12,580 results on '"apolipoprotein E4"'
Search Results
2. 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
3. Sex differences in interacting genetic and functional connectivity biomarkers in Alzheimer’s disease
- Author
-
Williamson, Jordan N, James, Shirley A, Mullen, Sean P, Sutton, Bradley P, Wszalek, Tracey, Mulyana, Beni, Mukli, Peter, Yabluchanskiy, Andriy, and Yang, Yuan
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Brain Disorders ,Acquired Cognitive Impairment ,Genetics ,Neurosciences ,Alzheimer's Disease ,Women's Health ,Neurodegenerative ,Biomedical Imaging ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Aging ,Clinical Research ,Dementia ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Neurological ,Humans ,Alzheimer Disease ,Female ,Male ,Aged ,Magnetic Resonance Imaging ,Hippocampus ,Aged ,80 and over ,Apolipoprotein E4 ,Biomarkers ,Genotype ,Sex Factors ,Case-Control Studies ,Alzheimer's disease ,Sex difference ,Apolipoprotein E ,Functional connectivity ,Alzheimer’s Disease Neuroimaging Initiative Consortium ,Alzheimer’s disease ,Clinical sciences - Abstract
As of 2023, it is estimated that 6.7 million individuals in the United States live with Alzheimer's disease (AD). Prior research indicates that AD disproportionality affects females; females have a greater incidence rate, perform worse on a variety of neuropsychological tasks, and have greater total brain atrophy. Recent research shows that hippocampal functional connectivity differs by sex and may be related to the observed sex differences in AD, and apolipoprotein E (ApoE) ε4 carriers have reduced hippocampal functional connectivity. The purpose of this study was to determine if the ApoE genotype plays a role in the observed sex differences in hippocampal functional connectivity in Alzheimer's disease. The resting state fMRI and T2 MRI of individuals with AD (n = 30, female = 15) and cognitively normal individuals (n = 30, female = 15) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were analyzed using the functional connectivity toolbox (CONN). Our results demonstrated intrahippocampal functional connectivity differed between those without an ε4 allele and those with at least one ε4 allele in each group. Additionally, intrahippocampal functional connectivity differed only by sex when Alzheimer's participants had at least one ε4 allele. These results improve our current understanding of the role of the interacting relationship between sex, ApoE genotype, and hippocampal function in AD. Understanding these biomarkers may aid in the development of sex-specific interventions for improved AD treatment.
- Published
- 2024
4. Impact of APOE ε4 and ε2 on plasma neurofilament light chain and cognition in autosomal dominant Alzheimers disease.
- Author
-
Langella, Stephanie, Bonta, Kyra, Chen, Yinghua, Su, Yi, Vasquez, Daniel, Aguillon, David, Acosta-Baena, Natalia, Baena, Ana, Garcia-Ospina, Gloria, Giraldo-Chica, Margarita, Tirado, Victoria, Muñoz, Claudia, Ríos-Romenets, Silvia, Guzman-Martínez, Claudia, Pruzin, Jeremy, Ghisays, Valentina, Arboleda-Velasquez, Joseph, Kosik, Kenneth, Tariot, Pierre, Reiman, Eric, Lopera, Francisco, and Quiroz, Yakeel
- Subjects
APOE ,Autosomal dominant Alzheimer’s disease ,Blood biomarkers ,Neurodegeneration ,PSEN1 ,Humans ,Alzheimer Disease ,Neurofilament Proteins ,Female ,Male ,Middle Aged ,Apolipoprotein E4 ,Aged ,Cross-Sectional Studies ,Apolipoprotein E2 ,Presenilin-1 ,Adult ,Cognition ,Biomarkers ,Neuropsychological Tests ,Mutation ,Heterozygote ,Genotype - Abstract
BACKGROUND: Apolipoprotein E (APOE) genotypes have been suggested to influence cognitive impairment and clinical onset in presenilin-1 (PSEN1) E280A carriers for autosomal dominant Alzheimers disease (ADAD). Less is known about their impact on the trajectory of biomarker changes. Neurofilament light chain (NfL), a marker of neurodegeneration, begins to accumulate in plasma about 20 years prior to the clinical onset of ADAD. In this study we investigated the impact of APOE ε4 and ε2 variants on age-related plasma NfL increases and cognition in PSEN1 E280A mutation carriers. METHODS: We analyzed cross-sectional data from PSEN1 E280A mutation carriers and non-carriers recruited from the Alzheimers Prevention Initiative Registry of ADAD. All participants over 18 years with available APOE genotype, plasma NfL, and neuropsychological evaluation were included in this study. APOE genotypes and plasma NfL concentrations were characterized for each participant. Cubic spline models using a Hamiltonian Markov chain Monte Carlo method were used to characterize the respective impact of at least one APOE ε4 or ε2 allele on age-related log-transformed plasma NfL increases. Linear regression models were estimated to explore the impact of APOE ε4 and ε2 variants and plasma NfL on a composite cognitive test score in the ADAD mutation carrier and non-carrier groups. RESULTS: Analyses included 788 PSEN1 E280A mutation carriers (169 APOE ε4 + , 114 ε2 +) and 650 mutation non-carriers (165 APOE ε4 + , 80 ε2 +), aged 18-75 years. APOE ε4 allele carriers were distinguished from ε4 non-carriers by greater age-related NfL elevations in the ADAD mutation carrier group, beginning about three years after the mutation carriers estimated median age at mild cognitive impairment onset. APOE ε2 allele carriers had lower plasma NfL concentrations than ε2 non-carriers in both the ADAD mutation carrier and non-carrier groups, unrelated to age, and an attenuated relationship between higher NfL levels on cognitive decline in the ADAD mutation carrier group. CONCLUSIONS: APOE ε4 accelerates age-related plasma NfL increases and APOE ε2 attenuates the relationship between higher plasma NfL levels and cognitive decline in ADAD. NfL may be a useful biomarker to assess clinical efficacy of APOE-modifying drugs with the potential to help in the treatment and prevention of ADAD.
- Published
- 2024
5. Associations of cerebral amyloid beta and tau with cognition from midlife.
- Author
-
Gonzales, Mitzi, ODonnell, Adrienne, Ghosh, Saptaparni, Thibault, Emma, Tanner, Jeremy, Satizabal, Claudia, Decarli, Charles, Fakhri, Georges, Johnson, Keith, Beiser, Alexa, Seshadri, Sudha, and Pase, Matthew
- Subjects
PET imaging ,amyloid beta ,cognition ,midlife ,tau ,Aged ,Female ,Humans ,Male ,Middle Aged ,Amyloid beta-Peptides ,Aniline Compounds ,Apolipoprotein E4 ,Brain ,Cognition ,Cohort Studies ,Neuropsychological Tests ,Positron-Emission Tomography ,tau Proteins - Abstract
INTRODUCTION: Understanding early neuropathological changes and their associations with cognition may aid dementia prevention. This study investigated associations of cerebral amyloid and tau positron emission tomography (PET) retention with cognition in a predominately middle-aged community-based cohort and examined factors that may modify these relationships. METHODS: 11C-Pittsburgh compound B amyloid and 18F-flortaucipir tau PET imaging were performed. Associations of amyloid and tau PET with cognition were evaluated using linear regression. Interactions with age, apolipoprotein E (APOE) ε4 status, and education were examined. RESULTS: Amyloid and tau PET were not associated with cognition in the overall sample (N = 423; mean: 57 ± 10 years; 50% female). However, younger age (
- Published
- 2024
6. Subjective cognitive concerns, APOE ε4, PTSD symptoms, and risk for dementia among older veterans.
- Author
-
Neale, Zoe, Fonda, Jennifer, Miller, Mark, Wolf, Erika, Zhang, Rui, Sherva, Richard, Harrington, Kelly, Merritt, Victoria, Panizzon, Matthew, Hauger, Richard, Gaziano, J, and Logue, Mark
- Subjects
APOE ε4 ,Dementia ,PTSD ,Survival analysis ,TBI ,Aged ,Aged ,80 and over ,Female ,Humans ,Male ,Apolipoprotein E4 ,Brain Injuries ,Traumatic ,Dementia ,Retrospective Studies ,Risk Factors ,Stress Disorders ,Post-Traumatic ,United States ,Veterans - Abstract
BACKGROUND: Posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are associated with self-reported problems with cognition as well as risk for Alzheimers disease and related dementias (ADRD). Overlapping symptom profiles observed in cognitive disorders, psychiatric disorders, and environmental exposures (e.g., head injury) can complicate the detection of early signs of ADRD. The interplay between PTSD, head injury, subjective (self-reported) cognitive concerns and genetic risk for ADRD is also not well understood, particularly in diverse ancestry groups. METHODS: Using data from the U.S. Department of Veterans Affairs (VA) Million Veteran Program (MVP), we examined the relationship between dementia risk factors (APOE ε4, PTSD, TBI) and subjective cognitive concerns (SCC) measured in individuals of European (n = 140,921), African (n = 15,788), and Hispanic (n = 8,064) ancestry (EA, AA, and HA, respectively). We then used data from the VA electronic medical record to perform a retrospective survival analysis evaluating PTSD, TBI, APOE ε4, and SCC and their associations with risk of conversion to ADRD in Veterans aged 65 and older. RESULTS: PTSD symptoms (B = 0.50-0.52, p
- Published
- 2024
7. Older adults at greater risk for Alzheimers disease show stronger associations between sleep apnea severity in REM sleep and verbal memory.
- Author
-
Lui, Kitty, Dave, Abhishek, Sprecher, Kate, Chappel-Farley, Miranda, Riedner, Brady, Heston, Margo, Taylor, Chase, Carlsson, Cynthia, Okonkwo, Ozioma, Asthana, Sanjay, Johnson, Sterling, Bendlin, Barbara, Benca, Ruth, and Mander, Bryce
- Subjects
Humans ,Female ,Male ,Alzheimer Disease ,Middle Aged ,Sleep ,REM ,Aged ,Sleep Apnea ,Obstructive ,Polysomnography ,Risk Factors ,Verbal Learning ,Apolipoprotein E4 ,Memory ,Severity of Illness Index ,Sleep Apnea Syndromes - Abstract
BACKGROUND: Obstructive sleep apnea (OSA) increases risk for cognitive decline and Alzheimers disease (AD). While the underlying mechanisms remain unclear, hypoxemia during OSA has been implicated in cognitive impairment. OSA during rapid eye movement (REM) sleep is usually more severe than in non-rapid eye movement (NREM) sleep, but the relative effect of oxyhemoglobin desaturation during REM versus NREM sleep on memory is not completely characterized. Here, we examined the impact of OSA, as well as the moderating effects of AD risk factors, on verbal memory in a sample of middle-aged and older adults with heightened AD risk. METHODS: Eighty-one adults (mean age:61.7 ± 6.0 years, 62% females, 32% apolipoprotein E ε4 allele (APOE4) carriers, and 70% with parental history of AD) underwent clinical polysomnography including assessment of OSA. OSA features were derived in total, NREM, and REM sleep. REM-NREM ratios of OSA features were also calculated. Verbal memory was assessed with the Rey Auditory Verbal Learning Test (RAVLT). Multiple regression models evaluated the relationships between OSA features and RAVLT scores while adjusting for sex, age, time between assessments, education years, body mass index (BMI), and APOE4 status or parental history of AD. The significant main effects of OSA features on RAVLT performance and the moderating effects of AD risk factors (i.e., sex, age, APOE4 status, and parental history of AD) were examined. RESULTS: Apnea-hypopnea index (AHI), respiratory disturbance index (RDI), and oxyhemoglobin desaturation index (ODI) during REM sleep were negatively associated with RAVLT total learning and long-delay recall. Further, greater REM-NREM ratios of AHI, RDI, and ODI (i.e., more events in REM than NREM) were related to worse total learning and recall. We found specifically that the negative association between REM ODI and total learning was driven by adults 60 + years old. In addition, the negative relationships between REM-NREM ODI ratio and total learning, and REM-NREM RDI ratio and long-delay recall were driven by APOE4 carriers. CONCLUSION: Greater OSA severity, particularly during REM sleep, negatively affects verbal memory, especially for people with greater AD risk. These findings underscore the potential importance of proactive screening and treatment of REM OSA even if overall AHI appears low.
- Published
- 2024
8. APOE4/4 is linked to damaging lipid droplets in Alzheimers disease microglia.
- Author
-
Haney, Michael, Pálovics, Róbert, Munson, Christy, Long, Chris, Johansson, Patrik, Yip, Oscar, Dong, Wentao, Rawat, Eshaan, Tsai, Andy, Guldner, Ian, Lamichhane, Bhawika, Smith, Amanda, Schaum, Nicholas, Calcuttawala, Kruti, Shin, Andrew, Wang, Yung-Hua, Wang, Chengzhong, Koutsodendris, Nicole, Serrano, Geidy, Beach, Thomas, Reiman, Eric, Glass, Christopher, Abu-Remaileh, Monther, Enejder, Annika, Huang, Yadong, Wyss-Coray, Tony, Schlachetzki, Johannes, and West, Elizabeth
- Subjects
Animals ,Female ,Humans ,Male ,Mice ,Alzheimer Disease ,Amyloid beta-Peptides ,Apolipoprotein E4 ,Induced Pluripotent Stem Cells ,Lipid Droplets ,Microglia ,Triglycerides ,tau Proteins ,Culture Media ,Conditioned ,Phosphorylation ,Genetic Predisposition to Disease - Abstract
Several genetic risk factors for Alzheimers disease implicate genes involved in lipid metabolism and many of these lipid genes are highly expressed in glial cells1. However, the relationship between lipid metabolism in glia and Alzheimers disease pathology remains poorly understood. Through single-nucleus RNA sequencing of brain tissue in Alzheimers disease, we have identified a microglial state defined by the expression of the lipid droplet-associated enzyme ACSL1 with ACSL1-positive microglia being most abundant in patients with Alzheimers disease having the APOE4/4 genotype. In human induced pluripotent stem cell-derived microglia, fibrillar Aβ induces ACSL1 expression, triglyceride synthesis and lipid droplet accumulation in an APOE-dependent manner. Additionally, conditioned media from lipid droplet-containing microglia lead to Tau phosphorylation and neurotoxicity in an APOE-dependent manner. Our findings suggest a link between genetic risk factors for Alzheimers disease with microglial lipid droplet accumulation and neurotoxic microglia-derived factors, potentially providing therapeutic strategies for Alzheimers disease.
- Published
- 2024
9. The impact of apolipoprotein E, type ∊4 allele on Alzheimer's disease pathological biomarkers: a comprehensive post-mortem pilot-analysis.
- Author
-
Wan, Ziyu and Ma, Tao
- Subjects
- *
GENETIC risk score , *DISEASE risk factors , *PATHOLOGICAL physiology , *ALZHEIMER'S disease , *APOLIPOPROTEIN E , *APOLIPOPROTEIN E4 - Abstract
The apolipoprotein E type ∊4 allele (ApoE4) is known as the strongest genetic risk factor for Alzheimer's Disease (AD). Meanwhile, many aspects of its impact on AD pathology remain underexplored. This study conducts a systematic data analysisof donor data from the Seattle Alzheimer's Disease Brain Cell Atlas. Our investigation delves into the intricate interplay between identified biomarkers and their correlation with ApoE4 across all severities of AD. Employing Pearson R correlation, and one-way and two-way ANOVA tests, we elucidate the pathological changes in biomarkers and the altering effects of ApoE4. Remarkably, the phosphorylation of tau observed in neurofibrillary tangles (NFTs) marked by the AT8 antibody, emerges as the most correlated factor with other pathological biomarkers. This correlation is mediated by both tau and amyloid pathology, suggesting a higher hierarchical role in determining AD pathological effects than other biomarkers. However, non-ApoE4 carriers exhibit a more significant correlation with disease progression severity compared to ApoE4 carriers, though ApoE4 carriers demonstrate significance in exacerbating the effect of accumulating phosphorylated tau and amyloid plaques assessed by AT8 and 6E10 antibodies. Furthermore, our analysis does not observe dramatic neuronal changes in grey matter across the span of AD pathology. Glia activation, measured by Iba1 and GFAP, demonstrates an amyloid-specific correlation. This research marks the first human post-mortem analysis providing a comprehensive examination of prevailing AD biomarkers and their interconnectedness with pathology and ApoE4 genetic factor. Limitations in the study are acknowledged, underscoring the need for further exploration and refinement in future research endeavors. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
10. Sex differences in cognition, anxiety-phenotype and therapeutic effect of metformin in the aged apoE-TR mice.
- Author
-
Lin, Yingbin, Luo, Xinqun, Wang, Fangyu, Cai, Huange, Lin, Yuanxiang, Kang, Dezhi, and Fang, Wenhua
- Subjects
- *
DISEASE risk factors , *APOLIPOPROTEIN E4 , *HYPOGLYCEMIA , *MEDICAL sciences , *APOLIPOPROTEIN E , *METFORMIN - Abstract
Background: Apolipoprotein E4 (ApoE4) is associated with an increased risk of Alzheimer's disease (AD), depression, and anxiety, which were reported to improve after the administration of metformin. However, sex influence on the effect of ApoE4 and metformin on cognition and mental health is poorly understood. Methods: ApoE3-TR and apoE4-TR mice of both sexes were randomly assigned to the normal saline and metformin groups from 13 months to 18 months of age. Behavior tests (MWM, EPM, OFT, TST, FST) were conducted to assess cognition, anxiety, and depression-like behaviors. The mice's blood glucose was also recorded. Results: Male aged apoE4-TR mice are more vulnerable to cognitive decline than females. Metformin improves the spatial memory of female, but not male apoE3-TR mice and female apoE4-TR mice while aggravating the cognitive impairment of male apoE4-TR mice. The anxiety-like phenotypes in male apoE4-TR mice are more severe than in male apoE3-TR mice, while metformin ameliorates the anxiety-like behaviors in the male apoE4-TR mice but not in male apoE3-TR mice. In addition, metformin alleviates depression-like behaviors in male and female apoE4-TR mice. The hypoglycemic effect of metformin is insignificant in both male and female apoE4-TR mice. Conclusions: Male sex exacerbates APOE4-related cognitive impairment and anxiety in aged mice and is insensitive to the cognition improvement effect of metformin in the aged apoE3 mice. Male sex with APOE4 may experience more severe cognitive impairment after treatment with metformin while sensitive to the anti-anxiety effects of metformin. These findings identify sex-specific effects on ApoE4-based dementia, anxiety prevention, and therapy, emphasizing the importance of further sex dimension analyses in vivo and clinical studies. Plain language summary: The apolipoprotein E4 (APOE4) gene increases the risk of Alzheimer's disease, depression, and anxiety. The present study examined how ApoE4 and a hypoglycemic drug, metformin, affected male and female mice's cognition and mental health. The mice were carriers of the APOE3 gene (which is less risky) or the APOE4 gene. They were given normal saline or metformin from 13 to 18 months of age. We found that: (1) Male apoE4 mice showed more cognitive decline than female apoE4 mice as they aged. (2) Metformin improved the spatial cognition of female apoE3 mice but not male apoE3 mice or female apoE4 mice. Metformin worsened the cognitive problems of male apoE4 mice. (3) Male apoE4 mice showed more anxiety-like behavior than male apoE3 mice. However, metformin reduced the anxiety in male apoE4 mice but not in male apoE3 mice. (4) Metformin reduced depression-like phenomenon in both male and female apoE4 mice. (5) ApoE4 reduces the ability of metformin to lower blood sugar in mice, regardless of gender. These findings highlight the importance of considering sex differences in studies of ApoE4-related dementia, anxiety, and treatment. Highlights: ApoE4 could aggravate memory decline and anxiety-like phenotype in male aged mice but not females. Metformin was observed to have a sex-dependent improvement in the spatial memory of aged apoE3-TR mice. Metformin leads to lower cognitive performance in male apoE4-TR mice and has anti-anxiety effects on them. ApoE4 blunts metformin's hypoglycemic impact in aged male and female mice. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
11. The effect of the APOE4 genotype on physiological and cognitive health in randomised controlled trials with an exercise intervention: a systematic review and meta-analysis.
- Author
-
Spencer, Felicity S. E., Elsworthy, Richard J., Breen, Leigh, Bishop, Jon R. B., Dunleavy, Connor, and Aldred, Sarah
- Subjects
- *
GENETIC risk score , *DISEASE risk factors , *EXERCISE physiology , *RANDOMIZED controlled trials , *APOLIPOPROTEIN E4 - Abstract
Background: Alzheimer's disease is caused by modifiable and non-modifiable risk factors. Randomised controlled trials have investigated whether the strongest genetic risk factor for Alzheimer's disease, APOE4, impacts the effectiveness of exercise on health. Systematic reviews are yet to evaluate the effect of exercise on physical and cognitive outcomes in APOE genotyped participants. A quality assessment of these randomised controlled trials is needed to understand the impact genotype has on the potential success of intervention. This systematic review aimed to determine if the APOE4 genotype influences the effectiveness of exercise-based randomised controlled trials. Method: Searches on MEDLINE, EMBASE, and PsycINFO identified eligible exercise based randomised controlled trials incorporating participants with varied cognitive abilities. Quality assessments were conducted. Results: Nineteen studies met the inclusion criteria for systematic review, and 3 for the meta-analysis. Very low to moderate quality evidence showed that APOE4 carriers benefitted more than APOE4 non-carriers on cognitive (e.g. executive function, learning) and physical (e.g. relative telomere length) outcomes after exercise; and that APOE4 non-carriers benefited over carriers for physical (serum BDNF, gait speed) and cognitive (global cognition, verbal memory) markers. Very low quality evidence indicated that there was no evidence of difference between APOE4 carriers and non-carriers on physical function outcomes in meta-analysis. Several areas of study design and reporting, including maintenance of relative exercise intensity and complete statistical reporting, were identified as needing improvement. Discussion: This systematic review found very limited evidence to suggest that exercise interventions can benefit APOE4 carriers and non-carriers equally, though conclusions were limited by evidence quality. Further randomised controlled trials, stratifying participants by APOE status are required to better understand the relationship between APOE genotype and the effect of exercise on health-related outcomes. Trial registration: This review was registered with PROSPERO (CRD42023436842). Registered on June 16, 2023. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
12. APOE4 and infectious diseases jointly contribute to brain glucose hypometabolism, a biomarker of Alzheimer's pathology: New findings from the ADNI.
- Author
-
Lathika Rajendrakumar, Aravind, Arbeev, Konstantin G., Bagley, Olivia, Duan, Matt, Yashin, Anatoliy I., and Ukraintseva, Svetlana
- Subjects
- *
GENETIC risk score , *ALZHEIMER'S disease , *MILD cognitive impairment , *APOLIPOPROTEIN E4 ,BRAIN metabolism - Abstract
Background: Impaired brain glucose metabolism is a preclinical feature of neurodegenerative diseases such as Alzheimer's disease (AD). Infections may promote AD-related pathology. Therefore, we investigated the interplay between infections and APOE4, a strong genetic risk factor for AD. Methods: We analyzed data on 1,509 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database using multivariate linear regression models. The outcomes were rank-normalized hypometabolic convergence index (HCI), statistical regions of interest (SROI) for AD, and mild cognitive impairment (MCI). Marginal mean estimates for infections, stratified by APOE4 carrier status, were then computed. Results: Prior infections were associated with greater HCI [β = 0.15, 95% CI: 0.03, 0.27, p = 0.01]. The combined effects of infections and APOE4 carriers on HCI levels were significantly greater than either variable alone. Among APOE4 carriers, the estimated marginal mean was 0.62, rising to 0.77, with infections (p<0.001), indicating an interaction effect. Carriers with multiple infections showed greater hypometabolism (higher HCI), with an estimate of 0.44 (p = 0.01) compared to 0.11 (p = 0.08) for those with a single infection, revealing a dose-response relationship. The estimates for the association of infections with SROI AD and SROI MCI were β = -0.01 (p = 0.02) and β = -0.01 (p = 0.04), respectively. Conclusion: Our findings suggest that infections and APOE4 jointly contribute to brain glucose hypometabolism and AD pathology, supporting a "multi-hit" mechanism in AD development. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
13. Biochemical Investigation of the Association of Apolipoprotein E Gene Allele Variations with Insulin Resistance and Amyloid‐β Aggregation in Cardiovascular Disease.
- Author
-
Jabeen, Komal, Rehman, Kanwal, Akash, Muhammad Sajid Hamid, Hussain, Amjad, Shahid, Mudassar, and Sadaf, Bushra
- Subjects
- *
TYPE 2 diabetes , *ALZHEIMER'S disease , *CARDIOVASCULAR diseases risk factors , *INSULIN resistance , *LIVER enzymes , *APOLIPOPROTEIN E , *APOLIPOPROTEIN E4 - Abstract
This article investigates the intricate associations between apolipoprotein E (APOE) gene alleles variation, insulin resistance (IR) and amyloid‐β aggregation in cardiovascular disease (CVD) patients. A cohort of 250 patients exhibiting the symptoms of CVD and 50 control subjects participated in this study. After applying the stringent inclusion and exclusion criteria, the diseased group was further stratified into three categories: CVD+ (Alzheimer's disease) AD, CVD + (diabetes mellitus) DM and CVD + DM + AD. Blood samples were collected from all recruited participants for the biochemical analyses of lipid profile, glycaemic status, liver function enzymes, inflammatory and oxidative stress biomarkers. Tetra amplification‐refractory mutation system‐polymerase chain reaction (ARMS‐PCR) was employed for APOE gene analysis. Biochemical evaluations revealed the significant elevations in the serum levels of glucose, liver enzymes, interleukin‐6 (IL‐6), cholesterol, low‐density lipoproteins (LDL), triglycerides (TG) and malondialdehyde (MDA) in CVD + DM + AD group. Conversely, the serum levels of insulin, HDL and hexokinase decreased in CVD + DM + AD group compared to the controls and other CVD groups. Tetra ARMS‐PCR results indicated a higher percentage of the risk allele in CVD + DM + AD group when compared with the other groups. Our study elucidates the multifaceted cardiovascular risk factors contributing to IR and AD in CVD patients. Age‐related risk factors, prevalence of APOE risk alleles and the impact of statin use on AD incidences were identified. These findings underscore the need for tailored preventive measures, particularly in APOEε4 and ε3/ε4 carriers with CVD. Further studies should delve into the knowledge‐based protocols to comprehend the underlying mechanisms. Focusing on the therapeutic targets to prevent or delay DM and AD progression in CVDs, especially in APOEε4 carriers, is essential. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
14. 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
15. APOE genotype and brain amyloid are associated with changes in the plasma proteome in elderly subjects without dementia.
- Author
-
Philippi, Sarah M., BP, Kailash, Raj, Towfique, and Castellano, Joseph M.
- Subjects
- *
BLOOD proteins , *ALZHEIMER'S disease , *PROTEIN-tyrosine kinases , *PROTEOMICS , *APOLIPOPROTEIN E4 - Abstract
Objective Methods Results Interpretation Recent work has bolstered the possibility that peripheral changes may be relevant to Alzheimer's disease pathogenesis in the brain. While age‐associated blood‐borne proteins have been targeted to restore function to the aged brain, it remains unclear whether other dysfunctional systemic states can be exploited for similar benefits. Here, we investigate whether APOE allelic variation or presence of brain amyloid are associated with plasma proteomic changes and the molecular processes associated with these changes.Using the SOMAscan assay, we measured 1305 plasma proteins from 53 homozygous, APOE3 and APOE4 subjects without dementia. We investigated the relationship of either the APOE‐ε4 allele or amyloid positivity with plasma proteome changes by linear mixed effects modeling and ontology‐based pathway and module–trait correlation analyses.APOE4 is associated with plasma protein differences linked to atherosclerosis, tyrosine kinase activity, cholesterol transport, extracellular matrix, and synaptogenesis pathways. Independent of APOE4, we found that subjects likely harboring brain amyloid exhibit plasma proteome signatures associated with AD‐linked pathways, including neurovascular dysfunction.Our results indicate that APOE4 status or presence of brain amyloid are associated with plasma proteomic shifts prior to the onset of symptoms, suggesting that systemic pathways in certain risk contexts may be plausible targets for disease modification. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Inhibiting the Cholesterol Storage Enzyme ACAT1/SOAT1 in Aging Apolipoprotein E4 Mice Alters Their Brains' Inflammatory Profiles.
- Author
-
Huynh, Thao N., Fikse, Emma N., De La Torre, Adrianna L., Havrda, Matthew C., Chang, Catherine C. Y., and Chang, Ta Yuan
- Subjects
- *
APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E , *LIPID rafts , *ALZHEIMER'S disease , *MACROMOLECULES , *ACYLTRANSFERASES - Abstract
Aging and apolipoprotein E4 (APOE4) are the two most significant risk factors for late-onset Alzheimer's disease (LOAD). Compared to APOE3, APOE4 disrupts cholesterol homeostasis, increases cholesteryl esters (CEs), and exacerbates neuroinflammation in brain cells, including microglia. Targeting CEs and neuroinflammation could be a novel strategy to ameliorate APOE4-dependent phenotypes. Toll-like receptor 4 (TLR4) is a key macromolecule in inflammation, and its regulation is associated with the cholesterol content of lipid rafts in cell membranes. We previously demonstrated that in normal microglia expressing APOE3, inhibiting the cholesterol storage enzyme acyl-CoA:cholesterol acyltransferase 1 (ACAT1/SOAT1) reduces CEs, dampened neuroinflammation via modulating the fate of TLR4. We also showed that treating myelin debris-loaded normal microglia with ACAT inhibitor F12511 reduced cellular CEs and activated ABC transporter 1 (ABCA1) for cholesterol efflux. This study found that treating primary microglia expressing APOE4 with F12511 also reduces CEs, activates ABCA1, and dampens LPS-dependent NFκB activation. In vivo, two-week injections of nanoparticle F12511, which consists of DSPE-PEG2000, phosphatidylcholine, and F12511, to aged female APOE4 mice reduced TLR4 protein content and decreased proinflammatory cytokines, including IL-1β in mice brains. Overall, our work suggests nanoparticle F12511 is a novel agent to ameliorate LOAD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Amyloid‐related imaging abnormalities in a woman with apolipoprotein E ε4 homozygotes treated with lecanemab for Alzheimer's disease.
- Author
-
Noguchi‐Shinohara, Moeko, Komatsu, Junji, and Ono, Kenjiro
- Subjects
- *
APOLIPOPROTEIN E , *ALZHEIMER'S disease , *MILD cognitive impairment , *LECANEMAB , *APOLIPOPROTEIN E4 - Abstract
Background Case Presentation Conclusion Lecanemab (Leqembi®) is an anti‐amyloid monoclonal antibody used for the treatment of Alzheimer's disease (AD). However, side effects may occur with lecanemab, including amyloid‐related imaging abnormalities (ARIA), especially in patients with apolipoprotein E ε4 (APOE4) homozygous.A 69‐year‐old woman had a 2‐year history of worsening memory symptoms and was diagnosed with mild cognitive impairment due to AD. Because she carries two copies of the E4 allele of APOE, her doctor did not recommend lecanemab treatment. However, she strongly desired lecanemab treatment and received four infusions of lecanemab. She had no symptoms or neurological abnormalities, but a head MRI before the fifth infusion showed moderate radiographic ARIA; therefore, she was admitted and treated with steroids. One month later, a head MRI showed the ARIA had disappeared.The indications of lecanemab treatment for patients with APOE4 homozygous must be carefully considered due to the higher risk of ARIA. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Inhibition of Bone Morphogenetic Protein Signaling Prevents Tau Pathology in iPSC Derived Neurons and PS19 Mice.
- Author
-
Affaneh, Amira, Linden, Anne K., Tunc‐Ozcan, Elif, Tsai, Yung‐Hsu, Peng, Chian‐Yu, and Kessler, John A.
- Subjects
- *
BONE morphogenetic proteins , *TAU proteins , *APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E , *PREVENTIVE medicine - Abstract
Objective Methods Results Interpretation Many neurodegenerative disorders share a common pathologic feature involving the deposition of abnormal tau protein in the brain (tauopathies). This suggests that there may be some shared pathophysiologic mechanism(s). The largest risk factor for the majority of these disorders is aging, suggesting involvement of the aging process in the shared pathophysiology. We test the hypothesis that an increase in bone morphogenetic protein (BMP) signaling that occurs during aging contributes to the onset and progression of tauopathies.Human induced pluripotent stem cell (iPSC)‐derived neurons from patients with Alzheimer's disease (AD) were used to investigate the effects of BMP signaling on tau phosphorylation and release and the mechanisms underlying these effects. Wildtype mice were used to examine effects of BMP signaling
in vivo . P301S (PS19) mice were examined for the effects of BMP signaling in a model of tauopathy.Here, we show that BMP signaling, mediated by non‐canonical p38 signaling, increases tau phosphorylation and release of p‐tau in human iPSC‐derived AD neurons. Further, there is an interaction between BMP signaling and apolipoprotein E4 (ApoE4) that significantly increases tau phosphorylation and release compared with ApoE3 neurons. Inhibiting BMP signaling reduces the changes in tau in the cultured human neurons, and it limits tau pathology and prevents cognitive decline in PS19 mice.Our study suggests that the age‐related increase in BMP signaling may participate in the onset and progression of tau pathology. Thus, therapeutic interventions that reduce BMP signaling in the aging brain could potentially slow or prevent development of diseases involving tau hyperphosphorylation. ANN NEUROL 2024 [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
19. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model.
- Author
-
Carling, Gillian K., Fan, Li, Foxe, Nessa R., Norman, Kendra, Wong, Man Ying, Zhu, Daphne, Corona, Carlo, Razzoli, Agnese, Yu, Fangmin, Yarahmady, Allan, Ye, Pearly, Chen, Hao, Huang, Yige, Amin, Sadaf, Sereda, Rebecca, Lopez-Lee, Chloe, Zacharioudakis, Emmanouil, Chen, Xiaoying, Xu, Jielin, and Cheng, Feixiong
- Subjects
- *
ALZHEIMER'S disease , *CYCLIC guanylic acid , *MYELOID cells , *TAUOPATHIES , *TAU proteins , *APOLIPOPROTEIN E4 - Abstract
The strongest risk factors for late-onset sporadic Alzheimer's disease (AD) include the ε4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2 R47H (R47H) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting detrimental disease mechanisms. We find that R47H induces neurodegeneration in 9- to 10-month-old female APOE4 tauopathy mice. The combination of APOE4 and R47H (APOE4-R47H) worsened hyperphosphorylated tau pathology in the frontal cortex and amplified tauopathy-induced microglial cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling and downstream interferon response. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk. [Display omitted] • AD risk gene TREM2-R47H worsens tau-induced neurodegeneration in APOE4 female mice • TREM2-R47H increases tau pathology in the frontal cortex of APOE4 female mice • APOE4 and R47H amplify tau-induced microglial cGAS-STING and IFN-I in female mice • cGAS- and BAX-dependent microglial senescence is upregulated by APOE4 and R47H Carling et al. combine Alzheimer's disease (AD) risk factors APOE4 and R47H in a tauopathy model, uncovering amplification of microglial cGAS-STING, interferon, and cGAS-related senescence as disease-enhancing mechanisms in females. Their findings emphasize the importance of cGAS-dependent senescence as a potential driver of tauopathy in AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Pathways to Alzheimer's Disease: The Intersecting Roles of Clusterin and Apolipoprotein E in Amyloid-β Regulation and Neuronal Health.
- Author
-
Laslo, Alexandru, Laslo, Laura, Arbănași, Eliza-Mihaela, Ujlaki-Nagi, Alexandru-Andrei, Chinezu, Laura, Ivănescu, Adrian Dumitru, Arbănași, Emil-Marian, Cărare, Roxana Octavia, Cordoș, Bogdan Andrei, Popa, Ioana Adriana, and Brînzaniuc, Klara
- Subjects
- *
CEREBRAL amyloid angiopathy , *ALZHEIMER'S disease , *CLUSTERIN , *NEUROLOGICAL disorders , *EXTRACELLULAR space , *APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E - Abstract
One of the hallmarks of Alzheimer's disease (AD) is the deposition of amyloid-β (Aβ) within the extracellular spaces of the brain as plaques and along the blood vessels in the brain, a condition also known as cerebral amyloid angiopathy (CAA). Clusterin (CLU), or apolipoprotein J (APOJ), is a multifunctional glycoprotein that has a role in many physiological and neurological conditions, including AD. The apolipoprotein E (APOE) is a significant genetic factor in AD, and while the primary physiological role of APOE in the brain and peripheral tissues is to regulate lipid transport, it also participates in various other biological processes, having three basic human forms: APOE2, APOE3, and APOE4. Notably, the APOE4 allele substantially increases the risk of developing late-onset AD. The main purpose of this review is to examine the roles of CLU and APOE in AD pathogenesis in order to acquire a better understanding of AD pathogenesis from which to develop targeted therapeutic approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Long‐Term Monitoring of ApoE4 within Aqueous Humor Using Intraocular Lenses Containing Target‐Responsive Inverse Opal Hydrogel.
- Author
-
Lee, Hyo, Lim, Jaewook, Shin, Moo‐Kwang, Moon, Chae‐Eun, Lee, Jun‐Ki, Kim, Jiwon, Kang, Yujin, Woo Ji, Yong, and Haam, Seungjoo
- Subjects
- *
AQUEOUS humor , *ALZHEIMER'S disease , *MEDICAL equipment , *NEURODEGENERATION , *APOLIPOPROTEIN E4 - Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), pose significant challenges to global healthcare because of their irreversible postsymptomatic progression. Conventional radiographic techniques have limitations in early detection, owing to the time lapse between symptom recognition by individuals and precise inspection using medical instruments. Current research explores ophthalmic approaches to address this gap, investigating the physiological link between ocular health and brain diseases. Intraocular lenses (IOLs), widely used in cataract and refractive surgeries, provide a safe and minimally invasive platform for the continuous monitoring of neurodegenerative disease biomarkers, including those associated with AD. By integrating biomolecule‐responsive sensors with IOLs, it becomes feasible to achieve early diagnosis and long‐term, noninvasive monitoring postimplantation. Herein, a fluorescently labeled inverse opal hydrogel (FIOH) is merged with IOLs for ApoE4 detection, a key risk factor for AD. The permeability and inherent photonic properties of FIOH facilitate the cumulative enhancement of the fluorescence signal. In a simulation of the aqueous humor circulation, the detection limit for ApoE4 is determined to be 0.1 nM. Therefore, FIOH‐integrated IOLs hold promise not only for early detection but also for providing a reliable platform for the continuous, noninvasive monitoring of neurodegenerative diseases after the initial, minimally invasive implantation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Kidney damage associated with COVID-19: from the acute to the chronic phase.
- Author
-
Nlandu, Yannick, Tannor, Elliot Koranteng, Bafemika, Titilope, and Makulo, Jean-Robert
- Subjects
- *
SARS-CoV-2 , *POST-acute COVID-19 syndrome , *COVID-19 , *APOLIPOPROTEIN E4 , *ACUTE kidney failure , *ANGIOTENSIN converting enzyme , *RENAL tubular transport disorders - Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-COV-2) infection is well established as a systemic disease including kidney damage. The entry point into the renal cell remains the angiotensin-converting enzyme 2 (ACE-2) receptor and the spectrum of renal lesions is broad, with a clear predominance of structural and functional tubular lesions. The most common form of glomerular injury is collapsing glomerulopathy (CG), which is strongly associated with apolipoprotein L1(APOL-1) risk variants. These acute lesions, which are secondary to the direct or indirect effects of SARS-CoV-2, can progress to chronicity and are specific to long COVID-19 in the absence of any other cause. Residual inflammation associated with SARS-CoV-2 infection, in addition to acute kidney injury (AKI) as a transitional state with or without severe histological lesions, may be responsible for greater kidney function decline in mild-to-moderate COVID-19. This review discusses the evidence for renal histological markers of chronicity in COVID-19 patients and triggers of low-grade inflammation that may explain the decline in kidney function in the post-COVID-19 period. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Advancements and challenges in mouse models of Alzheimer's disease.
- Author
-
Qian, Zhengjiang, Li, Yanjiao, and Ye, Keqiang
- Subjects
- *
ALZHEIMER'S disease , *GENETIC mutation , *LABORATORY mice , *ANIMAL disease models , *CLINICAL trials , *APOLIPOPROTEIN E4 - Abstract
Alzheimer's disease (AD) mouse models are indispensable tools to understand the regulatory mechanisms of AD pathogenesis and to evaluate the potential therapeutic strategies in preclinical studies, although they rarely recapitulate the entire spectrum of AD pathological features. Most of the commonly used AD mouse models are developed by overexpressing genetic mutations implicated in familial AD, which accounts for less than 1% of all AD cases, and these models represent an extreme condition that would not occur in human patients with AD. On the basis of multiple humanized sporadic AD genetic risk factors, novel mouse models have increasingly been developed in an attempt to capture the trajectory and progression of AD more accurately. Despite numerous advantages, AD mouse models have their intrinsic limitations in translating preclinical findings to human clinical trials. Alzheimer's disease (AD) poses a significant health challenge worldwide, and the development of effective treatments necessitates a comprehensive understanding of its pathophysiology. Mouse models have been instrumental in offering insights into the crucial pathogenesis of AD. However, current models rarely recapitulate all aspects of AD pathology in patients; thus, translating the findings from mouse to human clinical trials has proved to be complex. In this review, we outline the development of some prevalently used AD mice, with a particular emphasis on the latest advances in newly generated models. In addition, we discuss the advantages and limitations in mouse models of AD and their applications in blood-based biomarkers. Finally, we speculate on potential future research directions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Prevalence of ApoE Alleles in a Spanish Population of Patients with a Clinical Diagnosis of Alzheimer's Disease: An Observational Case-Control Study.
- Author
-
Bello-Corral, Laura, Seco-Calvo, Jesús, Molina Fresno, Angela, González, Ana Isabel, Llorente, Ana, Fernández-Lázaro, Diego, and Sánchez-Valdeón, Leticia
- Subjects
DISEASE risk factors ,ALZHEIMER'S disease ,APOLIPOPROTEIN E ,NEURODEGENERATION ,APOLIPOPROTEIN E4 ,POPULATION of China - Abstract
Background and Objectives: Alzheimer's dementia is a progressive neurodegenerative disease that affects memory abilities due to genetic and environmental factors. A well-known gene that influences the risk of Alzheimer's disease is the apolipoprotein E (APOE) gene. The APOE gene is involved in the production of a protein that helps transport cholesterol and other types of fat in the bloodstream. Problems in this process are thought to contribute to the development of Alzheimer's disease. APOE comes in several forms, which are called alleles (ε2, ε3, ε4). Materials and Methods: Therefore, our study aims to identify those subjects with a higher genetic risk through the polymorphism of the APOE gene, using a population screening in patients with a clinical diagnosis of AD in a region of Spain, Castilla y León, as potential biomarkers and to identify individuals at increased genetic risk by polymorphism of the APOE gene. An observational case-control study was conducted in Castilla y León (Spain). Saliva samples were collected and the ApoE gene was analyzed by PCR and agarose gel electrophoresis, respecting ethical criteria. Results: In the Alzheimer's population in Castilla y León, a high prevalence of ApoE3 (74%) was found, followed by ApoE4 (22%); in addition, a higher presence of the ε4 allele was found in the Alzheimer's disease (AD) group than in the control group. It was also observed that the ε2/ε2 genotype was not found in any individual with AD but was found in healthy subjects and that the opposite was observed for the ε4/ε4 genotype. The odds ratio (OR) indicated a risk four times greater of having AD if having the ε4 allele. Conclusions: The demonstrated relation between the different isoforms and the likelihood of developing AD has led to its consideration as a biomarker and a potential pre-symptomatic therapy. The molecular mechanisms that confer a disruptive and protective role to ApoE4 and ApoE2, respectively, are still being studied. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Cell type-specific roles of APOE4 in Alzheimer disease.
- Author
-
Blumenfeld, Jessica, Yip, Oscar, Kim, Min, and Huang, Yadong
- Subjects
Animals ,Alzheimer Disease ,Apolipoprotein E4 ,Neurons ,Neuroglia ,Astrocytes - Abstract
The ɛ4 allele of the apolipoprotein E gene (APOE), which translates to the APOE4 isoform, is the strongest genetic risk factor for late-onset Alzheimer disease (AD). Within the CNS, APOE is produced by a variety of cell types under different conditions, posing a challenge for studying its roles in AD pathogenesis. However, through powerful advances in research tools and the use of novel cell culture and animal models, researchers have recently begun to study the roles of APOE4 in AD in a cell type-specific manner and at a deeper and more mechanistic level than ever before. In particular, cutting-edge omics studies have enabled APOE4 to be studied at the single-cell level and have allowed the identification of critical APOE4 effects in AD-vulnerable cellular subtypes. Through these studies, it has become evident that APOE4 produced in various types of CNS cell - including astrocytes, neurons, microglia, oligodendrocytes and vascular cells - has diverse roles in AD pathogenesis. Here, we review these scientific advances and propose a cell type-specific APOE4 cascade model of AD. In this model, neuronal APOE4 emerges as a crucial pathological initiator and driver of AD pathogenesis, instigating glial responses and, ultimately, neurodegeneration. In addition, we provide perspectives on future directions for APOE4 research and related therapeutic developments in the context of AD.
- Published
- 2024
26. CYP1B1-RMDN2 Alzheimer’s disease endophenotype locus identified for cerebral tau PET
- Author
-
Nho, Kwangsik, Risacher, Shannon L, Apostolova, Liana G, Bice, Paula J, Brosch, Jared R, Deardorff, Rachael, Faber, Kelley, Farlow, Martin R, Foroud, Tatiana, Gao, Sujuan, Rosewood, Thea, Kim, Jun Pyo, Nudelman, Kelly, Yu, Meichen, Aisen, Paul, Sperling, Reisa, Hooli, Basavaraj, Shcherbinin, Sergey, Svaldi, Diana, Jack, Clifford R, Jagust, William J, Landau, Susan, Vasanthakumar, Aparna, Waring, Jeffrey F, Doré, Vincent, Laws, Simon M, Masters, Colin L, Porter, Tenielle, Rowe, Christopher C, Villemagne, Victor L, Dumitrescu, Logan, Hohman, Timothy J, Libby, Julia B, Mormino, Elizabeth, Buckley, Rachel F, Johnson, Keith, Yang, Hyun-Sik, Petersen, Ronald C, Ramanan, Vijay K, Ertekin-Taner, Nilüfer, Vemuri, Prashanthi, Cohen, Ann D, Fan, Kang-Hsien, Kamboh, M Ilyas, Lopez, Oscar L, Bennett, David A, Ali, Muhammad, Benzinger, Tammie, Cruchaga, Carlos, Hobbs, Diana, De Jager, Philip L, Fujita, Masashi, Jadhav, Vaishnavi, Lamb, Bruce T, Tsai, Andy P, Castanho, Isabel, Mill, Jonathan, Weiner, Michael W, and Saykin, Andrew J
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Dementia ,Neurodegenerative ,Acquired Cognitive Impairment ,Alzheimer's Disease ,Human Genome ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Brain Disorders ,Aging ,2.1 Biological and endogenous factors ,Neurological ,Aged ,Aged ,80 and over ,Animals ,Female ,Humans ,Male ,Mice ,Alzheimer Disease ,Amyloid beta-Peptides ,Apolipoprotein E4 ,Cytochrome P-450 CYP1B1 ,Disease Models ,Animal ,Endophenotypes ,Genome-Wide Association Study ,Polymorphism ,Single Nucleotide ,Positron-Emission Tomography ,tau Proteins ,Alzheimer’s Disease Neuroimaging Initiative ,Department of Defense Alzheimer’s Disease Neuroimaging Initiative ,Anti-Amyloid Treatment in Asymptomatic Alzheimer’s Study (A4 Study) and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration ,Australian Imaging ,Biomarker & Lifestyle Study - Abstract
Determining the genetic architecture of Alzheimer's disease pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we perform a genome-wide association study of cortical tau quantified by positron emission tomography in 3046 participants from 12 independent studies. The CYP1B1-RMDN2 locus is associated with tau deposition. The most significant signal is at rs2113389, explaining 4.3% of the variation in cortical tau, while APOE4 rs429358 accounts for 3.6%. rs2113389 is associated with higher tau and faster cognitive decline. Additive effects, but no interactions, are observed between rs2113389 and diagnosis, APOE4, and amyloid beta positivity. CYP1B1 expression is upregulated in AD. rs2113389 is associated with higher CYP1B1 expression and methylation levels. Mouse model studies provide additional functional evidence for a relationship between CYP1B1 and tau deposition but not amyloid beta. These results provide insight into the genetic basis of cerebral tau deposition and support novel pathways for therapeutic development in AD.
- Published
- 2024
27. Pure LATE-NC: Frequency, clinical impact, and the importance of considering APOE genotype when assessing this and other subtypes of non-Alzheimer’s pathologies
- Author
-
Katsumata, Yuriko, Wu, Xian, Aung, Khine Zin, Fardo, David W, Woodworth, Davis C, Sajjadi, S Ahmad, Tomé, Sandra O, Thal, Dietmar Rudolf, Troncoso, Juan C, Chang, Koping, Mock, Charles, and Nelson, Peter T
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Acquired Cognitive Impairment ,Brain Disorders ,Alzheimer's Disease ,Neurodegenerative ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Genetics ,Alzheimer's Disease Related Dementias (ADRD) ,Aging ,Dementia ,2.1 Biological and endogenous factors ,Neurological ,Humans ,Female ,Male ,Aged ,80 and over ,Aged ,Genotype ,Apolipoproteins E ,Alzheimer Disease ,TDP-43 Proteinopathies ,Brain ,Apolipoprotein E4 ,Community-based ,DLB ,Epidemiology ,FTD ,FTLD ,Prevalence ,Clinical Sciences ,Neurology & Neurosurgery - Abstract
Pure limbic-predominant age-related TDP-43 encephalopathy neuropathologic changes (pure LATE-NC) is a term used to describe brains with LATE-NC but lacking intermediate or severe levels of Alzheimer's disease neuropathologic changes (ADNC). Focusing on pure LATE-NC, we analyzed data from the National Alzheimer's Coordinating Center (NACC) Neuropathology Data Set, comprising clinical and pathological information aggregated from 32 NIH-funded Alzheimer's Disease Research Centers (ADRCs). After excluding subjects dying with unusual conditions, n = 1,926 autopsied subjects were included in the analyses. For > 90% of these participants, apolipoprotein E (APOE) allele status was known; 46.5% had at least one APOE 4 allele. In most human populations, only 15-25% of people are APOE ε4 carriers. ADRCs with higher documented AD risk allele (APOE or BIN1) rates had fewer participants lacking ADNC, and correspondingly low rates of pure LATE-NC. Among APOE ε4 non-carries, 5.3% had pure LATE-NC, 37.0% had pure ADNC, and 3.6% had pure neocortical Lewy body pathology. In terms of clinical impact, participants with pure LATE-NC tended to die after having received a diagnosis of dementia: 56% died with dementia among APOE ε4 non-carrier participants, comparable to 61% with pure ADNC. LATE-NC was associated with increased Clinical Dementia Rating Sum of Boxes (CDR-SOB) scores, i.e. worsened global cognitive impairments, in participants with no/low ADNC and no neocortical Lewy body pathology (p = 0.0023). Among pure LATE-NC cases, there was a trend for higher LATE-NC stages to be associated with worse CDR-SOB scores (p = 0.026 for linear trend of LATE-NC stages). Pure LATE-NC was not associated with clinical features of disinhibition or primary progressive aphasia. In summary, LATE-NC with no or low levels of ADNC was less frequent than pure ADNC but was not rare, particularly among individuals who lacked the APOE 4 allele, and in study cohorts with APOE 4 frequencies similar to those in most human populations.
- Published
- 2024
28. Oxylipin transport by lipoprotein particles and its functional implications for cardiometabolic and neurological disorders
- Author
-
Liang, Nuanyi, Harsch, Brian A, Zhou, Sitong, Borkowska, Alison, Shearer, Gregory C, Kaddurah-Daouk, Rima, Newman, John W, and Borkowski, Kamil
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Brain Disorders ,Neurosciences ,2.1 Biological and endogenous factors ,Neurological ,Humans ,Oxylipins ,Apolipoprotein E4 ,Lipoproteins ,Nervous System Diseases ,Cardiovascular Diseases ,Inflammation ,Cardiometabolic disorders ,Neurological disorders ,Neurodegenerative disorders ,Alzheimer's disease ,COVID-19 ,Immune memory ,Trained immunity ,Medical Biochemistry and Metabolomics ,Nutrition and Dietetics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics - Abstract
Lipoprotein metabolism is critical to inflammation. While the periphery and central nervous system (CNS) have separate yet connected lipoprotein systems, impaired lipoprotein metabolism is implicated in both cardiometabolic and neurological disorders. Despite the substantial investigation into the composition, structure and function of lipoproteins, the lipoprotein oxylipin profiles, their influence on lipoprotein functions, and their potential biological implications are unclear. Lipoproteins carry most of the circulating oxylipins. Importantly, lipoprotein-mediated oxylipin transport allows for endocrine signaling by these lipid mediators, long considered to have only autocrine and paracrine functions. Alterations in plasma lipoprotein oxylipin composition can directly impact inflammatory responses of lipoprotein metabolizing cells. Similar investigations of CNS lipoprotein oxylipins are non-existent to date. However, as APOE4 is associated with Alzheimer's disease-related microglia dysfunction and oxylipin dysregulation, ApoE4-dependent lipoprotein oxylipin modulation in neurological pathologies is suggested. Such investigations are crucial to bridge knowledge gaps linking oxylipin- and lipoprotein-related disorders in both periphery and CNS. Here, after providing a summary of existent literatures on lipoprotein oxylipin analysis methods, we emphasize the importance of lipoproteins in oxylipin transport and argue that understanding the compartmentalization and distribution of lipoprotein oxylipins may fundamentally alter our consideration of the roles of lipoprotein in cardiometabolic and neurological disorders.
- Published
- 2024
29. Effects of APOE2 and APOE4 on brain microstructure in older adults: modification by age, sex, and cognitive status
- Author
-
Reas, Emilie T, Triebswetter, Curtis, Banks, Sarah J, and McEvoy, Linda K
- Subjects
Health Services and Systems ,Health Sciences ,Biomedical Imaging ,Dementia ,Alzheimer's Disease ,Brain Disorders ,Aging ,Prevention ,Acquired Cognitive Impairment ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Clinical Research ,Neurodegenerative ,Neurosciences ,2.1 Biological and endogenous factors ,Neurological ,Aged ,Female ,Humans ,Male ,Alzheimer Disease ,Apolipoprotein E2 ,Apolipoprotein E4 ,Brain ,Cognition ,Cognitive Dysfunction ,Aged ,80 and over ,APOE ,Diffusion MRI ,Brain microstructure ,Genetics ,Brain aging ,Neuroimaging ,Sex differences ,Medical and Health Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundAPOE4 is the strongest genetic risk factor for sporadic Alzheimer's disease (AD), whereas APOE2 confers protection. However, effects of APOE on neurodegeneration in cognitively intact individuals, and how these associations evolve with cognitive decline, are unclear. Furthermore, few studies have evaluated whether effects of APOE on neurodegenerative changes are modified by other AD key risk factors including age and sex.MethodsParticipants included older adults (57% women; 77 ± 7 years) from the Rancho Bernardo Study of Health Aging and the University of California San Diego Alzheimer's Disease Research Center, including 192 cognitively normal (CN) individuals and 33 with mild cognitive impairment. Participants underwent diffusion MRI, and multicompartment restriction spectrum imaging (RSI) metrics were computed in white matter, gray matter, and subcortical regions of interest. Participants were classified as APOE4 carriers, APOE2 carriers, and APOE3 homozygotes. Analysis of covariance among CN (adjusting for age, sex, and scanner) assessed differences in brain microstructure by APOE, as well as interactions between APOE and sex. Analyses across all participants examined interactions between APOE4 and cognitive status. Linear regressions assessed APOE by age interactions.ResultsAmong CN, APOE4 carriers showed lower entorhinal cortex neurite density than non-carriers, whereas APOE2 carriers showed lower cingulum neurite density than non-carriers. Differences in entorhinal microstructure by APOE4 and in entorhinal and cingulum microstructure by APOE2 were present for women only. Age correlated with lower entorhinal restricted isotropic diffusion among APOE4 non-carriers, whereas age correlated with lower putamen restricted isotropic diffusion among APOE4 carriers. Differences in microstructure between cognitively normal and impaired participants were stronger for APOE4-carriers in medial temporal regions, thalamus, and global gray matter, but stronger for non-carriers in caudate.ConclusionsThe entorhinal cortex may be an early target of neurodegenerative changes associated with APOE4 in presymptomatic individuals, whereas APOE2 may support beneficial white matter and entorhinal microstructure, with potential sex differences that warrant further investigation. APOE modifies microstructural patterns associated with aging and cognitive impairment, which may advance the development of biomarkers to distinguish microstructural changes characteristic of normal brain aging, APOE-dependent pathways, and non-AD etiologies.
- Published
- 2024
30. Synthesis and Preclinical Evaluation of 22-[18F]Fluorodocosahexaenoic Acid as a Positron Emission Tomography Probe for Monitoring Brain Docosahexaenoic Acid Uptake Kinetics.
- Author
-
Duro, Marlon, Van Valkenburgh, Juno, Ingles, Diana, Tran, Jenny, Cai, Zhiheng, Ebright, Brandon, Wang, Shaowei, Kerman, Bilal, Galvan, Jasmin, Hwang, Sung Hee, Sta Maria, Naomi, Zanderigo, Francesca, Croteau, Etienne, Cunnane, Stephen, Rapoport, Stanley, Louie, Stan, Jacobs, Russell, Yassine, Hussein, and Chen, Kai
- Subjects
docosahexaenoic acid ,incorporation coefficient ,polyunsaturated fatty acid ,positron emission tomography ,radiofluorination ,Humans ,Mice ,Animals ,Docosahexaenoic Acids ,Apolipoprotein E4 ,Brain ,Positron-Emission Tomography ,Biological Transport ,Alzheimer Disease - Abstract
Docosahexaenoic acid [22:6(n-3), DHA], a polyunsaturated fatty acid, has an important role in regulating neuronal functions and in normal brain development. Dysregulated brain DHA uptake and metabolism are found in individuals carrying the APOE4 allele, which increases the genetic risk for Alzheimers disease (AD), and are implicated in the progression of several neurodegenerative disorders. However, there are limited tools to assess brain DHA kinetics in vivo that can be translated to humans. Here, we report the synthesis of an ω-radiofluorinated PET probe of DHA, 22-[18F]fluorodocosahexaenoic acid (22-[18F]FDHA), for imaging the uptake of DHA into the brain. Using the nonradiolabeled 22-FDHA, we confirmed that fluorination of DHA at the ω-position does not significantly alter the anti-inflammatory effect of DHA in microglial cells. Through dynamic PET-MR studies using mice, we observed the accumulation of 22-[18F]FDHA in the brain over time and estimated DHAs incorporation coefficient (K*) using an image-derived input function. Finally, DHA brain K* was validated using intravenous administration of 15 mg/kg arecoline, a natural product known to increase the DHA K* in rodents. 22-[18F]FDHA is a promising PET probe that can reveal altered lipid metabolism in APOE4 carriers, AD, and other neurologic disorders. This new probe, once translated into humans, would enable noninvasive and longitudinal studies of brain DHA dynamics by guiding both pharmacological and nonpharmacological interventions for neurodegenerative diseases.
- Published
- 2023
31. 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
32. Association between APOE-ε4 allele and cognitive function is mediated by Alzheimers disease pathology: a population-based autopsy study in an admixed sample.
- Author
-
Naslavsky, Michel, Zatz, Mayana, Nitrini, Ricardo, Jacob-Filho, Wilson, Suemoto, Claudia, Paradela, Regina, Justo, Alberto, Paes, Vítor, Leite, Renata, Pasqualucci, Carlos, and Grinberg, Lea
- Subjects
Apolipoprotein E ,Cognition ,Dementia ,Mediation analysis ,Neuritic plaques ,Neurofibrillary tangles ,Aged ,Aged ,80 and over ,Female ,Humans ,Male ,Middle Aged ,Alleles ,Alzheimer Disease ,Apolipoprotein E4 ,Apolipoproteins E ,Arteriosclerosis ,Autopsy ,Cerebral Amyloid Angiopathy ,Cognition ,DNA-Binding Proteins ,Genotype ,Lewy Body Disease ,Stroke ,Lacunar - Abstract
BACKGROUND: Apolipoprotein E ε4 allele (APOE-ε4) is the main genetic risk factor for late-onset Alzheimers disease (AD) and may impact cognitive function also via other neuropathological lesions. However, there is limited evidence available from diverse populations, as APOE associations with dementia seem to differ by race. Therefore, we aimed to evaluate the pathways linking APOE-ε4 to cognitive abilities through AD and non-AD neuropathology in an autopsy study with an admixed sample. METHODS: Neuropathological lesions were evaluated following international criteria using immunohistochemistry. Participants were classified into APOE-ε4 carriers (at least one ε4 allele) and non-carriers. Cognitive abilities were evaluated by the Clinical Dementia Rating Scale sum of boxes. Mediation analyses were conducted to assess the indirect association of APOE-ε4 with cognition through AD-pathology, lacunar infarcts, hyaline arteriosclerosis, cerebral amyloid angiopathy (CAA), Lewy body disease (LBD), and TAR DNA-binding protein 43 (TDP-43). RESULTS: We included 648 participants (mean age 75 ± 12 years old, mean education 4.4 ± 3.7 years, 52% women, 69% White, and 28% APOE-ε4 carriers). The association between APOE-ε4 and cognitive abilities was mediated by neurofibrillary tangles (β = 0.88, 95% CI = 0.45; 1.38, p
- Published
- 2023
33. Associations of dietary cholesterol and fat, blood lipids, and risk for dementia in older women vary by APOE genotype.
- Author
-
Dunk, Michelle, Li, Jie, Liu, Simin, Casanova, Ramon, Chen, Jiu-Chiuan, Espeland, Mark, Hayden, Kathleen, Manson, JoAnn, Rapp, Stephen, Shadyab, Aladdin, Snetselaar, Linda, Van Horn, Linda, Wild, Robert, and Driscoll, Ira
- Subjects
Alzheimers disease ,Mendelian randomization ,apolipoprotein E ,cholesterol ,dementia ,diet ,mild cognitive impairment ,womens health initiative memory study ,Aged ,Female ,Humans ,Apolipoprotein E4 ,Apolipoproteins E ,Cholesterol ,Cholesterol ,Dietary ,Dementia ,Genotype ,Risk Factors ,Triglycerides - Abstract
INTRODUCTION: Whether apolipoprotein Es (APOEs) involvement in lipid metabolism contributes to Alzheimers disease (AD) risk remains unknown. METHODS: Incident probable dementia and cognitive impairment (probable dementia+mild cognitive impairment) were analyzed in relation to baseline serum lipids (total, low-density lipoprotein [LDL], high-density lipoprotein [HDL], non-HDL cholesterol, total-to-HDL, LDL-to-HDL, remnant cholesterol, and triglycerides) using Mendelian randomization in 5358 postmenopausal women from the Womens Health Initiative Memory Study. We also examined associations of baseline dietary cholesterol and fat with lipids based on APOE status. RESULTS: After an average of 11.13 years, less favorable lipid levels related to greater dementia and cognitive impairment risk. Dementia (odds ratio [OR] = 3.13; 95% confidence interval [CI]: 2.31 to 4.24) and cognitive impairment (OR = 2.38; 95% CI: 1.85 to 3.06) risk were greatest in relation to higher remnant cholesterol levels. Greater cholesterol consumption related to poorer lipids in APOE4+ compared to APOE3 carriers. DISCUSSION: APOE4+ carriers consuming more cholesterol had less favorable lipids, which were associated with greater dementia and cognitive impairment risk. HIGHLIGHTS: Less favorable serum lipids were associated with higher dementia incidence. Mendelian randomization findings suggest causality between lipids and dementia. Lipid levels in older women may be clinical indicators of dementia risk. APOE4 carriers had poorest lipid profiles in relation to cholesterol consumption. APOE risk for dementia may be modifiable through lipid management.
- Published
- 2023
34. The APOE-R136S mutation protects against APOE4-driven Tau pathology, neurodegeneration and neuroinflammation.
- Author
-
Nelson, Maxine, Liu, Peng, Agrawal, Ayushi, Yip, Oscar, Blumenfeld, Jessica, Traglia, Michela, Kim, Min, Koutsodendris, Nicole, Rao, Antara, Grone, Brian, Hao, Yanxia, Yoon, Seo, Xu, Qin, De Leon, Samuel, Choenyi, Tenzing, Thomas, Reuben, Lopera, Francisco, Quiroz, Yakeel, Arboleda-Velasquez, Joseph, Reiman, Eric, Mahley, Robert, and Huang, Yadong
- Subjects
Animals ,Humans ,Mice ,Alzheimer Disease ,Apolipoprotein E3 ,Apolipoprotein E4 ,Mutation ,Neuroinflammatory Diseases ,Tauopathies - Abstract
Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for late-onset Alzheimers disease (LOAD), leading to earlier age of clinical onset and exacerbating pathologies. There is a critical need to identify protective targets. Recently, a rare APOE variant, APOE3-R136S (Christchurch), was found to protect against early-onset AD in a PSEN1-E280A carrier. In this study, we sought to determine if the R136S mutation also protects against APOE4-driven effects in LOAD. We generated tauopathy mouse and human iPSC-derived neuron models carrying human APOE4 with the homozygous or heterozygous R136S mutation. We found that the homozygous R136S mutation rescued APOE4-driven Tau pathology, neurodegeneration and neuroinflammation. The heterozygous R136S mutation partially protected against APOE4-driven neurodegeneration and neuroinflammation but not Tau pathology. Single-nucleus RNA sequencing revealed that the APOE4-R136S mutation increased disease-protective and diminished disease-associated cell populations in a gene dose-dependent manner. Thus, the APOE-R136S mutation protects against APOE4-driven AD pathologies, providing a target for therapeutic development against AD.
- Published
- 2023
35. 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
36. Influence of APOE4 genotype on PCSK9-lipids association in cerebrospinal fluid and serum of patients in the Alzheimer's disease continuum.
- Author
-
Papotti, Bianca, Palumbo, Marcella, Adorni, Maria Pia, Elviri, Lisa, Chiari, Annalisa, Tondelli, Manuela, Bedin, Roberta, Baldelli, Enrica, Lancellotti, Giulia, Lupo, Maria Giovanna, Ferri, Nicola, Bertolotti, Marco, Bernini, Franco, Mussi, Chiara, and Zimetti, Francesca
- Subjects
- *
APOLIPOPROTEIN E4 , *BLOOD lipids , *APOLIPOPROTEIN E , *ALZHEIMER'S patients , *MILD cognitive impairment - Abstract
Background: Alterations in factors involved in cholesterol homeostasis are critical in Alzheimer's disease (AD), but the stage of occurrence, their specific association, and a possible relationship with the APOE4 genotype are not clarified. Objective: We aimed to quantify and correlate specific lipid factors in patients with different degrees of cognitive decline, namely patients with AD and patients with mild cognitive impairment due to AD (MCI-AD), carriers or non-carriers of the APOE4 genotype. Methods: We evaluated Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9), cholesterol and the oxidative metabolites 24-, 25-, 27-hydroxycholesterol (HC) in the cerebrospinal fluid (CSF) and serum of AD (n = 28) and MCI-AD (n = 27) patients. Results: CSF and serum PCSK9 and lipids were similar, except for higher serum PCSK9 and triglycerides in MCI-AD compared to AD. In CSF, AD APOE4 carriers showed higher PCSK9 and 24-HC (+61.3%, p = 0.027 and +32.7%, p = 0.037), compared to non-carriers. There was a negative association between CSF PCSK9 and 27-HC in AD (r = −0.444, p = 0.049) and, exclusively among AD APOE4 carriers, a negative association between CSF PCSK9 and 24-HC (r = −0.786, p = 0.028). A positive correlation was observed between CSF and serum PCSK9 in AD (r = 0.520, p = 0.004), driven by APOE4 carriers (r = 0.544, p = 0.038), suggesting PCSK9 exchange between brain and periphery. A positive correlation was detected between serum and CSF 27-HC (r = 0.465, p = 0.039) in AD. None of these results were found in MCI-AD patients. Conclusions: PCSK9 and 24-HC might be specific markers of ApoE4-associated lipid alterations in AD, possibly contributing to clinical progression in the AD continuum. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Added value of inflammatory plasma biomarkers to pathologic biomarkers in predicting preclinical Alzheimer's disease.
- Author
-
Leclerc, Haley, Lee, Athene KW, Kunicki, Zachary J, and Alber, Jessica
- Subjects
- *
ALZHEIMER'S disease , *OLDER people , *APOLIPOPROTEIN E4 , *RANDOM forest algorithms , *TAU proteins , *BIOMARKERS - Abstract
Background: Plasma biomarkers have recently emerged for the diagnosis, assessment, and disease monitoring of Alzheimer's disease (AD), but have yet to be fully validated in preclinical AD. In addition to AD pathologic plasma biomarkers (amyloid-β (Aβ) and phosphorylated tau (p-tau) species), a proteomic panel can discriminate between symptomatic AD and cognitively unimpaired older adults in a dementia clinic population. Objective: Examine the added value of a plasma proteomic panel, validated in symptomatic AD, over standard AD pathologic plasma biomarkers and demographic and genetic (apolipoprotein (APOE) ɛ4 status) risk factors in detecting preclinical AD. Methods: 125 cognitively unimpaired older adults (mean age = 66 years) who completed Aβ PET and plasma draw were analyzed using multiple regression with Aβ PET status (positive versus negative) as the outcome to determine the best fit for predicting preclinical AD. Model 1 included age, education, and gender. Model 2 and 3 added predictors APOE ɛ4 status (carrier versus non-carrier) and AD pathologic blood biomarkers (Aβ42/40 ratio, p-tau181), respectively. Random forest modeling established the 5 proteomic markers from the proteomic panel that best predicted Aβ PET status, and these markers were added in Model 4. Results: The best model for predicting Aβ PET status included age, years of education, APOE ɛ4 status, Aβ42/40 ratio, and p-tau181. Adding the top 5 proteomic markers did not significantly improve the model. Conclusions: Proteomic markers in plasma did not add predictive value to standard AD pathologic plasma biomarkers in predicting preclinical AD in this sample. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Therapeutic Challenges Derived from the Interaction Among Apolipoprotein E, Cholesterol, and Amyloid in Alzheimer's Disease.
- Author
-
Menendez-Gonzalez, Manuel
- Subjects
- *
ALZHEIMER'S disease , *CHOLESTEROL metabolism , *APOLIPOPROTEIN E , *APOLIPOPROTEIN E4 , *AMYLOID plaque , *TAU proteins - Abstract
The isoform E4 of the Apolipoprotein E (ApoE) represents one of the strongest genetic risk factors for late-onset Alzheimer's disease (AD). ApoE has key roles in cholesterol transport and amyloid-β (Aβ) metabolism, which are both central to AD pathogenesis. The E4 isoform has been implicated in reduced cholesterol homeostasis, increased Aβ aggregation, and heightened tau phosphorylation, contributing to amyloid plaques and neurodegeneration. This manuscript examines the complex interactions among ApoE isoforms, cholesterol metabolism, and amyloid pathology. Moreover, the therapeutic challenges associated with lipid-lowering agents (e.g., statins, PCSK9 inhibitors), anti-amyloid immunotherapies, and anticoagulants are described, focusing on ApoE4 carriers. Decision-making challenges are discussed by analyzing the pros and cons of these therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. Apolipoprotein E aggregation in microglia initiates Alzheimer's disease pathology by seeding β-amyloidosis.
- Author
-
Kaji, Seiji, Berghoff, Stefan A., Spieth, Lena, Schlaphoff, Lennart, Sasmita, Andrew O., Vitale, Simona, Büschgens, Luca, Kedia, Shreeya, Zirngibl, Martin, Nazarenko, Taisiia, Damkou, Alkmini, Hosang, Leon, Depp, Constanze, Kamp, Frits, Scholz, Patricia, Ewers, David, Giera, Martin, Ischebeck, Till, Wurst, Wolfgang, and Wefers, Benedikt
- Subjects
- *
ALZHEIMER'S disease , *LIPID metabolism , *PATHOLOGY , *CHOLESTEROL metabolism , *MICROGLIA , *APOLIPOPROTEIN E4 - Abstract
The seeded growth of pathogenic protein aggregates underlies the pathogenesis of Alzheimer's disease (AD), but how this pathological cascade is initiated is not fully understood. Sporadic AD is linked genetically to apolipoprotein E (APOE) and other genes expressed in microglia related to immune, lipid, and endocytic functions. We generated a transgenic knockin mouse expressing HaloTag-tagged APOE and optimized experimental protocols for the biochemical purification of APOE, which enabled us to identify fibrillary aggregates of APOE in mice with amyloid-β (Aβ) amyloidosis and in human AD brain autopsies. These APOE aggregates that stained positive for β sheet-binding dyes triggered Aβ amyloidosis within the endo-lysosomal system of microglia, in a process influenced by microglial lipid metabolism and the JAK/STAT signaling pathway. Taking these observations together, we propose a model for the onset of Aβ amyloidosis in AD, suggesting that the endocytic uptake and aggregation of APOE by microglia can initiate Aβ plaque formation. [Display omitted] • Apolipoprotein E aggregates can serve as seeds for Aβ plaque pathology • APOE aggregation occurs within the endo-lysosomal system of microglia • APOE internalization is regulated by cholesterol metabolism in microglia • Interferon-activated microglia facilitate the aggregation process Pathogenic protein aggregation drives Alzheimer's disease, but how it is initiated remains unclear. By generating a APOE-HaloTag knockin mouse model and optimizing APOE purification techniques, Kaji et al. identify fibrillary APOE aggregates that can serve as seeds for Aβ plaque formation. This aggregation occurs within the endo-lysosomal system of microglia and is regulated by cholesterol metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. Hormone Replacement Therapy and Alzheimer's Disease: Current State of Knowledge and Implications for Clinical Use.
- Author
-
Sayfullaeva, Jessica, McLoughlin, John, and Kwakowsky, Andrea
- Subjects
- *
HORMONE therapy , *ALZHEIMER'S disease , *ESTROGEN replacement therapy , *DISEASE risk factors , *NEURODEGENERATION , *APOLIPOPROTEIN E4 - Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder responsible for over half of dementia cases, with two-thirds being women. Growing evidence from preclinical and clinical studies underscores the significance of sex-specific biological mechanisms in shaping AD risk. While older age is the greatest risk factor for AD, other distinct biological mechanisms increase the risk and progression of AD in women including sex hormones, brain structural differences, genetic background, immunomodulation and vascular disorders. Research indicates a correlation between declining estrogen levels during menopause and an increased risk of developing AD, highlighting a possible link with AD pathogenesis. The neuroprotective effects of estrogen vary with the age of treatment initiation, menopause stage, and type. This review assesses clinical and observational studies conducted in women, examining the influence of estrogen on cognitive function or addressing the ongoing question regarding the potential use of hormone replacement therapy (HRT) as a preventive or therapeutic option for AD. This review covers recent literature and discusses the working hypothesis, current use, controversies and challenges regarding HRT in preventing and treating age-related cognitive decline and AD. The available evidence indicates that estrogen plays a significant role in influencing dementia risk, with studies demonstrating both beneficial and detrimental effects of HRT. Recommendations regarding HRT usage should carefully consider the age when the hormonal supplementation is initiated, baseline characteristics such as genotype and cardiovascular health, and treatment duration until this approach can be more thoroughly investigated or progress in the development of alternative treatments can be made. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. The Relationship Between Physical Activity and Non-Modifiable Risk Factors on Alzheimer's Disease and Brain Health Markers: A UK Biobank Study.
- Author
-
Spencer, Felicity S.E., Elsworthy, Richard J., Breen, Leigh, Bishop, Jonathan, Morrissey, Sol, and Aldred, Sarah
- Subjects
- *
COGNITIVE processing speed , *DISEASE risk factors , *PHYSICAL activity , *APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E - Abstract
Background: Modifiable (physical activity) and non-modifiable (sex and genotype) risk factors interact to affect Alzheimer's disease (AD) risk. Further investigation is necessary to understand if these factors influence brain volume and cognition. Objective: The study aimed to assess the effect of physical activity, APOE genotype, and sex on AD risk, brain volume, and cognition. Methods: UK Biobank data from 2006 to 2023 was accessed. Physical activity was measured by accelerometers, and International Physical Activity Questionnaire. Outcomes were AD incidence; brain volume (ventricular cerebrospinal fluid and total brain); and cognition (executive function, memory, visuospatial ability, processing speed, and reaction time). Logistic and linear regression models were conducted. Results: 69,060 participants met inclusion criteria (mean age: 62.28 years, SD: 7.84; 54.64% female). Higher self-reported (OR = 0.63, 95% CI [0.40, 1.00], p = 0.047) and accelerometer-assessed (OR = 0.96 [0.93, 0.98], p = 0.002) physical activity was associated with lower disease incidence. Smaller ventricular cerebrospinal fluid volume (β= – 65.43 [– 109.68, – 17.40], p = 0.007), and larger total brain volume (β= 4398.46 [165.11, 8631.82], p < 0.001) was associated with increased accelerometer-assessed and self-reported physical activity respectively. Both brain volume analyses were moderated by sex. Increased accelerometer-assessed physical activity levels were associated with faster reaction time (β= – 0.43 [– 0.68, – 0.18], p = 0.001); though poorer visuospatial ability (β= – 0.06 [– 0.09, – 0.03], p < 0.001), and executive function (β= 0.49 [0.31, 0.66], p < 0.001; β= 0.27 [0.10, 0.45], p = 0.002) was related to self-reported physical activity levels. Conclusions: Higher levels of physical activity reduce AD risk independently of non-modifiable risk factors. Moderation of sex on brain volume highlighted the importance of incorporating non-modifiable risk factors in analysis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Contrasting association pattern of plasma low-density lipoprotein with white matter integrity in APOE4 carriers versus non-carriers.
- Author
-
Ye, Zhenyao, Pan, Yezhi, McCoy, Rozalina G., Bi, Chuan, Mo, Chen, Feng, Li, Yu, Jiaao, Lu, Tong, Liu, Song, Carson Smith, J., Duan, Minxi, Gao, Si, Ma, Yizhou, Chen, Chixiang, Mitchell, Braxton D., Thompson, Paul M., Elliot Hong, L., Kochunov, Peter, Ma, Tianzhou, and Chen, Shuo
- Subjects
- *
DIFFUSION magnetic resonance imaging , *NUCLEAR magnetic resonance spectroscopy , *DISEASE risk factors , *APOLIPOPROTEIN E4 , *APOLIPOPROTEIN E - Abstract
Apolipoprotein E ε4 (APOE4) is a strong genetic risk factor of Alzheimer's disease and metabolic dysfunction. However, whether APOE4 and markers of metabolic dysfunction synergistically impact the deterioration of white matter (WM) integrity in older adults remains unknown. In the UK Biobank data, we conducted a multivariate analysis to investigate the interactions between APOE4 and 249 plasma metabolites (measured using nuclear magnetic resonance spectroscopy) with whole-brain WM integrity (measured by diffusion-weighted magnetic resonance imaging) in a cohort of 1917 older adults (aged 65.0–81.0 years; 52.4 % female). Although no main association was observed between either APOE4 or metabolites with WM integrity (adjusted P > 0.05), significant interactions between APOE4 and metabolites with WM integrity were identified. Among the examined metabolites, higher concentrations of low-density lipoprotein and very low-density lipoprotein were associated with a lower level of WM integrity (b= − 0.12 , CI= − 0.14 , − 0.10 ) among APOE4 carriers. Conversely, among non-carriers, they were associated with a higher level of WM integrity (b=0.05, CI= 0.04,0.07 ), demonstrating a significant moderation role of APOE4 (b = − 0.18 , CI= − 0.20 , − 0.15 , P<0.00001). • LDL is negatively associated with white matter integrity in older APOE4 carriers. • This association, although weak, is positive in APOE4 non-carriers. • APOE4 carriers with high LDL show a stronger negative age-cognition correlation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Apolipoprotein E Induces Lipid Accumulation Through Dgat2 That Is Prevented with Time-Restricted Feeding in Drosophila.
- Author
-
Moraes, Ruan C. M., Roth, Jonathan R., Mao, Hailey, Crawley, Savannah R., Xu, Brittney P., Watson, John C., and Melkani, Girish C.
- Subjects
- *
LIPID metabolism , *APOLIPOPROTEIN E , *ALZHEIMER'S disease , *APOLIPOPROTEIN E4 , *LIPIDS - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. 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
45. Exercise as medicine in Parkinson's disease.
- Author
-
Langeskov-Christensen, Martin, Franzén, Erika, Hvid, Lars Grøndahl, and Dalgas, Ulrik
- Subjects
EXERCISE physiology ,CENTRAL nervous system diseases ,ALZHEIMER'S disease ,RAPID eye movement sleep ,PHYSICAL activity ,TREMOR ,APOLIPOPROTEIN E4 - Published
- 2024
- Full Text
- View/download PDF
46. The influences of ApoE isoforms on endothelial adherens junctions and actin cytoskeleton responding to mCRP.
- Author
-
Zhang, Zhengrong, Lin, Weiwei, Gan, Qini, Lei, Maohua, Gong, Bin, Zhang, Chao, Henrique, Jessica Salles, Han, Jingyan, Tian, Hua, Tao, Qiushan, Potempa, Lawrence A., Stein, Thor D., Emili, Andrew, and Qiu, Wei Qiao
- Subjects
RECOMBINANT proteins ,APOLIPOPROTEIN E4 ,ADHERENS junctions ,APOLIPOPROTEIN E ,CYTOSKELETON ,CYTOSKELETAL proteins - Abstract
Apolipoprotein E4 (ApoE4) plays an important role responding to monomeric C-reactive protein (mCRP) via binding to CD31 leading to cerebrovascular damage and Alzheimer's disease (AD). Using phosphor-proteomic profiling, we found altered cytoskeleton proteins in the microvasculature of AD brains, including increased levels of hyperphosphorylated tau (pTau) and the actin-related protein, LIMA1. To address the hypothesis that cytoskeletal changes serve as early pathological signatures linked with CD31 in brain endothelia in ApoE4 carriers, ApoE4 knock-in mice intraperitoneal injected with mCRP revealed that mCRP increased the expressions of phosphorylated CD31 (pCD31) and LIMA1, and facilitate the binding of pCD31 to LIMA1. mCRP combined with recombinant APOE4 protein decreased interaction of CD31 and VE-Cadherin at adherens junctions (AJs), along with altered the expression of various actin cytoskeleton proteins, causing microvasculature damage. Notably, the APOE2 protein attenuated these changes. Overall, our study demonstrates that ApoE4 responds to mCRP to disrupt the endothelial AJs which link with the actin cytoskeleton and this pathway could play a key role in the barrier dysfunction leading to AD risk. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. 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 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. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Network dynamics-based subtyping of Alzheimer's disease with microglial genetic risk factors.
- Author
-
Choi, Jae Hyuk, Lee, Jonghoon, Kang, Uiryong, Chang, Hongjun, and Cho, Kwang-Hyun
- Subjects
- *
DISEASE risk factors , *ALZHEIMER'S disease , *SYSTEMS biology , *RNA sequencing , *DRUG target , *APOLIPOPROTEIN E4 - Abstract
Background: The potential of microglia as a target for Alzheimer's disease (AD) treatment is promising, yet the clinical and pathological diversity within microglia, driven by genetic factors, poses a significant challenge. Subtyping AD is imperative to enable precise and effective treatment strategies. However, existing subtyping methods fail to comprehensively address the intricate complexities of AD pathogenesis, particularly concerning genetic risk factors. To address this gap, we have employed systems biology approaches for AD subtyping and identified potential therapeutic targets. Methods: We constructed patient-specific microglial molecular regulatory network models by utilizing existing literature and single-cell RNA sequencing data. The combination of large-scale computer simulations and dynamic network analysis enabled us to subtype AD patients according to their distinct molecular regulatory mechanisms. For each identified subtype, we suggested optimal targets for effective AD treatment. Results: To investigate heterogeneity in AD and identify potential therapeutic targets, we constructed a microglia molecular regulatory network model. The network model incorporated 20 known risk factors and crucial signaling pathways associated with microglial functionality, such as inflammation, anti-inflammation, phagocytosis, and autophagy. Probabilistic simulations with patient-specific genomic data and subsequent dynamics analysis revealed nine distinct AD subtypes characterized by core feedback mechanisms involving SPI1, CASS4, and MEF2C. Moreover, we identified PICALM, MEF2C, and LAT2 as common therapeutic targets among several subtypes. Furthermore, we clarified the reasons for the previous contradictory experimental results that suggested both the activation and inhibition of AKT or INPP5D could activate AD through dynamic analysis. This highlights the multifaceted nature of microglial network regulation. Conclusions: These results offer a means to classify AD patients by their genetic risk factors, clarify inconsistent experimental findings, and advance the development of treatments tailored to individual genotypes for AD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta.
- Author
-
Ding, Yunfeng, Palecek, Sean P., and Shusta, Eric V.
- Subjects
- *
BLOOD-brain barrier disorders , *RECOMBINANT proteins , *PLURIPOTENT stem cells , *ALZHEIMER'S disease , *APOLIPOPROTEIN E , *BLOOD-brain barrier , *APOLIPOPROTEIN E4 - Abstract
Background: Three common isoforms of the apolipoprotein E (APOE) gene - APOE2, APOE3, and APOE4 - hold varying significance in Alzheimer's Disease (AD) risk. The APOE4 allele is the strongest known genetic risk factor for late-onset Alzheimer's Disease (AD), and its expression has been shown to correlate with increased central nervous system (CNS) amyloid deposition and accelerated neurodegeneration. Conversely, APOE2 is associated with reduced AD risk and lower CNS amyloid burden. Recent clinical data have suggested that increased blood-brain barrier (BBB) leakage is commonly observed among AD patients and APOE4 carriers. However, it remains unclear how different APOE isoforms may impact AD-related pathologies at the BBB. Methods: To explore potential impacts of APOE genotypes on BBB properties and BBB interactions with amyloid beta, we differentiated isogenic human induced pluripotent stem cell (iPSC) lines with different APOE genotypes into both brain microvascular endothelial cell-like cells (BMEC-like cells) and brain pericyte-like cells. We then compared the effect of different APOE isoforms on BBB-related and AD-related phenotypes. Statistical significance was determined via ANOVA with Tukey's post hoc testing as appropriate. Results: Isogenic BMEC-like cells with different APOE genotypes had similar trans-endothelial electrical resistance, tight junction integrity and efflux transporter gene expression. However, recombinant APOE4 protein significantly impeded the "brain-to-blood" amyloid beta 1–40 (Aβ40) transport capabilities of BMEC-like cells, suggesting a role in diminished amyloid clearance. Conversely, APOE2 increased amyloid beta 1–42 (Aβ42) transport in the model. Furthermore, we demonstrated that APOE-mediated amyloid transport by BMEC-like cells is dependent on LRP1 and p-glycoprotein pathways, mirroring in vivo findings. Pericyte-like cells exhibited similar APOE secretion levels across genotypes, yet APOE4 pericyte-like cells showed heightened extracellular amyloid deposition, while APOE2 pericyte-like cells displayed the least amyloid deposition, an observation in line with vascular pathologies in AD patients. Conclusions: While APOE genotype did not directly impact general BMEC or pericyte properties, APOE4 exacerbated amyloid clearance and deposition at the model BBB. Conversely, APOE2 demonstrated a potentially protective role by increasing amyloid transport and decreasing deposition. Our findings highlight that iPSC-derived BBB models can potentially capture amyloid pathologies at the BBB, motivating further development of such in vitro models in AD modeling and drug development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. Introduction.
- Author
-
Colonna, Marco
- Subjects
- *
DISEASE risk factors , *PATHOLOGY , *PATHOLOGICAL physiology , *TAU proteins , *MATERNAL immune activation , *PSYCHONEUROIMMUNOLOGY , *CEREBRAL amyloid angiopathy , *APOLIPOPROTEIN E4 - 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.