1
|
Hu H, Wan S, Hu Y, Wang Q, Li H, Zhang N. Deciphering the role of APOE in cerebral amyloid angiopathy: from genetic insights to therapeutic horizons. Ann Med 2025; 57:2445194. [PMID: 39745195 DOI: 10.1080/07853890.2024.2445194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid-β (Aβ) peptides in the walls of medium and small vessels of the brain and leptomeninges, is a major cause of lobar hemorrhage in elderly individuals. Among the genetic risk factors for CAA that continue to be recognized, the apolipoprotein E (APOE) gene is the most significant and prevalent, as its variants have been implicated in more than half of all patients with CAA. While the presence of the APOE ε4 allele markedly increases the risk of CAA, the ε2 allele confers a protective effect relative to the common ε3 allele. These allelic variants encode three APOE isoforms that differ at two amino acid positions. The primary physiological role of APOE is to mediate lipid transport in the brain and periphery; however, it has also been shown to be involved in a wide array of biological functions, particularly those involving Aβ, in which it plays a known role in processing, production, aggregation, and clearance. The challenges posed by the reliance on postmortem histological analyses and the current absence of an effective intervention underscore the urgency for innovative APOE-targeted strategies for diagnosing CAA. This review not only deepens our understanding of the impact of APOE on the pathogenesis of CAA but can also help guide the exploration of targeted therapies, inspiring further research into the therapeutic potential of APOE.
Collapse
Affiliation(s)
- Hantian Hu
- Tianjin Medical University, Tianjin, China
| | - Siqi Wan
- Tianjin Medical University, Tianjin, China
| | - Yuetao Hu
- Tianjin Medical University, Tianjin, China
| | - Qi Wang
- Tianjin Medical University, Tianjin, China
| | - Hanyu Li
- Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Lucassen PJ, Korosi A, de Rooij SR, Smit AB, Van Dam AM, Daskalakis NP, Van Kesteren RE, Verheijen MHG, Lesuis SL, Kessels HW, Krugers HJ. How Can Early Stress Influence Later Alzheimer's Disease Risk? Possible Mediators and Underlying Mechanisms. Biol Psychiatry 2025; 97:372-381. [PMID: 39577793 DOI: 10.1016/j.biopsych.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Alzheimer's disease (AD) is a progressive, age-related neurodegenerative disorder to which genetic mutations and risk factors contribute. Evidence is increasing that environmental and lifestyle-related factors, such as exercise, nutrition, education, and exposure to (early-life) stress modify the onset, incidence, and progression of AD. Here, we discuss recent preclinical findings on putative substrates that can explain or contribute to the effects of stress early in life on the risk of developing AD. We focus in particular on stress hormones, neural networks, synapses, mitochondria, nutrient and lipid metabolism, adult neurogenesis, engram cell ensembles, and neuroinflammation. We discuss the idea that stress exposure early in life can alter these processes, either combined or in isolation, thereby reducing the capacity of the brain to resist deleterious consequences of, for example, amyloid-β accumulation, thereby accelerating cognitive decline and progression of Alzheimer-related changes in model systems of the disease. A better understanding of whether experiences early in life also modify trajectories of cognitive decline and pathology in AD and how the substrates discussed translate to humans may help develop novel preventive and/or therapeutic strategies to mitigate the consequences of stressors early in life and increase resilience to developing dementia.
Collapse
Affiliation(s)
- Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.
| | - Aniko Korosi
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anne-Marie Van Dam
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Nikolaos P Daskalakis
- Neurogenomics and Translational Bioinformatics Laboratory, McLean Hospital, Harvard University, Boston, Massachusetts
| | - Ronald E Van Kesteren
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Centre for Neurogenomics and Cognitive Research, Department of Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sylvie L Lesuis
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Helmut W Kessels
- Department of Cellular & Computational Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
3
|
Kim J, Ji E, Bae JB, Han JW, Kim TH, Kwak KP, Kim BJ, Kim SG, Kim JL, Moon SW, Park JH, Ryu SH, Youn JC, Lee DY, Lee DW, Lee SB, Lee JJ, Jhoo JH, Song J, Lee K, Kim KW. Vitamin D deficiency may accelerate cognitive decline in female apolipoprotein E ε4 non-carriers. Clin Nutr 2025; 45:167-173. [PMID: 39826240 DOI: 10.1016/j.clnu.2024.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND & AIMS The impact of vitamin D deficiency (VDD) on cognition remains controversial. Evidences suggest that variability based on apolipoprotein E (APOE) ε4 status and gender, given APOE ε4's influence on vitamin D metabolism and women's heightened vitamin D sensitivity. We investigated the interplay between APOE ε4, gender, and VDD in cognitive decline among older adults. METHODS In a population-based cohort of 1547 cognitively normal Koreans aged ≥60 years, Mini Mental State Examination (MMSE) changes were tracked biennially (2010-2020). VDD was defined as serum 25-hydroxyvitamin D < 10 ng/mL. Linear mixed models analyzed VDD effects, with subgroup analyses for APOE ε4 status and gender. RESULTS VDD was present in 21.3 % at baseline and was linked to faster MMSE decline (estimate = -0.054, 95 % CI [-0.091, -0.017], p = 0.004), particularly in APOE ε4 non-carriers (estimate = -0.070, 95 % CI [-0.112, -0.029], p = 0.001). A gender-based analysis revealed that this effect was significant only in female non-carriers (estimate = -0.097, 95 % CI [-0.156, -0.038], p = 0.001). Conversely, male non-carriers demonstrated an absence of a statistically significant association (estimate = -0.017, 95 % CI [-0.076, 0.041], p = 0.562). CONCLUSIONS VDD accelerates cognitive decline in cognitively normal APOE ε4 non-carriers, particularly women, underscoring the importance of tailored prevention strategies.
Collapse
Affiliation(s)
- Jiwon Kim
- Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea
| | - Eunjeong Ji
- Medical Research Collaborating Center, Seoul National University Bundang Hospital, Republic of Korea
| | - Jong Bin Bae
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Tae Hui Kim
- Department of Psychiatry, Yonsei University Wonju Severance Christian Hospital, Wonju, Republic of Korea
| | - Kyung Phil Kwak
- Department of Psychiatry, Dongguk University Gyeongju Hospital, Gyeongju, Republic of Korea
| | - Bong Jo Kim
- Department of Psychiatry, Gyeongsang National University School of Medicine, Jinju, Republic of Korea
| | - Shin Gyeom Kim
- Department of Neuropsychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Jeong Lan Kim
- Department of Psychiatry, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seok Woo Moon
- Department of Psychiatry, School of Medicine, Konkuk University, Konkuk University Chungju Hospital, Chungju, Republic of Korea
| | - Joon Hyuk Park
- Department of Neuropsychiatry, Jeju National University Hospital, Jeju, Republic of Korea
| | - Seung-Ho Ryu
- Department of Psychiatry, School of Medicine, Konkuk University, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Jong Chul Youn
- Department of Neuropsychiatry, Kyunggi Provincial Hospital for the Elderly, Yongin, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Psychiatry, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Dong Woo Lee
- Department of Neuropsychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Seok Bum Lee
- Department of Psychiatry, Dankook University Hospital, Cheonan, Republic of Korea
| | - Jung Jae Lee
- Department of Psychiatry, Dankook University Hospital, Cheonan, Republic of Korea
| | - Jin Hyeong Jhoo
- Department of Psychiatry, Kangwon National University, School of Medicine, Chuncheon, Republic of Korea
| | - Junghan Song
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Kyunghoon Lee
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ki Woong Kim
- Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Psychiatry, Seoul National University, College of Medicine, Seoul, Republic of Korea; Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Elhefnawy ME, Patson N, Mouksassi S, Pillai G, Shcherbinin S, Chigutsa E, Gueorguieva I. Quantifying natural amyloid plaque accumulation in the continuum of Alzheimer's disease using ADNI. J Pharmacokinet Pharmacodyn 2025; 52:15. [PMID: 39862333 DOI: 10.1007/s10928-024-09959-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Brain amyloid beta neuritic plaque accumulation is associated with an increased risk of progression to Alzheimer's disease (AD) [Pfeil, J., et al. in Neurobiol Aging 106: 119-129, 2021]. Several studies estimate rates of change in amyloid plaque over time in clinically heterogeneous cohorts with different factors impacting amyloid plaque accumulation from ADNI and AIBL [Laccarino, L., et al. in Annals Clin and Trans Neurol 6: 1113 1120, 2019, Vos, S.J., et al. in Brain 138: 1327-1338, 2015, Lim, Y.Y., et al. in Alzheimer's Dementia 9: 538-545, 2013], but there are no reports using non-linear mixed effect model for amyloid plaque progression over time similar to that existing of disease-modifying biomarkers for other diseases [Cook, S.F. and R.R. Bies in Current Pharmacol Rep 2: 221-230, 2016, Gueorguieva, I., et al. in Alzheimer's Dementia 19: 2253-2264, 2023]. This study describes the natural progression of amyloid accumulation with population mean and between-participant variability for baseline and intrinsic progression rates quantified across the AD spectrum. 1340 ADNI participants were followed over a 10-year period with 18F-florbetapir PET scans used for amyloid plaque detection. Non-linear mixed effect with stepwise covariate modelling (scm) was used. Change in natural amyloid plaque levels over 10 year period followed an exponential growth model with an intrinsic rate of approx. 3 centiloid units/year. Age, gender, APOE4 genotype and disease stage were important factors on the baseline in the natural amyloid model. In APOE4 homozygous carriers mean baseline amyloid was increased compared to APOE4 non carriers. These results demonstrate natural progression of amyloid plaque in the continuum of AD.
Collapse
Affiliation(s)
- Marwa E Elhefnawy
- Applied Pharmacometrics Training-Africa Program, c/o Pharmacometrics Africa NPC, Cape Town, South Africa
- Pumas-AI, Inc, Dover, Delaware, USA
| | - Noel Patson
- Applied Pharmacometrics Training-Africa Program, c/o Pharmacometrics Africa NPC, Cape Town, South Africa
- School of Global and Public Health, Kamuzu University of Health Sciences, Chichiri BT3, Blantyre, Malawi
| | - Samer Mouksassi
- Applied Pharmacometrics Training-Africa Program, c/o Pharmacometrics Africa NPC, Cape Town, South Africa
- Integrated Drug Development, Certara, 100 Overlook Ctr Site 101, Princeton, NJ, USA
| | - Goonaseelan Pillai
- Applied Pharmacometrics Training-Africa Program, c/o Pharmacometrics Africa NPC, Cape Town, South Africa
- Division of Clinical Pharmacology, University of Cape Town, Rondebosch, 7701, South Africa
| | - Sergey Shcherbinin
- Eli Lilly and Company, 16 893 South Delaware Street, Indianapolis, IN, USA
| | - Emmanuel Chigutsa
- Eli Lilly and Company, 16 893 South Delaware Street, Indianapolis, IN, USA
| | - Ivelina Gueorguieva
- Global PK/PD/PMx, Eli Lilly and Company, 8 Arlington Square West, Downshire Way, Bracknell, Berkshire, RG12 1PU, UK.
| |
Collapse
|
5
|
Ramsden CE, Cutler RG, Li X, Keyes GS. Lipid-protecting disulfide bridges are the missing molecular link between ApoE4 and sporadic Alzheimer's disease in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633633. [PMID: 39868210 PMCID: PMC11761642 DOI: 10.1101/2025.01.17.633633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
As the principal lipid transporter in the human brain, apolipoprotein E (ApoE) is tasked with the transport and protection of highly vulnerable lipids required to support and remodel neuronal membranes, in a process that is dependent on ApoE receptors. Human APOE allele variants that encode proteins differing only in the number of cysteine (Cys)-to-arginine (Arg) exchanges (ApoE2 [2 Cys], ApoE3 [1 Cys], ApoE4 [0 Cys]) comprise the strongest genetic risk factor for sporadic Alzheimer's disease (AD); however, the specific molecular feature(s) and resultant mechanisms that underlie these isoform-dependent effects are unknown. One signature feature of Cys is the capacity to form disulfide (Cys-Cys) bridges, which are required to form disulfide bridge-linked dimers and multimers. Here we propose the overarching hypothesis that the super-ability (for ApoE2), intermediate ability (for ApoE3) or inability (for ApoE4) to form lipid-protecting intermolecular disulfide bridges, is the central molecular determinant accounting for the disparate effects of APOE alleles on AD risk and amyloid-β and Tau pathologies in humans. We posit that presence and abundance of Cys in human ApoE3 and ApoE2 respectively, conceal and protect vulnerable lipids transported by ApoE from peroxidation by enabling formation of ApoE homo-dimers/multimers and heteromeric ApoE complexes such as ApoE-ApoJ and ApoE-ApoD. We thus propose that the inability to form intermolecular disulfide bridges makes ApoE4-containing lipoproteins uniquely vulnerable to peroxidation and its downstream consequences. Consistent with our model, we found that brain-enriched polyunsaturated fatty acid-containing phospholipids induce disulfide-dependent dimerization and multimerization of ApoE3 and ApoE2 (but not ApoE4). By contrast, incubation with the peroxidation-resistant lipid DMPC or cholesterol alone had minimal effects on dimerization. These novel concepts and findings are integrated into our unifying model implicating peroxidation of ApoE-containing lipoproteins, with consequent ApoE receptor-ligand disruption, as the initiating molecular events that ultimately lead to AD in humans. Highlights APOE alleles are the strongest genetic risk factor for sporadic Alzheimer's disease (AD) APOE alleles encode proteins that differ only in the number of Cys⟶Arg exchanges Despite 30 years of inquiry, mechanisms linking Cys⟶Arg exchanges to AD remain unknownPUFA-phospholipids induced disulfide bridge formation in ApoE3 and ApoE2 (but not ApoE4)We hypothesize that disulfide bridges in ApoE protect vulnerable lipids from peroxidation We propose that lipid-protecting disulfide bridges explain APOE allele-dependent AD risks.
Collapse
|
6
|
Rajmohan R, Al-Darsani Z, Ho CC, Wong J, Paganini-Hill A, Montine T, Corrada M, Kawas C. Vascular risk factors and cerebrovascular pathologic changes on autopsy: The 90+ Study. Alzheimers Dement 2025. [PMID: 39777966 DOI: 10.1002/alz.14454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Cerebrovascular pathologic changes (CVPC) are prevalent and associated with dementia in those ≥ 90 years. However, CVPC associations to traditional risk factors (hypertension, diabetes, and hyperlipidemia) are variable. We hypothesized that neither traditional risk factors nor related medications would be associated with CVPC presence. METHODS In autopsy volunteers from The 90+ Study, odds ratios (OR) of CVPC presence to self-reported vascular risk factors and cardiovascular medication classes were calculated using logistic regressions adjusted for age of death, sex, and education. RESULTS In 267 participants (mean age at death 98 (±3.5) years; 75% female), CVPC presence was not related to traditional risk factors. Lower odds of atherosclerosis with diuretics [OR 0.55] and lower odds of cerebral amyloid angiopathy (CAA) with B-blocker [OR 0.57] or vasodilator [OR 0.40] use were observed. DISCUSSION Our findings suggest that vascular risk factors are not risk factors for CVPC at this age, medications have mitigated risks, or survival bias obscures associations. HIGHLIGHTS Cardiovascular risk factors are thought to contribute to cerebrovascular diseases. Risk factors were generally not associated with cerebrovascular changes. Lower odds of certain cerebrovascular changes were associated with antihypertensive use.
Collapse
Affiliation(s)
- Ravi Rajmohan
- Department of Neurology, University of California, Irvine, California, USA
| | - Zeinah Al-Darsani
- Department of Epidemiology and Biostatistics, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| | - Chu-Ching Ho
- Institute for Memory Impairments and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Joey Wong
- School of Medicine, University of California, Irvine, California, USA
| | - Annlia Paganini-Hill
- Department of Neurology, University of California, Irvine, California, USA
- Institute for Memory Impairments and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Thomas Montine
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Maria Corrada
- Department of Neurology, University of California, Irvine, California, USA
- Institute for Memory Impairments and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
- School of Medicine, University of California, Irvine, California, USA
| | - Claudia Kawas
- Department of Neurology, University of California, Irvine, California, USA
- Institute for Memory Impairments and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
- School of Medicine, University of California, Irvine, California, USA
| |
Collapse
|
7
|
Wei DX, Chen Z. Current situation and challenge of exogenous 3-hydroxybutyrate derived from polyhydroxyalkanoates for elderly health: A review. Int J Biol Macromol 2025; 285:138328. [PMID: 39631604 DOI: 10.1016/j.ijbiomac.2024.138328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
3-Hydroxybutyrate (3HB), predominantly found in the liver, muscles, and brain, is the most important endogenous ketone body in humans. During prolonged fasting and starvation, 3HB can partially replace glucose to meet some of the body's energy needs. In recent years, the resurgence of the ketogenic diet (KD) and systematic exploration of the favorable biocompatibility of polyhydroxyalkanoates (PHAs), the precursor polymer to 3HB, have led to numerous reports indicating that the benefits of exogenous 3HB supplementation outweigh the drawbacks, particularly for middle-aged and elderly individuals. In this review, the physicochemical properties, physiological functions, and biosafety of 3HB in the elderly population are introduced. The effects of exogenous 3HB supplements, including KD, ketone esters, 1,3-butanediol, and ketone salts, on the elderly are compared. It is proposed that exogenous 3HB derived from PHAs is optimal for this population. Exogenous 3HB supplementation for elderly health maintenance and disease treatment is summarized, with an outline of four challenges related to the mechanistic and clinical research of exogenous 3HB supplementation for elderly health. This aims to explore its novel therapeutic potential as a small molecule in the context of aging.
Collapse
Affiliation(s)
- Dai-Xu Wei
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, Sichuan 610081, China.
| | - Zongcun Chen
- Department of Endocrinology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, China.
| |
Collapse
|
8
|
Pei Y, Mou Z, Jiang L, Yang J, Gu Y, Min J, Sunzhang L, Xiong N, Xu X, Chi H, Xu K, Liu S, Luo H. Aging and head and neck cancer insights from single cell and spatial transcriptomic analyses. Discov Oncol 2024; 15:801. [PMID: 39692961 DOI: 10.1007/s12672-024-01672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma(HNSCC) is the sixth most common malignancy worldwide, with more than 890,000 new cases and 450,000 deaths annually. Its major risk factors include smoking, alcohol abuse, aging, and poor oral hygiene. Due to the lack of early and effective detection and screening methods, many patients are diagnosed at advanced stages with a five-year survival rate of less than 50%. In this study, we deeply explored the expression of Aging-related genes(ARGs) in HNSCC and analyzed their prognostic significance using single-cell sequencing and spatial transcriptomics analysis. This research aims to provide new theoretical support and directions for personalized treatment. Annually, more than 890,000 new cases of head and neck squamous cell carcinoma (HNSCC) are diagnosed globally, leading to 450,000 deaths, making it the sixth most common malignancy worldwide. The primary risk factors for HNSCC include smoking, alcohol abuse, aging, and poor oral hygiene. Many patients are diagnosed at advanced stages due to the absence of early and effective detection and screening methods, resulting in a five-year survival rate of less than 50%. In this research, single cell sequencing and spatial transcriptome analysis were used to investigate the expression of Aging-related genes (ARGs) in HNSCC and to analyse their prognostic significance. This research aims to provide new theoretical support and directions for personalized treatment. METHODS In this study, we investigated the association between HNSCC and AGRs by utilizing the GSE139324 series in the GEO database alongside the TCGA database, combined with single-cell sequencing and spatial transcriptomics analysis. The data were analyzed using Seurat and tSNE tools to reveal intercellular communication networks. For the spatial transcriptome data, SCTransform and RunPCA were applied to examine the metabolic activities of the cells. Gene expression differences were determined through spacerxr and RCTD tools, while the limma package was employed to identify differentially expressed genes and to predict recurrence rates using Cox regression analysis and column line plots. These findings underscore the potential importance of molecular classification, prognostic assessment, and personalized treatment of HNSCC. RESULTS This study utilized HNSCC single-cell sequencing data to highlight the significance of ARGs in the onset and prognosis of HNSCC. It revealed that the proportion of monocytes and macrophages increased, while the proportion of B cells decreased. Notably, high expression of the APOE gene in monocytes was closely associated with patient prognosis. Additionally, a Cox regression model was developed based on GSTP1 and age to provide personalized prediction tools for clinical use in predicting patient survival. CONCLUSIONS We utilized single-cell sequencing and spatial transcriptomics to explore the cellular characteristics of HNSCC and its interaction with the tumor microenvironment. Our findings reveal that HNSCC tissues show increased mononuclear cells and demonstrate enhanced activity in ARGs, thereby advancing our understanding of HNSCC development mechanisms.
Collapse
Affiliation(s)
- Yi Pei
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Zhuying Mou
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jinyan Yang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Yuheng Gu
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jie Min
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lingyi Sunzhang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Nan Xiong
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xiang Xu
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| | - Sinian Liu
- Department of Pathology, Xichong People's Hospital, Nanchong, 637200, China.
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| |
Collapse
|
9
|
Li T, Fili M, Mohammadiarvejeh P, Dawson A, Hu G, Willette AA. Associations of Coffee and Tea Consumption on Neural Network Connectivity: Unveiling the Role of Genetic Factors in Alzheimer's Disease Risk. Nutrients 2024; 16:4303. [PMID: 39770924 PMCID: PMC11677865 DOI: 10.3390/nu16244303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Coffee and tea are widely consumed beverages, but their long-term effects on cognitive function and aging remain largely unexplored. Lifestyle interventions, particularly dietary habits, offer promising strategies for enhancing cognitive performance and preventing cognitive decline. METHODS This study utilized data from the UK Biobank cohort (n = 12,025) to examine the associations between filtered coffee, green tea, and standard tea consumption and neural network functional connectivity across seven resting-state networks. We focused on networks spanning prefrontal and occipital areas that are linked to complex cognitive and behavioral functions. Linear mixed models were used to assess the main effects of coffee and tea consumption, as well as their interactions with Apolipoprotein E (APOE) genetic risk-the strongest genetic risk factor for Alzheimer's disease (AD). RESULTS Higher filtered coffee consumption was associated with increased functional connectivity in several networks, including Motor Execution, Sensorimotor, Fronto-Cingular, and a Prefrontal + 'What' Pathway Network. Similarly, greater green tea intake was associated with enhanced connectivity in the Extrastriate Visual and Primary Visual Networks. In contrast, higher standard tea consumption was linked to reduced connectivity in networks such as Memory Consolidation, Motor Execution, Fronto-Cingular, and the "What" Pathway + Prefrontal Network. The APOE4 genotype and family history of AD influenced the relationship between coffee intake and connectivity in the Memory Consolidation Network. Additionally, the APOE4 genotype modified the association between standard tea consumption and connectivity in the Sensorimotor Network. CONCLUSIONS The distinct patterns of association between coffee, green tea, and standard tea consumption and resting-state brain activity may provide insights into AD-related brain changes. The APOE4 genotype, in particular, appears to play a significant role in modulating these relationships. These findings enhance our knowledge of how commonly consumed beverages may influence cognitive function and potentially AD risk among older adults.
Collapse
Affiliation(s)
- Tianqi Li
- Genetics and Genomics Program, Iowa State University, Ames, IA 50011, USA;
| | - Mohammad Fili
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
| | - Parvin Mohammadiarvejeh
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA 50011, USA
| | - Alice Dawson
- Chestnut Health Systems, Lighthouse Institute, Chicago, IL 60610, USA;
| | - Guiping Hu
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
| | - Auriel A. Willette
- Department of Neurology, Rutgers University, New Brunswick, NJ 08854, USA
| |
Collapse
|
10
|
Negi M, Amulya E, Phatale V, Abraham N, Hedaoo A, Srinivasarao DA, Srivastava S. Surface engineered nano architectonics: An evolving paradigm for tackling Alzheimer's disease. Life Sci 2024; 358:123155. [PMID: 39433085 DOI: 10.1016/j.lfs.2024.123155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/21/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As per the World Health Organization (WHO) estimation, Alzheimer's disease (AD) will affect 100 million population across the globe by 2050. AD is an incurable neurodegenerative disease that remains a mystery for neurologists owing to its complex pathophysiology. Currently, available therapeutic regimens will only cause symptomatic relief by improving the cognitive and behavioral functions of AD. However, the major pitfalls in managing AD include tight junctions in the endothelial cells of the blood-brain barrier (BBB), diminished neuronal bioavailability, enzymatic degradation and reduced stability of the therapeutic moiety. In an effort to surmount the drawbacks mentioned above, researchers shifted their focus toward nanocarriers (NCs). Nevertheless, non-specific targeting of NCs imparts toxicity to the peripheral organs, thereby reducing the bioavailability of therapeutic moiety at the target site. To unravel this unmet clinical need, scientists came up with the idea of a novel intriguing strategy of surface engineering by targeting ligands. Surface-decorated NCs provide targeted drug delivery, controlled drug release, enhanced penetration and bioavailability. In this state-of-the-art review, we have highlighted in detail various molecular signalling pathways involved in AD pathogenesis. The significance of surface functionalization and its application in AD management have been deliberated. We have elaborated on the regulatory bottlenecks and clinical hurdles faced during lab-to-industrial scale translation along with possible solutions.
Collapse
Affiliation(s)
- Mansi Negi
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Etikala Amulya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Noella Abraham
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Aachal Hedaoo
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
11
|
Di Rienzo M, Romagnoli A, Refolo G, Vescovo T, Ciccosanti F, Zuchegna C, Lozzi F, Occhigrossi L, Piacentini M, Fimia GM. Role of AMBRA1 in mitophagy regulation: emerging evidence in aging-related diseases. Autophagy 2024; 20:2602-2615. [PMID: 39113560 PMCID: PMC11587829 DOI: 10.1080/15548627.2024.2389474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Aging is a gradual and irreversible physiological process that significantly increases the risks of developing a variety of pathologies, including neurodegenerative, cardiovascular, metabolic, musculoskeletal, and immune system diseases. Mitochondria are the energy-producing organelles, and their proper functioning is crucial for overall cellular health. Over time, mitochondrial function declines causing an increased release of harmful reactive oxygen species (ROS) and DNA, which leads to oxidative stress, inflammation and cellular damage, common features associated with various age-related pathologies. The impairment of mitophagy, the selective removal of damaged or dysfunctional mitochondria by autophagy, is relevant to the development and progression of age-related diseases. The molecular mechanisms that regulates mitophagy levels in aging remain largely uncharacterized. AMBRA1 is an intrinsically disordered scaffold protein with a unique property of regulating the activity of both proliferation and autophagy core machineries. While the role of AMBRA1 during embryonic development and neoplastic transformation has been extensively investigated, its functions in post-mitotic cells of adult tissues have been limited due to the embryonic lethality caused by AMBRA1 deficiency. Recently, a key role of AMBRA1 in selectively regulating mitophagy in post-mitotic cells has emerged. Here we summarize and discuss these results with the aim of providing a comprehensive view of the mitochondrial roles of AMBRA1, and how defective activity of AMBRA1 has been functionally linked to mitophagy alterations observed in age-related degenerative disorders, including muscular dystrophy/sarcopenia, Parkinson diseases, Alzheimer diseases and age-related macular degeneration.Abbreviations: AD: Alzheimer disease; AMD: age-related macular degeneration; AMBRA1: autophagy and beclin 1 regulator 1; APOE4: apolipoprotein E4; ATAD3A: ATPase family AAA domain containing 3A; ATG: autophagy related; BCL2: BCL2 apoptosis regulator; BH3: BCL2-homology-3; BNIP3L/NIX: BCL2 interacting protein 3 like; CDK: cyclin dependent kinase; CHUK/IKKα: component of inhibitor of nuclear factor kappa B kinase complex; CRL2: CUL2-RING ubiquitin ligase; DDB1: damage specific DNA binding protein 1; ER: endoplasmic reticulum; FOXO: forkhead box O; FUNDC1: FUN14 domain containing 1; GBA/β-glucocerebrosidase: glucosylceramidase beta; HUWE1: HECT, UBA and WWE domain containing E3 ubiquitin protein ligase 1; IDR: intrinsically disordered region; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAVS: mitochondrial antiviral signaling protein; MCL1: MCL1 apoptosis regulator, BCL2 family member; MFN2: mitofusin 2; MTOR: mechanistic target of rapamycin kinase; MSA: multiple system atrophy; MYC: MYC proto-oncogene, bHLH transcription factor; NUMA1: nuclear mitotic apparatus protein 1; OMM; mitochondria outer membrane; PD: Parkinson disease; PHB2: prohibitin 2; PINK1: PTEN induced kinase 1; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PTK2/FAK: protein tyrosine kinase 2; ROS: reactive oxygen species; RPE: retinal pigment epithelium; SAD: sporadic AD; SOCS3: suppressor of cytokine signaling 3; SRC, SRC proto-oncogene, non-receptor tyrosine kinase; STAT3: signal transducer and activator of transcription 3; STING1: stimulator of interferon response cGAMP interactor 1; SQSTM1/p62: sequestosome 1; TBK1: TANK binding kinase 1; TGFB/TGFβ: transforming growth factor beta; TOMM: translocase of outer mitochondrial membrane; TRAF6: TNF receptor associated factor 6; TRIM32: tripartite motif containing 32; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Martina Di Rienzo
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Alessandra Romagnoli
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Giulia Refolo
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Tiziana Vescovo
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Fabiola Ciccosanti
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Candida Zuchegna
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Francesca Lozzi
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
| | - Luca Occhigrossi
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
- Department of Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy
| | - Mauro Piacentini
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
- Department of Biology, University of Rome ‘Tor Vergata’, Rome, Italy
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases IRCCS ‘L. Spallanzani’, Rome, Italy
- Department of Molecular Medicine, University of Rome “La Sapienza”, Rome, Italy
| |
Collapse
|
12
|
Sian-Hulsmann J, Riederer P. Virus-induced brain pathology and the neuroinflammation-inflammation continuum: the neurochemists view. J Neural Transm (Vienna) 2024; 131:1429-1453. [PMID: 38261034 PMCID: PMC11608394 DOI: 10.1007/s00702-023-02723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/18/2023] [Indexed: 01/24/2024]
Abstract
Fascinatingly, an abundance of recent studies has subscribed to the importance of cytotoxic immune mechanisms that appear to increase the risk/trigger for many progressive neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis, and multiple sclerosis. Events associated with the neuroinflammatory cascades, such as ageing, immunologic dysfunction, and eventually disruption of the blood-brain barrier and the "cytokine storm", appear to be orchestrated mainly through the activation of microglial cells and communication with the neurons. The inflammatory processes prompt cellular protein dyshomeostasis. Parkinson's and Alzheimer's disease share a common feature marked by characteristic pathological hallmarks of abnormal neuronal protein accumulation. These Lewy bodies contain misfolded α-synuclein aggregates in PD or in the case of AD, they are Aβ deposits and tau-containing neurofibrillary tangles. Subsequently, these abnormal protein aggregates further elicit neurotoxic processes and events which contribute to the onset of neurodegeneration and to its progression including aggravation of neuroinflammation. However, there is a caveat for exclusively linking neuroinflammation with neurodegeneration, since it's highly unlikely that immune dysregulation is the only factor that contributes to the manifestation of many of these neurodegenerative disorders. It is unquestionably a complex interaction with other factors such as genetics, age, and environment. This endorses the "multiple hit hypothesis". Consequently, if the host has a genetic susceptibility coupled to an age-related weakened immune system, this makes them more susceptible to the virus/bacteria-related infection. This may trigger the onset of chronic cytotoxic neuroinflammatory processes leading to protein dyshomeostasis and accumulation, and finally, these events lead to neuronal destruction. Here, we differentiate "neuroinflammation" and "inflammation" with regard to the involvement of the blood-brain barrier, which seems to be intact in the case of neuroinflammation but defect in the case of inflammation. There is a neuroinflammation-inflammation continuum with regard to virus-induced brain affection. Therefore, we propose a staging of this process, which might be further developed by adding blood- and CSF parameters, their stage-dependent composition and stage-dependent severeness grade. If so, this might be suitable to optimise therapeutic strategies to fight brain neuroinflammation in its beginning and avoid inflammation at all.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Human Anatomy and Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| | - Peter Riederer
- University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark, Winslows Vey 18, 5000, Odense, J.B, Denmark.
| |
Collapse
|
13
|
Andriambelo B, Vachon A, Dansereau MA, Laurent B, Plourde M. Providing lysophosphatidylcholine-bound omega-3 fatty acids increased eicosapentaenoic acid, but not docosahexaenoic acid, in the cortex of mice with the apolipoprotein E3 or E4 allele. Prostaglandins Leukot Essent Fatty Acids 2024; 204:102661. [PMID: 39642444 DOI: 10.1016/j.plefa.2024.102661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Several mechanisms have been proposed for the brain uptake of omega-3 fatty acids (n-3), including passive diffusion of the unesterified form and the use of Mfsd2a transporter for the lysophosphatidylcholine (LPC) form. We hypothesize that the accumulation of LPC n-3 in the brain is lower in mice carrying the apolipoprotein E ε4 allele (APOE4), a major genetic risk factor for developing sporadic Alzheimer's disease in humans. OBJECTIVE Determine whether two or four months of supplementation with LPC n-3 increases the levels of docosahexaenoic acids (DHA) and eicosapentaenoic acids (EPA) in the frontal cortex of APOE3 and APOE4 mice. METHODS APOE3 and APOE4 mice were administered LPC n-3 (9.6 mg DHA + 18.3 mg EPA) or sunflower oil (control) by oral gavage for two or four months (n = 5-8 per genotype, per treatment, and per treatment duration). At the end of the treatment period, frontal cortices were collected, and their FA profiles analyzed by gas chromatography with flame ionization detection. RESULTS After two months of gavage with LPC n-3, APOE3 mice showed increased levels of EPA in their cortex, but not DHA. In APOE4 mice, neither EPA nor DHA levels were significantly affected. After four months of LPC n-3, both APOE3 and APOE4 mice exhibited higher EPA levels, while changes in DHA levels were not statistically significant. CONCLUSION LPC n-3 supplementation increased EPA, but not DHA, levels in the frontal cortex of mice in a duration- and APOE genotype-dependent manner. Further research is needed to explore the implications for brain health.
Collapse
Affiliation(s)
- Bijou Andriambelo
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada; Institut de la Nutrition et des Aliments Fonctionnels, Université Laval, Québec, QC, Canada
| | - Annick Vachon
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada; Institut de la Nutrition et des Aliments Fonctionnels, Université Laval, Québec, QC, Canada
| | - Marc-André Dansereau
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Laurent
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada
| | - Mélanie Plourde
- Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada; Centre de Recherche sur le Vieillissement, CIUSSS de l'Estrie-CHUS, Sherbrooke, QC, Canada; Institut de la Nutrition et des Aliments Fonctionnels, Université Laval, Québec, QC, Canada.
| |
Collapse
|
14
|
Roy N, Haq I, Ngo JC, Bennett DA, Teich AF, De Jager PL, Olah M, Sher F. Elevated expression of the retrotransposon LINE-1 drives Alzheimer's disease-associated microglial dysfunction. Acta Neuropathol 2024; 148:75. [PMID: 39604588 PMCID: PMC11602836 DOI: 10.1007/s00401-024-02835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Aberrant activity of the retrotransposable element long interspersed nuclear element-1 (LINE-1) has been hypothesized to contribute to cellular dysfunction in age-related disorders, including late-onset Alzheimer's disease (LOAD). However, whether LINE-1 is differentially expressed in cell types of the LOAD brain, and whether these changes contribute to disease pathology is largely unknown. Here, we examined patterns of LINE-1 expression across neurons, astrocytes, oligodendrocytes, and microglia in human postmortem prefrontal cortex tissue from LOAD patients and cognitively normal, age-matched controls. We report elevated immunoreactivity of the open reading frame 1 protein (ORF1p) encoded by LINE-1 in microglia from LOAD patients and find that this immunoreactivity correlates positively with disease-associated microglial morphology. In human iPSC-derived microglia (iMG), we found that CRISPR-mediated transcriptional activation of LINE-1 drives changes in microglial morphology and cytokine secretion and impairs the phagocytosis of amyloid beta (Aβ). We also find LINE-1 upregulation in iMG induces transcriptomic changes genes associated with antigen presentation and lipid metabolism as well as impacting the expression of many AD-relevant genes. Our data posit that heightened LINE-1 expression may trigger microglial dysregulation in LOAD and that these changes may contribute to disease pathogenesis, suggesting a central role for LINE-1 activity in human LOAD.
Collapse
Affiliation(s)
- Nainika Roy
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Imdadul Haq
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Jason C Ngo
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Andrew F Teich
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Marta Olah
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Falak Sher
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA.
- Taub Institute for Research On Alzheimer's Disease and Aging Brain, Columbia University Medical Center, New York, NY, USA.
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
15
|
El Abiad E, Al-Kuwari A, Al-Aani U, Al Jaidah Y, Chaari A. Navigating the Alzheimer's Biomarker Landscape: A Comprehensive Analysis of Fluid-Based Diagnostics. Cells 2024; 13:1901. [PMID: 39594648 PMCID: PMC11593284 DOI: 10.3390/cells13221901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects a significant portion of the aging population, presenting a serious challenge due to the limited availability of effective therapies during its progression. The disease advances rapidly, underscoring the need for early diagnosis and the application of preventative measures. Current diagnostic methods for AD are often expensive and invasive, restricting access for the general public. One potential solution is the use of biomarkers, which can facilitate early detection and treatment through objective, non-invasive, and cost-effective evaluations of AD. This review critically investigates the function and role of biofluid biomarkers in detecting AD, with a specific focus on cerebrospinal fluid (CSF), blood-based, and saliva biomarkers. RESULTS CSF biomarkers have demonstrated potential for accurate diagnosis and valuable prognostic insights, while blood biomarkers offer a minimally invasive and cost-effective approach for diagnosing cognitive issues. However, while current biomarkers for AD show significant potential, none have yet achieved the precision needed to replace expensive PET scans and CSF assays. The lack of a single accurate biomarker underscores the need for further research to identify novel or combined biomarkers to enhance the clinical efficacy of existing diagnostic tests. In this context, artificial intelligence (AI) and deep-learning (DL) tools present promising avenues for improving biomarker analysis and interpretation, enabling more precise and timely diagnoses. CONCLUSIONS Further research is essential to confirm the utility of all AD biomarkers in clinical settings. Combining biomarker data with AI tools offers a promising path toward revolutionizing the personalized characterization and early diagnosis of AD symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (E.E.A.); (A.A.-K.); (U.A.-A.); (Y.A.J.)
| |
Collapse
|
16
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
17
|
Wiśniewska K, Rintz E, Żabińska M, Gaffke L, Podlacha M, Cyske Z, Węgrzyn G, Pierzynowska K. Comprehensive evaluation of pathogenic protein accumulation in fibroblasts from all subtypes of Sanfilippo disease patients. Biochem Biophys Res Commun 2024; 733:150718. [PMID: 39305572 DOI: 10.1016/j.bbrc.2024.150718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/06/2024]
Abstract
Sanfilippo disease is a lysosomal storage disorder from the group of mucopolysaccharidoses (MPS), characterized by storage of glycosaminoglycans (GAGs); thus, it is also called MPS type III. The syndrome is divided into 4 subtypes (MPS III A, B, C and D). Despite the storage of the same GAG, heparan sulfate (HS), the course of these subtypes can vary considerably. Here, we comprehensively evaluated the levels of protein aggregates (APP, β-amyloid, p-tau, α-synuclein, TDP43) in fibroblasts derived from patients with all MPS III subtypes, and tested whether lowering GAG levels results in a decrease in the levels of the investigated proteins and the number of aggregates they form. Elevated levels of APP, β-amyloid, tau, and TDP43 proteins were evident in all MPS III subtypes, and elevated levels of p-tau and α-synuclein were demonstrated in all subtypes except MPS IIIC. These findings were confirmed in the neural tissue of MPS IIIB mice. Fluorescence microscopy studies also indicated a high number of protein aggregates formed by β-amyloid and tau in all cell lines tested, and a high number of aggregates of p-tau, TDP43, and α-synuclein in all lines except MPS IIIC. Reduction of GAG levels by genistein led to the decrease of levels of all tested proteins and their aggregates except α-synuclein, indicating a relationship between GAG levels and those of some protein aggregates. This work describes for the first time the problem of deposited protein aggregates in all subtypes of Sanfilippo disease and suggests that GAGs are partly responsible for the formation of protein aggregates.
Collapse
Affiliation(s)
- Karolina Wiśniewska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Magdalena Żabińska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Magdalena Podlacha
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Zuzanna Cyske
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| |
Collapse
|
18
|
Kciuk M, Kruczkowska W, Gałęziewska J, Wanke K, Kałuzińska-Kołat Ż, Aleksandrowicz M, Kontek R. Alzheimer's Disease as Type 3 Diabetes: Understanding the Link and Implications. Int J Mol Sci 2024; 25:11955. [PMID: 39596023 PMCID: PMC11593477 DOI: 10.3390/ijms252211955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two prevalent conditions that present considerable public health issue in aging populations worldwide. Recent research has proposed a novel conceptualization of AD as "type 3 diabetes", highlighting the critical roles of insulin resistance and impaired glucose metabolism in the pathogenesis of the disease. This article examines the implications of this association, exploring potential new avenues for treatment and preventive strategies for AD. Key evidence linking diabetes to AD emphasizes critical metabolic processes that contribute to neurodegeneration, including inflammation, oxidative stress, and alterations in insulin signaling pathways. By framing AD within this metabolic context, we can enhance our understanding of its etiology, which in turn may influence early diagnosis, treatment plans, and preventive measures. Understanding AD as a manifestation of diabetes opens up the possibility of employing novel therapeutic strategies that incorporate lifestyle modifications and the use of antidiabetic medications to mitigate cognitive decline. This integrated approach has the potential to improve patient outcomes and deepen our comprehension of the intricate relationship between neurodegenerative diseases and metabolic disorders.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Weronika Kruczkowska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Julia Gałęziewska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Katarzyna Wanke
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| |
Collapse
|
19
|
Shao J, Deng Q, Feng S, Wu C, Liu X, Yang L. Role of astrocytes in Alzheimer's disease pathogenesis and the impact of exercise-induced remodeling. Biochem Biophys Res Commun 2024; 732:150418. [PMID: 39032410 DOI: 10.1016/j.bbrc.2024.150418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) is a prevalent and debilitating brain disorder that worsens progressively with age, characterized by cognitive decline and memory impairment. The accumulation of amyloid-beta (Aβ) leading to amyloid plaques and hyperphosphorylation of Tau, resulting in intracellular neurofibrillary tangles (NFTs), are primary pathological features of AD. Despite significant research investment and effort, therapies targeting Aβ and NFTs have proven limited in efficacy for treating or slowing AD progression. Consequently, there is a growing interest in non-invasive therapeutic strategies for AD prevention. Exercise, a low-cost and non-invasive intervention, has demonstrated promising neuroprotective potential in AD prevention. Astrocytes, among the most abundant glial cells in the brain, play essential roles in various physiological processes and are implicated in AD initiation and progression. Exercise delays pathological progression and mitigates cognitive dysfunction in AD by modulating astrocyte morphological and phenotypic changes and fostering crosstalk with other glial cells. This review aims to consolidate the current understanding of how exercise influences astrocyte dynamics in AD, with a focus on elucidating the molecular and cellular mechanisms underlying astrocyte remodeling. The review begins with an overview of the neuropathological changes observed in AD, followed by an examination of astrocyte dysfunction as a feature of the disease. Lastly, the review explores the potential therapeutic implications of exercise-induced astrocyte remodeling in the context of AD.
Collapse
Affiliation(s)
- Jie Shao
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Shu Feng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Xiaocao Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
20
|
Alagiakrishnan K, Halverson T. Role of Peripheral and Central Insulin Resistance in Neuropsychiatric Disorders. J Clin Med 2024; 13:6607. [PMID: 39518747 PMCID: PMC11547162 DOI: 10.3390/jcm13216607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/27/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Insulin acts on different organs, including the brain, which helps it regulate energy metabolism. Insulin signaling plays an important role in the function of different cell types. In this review, we have summarized the key roles of insulin and insulin receptors in healthy brains and in different brain disorders. Insulin signaling, as well as insulin resistance (IR), is a major contributor in the regulation of mood, behavior, and cognition. Recent evidence showed that both peripheral and central insulin resistance play a role in the pathophysiology, clinical presentation, and management of neuropsychiatric disorders like Cognitive Impairment/Dementia, Depression, and Schizophrenia. Many human studies point out Insulin Resistance/Metabolic Syndrome can increase the risk of dementia especially Alzheimer's dementia (AD). IR has been shown to play a role in AD development but also in its progression. This review article discusses the pathophysiological pathways and mechanisms of insulin resistance in major neuropsychiatric disorders. The extent of insulin resistance can be quantified using IR biomarkers like insulin levels, HOMA-IR index, and Triglyceride glucose-body mass index (TyG-BMI) levels. IR has been shown to precede neurodegeneration. Human trials showed current treatment with certain antidiabetic drugs, as well as life style management, like weight loss and exercise for IR, have shown promise in the management of cognitive/neuropsychiatric disorders. This may pave the pathway to the development of new therapeutic approaches to these challenging disorders of dementia and psychiatric diseases. Recent clinical trials are showing some encouraging evidence for these pharmacological and nonpharmacological approaches for IR in psychiatric and cognitive disorders, even though more research is needed to apply this evidence into clinical practice. Early identification and management of IR may help as a strategy to potentially alter neuropsychiatric disorders onset as well as its progression.
Collapse
Affiliation(s)
| | - Tyler Halverson
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada;
| |
Collapse
|
21
|
Deza‐Lougovski YI, Weiss LM, Horton HM, Sun A, Borbye‐Lorenzen N, Skogstrand K, Holmgaard S, Andersen‐Ranberg K, Lundmark VP, Börsch‐Supan A, Börsch‐Supan M, Rieckmann A. Circulating apoE4 protein levels from dried blood spots predict cognitive function in a large population-based survey setting. Alzheimers Dement 2024; 20:7613-7623. [PMID: 39234633 PMCID: PMC11567846 DOI: 10.1002/alz.14224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The apolipoprotein E (APOE) ε4 allele carries risk for cognitive impairment, but whether the level of circulating apoE4 protein in carriers affects cognition is unclear, as is how health and lifestyle impact circulating apoE4 levels. METHODS We assayed apoE4 protein levels in dried blood spots of 12,532 adults aged 50+. Regression analyses tested the likelihood of cognitive impairment between groups and within those with detected apoE4 protein. Predictors of circulating apoE4 were assessed. RESULTS We detected protein binding that indicates the presence of an APOE ε4 allele in 28.4% of this group. This group was more likely to have cognitive impairment, and this risk increases with age. However, higher apoE4 levels were associated with less likelihood of cognitive impairment within this group. Antihypertensive medication predicted apoE4 protein levels. DISCUSSION The apoE4 isoform is associated with a deficient protein and worse cognition. This association is modulated by the level of circulating apoE4 protein in ε4 carriers. HIGHLIGHTS An assay to quantify apoE4 levels from dried blood spot samples was applied. The apoE4 protein was detected as specific binding at ≥30,000 pg/mL in 28.4% of samples. Having the apoE4 protein was associated with worse cognitive performance. Higher apoE4 protein levels in those who have it were associated with better cognition. Cardiovascular factors influenced levels of apoE4 protein.
Collapse
Affiliation(s)
- Yacila I. Deza‐Lougovski
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Luzia M. Weiss
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| | - Hannah M. Horton
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Aijing Sun
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Nis Borbye‐Lorenzen
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Kristin Skogstrand
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Solveig Holmgaard
- Department for Congenital DisordersCenter for Neonatal Screening, Statens Serum InstitutCopenhagenDenmark
| | - Karen Andersen‐Ranberg
- Department of Public Health Epidemiology, Biostatistics and BiodemographyUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research Geriatric Research UnitUniversity of Southern DenmarkOdenseDenmark
| | - Vania Panes Lundmark
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Umeå Center for Functional Brain ImagingUmeå UniversityUmeåSweden
| | - Axel Börsch‐Supan
- Max Planck Institute for Social Law and Social PolicyMunichGermany
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
| | - Martina Börsch‐Supan
- Munich Research Institute for the Economics of Aging and SHARE Analyses (MEA)MunichGermany
- Survey of Health, Ageing and Retirement in Europe (SHARE Biomarker Project)MunichGermany
| | - Anna Rieckmann
- Institute of Psychology, University of the Bundeswehr MünchenNeubibergGermany
- Max Planck Institute for Social Law and Social PolicyMunichGermany
| |
Collapse
|
22
|
Shin MK, Schuck A, Kang M, Kim YS. Electrochemical Analysis of Amyloid Plaques and ApoE4 with Chitosan-Coated Gold Nanostars for Alzheimer's Detection. BIOSENSORS 2024; 14:510. [PMID: 39451723 PMCID: PMC11506205 DOI: 10.3390/bios14100510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Monitoring the progression of Alzheimer's disease (AD) is crucial for mitigating dementia symptoms, alleviating pain, and improving mobility. Traditionally, AD biomarkers like amyloid plaques are predominantly identified in cerebrospinal fluid (CSF) due to their concentrated presence. However, detecting these markers in blood is hindered by the blood-brain barrier (BBB), resulting in lower concentrations. To address this challenge and identify pertinent AD biomarkers-specifically amyloid plaques and apolipoprotein E4 (ApoE4)-in blood plasma, we propose an innovative approach. This involves enhancing a screen-printed carbon electrode (SPCE) with an immobilization matrix comprising gold nanostars (AuNSs) coated with chitosan. Morphological and electrical analyses confirmed superior dispersion and conductivity with 0.5% chitosan, supported by UV-Vis spectroscopy, cyclic voltammetry, and Nyquist plots. Subsequent clinical assays measured electrical responses to quantify amyloid-β 42 (Aβ42) (15.63-1000 pg/mL) and APoE4 levels (0.41 to 40 ng/mL) in human blood plasma samples. Differential pulse voltammetry (DPV) responses exhibited peak currents proportional to biomarker concentrations, demonstrating high linear correlations (0.985 for Aβ42 and 0.919 for APoE4) with minimal error bars. Cross-reactivity tests with mixed solutions of amyloid-β 40 (Aβ40), Aβ42, and ApoE4 indicated minimal interference between biomarkers (<3% variation), further confirming the high specificity of the developed sensor. Validation studies demonstrated a strong concurrence with the gold-standard enzyme-linked immunosorbent assay (ELISA), while interference tests indicated a minimal variation in peak currents. This improved device presents promising potential as a point-of-care system, offering a less invasive, cost-effective, and simplified approach to detecting and tracking the progression of AD. The substantial surface binding area further supports the efficacy of our method, offering a promising avenue for advancing AD diagnostics.
Collapse
Affiliation(s)
- Min-Kyung Shin
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Ariadna Schuck
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| | - Minhee Kang
- Biomedical Engineering Research Center, Smart Healthcare Research Institute, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul 06351, Republic of Korea;
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yong-Sang Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea;
| |
Collapse
|
23
|
Palmer JA, Kaufman CS, Whitaker-Hilbig AA, Billinger SA. APOE4 carriers display loss of anticipatory cerebral vascular regulation over AD progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315344. [PMID: 39417136 PMCID: PMC11482999 DOI: 10.1101/2024.10.11.24315344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Maintenance of cerebral blood flow during orthostasis is impaired with aging and associated with cognitive decline, but the effect of Apolipoprotein 4-allele (APOE4) is unknown. METHODS Older adults (n=108) (APOE4 carriers, n=47; noncarriers, n=61) diagnosed as cognitively-normal (NC), MCI, or AD participated. Middle cerebral artery blood velocity (MCAv), assessed using Transcranial Doppler ultrasound, and beat-to-beat mean arterial blood pressure (MAP) were continuously recorded during a sit-to-stand transition. Anticipatory and orthostatic-induced MCAv and MAP responses were compared between genotypes and across disease progression. RESULTS Cognitively-normal APOE4 carriers showed greater anticipatory MCAv increase, greater MCAv decrease with orthostasis, and shorter latency of peripheral MAP responses to orthostasis compared to noncarriers. MCAv and MAP responses were delayed and attenuated across the APOE4 disease progression, with no differences between genotypes in MCI and AD. DISCUSSION APOE4 carriers and noncarriers present with distinct phenotypes of cerebral vascular dysfunction during hemodynamic orthostatic challenge. Unique cerebral and peripheral vascular compensation observed in APOE4 carriers may be lost as AD progresses.
Collapse
Affiliation(s)
- Jacqueline A. Palmer
- Division of Physical Therapy and Rehabilitation Science, University of Minnesota Medical School, , Minneapolis, MN, United States of America
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Carolyn S. Kaufman
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States of America
- Department of Internal Medicine, Stanford Health Care, Stanford University, Palo Alto, CA, United States of America
| | - Alicen A. Whitaker-Hilbig
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, 123 Milwaukee Way, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Sandra A. Billinger
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| |
Collapse
|
24
|
Ding Y, Palecek SP, Shusta EV. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta. Fluids Barriers CNS 2024; 21:79. [PMID: 39394110 PMCID: PMC11468049 DOI: 10.1186/s12987-024-00580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
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.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
25
|
Laslo A, Laslo L, Arbănași EM, Ujlaki-Nagi AA, Chinezu L, Ivănescu AD, Arbănași EM, Cărare RO, Cordoș BA, Popa IA, Brînzaniuc K. Pathways to Alzheimer's Disease: The Intersecting Roles of Clusterin and Apolipoprotein E in Amyloid-β Regulation and Neuronal Health. PATHOPHYSIOLOGY 2024; 31:545-558. [PMID: 39449522 PMCID: PMC11503414 DOI: 10.3390/pathophysiology31040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
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.
Collapse
Affiliation(s)
- Alexandru Laslo
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Laura Laslo
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (L.L.); (B.A.C.)
| | - Eliza-Mihaela Arbănași
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Regenerative Medicine Laboratory, Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | | | - Laura Chinezu
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Adrian Dumitru Ivănescu
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
| | - Emil-Marian Arbănași
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Regenerative Medicine Laboratory, Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
| | | | - Bogdan Andrei Cordoș
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (L.L.); (B.A.C.)
- Centre for Experimental Medical and Imaging Studies, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Ioana Adriana Popa
- Clinic of Radiology, Mures County Emergency Hospital, 540136 Targu Mures, Romania;
| | - Klara Brînzaniuc
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
| |
Collapse
|
26
|
Reyes CDG, Mojgan A, Fowowe M, Onigbinde S, Daramola O, Lubman DM, Mechref Y. Differential expression of N-glycopeptides derived from serum glycoproteins in mild cognitive impairment (MCI) patients. Proteomics 2024; 24:e2300620. [PMID: 38602241 PMCID: PMC11749004 DOI: 10.1002/pmic.202300620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Mild cognitive impairment (MCI) is an early stage of memory loss that affects cognitive abilities with the aging of individuals, such as language or visual/spatial comprehension. MCI is considered a prodromal phase of more complicated neurodegenerative diseases such as Alzheimer's. Therefore, accurate diagnosis and better understanding of the disease prognosis will facilitate prevention of neurodegeneration. However, the existing diagnostic methods fail to provide precise and well-timed diagnoses, and the pathophysiology of MCI is not fully understood. Alterations of the serum N-glycoproteome expression could represent an essential contributor to the overall pathophysiology of neurodegenerative diseases and be used as a potential marker to assess MCI diagnosis using less invasive procedures. In this approach, we identified N-glycopeptides with different expressions between healthy and MCI patients from serum glycoproteins. Seven of the N-glycopeptides showed outstanding AUC values, among them the antithrombin-III Asn224 + 4-5-0-2 with an AUC value of 1.00 and a p value of 0.0004. According to proteomics and ingenuity pathway analysis (IPA), our data is in line with recent publications, and the glycoproteins carrying the identified N-sites play an important role in neurodegeneration.
Collapse
Affiliation(s)
| | - Atashi Mojgan
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Mojibola Fowowe
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Sherifdeen Onigbinde
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Oluwatosin Daramola
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - David M. Lubman
- Department of Surgery, The University of Michigan, Ann Arbor, MI 48109
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| |
Collapse
|
27
|
Tripathi A, Pandey VK, Sharma G, Sharma AR, Taufeeq A, Jha AK, Kim JC. Genomic Insights into Dementia: Precision Medicine and the Impact of Gene-Environment Interaction. Aging Dis 2024; 15:2113-2135. [PMID: 38607741 PMCID: PMC11346410 DOI: 10.14336/ad.2024.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
The diagnosis, treatment, and management of dementia provide significant challenges due to its chronic cognitive impairment. The complexity of this condition is further highlighted by the impact of gene-environment interactions. A recent strategy combines advanced genomics and precision medicine methods to explore the complex genetic foundations of dementia. Utilizing the most recent research in the field of neurogenetics, the importance of precise genetic data in explaining the variation seen in dementia patients can be investigated. Gene-environment interactions are important because they influence genetic susceptibilities and aid in the development and progression of dementia. Modified to each patient's genetic profile, precision medicine has the potential to detect groups at risk and make previously unheard-of predictions about the course of diseases. Precision medicine techniques have the potential to completely transform treatment and diagnosis methods. Targeted medications that target genetic abnormalities will probably appear, providing the possibility for more efficient and customized medical interventions. Investigating the relationship between genes and the environment may lead to preventive measures that would enable people to change their surroundings and minimize the risk of dementia, leading to the improved lifestyle of affected people. This paper provides a comprehensive overview of the genomic insights into dementia, emphasizing the pivotal role of precision medicine, and gene-environment interactions.
Collapse
Affiliation(s)
- Anjali Tripathi
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Vinay Kumar Pandey
- Division of Research & Innovation (DRI), School of Applied & Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Anam Taufeeq
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Kanpur, Uttar Pradesh, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Jin-Chul Kim
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
28
|
Stoessel MB, Stowell RD, Lowery RL, Le L, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614526. [PMID: 39386439 PMCID: PMC11463386 DOI: 10.1101/2024.09.25.614526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite the emerging consensus that microglia are critical to physiological and pathological brain function, it is unclear how microglial roles and their underlying mechanisms differ between brain regions. Microglia throughout the brain express common markers, such as the purinergic receptor P2Y12, that delineate them from peripheral macrophages. P2Y12 is a critical sensor of injury but also contributes to the sensing of neuronal activity and remodeling of synapses, with microglial loss of P2Y12 resulting in behavioral deficits. P2Y12 has largely been studied in cortical microglia, despite the fact that a growing body of evidence suggests that microglia exhibit a high degree of regional specialization. Cerebellar microglia, in particular, exhibit transcriptional, epigenetic, and functional profiles that set them apart from their better studied cortical and hippocampal counterparts. Here, we demonstrate that P2Y12 deficiency does not alter the morphology, distribution, or dynamics of microglia in the cerebellum. In fact, loss of P2Y12 does little to disturb the distinct transcriptomic profiles of cortical and cerebellar microglia. However, unlike in cortex, P2Y12 is not required for a full microglial response to focal injury, suggesting that cerebellar and cortical microglia use different cues to respond to injury. Finally, we show that P2Y12 deficiency impairs cerebellar learning in a delay eyeblink conditioning task, a common test of cerebellar plasticity and circuit function. Our findings suggest not only region-specific roles of microglial P2Y12 signaling in the focal injury response, but also indicate a conserved role for P2Y12 in microglial modulation of plasticity across regions.
Collapse
|
29
|
Misiura M, Munkombwe C, Igwe K, Verble DD, Likos KDS, Minto L, Bartlett A, Zetterberg H, Turner JA, Dotson VM, Brickman AM, Hu WT, Wharton W. Neuroimaging correlates of Alzheimer's disease biomarker concentrations in a racially diverse high-risk cohort of middle-aged adults. Alzheimers Dement 2024; 20:5961-5972. [PMID: 39136298 PMCID: PMC11497767 DOI: 10.1002/alz.14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION In this study, we investigated biomarkers in a midlife, racially diverse, at-risk cohort to facilitate early identification and intervention. We examined neuroimaging measures, including resting state functional magnetic resonance imaging (fMRI), white matter hyperintensity vo (WMH), and hippocampal volumes, alongside cerebrospinal fluid (CSF) markers. METHODS Our data set included 76 cognitively unimpaired, middle-aged, Black Americans (N = 29, F/M = 17/12) and Non-Hispanic White (N = 47, F/M = 27/20) individuals. We compared cerebrospinal fluid phosphorylated tau141 and amyloid beta (Aβ)42 to fMRI default mode network (DMN) subnetwork connectivity, WMH volumes, and hippocampal volumes. RESULTS Results revealed a significant race × Aβ42 interaction in Black Americans: lower Aβ42 was associated with reduced DMN connectivity and increased WMH volumes regions but not in non-Hispanic White individuals. DISCUSSION Our findings suggest that precuneus DMN connectivity and temporal WMHs may be linked to Alzheimer's disease risk pathology during middle age, particularly in Black Americans. HIGHLIGHTS Cerebrospinal fluid (CSF) amyloid beta (Aβ)42 relates to precuneus functional connectivity in Black, but not White, Americans. Higher white matter hyperintensity volume relates to lower CSF Aβ42 in Black Americans. Precuneus may be a hub for early Alzheimer's disease pathology changes detected by functional connectivity.
Collapse
Affiliation(s)
- Maria Misiura
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Tri‐Institutional Center for Translational Research in Neuroimaging & Data Science, Georgia State University, Georgia Institute of TechnologyEmory UniversityAtlantaGeorgiaUSA
| | | | - Kay Igwe
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Danielle D. Verble
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Kelly D. S. Likos
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Lex Minto
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
| | | | - Henrik Zetterberg
- The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and GothenburgUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative Disease, UCL Institute of NeurologyUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCL, Maple HouseLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Jessica A. Turner
- Department of Psychiatry and Mental Health, College of MedicineOhio State UniversityColumbusOhioUSA
| | - Vonetta M. Dotson
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Gerontology DepartmentGeorgia State UniversityAtlantaGeorgiaUSA
| | - Adam M. Brickman
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - William T. Hu
- Institute for Health, Health Care Policy, and Aging ResearchRutgers UniversityNew BrunswickNew JerseyUSA
| | - Whitney Wharton
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
30
|
Tsintzas E, Niccoli T. Using Drosophila amyloid toxicity models to study Alzheimer's disease. Ann Hum Genet 2024; 88:349-363. [PMID: 38517001 DOI: 10.1111/ahg.12554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 03/23/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia and is characterised by a progressive loss of neurons, which manifests as gradual memory decline, followed by cognitive loss. Despite the significant progress in identifying novel biomarkers and understanding the prodromal pathology and symptomatology, AD remains a significant unmet clinical need. Lecanemab and aducanumab, the only Food and Drug Administration approved drugs to exhibit some disease-modifying clinical efficacy, target Aβ amyloid, underscoring the importance of this protein in disease aetiology. Nevertheless, in the absence of a definitive cure, the utilisation of preclinical models remains imperative for the identification of novel therapeutic targets and the evaluation of potential therapeutic agents. Drosophila melanogaster is a model system that can be used as a research tool to investigate neurodegeneration and therapeutic interventions. The short lifespan, low price and ease of husbandry/rearing make Drosophila an advantageous model organism from a practical perspective. However, it is the highly conserved genome and similarity of Drosophila and human neurobiology which make flies a powerful tool to investigate neurodegenerative mechanisms. In addition, the ease of transgenic modifications allows for early proof of principle studies for future therapeutic approaches in neurodegenerative research. This mini review will specifically focus on utilising Drosophila as an in vivo model of amyloid toxicity in AD.
Collapse
Affiliation(s)
- Elli Tsintzas
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Teresa Niccoli
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| |
Collapse
|
31
|
Poblano J, Castillo-Tobías I, Berlanga L, Tamayo-Ordoñez MC, Del Carmen Rodríguez-Salazar M, Silva-Belmares SY, Aguayo-Morales H, Cobos-Puc LE. Drugs targeting APOE4 that regulate beta-amyloid aggregation in the brain: Therapeutic potential for Alzheimer's disease. Basic Clin Pharmacol Toxicol 2024; 135:237-249. [PMID: 39020526 DOI: 10.1111/bcpt.14055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/21/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024]
Abstract
Alzheimer's disease is characterized by progressive cognitive decline, and behavioural and psychological symptoms of dementia are common. The APOE ε4 allele, a genetic risk factor, significantly increases susceptibility to the disease. Despite efforts to effectively treat the disease, only seven drugs are approved for its treatment, and only two of these prevent its progression. This highlights the need to identify new pharmacological options. This review focuses on mimetic peptides, small molecule correctors and HAE-4 antibodies that target ApoE. These drugs reduce β-amyloid-induced neurodegeneration in preclinical models. In addition, loop diuretics such as bumetanide and furosemide show the potential to reduce the prevalence of Alzheimer's disease in humans, and antidepressants such as imipramine improve cognitive function in individuals diagnosed with Alzheimer's disease. Consistent with this, both classes of drugs have been shown to exert neuroprotective effects by inhibiting ApoE4-catalysed Aβ aggregation in preclinical models. Moreover, peroxisome proliferator-activated receptor ligands, particularly pioglitazone and rosiglitazone, reduce ApoE4-induced neurodegeneration in animal models. However, they do not prevent the cognitive decline in APOE ε4 allele carriers. Finally, ApoE4 impairs the integrity of the blood-brain barrier and haemostasis. On this basis, ApoE4 modulation is a promising avenue for the treatment of late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Joan Poblano
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Ileana Castillo-Tobías
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Lia Berlanga
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | | | | | | | - Hilda Aguayo-Morales
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| | - Luis E Cobos-Puc
- Faculty of Chemical Sciences, Autonomous University of Coahuila, Saltillo, Coahuila, Mexico
| |
Collapse
|
32
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
33
|
Bailey M, Ilchovska ZG, Hosseini AA, Jung J. Impact of Apolipoprotein E ε4 in Alzheimer's Disease: A Meta-Analysis of Voxel-Based Morphometry Studies. J Clin Neurol 2024; 20:469-477. [PMID: 39227329 PMCID: PMC11372214 DOI: 10.3988/jcn.2024.0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is the most-prevalent form of dementia and imposes substantial burdens at the personal and societal levels. The apolipoprotein E (APOE) ε4 allele is a genetic factor known to increase AD risk and exacerbate brain atrophy and its symptoms. We aimed to provide a comprehensive review of the impacts of APOE ε4 on brain atrophy in AD as well as in mild cognitive impairment (MCI) as a transitional stage of AD. METHODS We performed a coordinate-based meta-analysis of voxel-based morphometry studies to compare gray-matter atrophy patterns between carriers and noncarriers of APOE ε4. We obtained coordinate-based structural magnetic resonance imaging data from 1,135 individuals who met our inclusion criteria among 12 studies reported in PubMed and Google Scholar. RESULTS We found that atrophy of the hippocampus and parahippocampus was significantly greater in APOE ε4 carriers than in noncarriers, especially among those with AD and MCI, while there was no significant atrophy in these regions in healthy controls who were also carriers. CONCLUSIONS The present meta-analysis has highlighted the significant link between the APOE ε4 allele and hippocampal atrophy in both AD and MCI, which emphasizes the critical influence of the allele on neurodegeneration, especially in the hippocampus. These findings improve the understanding of AD pathology, potentially facilitating progress in early detection, targeted interventions, and personalized care strategies for individuals at risk of AD who carry the APOE ε4 allele.
Collapse
Affiliation(s)
- Madison Bailey
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Zlatomira G Ilchovska
- School of Psychology, University of Nottingham, Nottingham, UK
- School of Psychology, University of Birmingham, Birmingham, UK
| | - Akram A Hosseini
- School of Medicine, University of Nottingham, Nottingham, UK
- Department of Academic Neurology, Nottingham University Hospitals NHS Trust, Queens Medical Centre, Nottingham, UK
- Centre for Dementia, Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - JeYoung Jung
- School of Psychology, University of Nottingham, Nottingham, UK
- Centre for Dementia, Institute of Mental Health, University of Nottingham, Nottingham, UK
- Precision Imaging, University of Nottingham, Nottingham, UK.
| |
Collapse
|
34
|
Escamilla S, Salas-Lucia F. Thyroid Hormone and Alzheimer Disease: Bridging Epidemiology to Mechanism. Endocrinology 2024; 165:bqae124. [PMID: 39276028 DOI: 10.1210/endocr/bqae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, CSIC-Universidad Miguel Hernández, Alicante 03550, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Alicante 03550, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
| | | |
Collapse
|
35
|
Zuniga NR, Earls NE, Denos AEA, Elison JM, Jones BS, Smith EG, Moran NG, Brown KL, Romero GM, Hyer CD, Wagstaff KB, Almughamsi HM, Transtrum MK, Price JC. Quantitative and Kinetic Proteomics Reveal ApoE Isoform-dependent Proteostasis Adaptations in Mouse Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607719. [PMID: 39185235 PMCID: PMC11343127 DOI: 10.1101/2024.08.13.607719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Apolipoprotein E (ApoE) polymorphisms modify the risk of neurodegenerative disease with the ApoE4 isoform increasing and ApoE2 isoform decreasing risk relative to the 'wild-type control' ApoE3 isoform. To elucidate how ApoE isoforms alter the proteome, we measured relative protein abundance and turnover in transgenic mice expressing a human ApoE gene (isoform 2, 3, or 4). This data provides insight into how ApoE isoforms affect the in vivo synthesis and degradation of a wide variety of proteins. We identified 4849 proteins and tested for ApoE isoform-dependent changes in the homeostatic regulation of ~2700 ontologies. In the brain, we found that ApoE4 and ApoE2 both lead to modified regulation of mitochondrial membrane proteins relative to the wild-type control ApoE3. In ApoE4 mice, this regulation is not cohesive suggesting that aerobic respiration is impacted by proteasomal and autophagic dysregulation. ApoE2 mice exhibited a matching change in mitochondrial matrix proteins and the membrane which suggests coordinated maintenance of the entire organelle. In the liver, we did not observe these changes suggesting that the ApoE-effect on proteostasis is amplified in the brain relative to other tissues. Our findings underscore the utility of combining protein abundance and turnover rates to decipher proteome regulatory mechanisms and their potential role in biology.
Collapse
Affiliation(s)
- Nathan R. Zuniga
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Noah E. Earls
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Ariel E. A. Denos
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Jared M. Elison
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Benjamin S. Jones
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Ethan G. Smith
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Noah G. Moran
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Katie L. Brown
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Gerome M. Romero
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Chad D. Hyer
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Kimberly B. Wagstaff
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Haifa M. Almughamsi
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| | - Mark K. Transtrum
- Department of Physics and Astronomy, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - John C. Price
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| |
Collapse
|
36
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
37
|
Yadav JP, Verma A, Pathak P, Dwivedi AR, Singh AK, Kumar P, Khalilullah H, Jaremko M, Emwas AH, Patel DK. Phytoconstituents as modulators of NF-κB signalling: Investigating therapeutic potential for diabetic wound healing. Biomed Pharmacother 2024; 177:117058. [PMID: 38968797 DOI: 10.1016/j.biopha.2024.117058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
The NF-κB pathway plays a pivotal role in impeding the diabetic wound healing process, contributing to prolonged inflammation, diminished angiogenesis, and reduced proliferation. In contrast to modern synthetic therapies, naturally occurring phytoconstituents are well-studied inhibitors of the NF-κB pathway that are now attracting increased attention in the context of diabetic wound healing because of lower toxicity, better safety and efficacy, and cost-effectiveness. This study explores recent research on phytoconstituent-based therapies and delve into their action mechanisms targeting the NF-κB pathway and potential for assisting effective healing of diabetic wounds. For this purpose, we have carried out surveys of recent literature and analyzed studies from prominent databases such as Science Direct, Scopus, PubMed, Google Scholar, EMBASE, and Web of Science. The classification of phytoconstituents into various categorie such as: alkaloids, triterpenoids, phenolics, polyphenols, flavonoids, monoterpene glycosides, naphthoquinones and tocopherols. Noteworthy phytoconstituents, including Neferine, Plumbagin, Boswellic acid, Genistein, Luteolin, Kirenol, Rutin, Vicenin-2, Gamma-tocopherol, Icariin, Resveratrol, Mangiferin, Betulinic acid, Berberine, Syringic acid, Gallocatechin, Curcumin, Loureirin-A, Loureirin-B, Lupeol, Paeoniflorin, and Puerarin emerge from these studies as promising agents for diabetic wound healing through the inhibition of the NF-κB pathway. Extensive research on various phytoconstituents has revealed how they modulate signalling pathways, including NF-κB, studies that demonstrate the potential for development of therapeutic phytoconstituents to assist healing of chronic diabetic wounds.
Collapse
Affiliation(s)
- Jagat Pal Yadav
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; Pharmacology Research Laboratory, Faculty of Pharmaceutical Sciences, Rama University, Kanpur 209217, India; Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India.
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
| | - Prateek Pathak
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, GITAM School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, 502329, India
| | - Ashish R Dwivedi
- Department of Pharmaceutical Analysis, Quality Assurance and Pharmaceutical Chemistry, GITAM School of Pharmacy, GITAM (Deemed to be University), Hyderabad Campus, 502329, India
| | - Ankit Kumar Singh
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India; Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda 151401, India
| | - Habibullah Khalilullah
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Unayzah 51911, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Dinesh Kumar Patel
- Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India.
| |
Collapse
|
38
|
Falcon C, Montesinos P, Václavů L, Kassinopoulos M, Minguillon C, Fauria K, Cascales‐Lahoz D, Contador J, Fernández‐Lebrero A, Navalpotro I, Puig‐Pijoan A, Grau‐Rivera O, Kollmorgen G, Quijano‐Rubio C, Molinuevo JL, Zetterberg H, Blennow K, Suárez‐Calvet M, Van Osch MJP, Sanchez‐Gonzalez J, Gispert JD. Time-encoded ASL reveals lower cerebral blood flow in the early AD continuum. Alzheimers Dement 2024; 20:5183-5197. [PMID: 38958557 PMCID: PMC11350027 DOI: 10.1002/alz.14059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Cerebral blood flow (CBF) is reduced in cognitively impaired (CI) Alzheimer's disease (AD) patients. We checked the sensitivity of time-encoded arterial spin labeling (te-ASL) in measuring CBF alterations in individuals with positive AD biomarkers and associations with relevant biomarkers in cognitively unimpaired (CU) individuals. METHODS We compared te-ASL with single-postlabel delay (PLD) ASL in measuring CBF in 59 adults across the AD continuum, classified as CU amyloid beta (Aβ) negative (-), CU Aβ positive (+), and CI Aβ+. We sought associations of CBF with biomarkers of AD, cerebrovascular disease, synaptic dysfunction, neurodegeneration, and cognition in CU participants. RESULTS te-ASL was more sensitive at detecting CBF reduction in the CU Aβ+ and CI Aβ+ groups. In CU participants, lower CBF was associated with altered biomarkers of Aβ, tau, synaptic dysfunction, and neurodegeneration. DISCUSSION CBF reduction occurs early in the AD continuum. te-ASL is more sensitive than single-PLD ASL at detecting CBF changes in AD. HIGHLIGHTS Lower CBF can be detected in CU subjects in the early AD continuum. te-ASL is more sensitive than single-PLD ASL at detecting CBF alterations in AD. CBF is linked to biomarkers of AD, synaptic dysfunction, and neurodegeneration.
Collapse
Affiliation(s)
- Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)Instituto de Salud Carlos IIIMadridSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
| | | | - Lena Václavů
- Department of Radiology, C. J. Gorter MRI CenterLeiden University Medical CenterLeidenNetherlands
| | | | - Carolina Minguillon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
| | - Diego Cascales‐Lahoz
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - José Contador
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - Aida Fernández‐Lebrero
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - Irene Navalpotro
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - Albert Puig‐Pijoan
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | | | | | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
| | - Henrik Zetterberg
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at University of GothenburgMölndalSweden
- UK Dementia Research Institute at University College London (UCL)LondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Kaj Blennow
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at University of GothenburgMölndalSweden
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)Instituto de Salud Carlos IIIMadridSpain
- Servei de NeurologiaHospital del MarPg. Marítim de la BarcelonetaBarcelonaSpain
| | - Matthias J. P. Van Osch
- Department of Radiology, C. J. Gorter MRI CenterLeiden University Medical CenterLeidenNetherlands
| | | | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)Instituto de Salud Carlos IIIMadridSpain
- Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento SaludableHospital del Mar Research InstituteBarcelonaSpain
| |
Collapse
|
39
|
Haddadi M, Haghi M, Rezaei N, Kiani Z, Akkülah T, Celik A. APOE and Alzheimer's disease: Pathologic clues from transgenic Drosophila melanogaster. Arch Gerontol Geriatr 2024; 123:105420. [PMID: 38537387 DOI: 10.1016/j.archger.2024.105420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/03/2024] [Accepted: 03/19/2024] [Indexed: 06/06/2024]
Abstract
Alzheimer's disease (AD) is one of the most common forms of neurodegenerative diseases. Apolipoprotein E4 (ApoE4) is the main genetic risk factor in the development of late-onset AD. However, the exact mechanism underlying ApoE4-mediated neurodegeneration remains unclear. We utilized Drosophila melanogaster to examine the neurotoxic effects of various human APOE isoforms when expressed specifically in glial and neural cells. We assessed impacts on mitochondrial dynamics, ER stress, lipid metabolism, and bio-metal ion concentrations in the central nervous system (CNS) of the transgenic flies. Dachshund antibody staining revealed a reduction in the number of Kenyon cells. Behavioral investigations including ethanol tolerance and learning and memory performance demonstrated neuronal dysfunction in APOE4-expressing larvae and adult flies. Transcription level of marf and drp-1 were found to be elevated in APOE4 flies, while atf4, atf6, and xbp-1 s showed down regulation. Enhanced concentrations of triglyceride and cholesterol in the CNS were observed in APOE4 transgenic flies, with especially pronounced effects upon glial-specific expression of the gene. Spectrophotometry of brain homogenate revealed enhanced Fe++ and Zn++ ion levels in conjunction with diminished Cu++ levels upon APOE4 expression. To explore therapeutic strategies, we subjected the flies to heat-shock treatment, aiming to activate heat-shock proteins (HSPs) and assess their potential to mitigate the neurotoxic effects of APOE isoforms. The results showed potential therapeutic benefits for APOE4-expressing flies, hinting at an ability to attenuate memory deterioration. Overall, our findings suggest that APOE4 can alter lipid metabolism, bio metal ion homeostasis, and disrupt the harmonious fission-fusion balance of neuronal and glial mitochondria, ultimately inducing ER stress. These alterations mirror the main clinical manifestations of AD in patients. Therefore, our work underscores the suitability of Drosophila as a fertile model for probing the pathological roles of APOE and furthering our understanding of diverse isoform-specific functions.
Collapse
Affiliation(s)
- Mohammad Haddadi
- Department of Biology, Faculty of Basic Sciences, University of Zabol, Zabol, Iran; Genetics and Non-communicable Diseases Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Mehrnaz Haghi
- Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Niloofar Rezaei
- Department of Biology, Faculty of Basic Sciences, University of Zabol, Zabol, Iran
| | - Zahra Kiani
- Department of Biology, Faculty of Basic Sciences, University of Zabol, Zabol, Iran
| | - Taha Akkülah
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkiye; Center for Life Sciences and Technologies, Bogazici University, Istanbul, Turkiye
| | - Arzu Celik
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkiye; Center for Life Sciences and Technologies, Bogazici University, Istanbul, Turkiye
| |
Collapse
|
40
|
Shin KC, Ali Moussa HY, Park Y. Cholesterol imbalance and neurotransmission defects in neurodegeneration. Exp Mol Med 2024; 56:1685-1690. [PMID: 39085348 PMCID: PMC11371908 DOI: 10.1038/s12276-024-01273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
The brain contains the highest concentration of cholesterol in the human body, which emphasizes the importance of cholesterol in brain physiology. Cholesterol is involved in neurogenesis and synaptogenesis, and age-related reductions in cholesterol levels can lead to synaptic loss and impaired synaptic plasticity, which potentially contribute to neurodegeneration. The maintenance of cholesterol homeostasis in the neuronal plasma membrane is essential for normal brain function, and imbalances in cholesterol distribution are associated with various neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, and Huntington's disease. This review aims to explore the molecular and pathological mechanisms by which cholesterol imbalance can lead to neurotransmission defects and neurodegeneration, focusing on four key mechanisms: (1) synaptic dysfunction, (2) alterations in membrane structure and protein clustering, (3) oligomers of amyloid beta (Aβ) protein, and (4) α-synuclein aggregation.
Collapse
Affiliation(s)
- Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Houda Yasmine Ali Moussa
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
- College of Health & Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| |
Collapse
|
41
|
Kounatidis D, Tentolouris N, Vallianou NG, Mourouzis I, Karampela I, Stratigou T, Rebelos E, Kouveletsou M, Stamatopoulos V, Tsaroucha E, Dalamaga M. The Pleiotropic Effects of Lipid-Modifying Interventions: Exploring Traditional and Emerging Hypolipidemic Therapies. Metabolites 2024; 14:388. [PMID: 39057711 PMCID: PMC11278853 DOI: 10.3390/metabo14070388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerotic cardiovascular disease poses a significant global health issue, with dyslipidemia standing out as a major risk factor. In recent decades, lipid-lowering therapies have evolved significantly, with statins emerging as the cornerstone treatment. These interventions play a crucial role in both primary and secondary prevention by effectively reducing cardiovascular risk through lipid profile enhancements. Beyond their primary lipid-lowering effects, extensive research indicates that these therapies exhibit pleiotropic actions, offering additional health benefits. These include anti-inflammatory properties, improvements in vascular health and glucose metabolism, and potential implications in cancer management. While statins and ezetimibe have been extensively studied, newer lipid-lowering agents also demonstrate similar pleiotropic effects, even in the absence of direct cardiovascular benefits. This narrative review explores the diverse pleiotropic properties of lipid-modifying therapies, emphasizing their non-lipid effects that contribute to reducing cardiovascular burden and exploring emerging benefits for non-cardiovascular conditions. Mechanistic insights into these actions are discussed alongside their potential therapeutic implications.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | | | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
42
|
Medegan Fagla B, York J, Christensen A, Dela Rosa C, Balu D, Pike CJ, Tai LM, Buhimschi IA. Apolipoprotein E polymorphisms and female fertility in a transgenic mouse model of Alzheimer's disease. Sci Rep 2024; 14:15873. [PMID: 38982272 PMCID: PMC11233746 DOI: 10.1038/s41598-024-66489-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024] Open
Abstract
Apolipoprotein E (APOE) is a major cholesterol carrier responsible for lipid transport and injury repair in the brain. The human APOE gene (h-APOE) has 3 naturally occurring alleles: ε3, the common allele; ε4, which increases Alzheimer's disease (AD) risk up to 15-fold; and ε2, the rare allele which protects against AD. Although APOE4 has negative effects on neurocognition in old age, its persistence in the population suggests a survival advantage. We investigated the relationship between APOE genotypes and fertility in EFAD mice, a transgenic mouse model expressing h-APOE. We show that APOE4 transgenic mice had the highest level of reproductive performance, followed by APOE3 and APOE2. Intriguingly, APOE3 pregnancies had more fetal resorptions and reduced fetal weights relative to APOE4 pregnancies. In conclusion, APOE genotypes impact fertility and pregnancy outcomes in female mice, in concordance with findings in human populations. These mouse models may help elucidate how h-APOE4 promotes reproductive fitness at the cost of AD in later life.
Collapse
Affiliation(s)
- Bani Medegan Fagla
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Amy Christensen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Cielo Dela Rosa
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, Chicago, IL, 60612, USA
| | - Irina A Buhimschi
- Department of Obstetrics Gynecology, University of Illinois at Chicago College of Medicine, 820 S. Wood Street, Chicago, IL, 60612, USA.
| |
Collapse
|
43
|
Constantinou C, Meliou K, Skouras A, Siafaka P, Christodoulou P. Liposomes against Alzheimer's Disease: Current Research and Future Prospects. Biomedicines 2024; 12:1519. [PMID: 39062092 PMCID: PMC11275096 DOI: 10.3390/biomedicines12071519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease, the most common neurodegenerative disease, affects more than 60 million people worldwide, a number that is estimated to double by 2050. Alzheimer's disease is characterized by progressive memory loss, the impairment of behavior, and mood changes, as well as the disturbed daily routine of the patient. Although there are some active molecules that can be beneficial by halting the progression of the disease, the blood-brain barrier and other physiological barriers hinder their delivery and, consequently, the appropriate management of the disease. Therefore, drug delivery systems that effectively target and overcome the blood-brain barrier to reach the targeted brain area would improve treatment effectiveness. Liposomes are lipophilic carriers that consist of a phospholipid bilayer structure, simulating the physiological lipidic layer of the blood-brain barrier and enabling better delivery of the drug to the brain. Given that pure liposomes may have less targeting affinity than functionalized liposomes, modification with groups such as lactoferrin, poly(ethylene glycol), and transferrin may improve specificity. In this mini-review, we summarize the literature on the use of liposomes for the treatment of Alzheimer's disease, focusing on the functionalization moieties of liposomes. In addition, challenges in brain delivery are also discussed.
Collapse
Affiliation(s)
- Christiana Constantinou
- Department of Life Sciences, School of Sciences, Pharmacy Program, European University Cyprus, 2404 Nicosia, Cyprus; (C.C.); (K.M.); (P.S.)
| | - Katerina Meliou
- Department of Life Sciences, School of Sciences, Pharmacy Program, European University Cyprus, 2404 Nicosia, Cyprus; (C.C.); (K.M.); (P.S.)
| | - Athanasios Skouras
- Department of Nursing, Faculty of Health Sciences, Hellenic Mediterranean University, 71004 Heraklion, Crete, Greece;
| | - Panoraia Siafaka
- Department of Life Sciences, School of Sciences, Pharmacy Program, European University Cyprus, 2404 Nicosia, Cyprus; (C.C.); (K.M.); (P.S.)
| | | |
Collapse
|
44
|
Singhaarachchi PH, Antal P, Calon F, Culmsee C, Delpech JC, Feldotto M, Geertsema J, Hoeksema EE, Korosi A, Layé S, McQualter J, de Rooij SR, Rummel C, Slayo M, Sominsky L, Spencer SJ. Aging, sex, metabolic and life experience factors: Contributions to neuro-inflammaging in Alzheimer's disease research. Neurosci Biobehav Rev 2024; 162:105724. [PMID: 38762130 DOI: 10.1016/j.neubiorev.2024.105724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease (AD) is prevalent around the world, yet our understanding of the disease is still very limited. Recent work suggests that the cornerstone of AD may include the inflammation that accompanies it. Failure of a normal pro-inflammatory immune response to resolve may lead to persistent central inflammation that contributes to unsuccessful clearance of amyloid-beta plaques as they form, neuronal death, and ultimately cognitive decline. Individual metabolic, and dietary (lipid) profiles can differentially regulate this inflammatory process with aging, obesity, poor diet, early life stress and other inflammatory factors contributing to a greater risk of developing AD. Here, we integrate evidence for the interface between these factors, and how they contribute to a pro-inflammatory brain milieu. In particular, we discuss the importance of appropriate polyunsaturated fatty acids (PUFA) in the diet for the metabolism of specialised pro-resolving mediators (SPMs); raising the possibility for dietary strategies to improve AD outlook.
Collapse
Affiliation(s)
| | - Peter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, 1111, Hungary
| | - Frédéric Calon
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec-Laval University, Quebec G1V0A6, Canada; International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, Marburg D-35032, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany
| | - Jean-Christophe Delpech
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Martin Feldotto
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Jorine Geertsema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Emmy E Hoeksema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Sophie Layé
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Jonathan McQualter
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, 1018, the Netherlands
| | - Christoph Rummel
- Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Mary Slayo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Luba Sominsky
- Barwon Health, Geelong, Victoria 3220, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria 3217, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
45
|
AmeliMojarad M, AmeliMojarad M. The neuroinflammatory role of microglia in Alzheimer's disease and their associated therapeutic targets. CNS Neurosci Ther 2024; 30:e14856. [PMID: 39031970 PMCID: PMC11259573 DOI: 10.1111/cns.14856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
INTRODUCTION Alzheimer's disease (AD), the main cause of dementia, is characterized by synaptic loss and neurodegeneration. Amyloid-β (Aβ) accumulation, hyperphosphorylation of tau protein, and neurofibrillary tangles (NFTs) in the brain are considered to be the initiating factors of AD. However, this hypothesis falls short of explaining many aspects of AD pathogenesis. Recently, there has been mounting evidence that neuroinflammation plays a key role in the pathophysiology of AD and causes neurodegeneration by over-activating microglia and releasing inflammatory mediators. METHODS PubMed, Web of Science, EMBASE, and MEDLINE were used for searching and summarizing all the recent publications related to inflammation and its association with Alzheimer's disease. RESULTS Our review shows how inflammatory dysregulation influences AD pathology as well as the roles of microglia in neuroinflammation, the possible microglia-associated therapeutic targets, top neuroinflammatory biomarkers, and anti-inflammatory drugs that combat inflammation. CONCLUSION In conclusion, microglial inflammatory reactions are important factors in AD pathogenesis and need to be discussed in more detail for promising therapeutic strategies.
Collapse
Affiliation(s)
- Melika AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| | - Mandana AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental BiotechnologyNational Institute of Genetic Engineering and BiotechnologyTehranIran
| |
Collapse
|
46
|
Wang N, Cai L, Pei X, Lin Z, Huang L, Liang C, Wei M, Shao L, Guo T, Huang F, Luo H, Zheng H, Chen XF, Leng L, Zhang YW, Wang X, Zhang J, Guo K, Wang Z, Zhang H, Zhao Y, Xu H. Microglial apolipoprotein E particles contribute to neuronal senescence and synaptotoxicity. iScience 2024; 27:110006. [PMID: 38868202 PMCID: PMC11167441 DOI: 10.1016/j.isci.2024.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/13/2023] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Apolipoprotein E (apoE) plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Microglia exhibit a substantial upregulation of apoE in AD-associated circumstances, despite astrocytes being the primary source of apoE expression and secretion in the brain. Although the role of astrocytic apoE in the brain has been extensively investigated, it remains unclear that whether and how apoE particles generated from astrocytes and microglia differ in biological characteristic and function. Here, we demonstrate the differences in size between apoE particles generated from microglia and astrocytes. Microglial apoE particles impair neurite growth and synapses, and promote neuronal senescence, whereas depletion of GPNMB (glycoprotein non-metastatic melanoma protein B) in microglial apoE particles mitigated these deleterious effects. In addition, human APOE4-expressing microglia are more neurotoxic than APOE3-bearing microglia. For the first time, these results offer concrete evidence that apoE particles produced by microglia are involved in neuronal senescence and toxicity.
Collapse
Affiliation(s)
- Na Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lujian Cai
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xinyu Pei
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Zhihao Lin
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lihong Huang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Chensi Liang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Min Wei
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lin Shao
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Tiantian Guo
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Fang Huang
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Hong Luo
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Honghua Zheng
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xiao-fen Chen
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Lige Leng
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Yun-wu Zhang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Xin Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen 361102, China
| | - Jie Zhang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Kai Guo
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Zhanxiang Wang
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Hongsheng Zhang
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing 400016, China
| | - Yingjun Zhao
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Huaxi Xu
- Center for Brain Sciences, First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of Medicine, Xiamen University, Xiamen, Fujian 361005, China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
47
|
Nacarelli GS, Fasolino T, Davis S. Dietary, macronutrient, micronutrient, and nutrigenetic factors impacting cardiovascular risk markers apolipoprotein B and apolipoprotein A1: a narrative review. Nutr Rev 2024; 82:949-962. [PMID: 37615981 DOI: 10.1093/nutrit/nuad102] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023] Open
Abstract
Genetic predisposition and dietary factors can impact cardiovascular disease (CVD) risk. Two important markers in assessing CVD risk are apolipoprotein (apo) B and apolipoprotein A1 plasma levels. These markers are measured as a ratio, with a high apoB:apoA1 ratio associated with increased CVD risk. Dietary and lifestyle recommendations are the cornerstone of managing primary and secondary CVD risk-mitigation strategies. One way to assess the impact of various dietary and lifestyle interventions on CVD risk is to evaluate the changes in CVD risk markers, such as apoB, apoA1, and apoB:apoA1 ratio. Various human studies have demonstrated the impact of dietary, macronutrient, and micronutrient interventions on apoB and apoA1 status. This review aims to elucidate dietary, macronutrient, micronutrient, and nutrigenetic considerations for impacting apoB and apoA1 levels. A low-carbohydrate, high-saturated-fat diet, low fiber intake, low vitamin and mineral intake, and zinc and iron deficiency are associated with an elevated apoB:apoA1 ratio. The Mediterranean diet, vegan diet, fermented dairy products, lower sugar intake, higher protein intake, higher polyunsaturated fat intake, and an omega-3-rich diet are associated with a decreased apoB:apoA1 ratio. Micronutrients associated with a decreased apoB:apoA1 ratio include vitamin D sufficiency, increased serum vitamin C, and magnesium. Variants in the APOE, APOA1, and FADS2 genes may alter the apoB:apoA1 ratio in response to various dietary interventions. When accounting for factors that may favorably alter the apoB:apoA1 ratio, researchers should consider a healthy diet sufficient in polyunsaturated fats, vitamins, minerals, trace minerals, and lower excess sugars.
Collapse
Affiliation(s)
| | - Tracy Fasolino
- Clemson School of Nursing, Clemson University, Clemson, South Carolina, USA
| | - Stephanie Davis
- Clemson School of Nursing, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
48
|
Rahman MS, Harrison E, Biggs H, Seikus C, Elliott P, Breen G, Kingston N, Bradley JR, Hill SM, Tom BDM, Chinnery PF. Dynamics of cognitive variability with age and its genetic underpinning in NIHR BioResource Genes and Cognition cohort participants. Nat Med 2024; 30:1739-1748. [PMID: 38745010 PMCID: PMC11186791 DOI: 10.1038/s41591-024-02960-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/28/2024] [Indexed: 05/16/2024]
Abstract
A leading explanation for translational failure in neurodegenerative disease is that new drugs are evaluated late in the disease course when clinical features have become irreversible. Here, to address this gap, we cognitively profiled 21,051 people aged 17-85 years as part of the Genes and Cognition cohort within the National Institute for Health and Care Research BioResource across England. We describe the cohort, present cognitive trajectories and show the potential utility. Surprisingly, when studied at scale, the APOE genotype had negligible impact on cognitive performance. Different cognitive domains had distinct genetic architectures, with one indicating brain region-specific activation of microglia and another with glycogen metabolism. Thus, the molecular and cellular mechanisms underpinning cognition are distinct from dementia risk loci, presenting different targets to slow down age-related cognitive decline. Participants can now be recalled stratified by genotype and cognitive phenotype for natural history and interventional studies of neurodegenerative and other disorders.
Collapse
Affiliation(s)
- Md Shafiqur Rahman
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Emma Harrison
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research BioResource, Cambridge, UK
| | - Heather Biggs
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research BioResource, Cambridge, UK
| | - Chloe Seikus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research BioResource, Cambridge, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- UK National Institute for Health Research Biomedical Research Centre for Mental Health, South London and Maudsley Hospital, London, UK
| | - Nathalie Kingston
- National Institute for Health and Care Research BioResource, Cambridge, UK
- Dept of Haematology, Cambridge University, Cambridge, UK
| | - John R Bradley
- National Institute for Health and Care Research BioResource, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Steven M Hill
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester, UK
| | - Brian D M Tom
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK.
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
- National Institute for Health and Care Research BioResource, Cambridge, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
49
|
Grenon MB, Papavergi MT, Bathini P, Sadowski M, Lemere CA. Temporal Characterization of the Amyloidogenic APPswe/PS1dE9;hAPOE4 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:5754. [PMID: 38891941 PMCID: PMC11172317 DOI: 10.3390/ijms25115754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating disorder with a global prevalence estimated at 55 million people. In clinical studies administering certain anti-beta-amyloid (Aβ) antibodies, amyloid-related imaging abnormalities (ARIAs) have emerged as major adverse events. The frequency of these events is higher among apolipoprotein ε4 allele carriers (APOE4) compared to non-carriers. To reflect patients most at risk for vascular complications of anti-Aβ immunotherapy, we selected an APPswe/PS1dE9 transgenic mouse model bearing the human APOE4 gene (APPPS1:E4) and compared it with the same APP/PS1 mouse model bearing the human APOE3 gene (APOE ε3 allele; APPPS1:E3). Using histological and biochemical analyses, we characterized mice at three ages: 8, 12, and 16 months. Female and male mice were assayed for general cerebral fibrillar and pyroglutamate (pGlu-3) Aβ deposition, cerebral amyloid angiopathy (CAA), microhemorrhages, apoE and cholesterol composition, astrocytes, microglia, inflammation, lysosomal dysfunction, and neuritic dystrophy. Amyloidosis, lipid deposition, and astrogliosis increased with age in APPPS1:E4 mice, while inflammation did not reveal significant changes with age. In general, APOE4 carriers showed elevated Aβ, apoE, reactive astrocytes, pro-inflammatory cytokines, microglial response, and neuritic dystrophy compared to APOE3 carriers at different ages. These results highlight the potential of the APPPS1:E4 mouse model as a valuable tool in investigating the vascular side effects associated with anti-amyloid immunotherapy.
Collapse
Affiliation(s)
- Martine B. Grenon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Section Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maria-Tzousi Papavergi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Praveen Bathini
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| | - Martin Sadowski
- Departments of Neurology, Psychiatry, and Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| |
Collapse
|
50
|
Zhu Z, Zhong X, Wang B, Lu H, Li L. Probing Protein Structural Changes in Alzheimer's Disease via Quantitative Cross-linking Mass Spectrometry. Anal Chem 2024; 96:7506-7515. [PMID: 38690851 PMCID: PMC11114074 DOI: 10.1021/acs.analchem.4c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder featuring abnormal protein aggregation in the brain, including the pathological hallmarks of amyloid plaques and hyperphosphorylated tau. Despite extensive research efforts, understanding the molecular intricacies driving AD development remains a formidable challenge. This study focuses on identifying key protein conformational changes associated with the progression of AD. To achieve this, we employed quantitative cross-linking mass spectrometry (XL-MS) to elucidate conformational changes in the protein networks in cerebrospinal fluid (CSF). By using isotopically labeled cross-linkers BS3d0 and BS3d4, we reveal a dynamic shift in protein interaction networks during AD progression. Our comprehensive analysis highlights distinct alterations in protein-protein interactions within mild cognitive impairment (MCI) states. This study accentuates the potential of cross-linked peptides as indicators of AD-related conformational changes, including previously unreported site-specific binding between α-1-antitrypsin (A1AT) and complement component 3 (CO3). Furthermore, this work enables detailed structural characterization of apolipoprotein E (ApoE) and reveals modifications within its helical domains, suggesting their involvement in MCI pathogenesis. The quantitative approach provides insights into site-specific interactions and changes in the abundance of cross-linked peptides, offering an improved understanding of the intricate protein-protein interactions underlying AD progression. These findings lay a foundation for the development of potential diagnostic or therapeutic strategies aimed at mitigating the negative impact of AD.
Collapse
Affiliation(s)
- Zexin Zhu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Xiaofang Zhong
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Department of Cellular and Molecular Pharmacology, University of California-San Francisco, San Francisco, CA 94158, USA
| | - Bin Wang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Haiyan Lu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|