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Alzheimer's disease genetic risk score and neuroimaging in the FINGER lifestyle trial.
- Author
-
Saadmaan, Gazi, Dalmasso, Maria Carolina, Ramirez, Alfredo, Hiltunen, Mikko, Kemppainen, Nina, Lehtisalo, Jenni, Mangialasche, Francesca, Ngandu, Tiia, Rinne, Juha, Soininen, Hilkka, Stephen, Ruth, Kivipelto, Miia, and Solomon, Alina
- Abstract
INTRODUCTION: We assessed a genetic risk score for Alzheimer's disease (AD‐GRS) and apolipoprotein E (APOE4) in an exploratory neuroimaging substudy of the FINGER trial. METHODS: 1260 at‐risk older individuals without dementia were randomized to multidomain lifestyle intervention or health advice. N = 126 participants underwent magnetic resonance imaging (MRI), and N = 47 positron emission tomography (PET) scans (Pittsburgh Compund B [PiB], Fluorodeoxyglucose) at baseline; N = 107 and N = 38 had repeated 2‐year scans. RESULTS: The APOE4 allele, but not AD‐GRS, was associated with baseline lower hippocampus volume (β = −0.27, p = 0.001), greater amyloid deposition (β = 0.48, p = 0.001), 2‐year decline in hippocampus (β = −0.27, p = 0.01), total gray matter volume (β = −0.25, p = 0.01), and cortical thickness (β = −0.28, p = 0.003). In analyses stratified by AD‐GRS (below vs above median), the PiB composite score increased less in intervention versus control in the higher AD‐GRS group (β = −0.60, p = 0.03). DISCUSSION: AD‐GRS and APOE4 may have different impacts on potential intervention effects on amyloid, that is, less accumulation in the higher‐risk group (AD‐GRS) versus lower‐risk group (APOE). Highlights: First study of neuroimaging and AD genetics in a multidomain lifestyle intervention.Possible intervention effect on brain amyloid deposition may rely on genetic risk.AD‐GRS and APOE4 allele may have different impacts on amyloid during intervention. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Characterizing molecular and synaptic signatures in mouse models of late‐onset Alzheimer's disease independent of amyloid and tau pathology.
- Author
-
Kotredes, Kevin P., Pandey, Ravi S., Persohn, Scott, Elderidge, Kierra, Burton, Charles P, Miner, Ethan W., Haynes, Kathryn A., Santos, Diogo Francisco S., Williams, Sean‐Paul, Heaton, Nicholas, Ingraham, Cynthia M., Lloyd, Christopher, Garceau, Dylan, O'Rourke, Rita, Herrick, Sarah, Rangel‐Barajas, Claudia, Maharjan, Surendra, Wang, Nian, Sasner, Michael, and Lamb, Bruce T.
- Abstract
INTRODUCTION: MODEL‐AD (Model Organism Development and Evaluation for Late‐Onset Alzheimer's Disease) is creating and distributing novel mouse models with humanized, clinically relevant genetic risk factors to capture the trajectory and progression of late‐onset Alzheimer's disease (LOAD) more accurately. METHODS: We created the LOAD2 model by combining apolipoprotein E4 (APOE4), Trem2*R47H, and humanized amyloid‐beta (Aβ). Mice were subjected to a control diet or a high‐fat/high‐sugar diet (LOAD2+HFD). We assessed disease‐relevant outcome measures in plasma and brain including neuroinflammation, Aβ, neurodegeneration, neuroimaging, and multi‐omics. RESULTS: By 18 months, LOAD2+HFD mice exhibited sex‐specific neuron loss, elevated insoluble brain Aβ42, increased plasma neurofilament light chain (NfL), and altered gene/protein expression related to lipid metabolism and synaptic function. Imaging showed reductions in brain volume and neurovascular uncoupling. Deficits in acquiring touchscreen‐based cognitive tasks were observed. DISCUSSION: The comprehensive characterization of LOAD2+HFD mice reveals that this model is important for preclinical studies seeking to understand disease trajectory and progression of LOAD prior to or independent of amyloid plaques and tau tangles. Highlights: By 18 months, unlike control mice (e.g., LOAD2 mice fed a control diet, CD), LOAD2+HFD mice presented subtle but significant loss of neurons in the cortex, elevated levels of insoluble Ab42 in the brain, and increased plasma neurofilament light chain (NfL).Transcriptomics and proteomics showed changes in gene/proteins relating to a variety of disease‐relevant processes including lipid metabolism and synaptic function.In vivo imaging revealed an age‐dependent reduction in brain region volume (MRI) and neurovascular uncoupling (PET/CT).LOAD2+HFD mice also demonstrated deficits in acquisition of touchscreen‐based cognitive tasks. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Editorial: APOE4-associated heterogeneity in the pathogenesis of Alzheimer's disease
- Author
-
Eileen Ruth Samson Torres, G. William Rebeck, and Tal Nuriel
- Subjects
Alzheimer's disease (AD) ,Apolipoprotein E ,APOE4 ,heterogeneity ,lipids ,high fat diet ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2024
- Full Text
- View/download PDF
39. Skeletal muscle proteome differs between young APOE3 and APOE4 targeted replacement mice in a sex-dependent manner
- Author
-
Chelsea N. Johnson, Colton R. Lysaker, Colin S. McCoin, Mara R. Evans, John P. Thyfault, Heather M. Wilkins, Jill K. Morris, and Paige C. Geiger
- Subjects
APOE4 ,Alzheimer's disease ,skeletal muscle ,mitochondria ,proteomics ,mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
IntroductionApolipoprotein E4 (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD), yet it's unclear how this allele mediates risk. APOE4 carriers experience reduced mobility and faster decline in muscle strength, suggesting skeletal muscle involvement. Mitochondria are critical for muscle function and although we have reported defects in muscle mitochondrial respiration during early cognitive decline, APOE4-mediated effects on muscle mitochondria are unknown.MethodsHere, we sought to determine the impact of APOE4 on skeletal muscle bioenergetics using young, male and female APOE3 (control) and APOE4 targeted replacement mice (n = 8 per genotype/sex combination). We examined the proteome, mitochondrial respiration, fiber size, and fiber-type distribution in skeletal muscle.ResultsWe found that APOE4 alters mitochondrial pathway expression in young mouse muscle in a sex-dependent manner without affecting respiration and fiber size or composition relative to APOE3. In both sexes, the expression of mitochondrial pathways involved in electron transport, ATP synthesis, and heat production by uncoupling proteins and mitochondrial dysfunction significantly differed between APOE4 and APOE3 muscle. For pathways with predicted direction of activation, electron transport and oxidative phosphorylation were upregulated while mitochondrial dysfunction and sirtuin signaling were downregulated in female APOE4 vs. APOE3 muscle. In males, sulfur amino acid metabolism was upregulated in APOE4 vs. APOE3 muscle.DiscussionThis work highlights early involvement of skeletal muscle in a mouse model of APOE4-linked AD, which may contribute to AD pathogenesis or serve as a biomarker for brain health.
- Published
- 2024
- Full Text
- View/download PDF
40. In 2024, the amyloid-cascade-hypothesis still remains a working hypothesis, no less but certainly no more
- Author
-
Christian Behl
- Subjects
Alzheimer ,amyloid ,APOE4 ,autophagy ,GWAS ,neurodegeneration ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The amyloid-cascade-hypothesis of the pathogenesis of Alzheimer’s disease (AD) was introduced 32 years ago, in 1992. From early on, this clear and straight forward hypothesis received a lot of attention, but also a lot of substantial criticism. Foremost, there have always been massive doubts that a complex age-associated disorder of the most intricate organ of the human body, the brain, can be explained by a linear, one-dimensional cause-and-effect model. The amyloid-cascade defines the generation, aggregation, and deposition of the amyloid beta peptide as the central pathogenic mechanism in AD, as the ultimate trigger of the disease, and, consequently, as the key pharmacological target. Certainly, the original 1992 version of this hypothesis has been refined by various means, and the ‘formulating fathers’ followed up with a few reappraisals and partly very open reflections in 2002, 2006, 2009, and 2016. However, up until today, for the supporters of this hypothesis, the central and initial steps of the cascade are believed to be driven by amyloid beta—even if now displayed somewhat more elaborate. In light of the recently published clinical results achieved with anti-amyloid antibodies, the controversy in the field about (1) the clinical meaningfulness of this approach, (2) the significance of clearance of the amyloid beta peptide, and last but not least (3) the relevance of the amyloid-cascade-hypothesis is gaining momentum. This review addresses the interesting manifestation of the amyloid-cascade-hypothesis as well as its ups and downs over the decades.
- Published
- 2024
- Full Text
- View/download PDF
41. TREM2-independent microgliosis promotes tau-mediated neurodegeneration in the presence of ApoE4.
- Author
-
Gratuze, Maud, Schlachetzki, Johannes, DOliveira Albanus, Ricardo, Jain, Nimansha, Novotny, Brenna, Brase, Logan, Rodriguez, Lea, Mansel, Clayton, Kipnis, Michal, OBrien, Sydney, Pasillas, Martina, Lee, Choonghee, Manis, Melissa, Colonna, Marco, Harari, Oscar, Glass, Christopher, Ulrich, Jason, and Holtzman, David
- Subjects
Alzheimer’s disease ,ApoE4 ,TREM2 ,microgliosis ,tau pathology ,tau-mediated neurodegeneration ,Mice ,Animals ,Apolipoprotein E4 ,Alzheimer Disease ,Inflammation ,Microglia ,Disease Models ,Animal ,Membrane Glycoproteins ,Receptors ,Immunologic - Abstract
In addition to tau and Aβ pathologies, inflammation plays an important role in Alzheimers disease (AD). Variants in APOE and TREM2 increase AD risk. ApoE4 exacerbates tau-linked neurodegeneration and inflammation in P301S tau mice and removal of microglia blocks tau-dependent neurodegeneration. Microglia adopt a heterogeneous population of transcriptomic states in response to pathology, at least some of which are dependent on TREM2. Previously, we reported that knockout (KO) of TREM2 attenuated neurodegeneration in P301S mice that express mouse Apoe. Because of the possible common pathway of ApoE and TREM2 in AD, we tested whether TREM2 KO (T2KO) would block neurodegeneration in P301S Tau mice expressing ApoE4 (TE4), similar to that observed with microglial depletion. Surprisingly, we observed exacerbated neurodegeneration and tau pathology in TE4-T2KO versus TE4 mice, despite decreased TREM2-dependent microgliosis. Our results suggest that tau pathology-dependent microgliosis, that is, TREM2-independent microgliosis, facilitates tau-mediated neurodegeneration in the presence of ApoE4.
- Published
- 2023
42. APOE4 dysregulates autophagy in cultured cells
- Author
-
Fote, Gianna M and Steffan, Joan S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Dementia ,Aging ,Alzheimer's Disease ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Brain Disorders ,Acquired Cognitive Impairment ,Neurodegenerative ,2.1 Biological and endogenous factors ,Neurological ,APOE ,APOE4 ,Alzheimer’s disease ,LAMP2A ,chaperone-mediated autophagy - Abstract
Human APOE4 (apolipoprotein E4 isoform) is a powerful genetic risk factor for late-onset Alzheimer disease (AD). Many groups have investigated the effect of APOE4 on the degradation of amyloid β (Aβ), the main component of plaques found in the brains of AD patients. However, few studies have focused on the degradation of APOE itself. We investigated the lysosomal trafficking of APOE in cells and found that APOE from the post-Golgi compartment is degraded through an autophagic process requiring the lysosomal membrane protein LAMP2A. We found that APOE4 accumulates in enlarged lysosomes, alters autophagic flux, and changes the proteomic contents of lysosomes following internalization. This dysregulated lysosomal trafficking may represent one of the mechanisms that contributes to AD pathogenesis.
- Published
- 2022
43. APOE4 genotype and aging impair injury-induced microglial behavior in brain slices, including toward Aβ, through P2RY12
- Author
-
Jordy Sepulveda, Jennifer Yejean Kim, Joseph Binder, Stefano Vicini, and G. William Rebeck
- Subjects
Microglia ,APOE4 ,Aging ,Alzheimer’s disease ,P2RY12 ,Ex-vivo imaging ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Microglia are highly dynamic cells that play a critical role in tissue homeostasis through the surveillance of brain parenchyma and response to cues associated with damage. Aging and APOE4 genotype are the strongest risk factors for Alzheimer’s disease (AD), but how they affect microglial dynamics remains unclear. Using ex vivo confocal microscopy, we analyzed microglial dynamic behaviors in the entorhinal cortex (EC) and hippocampus CA1 of 6-, 12-, and 21-month-old mice APOE3 or APOE4 knock-in mice expressing GFP under the CX3CR1 promoter. To study microglia surveillance, we imaged microglia baseline motility for 20 min and measured the extension and retraction of processes. We found that APOE4 microglia exhibited significantly less brain surveillance (27%) compared to APOE3 microglia in 6-month-old mice; aging exacerbated this deficit. To measure microglia response to damage, we imaged process motility in response to ATP, an injury-associated signal, for 30 min. We found APOE4 microglia extended their processes significantly slower (0.9 µm/min, p
- Published
- 2024
- Full Text
- View/download PDF
44. Radiosynthesis of 20-[18F]fluoroarachidonic acid for PET-MR imaging: Biological evaluation in ApoE4-TR mice.
- Author
-
Van Valkenburgh, Juno, Duro, Marlon, Burnham, Erica, Chen, Quan, Wang, Shaowei, Tran, Jenny, Kerman, Bilal, Liu, Xiaodan, Sta Maria, Naomi, Zanderigo, Francesca, Croteau, Etienne, Rapoport, Stanley, Cunnane, Stephen, Jacobs, Russell, Yassine, Hussein, Chen, Kai, and Hwang, Sung Hee
- Subjects
ApoE4 ,Arachidonic acid ,Brain imaging ,Fatty acid ,PET-MR ,Animals ,Mice ,Humans ,Apolipoprotein E4 ,Magnetic Resonance Imaging ,Brain ,Astrocytes ,Positron-Emission Tomography ,Alzheimer Disease ,Mice ,Transgenic - Abstract
Dysreglulated brain arachidonic acid (AA) metabolism is involved in chronic inflammation and is influenced by apolipoprotein E4 (APOE4) genotype, the strongest genetic risk factor of late-onset Alzheimers disease (AD). Visualization of AA uptake and distribution in the brain can offer insight into neuroinflammation and AD pathogenesis. Here we present a novel synthesis and radiosynthesis of 20-[18F]fluoroarachidonic acid ([18F]-FAA) for PET imaging using a convergent route and a one-pot, single-purification radiolabeling procedure, and demonstrate its brain uptake in human ApoE4 targeted replacement (ApoE4-TR) mice. By examining p38 phosphorylation in astrocytes, we found that fluorination of AA at the ω-position did not significantly alter its biochemical role in cells. The brain incorporation coefficient (K*) of [18F]-FAA was estimated via multiple methods by using an image-derived input function from the right ventricle of the heart as a proxy of the arterial input function and brain tracer concentrations assessed by dynamic PET-MR imaging. This new synthetic approach should facilitate the practical [18F]-FAA production and allow its translation into clinical use, making investigations of dysregulation of lipid metabolism more feasible in the study of neurodegenerative diseases.
- Published
- 2022
45. Human in vivo evidence of associations between herpes simplex virus and cerebral amyloid-beta load in normal aging
- Author
-
Cantero, Jose L., Atienza, Mercedes, Sastre, Isabel, and Bullido, María Jesús
- Published
- 2024
- Full Text
- View/download PDF
46. Three major effects of APOEε4 on Aβ immunotherapy induced ARIA.
- Author
-
Foley, Kate E. and Wilcock, Donna M.
- Subjects
ALZHEIMER'S disease risk factors ,RISK assessment ,IMMUNOTHERAPY ,NEUROINFLAMMATION ,IMMUNE system ,MONOCLONAL antibodies ,APOLIPOPROTEINS ,CEREBRAL amyloid angiopathy ,CEREBROVASCULAR disease ,AMYLOID beta-protein precursor - Abstract
The targeting of amyloid-beta (Aß) plaques therapeutically as one of the primary causes of Alzheimer's disease (AD) dementia has been an ongoing effort spanning decades. While some antibodies are extremely promising and have been moved out of clinical trials and into the clinic, most of these treatments show similar adverse effects in the form of cerebrovascular damage known as amyloid-related imaging abnormalities (ARIA). The two categories of ARIA are of major concern for patients, families, and prescribing physicians, with ARIA-E presenting as cerebral edema, and ARIA-H as cerebral hemorrhages (microand macro-). From preclinical and clinical trials, it has been observed that the greatest genetic risk factor for AD, APOE
ε4 , is also a major risk factor for anti-Aβ immunotherapy-induced ARIA. APOEe4 carriers represent a large population of AD patients, and, therefore, limits the broad adoption of these therapies across the AD population. In this review we detail three hypothesized mechanisms by which APOEe4 influences ARIA risk: (1) reduced cerebrovascular integrity, (2) increased neuroinflammation and immune dysregulation, and (3) elevated levels of CAA. The effects of APOEε4 on ARIA risk is clear, however, the underlying mechanisms require more research. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
47. Estradiol improves behavior in FAD transgenic mice that express APOE3 but not APOE4 after ovariectomy.
- Author
-
Balu, Deebika, Valencia-Olvera, Ana C., Deshpande, Ashwini, Narayanam, Saharsh, Konasani, Sravya, Pattisapu, Shreya, York, Jason M., Thatcher, Gregory R. J., LaDu, Mary Jo, and Tai, Leon M.
- Subjects
TRANSGENIC mice ,APOLIPOPROTEIN E4 ,MICE ,APOLIPOPROTEIN E ,OVARIECTOMY ,ALZHEIMER'S disease ,ESTRADIOL - Abstract
Increasing evidence suggests that female individuals have a higher Alzheimer's disease (AD) risk associated with post-menopausal loss of circulating estradiol (E
2 ). However, clinical data are conflicting on whether E2 lowers AD risk. One potential contributing factor is APOE. The greatest genetic risk factor for AD is APOE4, a factor that is pronounced in female individuals post-menopause. Clinical data suggests that APOE impacts the response of AD patients to E2 replacement therapy. However, whether APOE4 prevents, is neutral, or promotes any positive effects of E2 is unclear. Therefore, our goal was to determine whether APOE modulates the impact of E2 on behavior and AD pathology in vivo. To that end, mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ42 were ovariectomized at either 4 months (early) or 8 months (late) and treated with vehicle or E2 for 4 months. In E3FAD mice, we found that E2 mitigated the detrimental effect of ovariectomy on memory, with no effect on Aβ in the early paradigm and only improved learning in the late paradigm. Although E2 lowered Aβ in E4FAD mice in the early paradigm, there was no impact on learning or memory, possibly due to higher Aβ pathology compared to E3FAD mice. In the late paradigm, there was no effect on learning/memory and Aβ pathology in E4FAD mice. Collectively, these data support the idea that, in the presence of Aβ pathology, APOE impacts the response to E2 supplementation post-menopause. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
48. Impact of Short-Term Computerized Cognitive Training on Cognition in Older Adults With and Without Genetic Risk of Alzheimer's Disease: Outcomes From the START Randomized Controlled Trial.
- Author
-
Corbett, Anne, Williams, Gareth, Creese, Byron, Hampshire, Adam, Palmer, Abbie, Brooker, Helen, and Ballard, Clive
- Subjects
- *
ALZHEIMER'S disease risk factors , *GENETICS of Alzheimer's disease , *THERAPEUTICS , *COGNITIVE testing , *EVALUATION of human services programs , *STATISTICAL sampling , *EXECUTIVE function , *TREATMENT effectiveness , *RANDOMIZED controlled trials , *INTERNET , *GENETIC risk score , *ATTENTION , *COMPUTERS in medicine , *COGNITION disorders , *APOLIPOPROTEINS , *MEMORY , *COGNITIVE therapy , *INDIVIDUALIZED medicine , *ACTIVE aging , *BIOMARKERS , *OLD age - Abstract
To establish the impact of a 3-minute computerized cognitive training program (START) on cognition in older adults with and without genetic risk of Alzheimer's disease. Two-arm randomized controlled trial of the START program. Remote online trial in adults older than 50 taking part from home. The trial compared the START program with placebo in 6544 people older than 50. Primary outcome was executive function measured through Trailmaking B, with other secondary cognitive measures. Genetic risk profile and ApoE4 status were determined by Illumina Array. START conferred benefit to executive function, attention, memory, and a composite measure, including in people with the ApoE4 genotype. The 3-minute START task offers a means of supporting cognitive health in older adults and could be used at scale and within a precision medicine approach to reduce risk of cognitive decline in a targeted way. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. ApoE4 dysregulation incites depressive symptoms and mitochondrial impairments in mice.
- Author
-
Li, Weifen, Ali, Tahir, He, Kaiwu, Zheng, Chengyou, Li, Ningning, Yu, Zhi‐Jian, and Li, Shupeng
- Abstract
Apolipoprotein E4 (ApoE4) is involved in the stress‐response processes and is hypothesized to be a risk factor for depression by means of mitochondrial dysfunction. However, their exact roles and underlying mechanisms are largely unknown. ApoE4 transgenic mice (B6. Cg‐ApoEtm1UncCdh18Tg(GFAP−APOE i4)1Hol/J) were subjected to stress (lipopolysaccharides, LPS) to elucidate the aetiology of ApoE4‐induced depression. LPS treatment significantly aggravated depression‐like behaviours, concurrent with neuroinflammation and impaired mitochondrial changes, and melatonin/Urolithin A (UA) + 5‐aminoimidazole‐4‐carboxamide 1‐β‐D‐ribofuranoside (AICAR) reversed these effects in ApoE4 mice. Concurrently, ApoE4 mice exhibited mitophagy deficits, which could be further exacerbated by LPS stimulation, as demonstrated by reduced Atg5, Beclin‐1 and Parkin levels, while PINK1 levels were increased. However, these changes were reversed by melatonin treatment. Additionally, proteomic profiling suggested mitochondria‐related signalling and network changes in ApoE4 mice, which may underlie the exaggerated response to LPS. Furthermore, HEK 293T cells transfected with ApoE4 showed mitochondria‐associated protein and mitophagy defects, including PGC‐1α, TFAM, p‐AMPKα, PINK1 and LC3B impairments. Additionally, it aggravates mitochondrial impairment (particularly mitophagy), which can be attenuated by triggering autophagy. Collectively, ApoE4 dysregulation enhanced depressive behaviour upon LPS stimulation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Involvement of ApoE4 in dementia with Lewy bodies in the prodromal and demented stages: evaluation of the Strasbourg cohort.
- Author
-
Bousiges, Olivier, Cretin, Benjamin, Muller, Candice, Botzung, Anne, Sanna, Lea, Anthony, Pierre, Philippi, Nathalie, Demuynck, Catherine, and Blanc, Frédéric
- Subjects
LEWY body dementia ,APOLIPOPROTEIN E4 ,APOLIPOPROTEIN E ,ALZHEIMER'S disease ,DISEASE risk factors - Abstract
ApoE4 as a risk factor for dementia with Lewy bodies (DLB) is still an issue. We sought to determine the involvement of ApoE4 according to different clinical parameters in our cohort of patients from Strasbourg, France. ApoE genotyping was performed on the AlphaLewyMA cohort. In this cohort, 197 patients were genotyped: 105 DLB patients, 37 Alzheimer's disease (AD) patients, 29 patients with AD/DLB comorbidity, and 26 control subjects (CS). The groups of patients were also classified according to the stage of evolution of the disease: prodromal or demented. We analyzed other parameters in relation to ApoE4 status, such as years of education (YOE) and Alzheimer CSF biomarkers. We observed a higher proportion of ApoE4 carriers in the AD (51.4%) and AD/DLB (72.4%) groups compared to the DLB (25.7%) and CS (11.5%) groups (p < 0.0001). We found a correlation between age at disease onset and YOE in the AD group (p = 0.039) but not in the DLB group (p = 0.056). Interestingly, in the DLB group, the subgroup of patients with high YOE (≥ 11) had significantly more patients with ApoE4 than the subgroup with low YOE (< 11). AD biomarkers did not seem to be impacted by the presence of ApoE4, except for Aβ42: DLB ApoE4-positive demented patients showed a more marked Aβ42 decrease. ApoE4 does not appear to be a risk factor for "pure" DLB patients. These results suggest a strong link between ApoE4 and amyloidopathy and consequently with AD. Trial registration: AlphaLewyMa, Identifier: NCT01876459, date of registration: June 12, 2013. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.