1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Henningfield CM, Ngo M, Murray KM, Kwang NE, Tsourmas KI, Neumann J, Pashkutz ZA, Kawauchi S, Swarup V, Lane TE, MacGregor GR, Green KN. Generation of an Inducible Destabilized-Domain Cre Mouse Line to Target Disease Associated Microglia. Glia 2025; 73:1272-1287. [PMID: 39988890 DOI: 10.1002/glia.70004] [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: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed mouse lines that express either Cre recombinase (Cre) for constitutive targeting, or destabilized-domain Cre recombinase (DD-Cre) for inducible targeting from the Cst7 locus (Cst7 DD-Cre) to specifically manipulate disease associated microglia (DAM) and crossed with Ai14 tdTomato cre-reporter line mice. Cst7Cre was found to target all brain resident myeloid cells, due to transient developmental expression of Cst7, but no expression was found in the inducible Cst7 DD-Cre mice. Further crossing of this line with 5xFAD mice combined with dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in DAM without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. These models allow either complete cre-loxP targeting of all brain myeloid cells (Cst7Cre), or inducible targeting of DAMs, without leakiness (Cst7 DD-Cre).
Collapse
Affiliation(s)
- Caden M Henningfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Minh Ngo
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kaitlin M Murray
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Zachary A Pashkutz
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Shimako Kawauchi
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Thomas E Lane
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
- Center for Virus Research, University of California, Irvine, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
3
|
Lee H, Pearse RV, Lish AM, Pan C, Augur ZM, Terzioglu G, Gaur P, Liao M, Fujita M, Tio ES, Duong DM, Felsky D, Seyfried NT, Menon V, Bennett DA, De Jager PL, Young‐Pearse TL. Contributions of Genetic Variation in Astrocytes to Cell and Molecular Mechanisms of Risk and Resilience to Late-Onset Alzheimer's Disease. Glia 2025; 73:1166-1187. [PMID: 39901616 PMCID: PMC12012329 DOI: 10.1002/glia.24677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 02/05/2025]
Abstract
Reactive astrocytes are associated with Alzheimer's disease (AD), and several AD genetic risk variants are associated with genes highly expressed in astrocytes. However, the contribution of genetic risk within astrocytes to cellular processes relevant to the pathogenesis of AD remains ill-defined. Here, we present a resource for studying AD genetic risk in astrocytes using a large collection of induced pluripotent stem cell (iPSC) lines from deeply phenotyped individuals with a range of neuropathological and cognitive outcomes. IPSC lines from 44 individuals were differentiated into astrocytes followed by unbiased molecular profiling using RNA sequencing and tandem mass tag-mass spectrometry. We demonstrate the utility of this resource in examining gene- and pathway-level associations with clinical and neuropathological traits, as well as in analyzing genetic risk and resilience factors through parallel analyses of iPSC-astrocytes and brain tissue from the same individuals. Our analyses reveal that genes and pathways altered in iPSC-derived astrocytes from individuals with AD are concordantly dysregulated in AD brain tissue. This includes increased levels of prefoldin proteins, extracellular matrix factors, COPI-mediated trafficking components and reduced levels of proteins involved in cellular respiration and fatty acid oxidation. Additionally, iPSC-derived astrocytes from individuals resilient to high AD neuropathology show elevated basal levels of interferon response proteins and increased secretion of interferon gamma. Correspondingly, higher polygenic risk scores for AD are associated with lower levels of interferon response proteins in astrocytes. This study establishes an experimental system that integrates genetic information with a matched iPSC lines and brain tissue data from a large cohort of individuals to identify genetic contributions to molecular pathways affecting AD risk and resilience.
Collapse
Affiliation(s)
- Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Richard V. Pearse
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Alexandra M. Lish
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Cheryl Pan
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Zachary M. Augur
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Gizem Terzioglu
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Pallavi Gaur
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Meichen Liao
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Earvin S. Tio
- Department of Psychiatry and Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
| | - Duc M. Duong
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Daniel Felsky
- Department of Psychiatry and Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Nicholas T. Seyfried
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and the Taub Institute for the Study of Alzheimer's Disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Tracy L. Young‐Pearse
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Harvard Stem Cell InstituteHarvard UniversityCambridgeMassachusettsUSA
| |
Collapse
|
4
|
Huda TI, Fawcett TJ, Green AJ, Blanck G. Association of the HLA-DQB1*02 allele with lower tauopathy in Alzheimer's disease. Arch Gerontol Geriatr 2025; 133:105802. [PMID: 40068480 DOI: 10.1016/j.archger.2025.105802] [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: 12/18/2024] [Revised: 02/10/2025] [Accepted: 02/23/2025] [Indexed: 04/05/2025]
Abstract
Although many studies have investigated the influence of HLA on the risk of Alzheimer's Disease (AD), there have been inconsistent results. Part of this problem has been attributed to limitations of a clinical assessment in the absence of histopathological confirmation or other quantitative assessments. This study employed a subset of the AD Sequencing Project, representing 2663 cases with histopathological confirmation of AD and confirmation of 881 cognitively normal cases. Two HLA allelic subtypes, DQB1*02:01 and DQB1*02:02, were associated with lower Braak staging, a measure of tauopathy in the brain. These HLA subtypes were also associated with a later age of onset. There was a lower occurrence of HLA-DQB1*02:01 and HLA-DQB1*02:02 in AD cases compared to cognitively normal cases. For all of the above results, replicative sets were confirmatory. The above results were also maintained for both HLA-DQB1*02:01 and HLA-DQB1*02:02 when removing the effect of APOE4 or APOE2. Interestingly, the HLA-DQB1*02 allele binds tau better than all other HLA-DQB1 alleles tested, per an in silico assessment, raising the question of whether deletion of tau binding, auto-reactive T-cells in the thymus could reduce the likelihood of the onset of AD?
Collapse
Affiliation(s)
- Taha I Huda
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Timothy J Fawcett
- Research Computing, University of South Florida, Tampa, FL 33620, USA
| | - Anthony J Green
- Research Computing, University of South Florida, Tampa, FL 33620, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| |
Collapse
|
5
|
Paul JK, Malik A, Azmal M, Gulzar T, Afghan MTR, Talukder OF, Shahzadi S, Ghosh A. Advancing Alzheimer's Therapy: Computational strategies and treatment innovations. IBRO Neurosci Rep 2025; 18:270-282. [PMID: 39995567 PMCID: PMC11849200 DOI: 10.1016/j.ibneur.2025.02.002] [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: 08/12/2024] [Revised: 01/22/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative condition distinguished by the occurrence of memory impairment, cognitive deterioration, and neuronal impairment. Despite extensive research efforts, conventional treatment strategies primarily focus on symptom management, highlighting the need for innovative therapeutic approaches. This review explores the challenges of AD treatment and the integration of computational methodologies to advance therapeutic interventions. A comprehensive analysis of recent literature was conducted to elucidate the broad scope of Alzheimer's etiology and the limitations of conventional drug discovery approaches. Our findings underscore the critical role of computational models in elucidating disease mechanisms, identifying therapeutic targets, and expediting drug discovery. Through computational simulations, researchers can predict drug efficacy, optimize lead compounds, and facilitate personalized medicine approaches. Moreover, machine learning algorithms enhance early diagnosis and enable precision medicine strategies by analyzing multi-modal datasets. Case studies highlight the application of computational techniques in AD therapeutics, including the suppression of crucial proteins implicated in disease progression and the repurposing of existing drugs for AD management. Computational models elucidate the interplay between oxidative stress and neurodegeneration, offering insights into potential therapeutic interventions. Collaborative efforts between computational biologists, pharmacologists, and clinicians are essential to translate computational insights into clinically actionable interventions, ultimately improving patient outcomes and addressing the unmet medical needs of individuals affected by AD. Overall, integrating computational methodologies represents a promising paradigm shift in AD therapeutics, offering innovative solutions to overcome existing challenges and transform the landscape of AD treatment.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Abbeha Malik
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tooba Gulzar
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Muhammad Talal Rahim Afghan
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Samar Shahzadi
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|
6
|
Price R, Ramirez-Moreno M, Cooper A, Singh R, Ming Khaw Y, Mudiwa Mhaka A, Sivanantharajah L, Mudher A. Are we missing a trick by not exploiting fruit flies in inflammation-led drug discovery for neurodegeneration? Expert Opin Drug Discov 2025. [PMID: 40372417 DOI: 10.1080/17460441.2025.2498675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) remains a formidable challenge in neurodegeneration research, with limited therapeutic options despite decades of study. While Drosophila melanogaster has been instrumental in in modeling AD related Tau and amyloid beta toxicity, inflammation, a key driver of AD pathology, remains unexplored in fly models. Given the evolutionary conservation of innate immune pathways between flies and mammals, drosophila presents a powerful yet underutilized tool for inflammation led drug discovery in AD. AREAS COVERED This perspective highlights the relevance of Drosophila in studying neuroinflammatory processes, including microglial-like glial activation, systemic inflammation and gut-brain axis interactions. It further explores how fly models can be leveraged to screen anti-inflammatory compounds and dissect immune related genetic factors implicated in AD. EXPERT OPINION By integrating immune modulation in Drosophila-based drug discovery pipeline we can accelerate the identification of novel therapeutic strategies. Fully exploiting the potential of Drosophila in inflammation led drug screening may usher in a new era of AD therapeutics, bridging gaps between fundamental research and translational medicine.
Collapse
Affiliation(s)
- Ray Price
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Miguel Ramirez-Moreno
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Amber Cooper
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Rachita Singh
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | | | | | | | - Amrit Mudher
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
7
|
Kjeldsen EW, Frikke-Schmidt R. Causal cardiovascular risk factors for dementia: insights from observational and genetic studies. Cardiovasc Res 2025; 121:537-549. [PMID: 39498825 PMCID: PMC12054631 DOI: 10.1093/cvr/cvae235] [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: 07/18/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
The escalating prevalence of dementia worldwide necessitates preventive strategies to mitigate its extensive health, psychological, and social impacts. As the prevalence of dementia continues to rise, gaining insights into its risk factors and causes becomes paramount, given the absence of a definitive cure. Cardiovascular disease has emerged as a prominent player in the complex landscape of dementia. Preventing dyslipidaemia, unhealthy western-type diets, hypertension, diabetes, being overweight, physical inactivity, smoking, and high alcohol intake have the potential to diminish not only cardiovascular disease but also dementia. The purpose of this review is to present our current understanding of cardiovascular risk factors for Alzheimer's disease and vascular dementia (VaD) by using clinical human data from observational, genetic studies and clinical trials, while elaborating on potential mechanisms. Hypertension and Type 2 diabetes surface as significant causal risk factors for both Alzheimer's disease and VaD, as consistently illustrated in observational and Mendelian randomization studies. Anti-hypertensive drugs and physical activity have been shown to improve cognitive function in clinical trials. Important to note is that robust genome-wide association studies are lacking for VaD, and indeed more and prolonged clinical trials are needed to establish these findings and investigate other risk factors. Trials should strategically target individuals at the highest dementia risk, identified using risk charts incorporating genetic markers, biomarkers, and cardiovascular risk factors. Understanding causal risk factors for dementia will optimize preventive measures, and the implementation of well-known therapeutics can halt or alleviate dementia symptoms if started early. Needless to mention is that future health policies should prioritize primordial prevention from early childhood to prevent risk factors from even occurring in the first place. Together, understanding the role of cardiovascular risk factors in dementia, improving genome-wide association studies for VaD, and advancing clinical trials are crucial steps in addressing this significant public health challenge.
Collapse
Affiliation(s)
- Emilie Westerlin Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital—Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Zhao X, Li Y, Zhang S, Sudwarts A, Zhang H, Kozlova A, Moulton MJ, Goodman LD, Pang ZP, Sanders AR, Bellen HJ, Thinakaran G, Duan J. Alzheimer's disease protective allele of Clusterin modulates neuronal excitability through lipid-droplet-mediated neuron-glia communication. Mol Neurodegener 2025; 20:51. [PMID: 40319306 PMCID: PMC12049787 DOI: 10.1186/s13024-025-00840-1] [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: 01/17/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) of Alzheimer's disease (AD) have identified a plethora of risk loci. However, the disease variants/genes and the underlying mechanisms have not been extensively studied. METHODS Bulk ATAC-seq was performed in induced pluripotent stem cells (iPSCs) differentiated various brain cell types to identify allele-specific open chromatin (ASoC) SNPs. CRISPR-Cas9 editing generated isogenic pairs, which were then differentiated into glutamatergic neurons (iGlut). Transcriptomic analysis and functional studies of iGlut co-cultured with mouse astrocytes assessed neuronal excitability and lipid droplet formation. RESULTS We identified a putative causal SNP of CLU that impacted neuronal chromatin accessibility to transcription-factor(s), with the AD protective allele upregulating neuronal CLU and promoting neuron excitability. And, neuronal CLU facilitated neuron-to-glia lipid transfer and astrocytic lipid droplet formation coupled with reactive oxygen species (ROS) accumulation. These changes caused astrocytes to uptake less glutamate thereby altering neuron excitability. CONCLUSIONS For a strong AD-associated locus near Clusterin (CLU), we connected an AD protective allele to a role of neuronal CLU in promoting neuron excitability through lipid-mediated neuron-glia communication. Our study provides insights into how CLU confers resilience to AD through neuron-glia interactions.
Collapse
Affiliation(s)
- Xiaojie Zhao
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, 60637, USA
| | - Yan Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Siwei Zhang
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, 60637, USA
| | - Ari Sudwarts
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33160, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA
| | - Alena Kozlova
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, 60637, USA
| | - Matthew J Moulton
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lindsey D Goodman
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Johnson Medical School, Child Health Institute of New Jersey, Rutgers Robert Wood, New Brunswick, NJ, 08901, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, 60637, USA
| | - Hugo J Bellen
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gopal Thinakaran
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33160, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, Endeavor Health, Evanston, IL, 60201, USA.
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
9
|
Jia P, Hao Z, Yiu K, Tsoi K. Causal Effects Between Blood Pressure Variability and Alzheimer's Disease: A Two-Sample Mendelian Randomization Study. J Clin Hypertens (Greenwich) 2025; 27:e70066. [PMID: 40346856 PMCID: PMC12064930 DOI: 10.1111/jch.70066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/10/2025] [Accepted: 04/21/2025] [Indexed: 05/12/2025]
Abstract
Alzheimer's disease (AD), an escalating global public health concern, demonstrates complex pathogenesis involving both genetic predisposition and vascular components. Blood pressure variability (BPV) has been implicated in neurodegenerative diseases, but its causal relationship with AD remains unclear. This study aims to explore the causal relationship between BPV and AD by applying Mendelian randomization (MR) to genome-wide association study (GWAS) summary data. Genetic instruments were selected from BPV GWAS based on UK Biobank data, ensuring relevance and significance(p < 5 × 10⁻⁶). Genetic estimates on exposure were obtained from three databases: The The International Genomic of Alzheimer's Project (IGAP); Maternal family history of AD from UK Biobank (MFH-UKBB), and Paternal family history of AD from UK Biobank (PFH-UKBB). Proxy SNPs were manually selected if SNPs were not available in the exposure GWAS. Data harmonization was performed to ensure consistency in effect and reference alleles. Three MR statistical methods were employed to assess causal effects, including inverse variance weighting (IVW) with random or fixed effect, MR-Egger regression, and the Weighted Median Method. Sensitivity analyses to evaluate robustness were also employed. Six SNPs associated with systolic BPV and six SNPs associated with diastolic BPV were included. Significant causal effects of SBPV on AD were found on the PFH-UKBB dataset in all four methods. The odds ratios for AD per 10-unit increment in SBPV were 1.028, 1.015, and 1.015 for MR-Egger, IVW-MR, and weighted median, respectively. In contrast, only IVW methods found significant results for DBPV in the MFH-UKBB dataset. SBPV is a possible causal risk factor for AD, while the evidence for DBPV needs further study. BPV control should be an important treatment target in preventing dementia.
Collapse
Affiliation(s)
- Pingping Jia
- JC School of Public Health and Primary CareThe Chinese University of Hong KongHong KongHong Kong SAR
- Center for Clinical and Epidemiologic ResearchBeijing Anzhen Hospital, Capital Medical UniversityBeijingChina
- Lung and Blood Vessel DiseasesBeijing Institute of HeartBeijingChina
| | - Ziyu Hao
- JC School of Public Health and Primary CareThe Chinese University of Hong KongHong KongHong Kong SAR
- Stanley Ho Big Data Decision Analytics Research CentreThe Chinese University of Hong KongHong KongHong Kong SAR
| | - Karen Yiu
- Lung and Blood Vessel DiseasesBeijing Institute of HeartBeijingChina
| | - Kelvin Tsoi
- JC School of Public Health and Primary CareThe Chinese University of Hong KongHong KongHong Kong SAR
- Lung and Blood Vessel DiseasesBeijing Institute of HeartBeijingChina
| |
Collapse
|
10
|
Lei S, Liu Y. Identifying microglia-derived NFKBIA as a potential contributor to the pathogenesis of Alzheimer's disease and age-related macular degeneration. J Alzheimers Dis 2025; 105:134-146. [PMID: 40105475 DOI: 10.1177/13872877251326267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
BackgroundAlzheimer's disease (AD) and age-related macular degeneration (AMD) place considerable health burden on affected individuals and significant economic burden on society.ObjectiveThis study aims to explore the shared cellular and molecular mechanisms underlying the pathogenesis of AD and AMD.MethodsThe investigation in this study is conducted via single-cell and bulk tissue transcriptomic analysis. Transcriptomic datasets of AD and AMD were obtained from the GEO database. The shared differentially expressed genes (DEGs) in control and AD- and AMD-affected samples were identified. Functional enrichment analysis for DEGs was subsequently performed. Then, the protein-protein interaction (PPI) network of these DEGs was established via the STRING database and hub genes of this network were identified by Cytoscape software. Single-cell transcriptomic analysis was performed using Seurat R package to explore their expression in different cell types.ResultsDifferential analysis identified 127 shared DEGs of the two diseases, including 71 upregulated and 56 downregulated genes. Upregulated DEGs were enriched in inflammation, gliogenesis, cell apoptosis, and response to bacterial and viral infection and downregulated DEGs were enriched in mitochondrial function and energy production. PPI network and Cytoscape determined 10 hub genes, of which the NFKBIA gene was associated with the severity of both AD and AMD. Moreover, single-cell transcriptomic analysis showed that NFKBIA was highly expressed in microglia from disease-affected tissues.ConclusionsThe findings indicated that microglia with high NFKBIA expression were important contributors to the progression of both AD and AMD. Microglia-derived NFKBIA might serve as a potential therapeutic target for AD and AMD.
Collapse
Affiliation(s)
- Shizhen Lei
- Department of Ophthalmology, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Yani Liu
- Department of Otolaryngology & Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, Hubei, China
| |
Collapse
|
11
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
12
|
Liu S, Bush WS, Akinyemi RO, Byrd GS, Caban-Holt AM, Rajabli F, Reitz C, Kunkle BW, Tosto G, Vance JM, Pericak-Vance M, Haines JL, Williams SM, Crawford DC. Alzheimer disease is (sometimes) highly heritable: Drivers of variation in heritability estimates for binary traits, a systematic review. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326648. [PMID: 40343016 PMCID: PMC12060970 DOI: 10.1101/2025.04.29.25326648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Estimating heritability has been fundamental in understanding the genetic contributions to complex disorders like late-onset Alzheimer's disease (LOAD), and provides a rationale for identifying genetic factors associated with disease susceptibility. While numerous studies have established substantial genetic contribution for LOAD, the interpretation of heritability estimates remains challenging. These challenges are further complicated by the binary nature of LOAD status, where estimation and interpretation require additional considerations. Through a systematic review, we identified LOAD heritability estimates from 6 twin studies and 17 genome-wide association studies, all conducted in populations of European ancestry. We demonstrate that these heritability estimates for LOAD vary considerably. The variation reflects not only differences in study design and methodological approaches but also the underlying study population characteristics. Our findings indicate that commonly cited heritability estimates, often treated as universal values, should be interpreted within specific population contexts and methodological frameworks.
Collapse
Affiliation(s)
- Shiying Liu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rufus Olusola Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Goldie S Byrd
- Maya Angelou Center for Health Equity, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Allison Mercedes Caban-Holt
- Department of Behavioral Science, College of Medicine and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Farid Rajabli
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christiane Reitz
- Gertrude H. Sergievsky Center, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology Columbia University, Department of Epidemiology, Columbia University, New York, NY, USA
| | - Brian W Kunkle
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University; Department of Neurology, Columbia University Irving Medical Center, Columbia University; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY USA
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Margaret Pericak-Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
13
|
Bankolé A, Srivastava A, Shihavuddin A, Tighanimine K, Faucourt M, Koka V, Weill S, Nemazanyy I, Nelson AJ, Stokes MP, Delgehyr N, Genovesio A, Meunier A, Fumagalli S, Pende M, Spassky N. mTOR controls ependymal cell differentiation by targeting the alternative cell cycle and centrosomal proteins. EMBO Rep 2025:10.1038/s44319-025-00460-2. [PMID: 40307619 DOI: 10.1038/s44319-025-00460-2] [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: 10/21/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Ependymal cells are multiciliated glial cells lining the ventricles of the mammalian brain. Their differentiation from progenitor cells involves cell enlargement and progresses through centriole amplification phases and ciliogenesis. These phases are accompanied by the sharp up-regulation of mTOR Complex 1 activity (mTORC1), a master regulator of macromolecule biosynthesis and cell growth, whose function in ependymal cell differentiation is unknown. We demonstrate that mTORC1 inhibition by rapamycin preserves the progenitor pool by reinforcing quiescence and preventing alternative cell cycle progression for centriole amplification. Overexpressing E2F4 and MCIDAS circumvents mTORC1-regulated processes, enabling centriole amplification despite rapamycin, and enhancing mTORC1 activity through positive feedback. Acute rapamycin treatment in multicentriolar cells during the late phases of differentiation causes centriole regrouping, indicating a direct role of mTORC1 in centriole dynamics. By phosphoproteomic and phosphomutant analysis, we reveal that the mTORC1-mediated phosphorylation of GAS2L1, a centrosomal protein that links actin and microtubule cytoskeletons, participates in centriole disengagement. This multilayered and sequential control of ependymal development by mTORC1, from the progenitor pool to centriolar function, has implications for pathophysiological conditions like aging and hydrocephalus-prone genetic diseases.
Collapse
Affiliation(s)
- Alexia Bankolé
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Asm Shihavuddin
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
- Department of EEE, Presidency University, Dhaka, Bangladesh
| | - Khaled Tighanimine
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Vonda Koka
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Solene Weill
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Alissa J Nelson
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Matthew P Stokes
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Auguste Genovesio
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Stefano Fumagalli
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Mario Pende
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France.
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France.
| |
Collapse
|
14
|
Lin J, Lei L, Liang Q, Huang X, Ding Y, Pan L, Yang J, Li W. Assessment of causality association between serum adiponectin levels and the risk of Alzheimer's disease and Parkinson's disease: a Mendelian randomization study. Front Neurol 2025; 16:1395798. [PMID: 40371086 PMCID: PMC12075267 DOI: 10.3389/fneur.2025.1395798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Background Until recently, the association between circulating adiponectin (ADPN) levels and the risk of Alzheimer's disease (AD) and Parkinson's disease (PD) remained unclear. Methods We utilized public data from the IEU GWAS database to conduct a two-sample bidirectional Mendelian randomization (MR) analysis and multiple sensitivity analyses. The MR analysis was performed using the aggregated data, with the genetic risk score (GRS) serving as an instrumental variable. Results The MR analyses revealed no significant causal association between genetically determined ADPN levels and the risk of AD (ORIVW = 0.852, 95% confidence interval [CI]: 0.586-1.117, p = 0.235) or PD (ORIVW = 0.830, 95% CI: 0.780-1.156, p = 0.606). Conversely, neither AD nor PD demonstrated any causal association with ADPN levels. The GRS approach yielded similar results (p > 0.05). However, it exhibited a negative correlation with interleukin 1β (IL1β, βIVW = -0.31; 95% CI: -0.55 to -0.07, p = 0.011). The Cochrane's Q test and MR-PRESSO analysis revealed no evidence of pleiotropy. Conclusion Our findings provide no evidence to substantiate a causal relationship between ADPN levels and the risk of AD and PD or vice versa. However, elevated levels of ADPN may correlate with lower levels of IL1β.
Collapse
Affiliation(s)
- Jiali Lin
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Langhuan Lei
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qiuyu Liang
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Xiaozhi Huang
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yanping Ding
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Liuxian Pan
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Jianrong Yang
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Wei Li
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| |
Collapse
|
15
|
Colín-Martínez E, Arias C. Involvement of the VGF/BDNF axis in the neuropathology of Alzheimer's disease and its potential role in diagnosis and treatment. Rev Neurosci 2025; 36:267-278. [PMID: 39566031 DOI: 10.1515/revneuro-2024-0110] [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: 08/13/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
The brain is a highly plastic organ that continually receives and integrates signals to generate functional and structural changes and homeostatic adaptations throughout life. Alterations in some signaling pathways that mediate these responses can impact brain plasticity, accelerate brain aging and potentially lead to neurodegeneration. There is substantial evidence that two important signaling pathways activated by neurotrophins, nonacronymic (VGF) and brain-derived neurotrophic factor (BDNF), are involved in substantial functions stimulating neuronal growth, differentiation, and circuit establishment during development and neuronal maintenance and plasticity in the mature brain. In this review, we present evidence that these two pathways and their interactions are central players in cognitive performance and alterations in pathological aging, particularly in conditions such as Alzheimer's disease (AD). Finally, we suggest specific avenues for future research on the basis of recent findings suggesting these molecules are diagnostic biomarkers and putative therapeutic tools to prevent, delay or improve AD neuropathology.
Collapse
Affiliation(s)
- Elizabeth Colín-Martínez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| |
Collapse
|
16
|
Murphy KB, Ye Y, Tsalenchuk M, Nott A, Marzi SJ. CHAS infers cell type-specific signatures in bulk brain histone acetylation studies of neurological and psychiatric disorders. CELL REPORTS METHODS 2025:101032. [PMID: 40300607 DOI: 10.1016/j.crmeth.2025.101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 05/01/2025]
Abstract
Epigenomic profiling of the brain has largely been done on bulk tissues, limiting our understanding of cell type-specific epigenetic changes in disease states. Here, we introduce cell type-specific histone acetylation score (CHAS), a computational tool for inferring cell type-specific signatures in bulk brain H3K27ac profiles. We applied CHAS to >300 H3K27ac chromatin immunoprecipitation sequencing samples from studies of Alzheimer's disease, Parkinson's disease, autism spectrum disorder, schizophrenia, and bipolar disorder in bulk postmortem brain tissue. In addition to recapitulating known disease-associated shifts in cellular proportions, we identified cell type-specific biological insights into brain-disorder-associated regulatory variation. In most cases, genetic risk and epigenetic dysregulation targeted different cell types, suggesting independent mechanisms. For instance, genetic risk of Alzheimer's disease was exclusively enriched within microglia, while epigenetic dysregulation predominantly fell within oligodendrocyte-specific H3K27ac regions. In addition, reanalysis of the original datasets using CHAS enabled identification of biological pathways associated with each neurological and psychiatric disorder at cellular resolution.
Collapse
Affiliation(s)
- Kitty B Murphy
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK.
| | - Yuqian Ye
- Department of Brain Sciences, Imperial College London, London, UK; UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria Tsalenchuk
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, UK; UK Dementia Research Institute at Imperial College London, London, UK
| | - Sarah J Marzi
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
17
|
Zhang W, Lukacsovich D, Young JI, Gomez L, Schmidt MA, Martin ER, Kunkle BW, Chen XS, O'Shea DM, Galvin JE, Wang L. DNA methylation signature of a lifestyle-based resilience index for cognitive health. Alzheimers Res Ther 2025; 17:88. [PMID: 40264239 PMCID: PMC12016380 DOI: 10.1186/s13195-025-01733-7] [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: 11/16/2024] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
Cognitive resilience (CR) contributes to the variability in risk for developing and progressing in Alzheimer's disease (AD) among individuals. Beyond genetics, recent studies highlight the critical role of lifestyle factors in enhancing CR and delaying cognitive decline. DNA methylation (DNAm), an epigenetic mechanism influenced by both genetic and environmental factors, including CR-related lifestyle factors, offers a promising pathway for understanding the biology of CR. We studied DNAm changes associated with the Resilience Index (RI), a composite measure of lifestyle factors, using blood samples from the Healthy Brain Initiative (HBI) cohort. After corrections for multiple comparisons, our analysis identified 19 CpGs and 24 differentially methylated regions significantly associated with the RI, adjusting for covariates age, sex, APOE ε4, and immune cell composition. The RI-associated methylation changes are significantly enriched in pathways related to lipid metabolism, synaptic plasticity, and neuroinflammation, and highlight the connection between cardiovascular health and cognitive function. By identifying RI-associated DNAm, our study provided an alternative approach to discovering future targets and treatment strategies for AD, complementary to the traditional approach of identifying disease-associated variants directly. Furthermore, we developed a Methylation-based Resilience Score (MRS) that successfully predicted future cognitive decline in an external dataset from the Alzheimer's Disease Neuroimaging Initiative (ADNI), even after accounting for age, sex, APOE ε4, years of education, baseline diagnosis, and baseline MMSE score. Our findings are particularly relevant for a better understanding of epigenetic architecture underlying cognitive resilience. Importantly, the significant association between baseline MRS and future cognitive decline demonstrated that DNAm could be a predictive marker for AD, laying the foundation for future studies on personalized AD prevention.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Juan I Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michael A Schmidt
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Eden R Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Brian W Kunkle
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - X Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Deirdre M O'Shea
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33433, USA.
| | - James E Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33433, USA.
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
18
|
Jamal R, Shaikh MA, Taleuzzaman M, Haque Z, Albratty M, Alam MS, Makeen HA, Zoghebi K, Saleh SF. Key biomarkers in Alzheimer's disease: Insights for diagnosis and treatment strategies. J Alzheimers Dis 2025:13872877251330500. [PMID: 40255041 DOI: 10.1177/13872877251330500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Alzheimer's disease (AD) remains a significant global health challenge, characterized by its progressive neurodegeneration and cognitive decline. The urgent need for early diagnosis and effective treatment necessitates the identification of reliable biomarkers that can illuminate the underlying pathophysiology of AD. This review provides a comprehensive overview of the latest advancements in biomarker research, focusing on their applications in diagnosis, prognosis, and therapeutic development. We delve into the multifaceted landscape of AD biomarkers, encompassing molecular, imaging, and fluid-based markers. The integration of these biomarkers, including amyloid-β and tau proteins, neuroimaging modalities, cerebrospinal fluid analysis, and genetic risk factors, offers a more nuanced understanding of AD's complex etiology. By leveraging the power of precision medicine, biomarker-driven approaches can enable personalized treatment strategies and enhance diagnostic accuracy. Moreover, this review highlights the potential of biomarker research to accelerate drug discovery and development. By identifying novel therapeutic targets and monitoring disease progression, biomarkers can facilitate the evaluation of experimental treatments and ultimately improve patient outcomes. In conclusion, this review underscores the critical role of biomarkers in advancing our comprehension of AD and driving the development of effective interventions. By providing a comprehensive overview of the current state-of-the-art, this work aims to inspire future research and contribute to the goal of conquering AD.
Collapse
Affiliation(s)
- Ruqaiya Jamal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Maulana Azad University, Jodhpur, Rajasthan, India
| | | | - Mohamad Taleuzzaman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Maulana Azad University, Jodhpur, Rajasthan, India
| | - Ziyaul Haque
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Maulana Azad University, Jodhpur, Rajasthan, India
- Department of Pharmaceutical Chemistry, AIKTC School of Pharmacy, Mumbai, India
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Md Shamsher Alam
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Safaa Fathy Saleh
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
19
|
Chen J, Hadi F, Wen X, Zhao W, Xu M, Xue S, Lin P, Calandrelli R, Richard JLC, Song Z, Li J, Amani A, Liu Y, Chen X, Zhong S. Transcriptional regulation by PHGDH drives amyloid pathology in Alzheimer's disease. Cell 2025:S0092-8674(25)00397-6. [PMID: 40273909 DOI: 10.1016/j.cell.2025.03.045] [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: 04/12/2024] [Revised: 02/06/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Virtually all individuals aged 65 or older develop at least early pathology of Alzheimer's disease (AD), yet most lack disease-causing mutations in APP, PSEN, or MAPT, and many do not carry the APOE4 risk allele. This raises questions about AD development in the general population. Although transcriptional dysregulation has not traditionally been a hallmark of AD, recent studies reveal significant epigenomic changes in late-onset AD (LOAD) patients. We show that altered expression of the LOAD biomarker phosphoglycerate dehydrogenase (PHGDH) modulates AD pathology in mice and human brain organoids independent of its enzymatic activity. PHGDH has an uncharacterized role in transcriptional regulation, promoting the transcription of inhibitor of nuclear factor kappa-B kinase subunit alpha (IKKa) and high-mobility group box 1 (HMGB1) in astrocytes, which suppress autophagy and accelerate amyloid pathology. A blood-brain-barrier-permeable small-molecule inhibitor targeting PHGDH's transcriptional function reduces amyloid pathology and improves AD-related behavioral deficits. These findings highlight transcriptional regulation in LOAD and suggest therapeutic strategies beyond targeting familial mutations.
Collapse
Affiliation(s)
- Junchen Chen
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Fatemeh Hadi
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Xingzhao Wen
- Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Wenxin Zhao
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Ming Xu
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Shuanghong Xue
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Riccardo Calandrelli
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | | | - Zhixuan Song
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Jessica Li
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Alborz Amani
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Yang Liu
- School of Biological Sciences, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Xu Chen
- Department of Neurosciences, University of California, San Diego, La Jolla, San Diego, CA, USA; Neuroscience Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, San Diego, CA, USA; Institute of Engineering in Medicine, University of California, San Diego, La Jolla, San Diego, CA, USA; Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, San Diego, CA, USA; Neuroscience Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
20
|
Liu H, Xie Y, Ji Y, Zhou Y, Xu J, Tang J, Liu N, Ding H, Qin W, Liu F, Yu C. Identification of genetic architecture shared between schizophrenia and Alzheimer's disease. Transl Psychiatry 2025; 15:150. [PMID: 40240757 PMCID: PMC12003746 DOI: 10.1038/s41398-025-03348-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Both schizophrenia (SCZ) and Alzheimer's disease (AD) are highly heritable brain disorders. Despite of the observed comorbidity and shared psychosis and cognitive decline between the two disorders, the genetic risk architecture shared by SCZ and AD remains largely unknown. Based on summary statistics of the currently available largest genome-wide association studies for SCZ (n = 130,644) and AD (n = 455,258) in individuals of European ancestry, we conducted conditional/conjunctional false discovery rate (FDR) analysis to enhance the statistical power for discovering more genetic associations with SCZ or AD and to detect the common genetic variants shared by both disorders. We found shared genetic architecture in SCZ conditioned on AD and vice versa across different levels of significance, indicating polygenic overlap. We found 268 (78 novel) SCZ-only and 125 (55 novel) AD-only SNPs at conditional FDR < 0.01, and 16 lead SNPs shared by SCZ and AD at conjunctional FDR < 0.05. Only half of the shared SNPs showed concordant effect direction, which was consistent with the modest genetic correlation (r = 0.097; P = 0.026) between the two disorders. This study provides evidence for polygenic overlap between SCZ and AD, suggesting the existence of the shared molecular genetic mechanisms, which may inform therapeutic targets that are applicable for both disorders.
Collapse
Affiliation(s)
- Huaigui Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingying Xie
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Ji
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yujing Zhou
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Tang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nana Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Ding
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, China.
- State Key Laboratory of Experimental Hematology, Tianjin, China.
| |
Collapse
|
21
|
Korbmacher M, Vidal‐Pineiro D, Wang M, van der Meer D, Wolfers T, Nakua H, Eikefjord E, Andreassen OA, Westlye LT, Maximov II. Cross-Sectional Brain Age Assessments Are Limited in Predicting Future Brain Change. Hum Brain Mapp 2025; 46:e70203. [PMID: 40235434 PMCID: PMC12000824 DOI: 10.1002/hbm.70203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
The concept of brain age (BA) describes an integrative imaging marker of brain health, often suggested to reflect aging processes. However, the degree to which cross-sectional MRI features, including BA, reflect past, ongoing, and future brain changes across different tissue types from macro- to microstructure remains controversial. Here, we use multimodal imaging data of 39,325 UK Biobank participants, aged 44-82 years at baseline and 2,520 follow-ups within 1.12-6.90 years to examine BA changes and their relationship to anatomical brain changes. We find insufficient evidence to conclude that BA reflects the rate of brain aging. However, modality-specific differences in brain ages reflect the state of the brain, highlighting diffusion and multimodal MRI brain age as potentially useful cross-sectional markers.
Collapse
Affiliation(s)
- Max Korbmacher
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
- Department of NeurologyNeuro‐SysMed Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University HospitalBergenNorway
- Mohn Medical Imaging and Visualization Centre (MMIV)BergenNorway
| | - Didac Vidal‐Pineiro
- Center for Lifespan Changes in Brain and Cognition, Department of PsychologyUniversity of OsloOsloNorway
| | - Meng‐Yun Wang
- Max Planck Institute for PsycholinguisticsNijmegenthe Netherlands
| | - Dennis van der Meer
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Thomas Wolfers
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental HealthUniversity of TübingenTübingenGermany
| | - Hajer Nakua
- Columbia University Irving Medical CentreColumbia UniversityNew York CityUSA
| | - Eli Eikefjord
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
- Department of NeurologyNeuro‐SysMed Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University HospitalBergenNorway
| | - Ole A. Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical MedicineUniversity of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
| | - Lars T. Westlye
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical MedicineUniversity of OsloOsloNorway
- KG Jebsen Centre for Neurodevelopmental DisordersUniversity of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Ivan I. Maximov
- Department of Health and FunctioningWestern Norway University of Applied SciencesBergenNorway
| |
Collapse
|
22
|
Pacherille AM, Viernes DR, Pedicone C, Fernandes S, Dungan OM, Dormann S, Wallach DR, Sudan R, Brooks R, Russo CM, Howard KT, Cosgrove MS, Kerr WG, Chisholm JD. Aminocholestane and Aminoandrostane Inhibitors of the SH2 Domain-Containing Inositol 5'-Phosphatase (SHIP). ChemMedChem 2025; 20:e202400597. [PMID: 39843392 PMCID: PMC12046972 DOI: 10.1002/cmdc.202400597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/03/2025] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
The SH2-containing inositol phosphatase (SHIP) has become an actively researched therapeutic target for a number of disorders, including Alzheimer's Disease, Graft-vs-Host disease, obesity and cancer. Analogs of the aminosteroid SHIP inhibitor 3α-aminocholestane (3AC) have been synthesized and tested. Analogs with improved water solubility have been identified. Deletion of the C17 alkyl group from the cholestane skeleton improves water solubility, however these compounds inhibit both SHIP1 and SHIP2. Enzyme kinetics imply that these molecules are competitive inhibitors of SHIP, binding at a site near where the substrate binds to the phosphatase. A model of the binding of the inhibitors within the active site of SHIP1 is proposed to explain the structure activity studies. Overall this work provides more water soluble aminosteroid pan-SHIP1/2 inhibitors that can be used for future studies of SHIP activity.
Collapse
Affiliation(s)
- Angela M Pacherille
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Dennis R Viernes
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Chiara Pedicone
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Sandra Fernandes
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Otto M Dungan
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Shawn Dormann
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Daniel R Wallach
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Raki Sudan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Robbie Brooks
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - Christopher M Russo
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Kyle T Howard
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| | - Michael S Cosgrove
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - William G Kerr
- Department of Microbiology & Immunology, SUNY Upstate Medical University, 766 Irving Avenue, Syracuse, NY, 13210, USA
| | - John D Chisholm
- Department of Chemistry, Syracuse University, 111 College Place, Syracuse, NY, 13244, USA
| |
Collapse
|
23
|
Brown AR, Fox GA, Kaplow IM, Lawler AJ, Phan BN, Gadey L, Wirthlin ME, Ramamurthy E, May GE, Chen Z, Su Q, McManus CJ, van de Weerd R, Pfenning AR. An in vivo systemic massively parallel platform for deciphering animal tissue-specific regulatory function. Front Genet 2025; 16:1533900. [PMID: 40270544 PMCID: PMC12016043 DOI: 10.3389/fgene.2025.1533900] [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: 11/25/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction: Transcriptional regulation is an important process wherein non-protein coding enhancer sequences play a key role in determining cell type identity and phenotypic diversity. In neural tissue, these gene regulatory processes are crucial for coordinating a plethora of interconnected and regionally specialized cell types, ensuring their synchronized activity in generating behavior. Recognizing the intricate interplay of gene regulatory processes in the brain is imperative, as mounting evidence links neurodevelopment and neurological disorders to non-coding genome regions. While genome-wide association studies are swiftly identifying non-coding human disease-associated loci, decoding regulatory mechanisms is challenging due to causal variant ambiguity and their specific tissue impacts. Methods: Massively parallel reporter assays (MPRAs) are widely used in cell culture to study the non-coding enhancer regions, linking genome sequence differences to tissue-specific regulatory function. However, widespread use in animals encounters significant challenges, including insufficient viral library delivery and library quantification, irregular viral transduction rates, and injection site inflammation disrupting gene expression. Here, we introduce a systemic MPRA (sysMPRA) to address these challenges through systemic intravenous AAV viral delivery. Results: We demonstrate successful transduction of the MPRA library into diverse mouse tissues, efficiently identifying tissue specificity in candidate enhancers and aligning well with predictions from machine learning models. We highlight that sysMPRA effectively uncovers regulatory effects stemming from the disruption of MEF2C transcription factor binding sites, single-nucleotide polymorphisms, and the consequences of genetic variations associated with late-onset Alzheimer's disease. Conclusion: SysMPRA is an effective library delivering method that simultaneously determines the transcriptional functions of hundreds of enhancers in vivo across multiple tissues.
Collapse
Affiliation(s)
- Ashley R. Brown
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Grant A. Fox
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Irene M. Kaplow
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Alyssa J. Lawler
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - BaDoi N. Phan
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Lahari Gadey
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Morgan E. Wirthlin
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Easwaran Ramamurthy
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Gemma E. May
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Ziheng Chen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Qiao Su
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - C. Joel McManus
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Robert van de Weerd
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Andreas R. Pfenning
- Ray and Stephanie Lane Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, United States
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Grabowska ME, Vaidya AU, Zhong X, Guardo C, Dickson AL, Babanejad M, Yan C, Xin Y, Mundo S, Peterson JF, Feng Q, Eaton J, Wen Z, Li B, Wei WQ. Multi-omics analysis reveals aspirin is associated with reduced risk of Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.07.25325038. [PMID: 40297415 PMCID: PMC12036415 DOI: 10.1101/2025.04.07.25325038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The urgent need for safe and effective therapies for Alzheimer's disease (AD) has spurred a growing interest in repurposing existing drugs to treat or prevent AD. In this study, we combined multi-omics and clinical data to investigate possible repurposing opportunities for AD. We performed transcriptome-wide association studies (TWAS) to construct gene expression signatures of AD from publicly available GWAS summary statistics, using both transcriptome prediction models for 49 tissues from the Genotype-Tissue Expression (GTEx) project and microglia-specific models trained on eQTL data from the Microglia Genomic Atlas (MiGA). We then identified compounds capable of reversing the AD-associated changes in gene expression observed in these signatures by querying the Connectivity Map (CMap) drug perturbation database. Out of >2,000 small-molecule compounds in CMap, aspirin emerged as the most promising AD repurposing candidate. To investigate the longitudinal effects of aspirin use on AD, we collected drug exposure and AD coded diagnoses from three independent sources of real-world data: electronic health records (EHRs) from Vanderbilt University Medical Center (VUMC) and the National Institutes of Health All of Us Research Program, along with national healthcare claims from the MarketScan Research Databases. In meta-analysis of EHR data from VUMC and All of Us , we found that aspirin use before age 65 was associated with decreased risk of incident AD (hazard ratio=0.76, 95% confidence interval [CI]: 0.64-0.89, P =0.001). Consistent with the findings utilizing EHR data, analysis of claims data from MarketScan revealed significantly lower odds of aspirin exposure among AD cases compared to matched controls (odds ratio=0.32, 95% CI: 0.28-0.38, P <0.001). Our results demonstrate the value of integrating genetic and clinical data for drug repurposing studies and highlight aspirin as a promising repurposing candidate for AD, warranting further investigation in clinical trials.
Collapse
|
25
|
Mamde S, Rose SE, Prater KE, Cochoit A, Lin YF, Smith I, Johnson CS, Reid AN, Qiu W, Strohbehn S, Keene CD, Rolf BA, Lin KZ, Lee SI, Garden GA, Blue EE, Young JE, Jayadev S. Genetic risk in endolysosomal network genes correlates with endolysosomal dysfunction across neural cell types in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643481. [PMID: 40166227 PMCID: PMC11956971 DOI: 10.1101/2025.03.16.643481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Late-onset Alzheimer's disease (LOAD) has a complex genomic architecture with risk variants in multiple pathways, including the endolysosomal network (ELN). Whether genetic risk in specific pathways correlates with corresponding biological dysfunction remains largely unknown. We developed an endolysosomal pathway-specific polygenic risk score (ePRS) using 13 established AD GWAS loci containing ELN genes. We investigated the association between ePRS and AD neuropathology, then examined cell-specific endolysosomal morphology and transcriptomic profiles in post-mortem dorsolateral prefrontal cortex samples from donors stratified by ePRS burden. We found that the ePRS was significantly associated with AD diagnosis and neuropathological measures, comparable to a pathway-agnostic PRS despite representing far fewer loci. High ePRS correlated with increased neuronal endosome volume, number and perinuclear aggregation, as well as enlarged microglial lysosomes, independent of AD pathology. Single-nucleus RNA sequencing revealed cell-type transcriptomic changes associated with ePRS status, including glutamatergic signaling, protein homeostasis, responses to DNA damage and immune function. Neurons, astrocytes, oligodendrocytes, and microglia showed varied gene expression patterns associated with ePRS burden. Conclusions: This study provides evidence that AD genetic risk variants harboring ELN genes correlate with endolysosomal dysfunction in human brain tissue. These findings suggest that pathway-specific genetic risk contributes to corresponding cellular pathology in AD and nominates candidate mechanisms by which ELN AD variants contribute to pathogenesis.
Collapse
|
26
|
Macías M, Alba-Linares JJ, Acha B, Blanco-Luquin I, Fernández AF, Álvarez-Jiménez J, Urdánoz-Casado A, Roldan M, Robles M, Cabezon-Arteta E, Alcolea D, de Gordoa JSR, Corroza J, Cabello C, Erro ME, Jericó I, Fraga MF, Mendioroz M. Advancing Personalized Medicine in Alzheimer's Disease: Liquid Biopsy Epigenomics Unveil APOE ε4-Linked Methylation Signatures. Int J Mol Sci 2025; 26:3419. [PMID: 40244264 PMCID: PMC11989983 DOI: 10.3390/ijms26073419] [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: 01/28/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Recent studies show that patients with Alzheimer's disease (AD) harbor specific methylation marks in the brain that, if accessible, could be used as epigenetic biomarkers. Liquid biopsy enables the study of circulating cell-free DNA (cfDNA) fragments originated from dead cells, including neurons affected by neurodegenerative processes. Here, we isolated and epigenetically characterized plasma cfDNA from 35 patients with AD and 35 cognitively healthy controls by using the Infinium® MethylationEPIC BeadChip array. Bioinformatics analysis was performed to identify differential methylation positions (DMPs) and regions (DMRs), including APOE ε4 genotype stratified analysis. Plasma pTau181 (Simoa) and cerebrospinal fluid (CSF) core biomarkers (Fujirebio) were also measured and correlated with differential methylation marks. Validation was performed with bisulfite pyrosequencing and bisulfite cloning sequencing. Epigenome-wide cfDNA analysis identified 102 DMPs associated with AD status. Most DMPs correlated with clinical cognitive and functional tests including 60% for Mini-Mental State Examination (MMSE) and 80% for Global Deterioration Scale (GDS), and with AD blood and CSF biomarkers. In silico functional analysis connected 30 DMPs to neurological processes, identifying key regulators such as SPTBN4 and APOE genes. Several DMRs were annotated to genes previously reported to harbor epigenetic brain changes in AD (HKR1, ZNF154, HOXA5, TRIM40, ATG16L2, ADAMST2) and were linked to APOE ε4 genotypes. Notably, a DMR in the HKR1 gene, previously shown to be hypermethylated in the AD hippocampus, was validated in cfDNA from an orthogonal perspective. These results support the feasibility of studying cfDNA to identify potential epigenetic biomarkers in AD. Thus, liquid biopsy could improve non-invasive AD diagnosis and aid personalized medicine by detecting epigenetic brain markers in blood.
Collapse
Affiliation(s)
- Mónica Macías
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Juan José Alba-Linares
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
| | - Blanca Acha
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Idoia Blanco-Luquin
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Agustín F. Fernández
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
| | - Johana Álvarez-Jiménez
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Amaya Urdánoz-Casado
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Miren Roldan
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Maitane Robles
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Eneko Cabezon-Arteta
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, 28029 Madrid, Spain
| | - Javier Sánchez Ruiz de Gordoa
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Jon Corroza
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Carolina Cabello
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - María Elena Erro
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Ivonne Jericó
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Mario F. Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, 33006 Oviedo, Spain
| | - Maite Mendioroz
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
27
|
Mishra S, Morshed N, Sidhu SB, Kinoshita C, Stevens B, Jayadev S, Young JE. The Alzheimer's Disease Gene SORL1 Regulates Lysosome Function in Human Microglia. Glia 2025. [PMID: 40183375 DOI: 10.1002/glia.70009] [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: 06/14/2024] [Revised: 12/31/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025]
Abstract
The SORL1 gene encodes the sortilin-related receptor protein SORLA, a sorting receptor that regulates endo-lysosomal trafficking of various substrates. Loss of function variants in SORL1 are causative for Alzheimer's disease (AD) and decreased expression of SORLA has been repeatedly observed in human AD brains. SORL1 is highly expressed in the central nervous system, including in microglia, the tissue-resident immune cells of the brain. Loss of SORLA leads to enlarged lysosomes in hiPSC-derived microglia-like cells (hMGLs). However, how SORLA deficiency contributes to lysosomal dysfunction in microglia and how this contributes to AD pathogenesis is not known. In this study, we show that loss of SORLA results in decreased lysosomal degradation and lysosomal enzyme activity due to altered trafficking of lysosomal enzymes in hMGLs. Phagocytic uptake of fibrillar amyloid beta 1-42 and synaptosomes is increased in SORLA-deficient hMGLs, but due to reduced lysosomal degradation, these substrates aberrantly accumulate in lysosomes. An alternative mechanism of lysosome clearance, lysosomal exocytosis, is also impaired in SORL1-deficient microglia, which may contribute to an altered immune response. Overall, these data suggest that SORLA has an important role in the proper trafficking of lysosomal hydrolases in hMGLs, which is critical for microglial function. This further substantiates the microglial endo-lysosomal network as a potential novel pathway through which SORL1 may increase AD risk and contribute to the development of AD. Additionally, our findings may inform the development of novel lysosome and microglia-associated drug targets for AD.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Nader Morshed
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Sonia Beant Sidhu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| | - Suman Jayadev
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Jessica E Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
28
|
Wang Y, Li F, Qin Q, Li T, Wang Q, Li Y, Li Y, Jia J. Influence of APOE ε4 on performance of CSF biomarkers in differentiating clinical Alzheimer's disease. J Prev Alzheimers Dis 2025; 12:100065. [PMID: 39827005 DOI: 10.1016/j.tjpad.2025.100065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Apolipoprotein E ε4 (APOE ε4) bring the higher risk of Alzheimer' Disease (AD). It is essential to evaluate whether the diagnostic performances and critical values of cerebrospinal fluid (CSF) biomarkers are influenced by APOE ε4, which has guiding significance for the clinical practical application. METHODS The differences in CSF biomarkers and their performances between APOE ε4 carriers and non-carriers in distinguishing AD, mild cognitive impairment (MCI) and preclinical AD from normal controls (NCs) were analyzed. The receiver operating characteristic (ROC) curves were generated to compare the area under the curve (AUC) between APOE ε4 carriers and non-carriers, as well as the critical values corresponding Youden Index. RESULTS In a cross sectional convenience sample of 1610 participants, lower Aβ42 and Aβ42/Aβ40 and higher p-Tau 181/Aβ42 in CSF were observed among APOE ε4 carriers than non-carriers in NC, MCI, and AD groups (P< 0.05). The performance of CSF p-tau/Aβ42 in distinguishing MCI from NC among APOE ε4 carriers was superior to non-carriers [AUC: 0.714 (95%CI: 0.673- 0.752) vs 0.600 (95%CI: 0.564- 0.634), P< 0.001], although it was similar in distinguishing AD from NC between APOE ε4 carriers and non-carriers [AUC: 0.874 (95%CI: 0.835-0.906) vs 0.876 (95%CI: 0.843- 0.904)]. In the longitudinal cohort of 254 participants, the association of CSF Aβ42, Aβ42/Aβ40 and p-Tau181/Aβ42 with cognitive decline were stronger in APOE ε4 carriers compared to non-carriers (P< 0.05). Meanwhile, the critical values were different depending on APOE genotype. DISCUSSION The CSF level of p-Tau181/Aβ42 was significantly different between APOE ε4 carriers and non-carriers at different stages of AD. The results indicate that the performances of CSF biomarkers are influenced by APOE ε4, which should be considered in the practical application.
Collapse
Affiliation(s)
- Yan Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Qi Qin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Tingting Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Ying Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases; Beijing Key Laboratory of Geriatric Cognitive Disorders; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University; Key Laboratory of Neurodegenerative Diseases, Ministry of Education.
| |
Collapse
|
29
|
Ovesen PL, Juul‐Madsen K, Telugu NS, Schmidt V, Frahm S, Radbruch H, Louth EL, Korshøj AR, Heppner FL, Diecke S, Kettenmann H, Willnow TE. Alzheimer's Disease Risk Gene SORL1 Promotes Receptiveness of Human Microglia to Pro-Inflammatory Stimuli. Glia 2025; 73:857-872. [PMID: 39688327 PMCID: PMC11845846 DOI: 10.1002/glia.24659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024]
Abstract
Sorting protein-related receptor containing class A repeats (SORLA) is an intracellular trafficking receptor encoded by the Alzheimer's disease (AD) gene SORL1 (sortilin-related receptor 1). Recent findings argue that altered expression in microglia may underlie the genome-wide risk of AD seen with some SORL1 gene variants, however, the functional significance of the receptor in microglia remains poorly explained. Using unbiased omics and targeted functional analyses in iPSC-based human microglia, we identified a crucial role for SORLA in sensitizing microglia to pro-inflammatory stimuli. We show that SORLA acts as a sorting factor for the pattern recognition receptor CD14, directing CD14 exposure on the cell surface and priming microglia to stimulation by pro-inflammatory factors. Loss of SORLA in gene-targeted microglia impairs proper CD14 sorting and blunts pro-inflammatory responses. Our studies indicate an important role for SORLA in shaping the inflammatory brain milieu, a biological process important to local immune responses in AD.
Collapse
Affiliation(s)
- Peter Lund Ovesen
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Kristian Juul‐Madsen
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Narasimha S. Telugu
- Technology Platform for Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Vanessa Schmidt
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
| | - Silke Frahm
- Technology Platform for Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Helena Radbruch
- Department of Neuropathology, Charité – Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
| | | | - Anders Rosendal Korshøj
- Department of NeurosurgeryAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Frank L. Heppner
- Department of Neuropathology, Charité – Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität Zu BerlinBerlinGermany
- Cluster of Excellence, NeuroCureBerlinGermany
- German Center for Neurodegenerative Diseases (DZNE) BerlinBerlinGermany
| | - Sebastian Diecke
- Technology Platform for Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz AssociationBerlinGermany
| | - Helmut Kettenmann
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Shenzhen University of Advanced Technology, Shenzhen Key Laboratory of Immunomodulation for Neurological DiseasesShenzhenChina
| | - Thomas E. Willnow
- Max‐Delbrück‐Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Department of BiomedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
30
|
Choi M, Zimmerman SC, Buto PT, Wang J, Brenowitz WD, Hoffmann TJ, Hazzouri AZA, Kezios K, Glymour MM. Association of genetic risk score for Alzheimer's disease with late-life body mass index in all of us: Evaluating reverse causation. Alzheimers Dement 2025; 21:e14598. [PMID: 40189781 PMCID: PMC11972977 DOI: 10.1002/alz.14598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Decreases in body mass index (BMI) may be early consequences of Alzheimer's disease (AD) pathophysiological changes. Previous research in the UK Biobank estimated that AD-related genes began affecting BMI around age 47. We assessed whether this result could be replicated using longitudinal data in an independent cohort. METHODS Using All of Us (AOU) (N = 197,619, aged 30+) data, we estimated linear mixed models for associations of Z-scored AD-Genetic Risk Score (AD-GRS) with BMI, stratified by decade of age. We calculated the earliest age at which AD-GRS was associated with differences in BMI using cross-validated models adjusted for demographics. RESULTS Higher AD-GRS was statistically associated with lower BMI in participants aged 60-70 (b = -0.060 [-0.113, -0.007]). Best fitting models suggested the inverse association of AD-GRS and BMI emerged beginning at ages 47-54. DISCUSSION AD genes accelerate age-related weight loss starting in middle age. HIGHLIGHTS Understanding when physiological changes from amyloid pathology begin is key for AD prevention. Our findings indicate that AD-associated genes accelerate midlife weight loss, starting between 47 and 54 years. AD prevention research should consider that disease pathology likely begins by middle age.
Collapse
Affiliation(s)
- Minhyuk Choi
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Scott C. Zimmerman
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Peter T. Buto
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Jingxuan Wang
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Willa D. Brenowitz
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Kaiser Permanente Center for Health ResearchPortlandOregonUSA
| | - Thomas J. Hoffmann
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Institute for Human GeneticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Katrina Kezios
- Department of EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| | - M. Maria Glymour
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
31
|
Dilliott AA, Costanzo MC, Bandres-Ciga S, Blauwendraat C, Casey B, Hoang Q, Iwaki H, Jang D, Kim JJ, Leonard HL, Levine KS, Makarious M, Nguyen TT, Rouleau GA, Singleton AB, Smadbeck P, Solle J, Vitale D, Nalls M, Flannick J, Burtt NP, Farhan SMK. The Neurodegenerative Disease Knowledge Portal: Propelling Discovery Through the Sharing of Neurodegenerative Disease Genomic Resources. Neurol Genet 2025; 11:e200246. [PMID: 39996130 PMCID: PMC11849525 DOI: 10.1212/nxg.0000000000200246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/02/2025] [Indexed: 02/26/2025]
Abstract
Although large-scale genetic association studies have proven useful for the delineation of neurodegenerative disease processes, we still lack a full understanding of the pathologic mechanisms of these diseases, resulting in few appropriate treatment options and diagnostic challenges. To mitigate these gaps, the Neurodegenerative Disease Knowledge Portal (NDKP) was created as an open-science initiative with the aim to aggregate, enable analysis, and display all available genomic datasets of neurodegenerative disease, while protecting the integrity and confidentiality of the underlying datasets. The portal contains 218 genomic datasets, including genotyping and sequencing studies, of individuals across 10 different phenotypic groups, including neurologic conditions such as Alzheimer disease, amyotrophic lateral sclerosis, Lewy body dementia, and Parkinson disease. In addition to securely hosting large genomic datasets, the NDKP provides accessible workflows and tools to effectively use the datasets and assist in the facilitation of customized genomic analyses. Here, we summarize the genomic datasets currently included within the portal, the bioinformatics processing of the datasets, and the variety of phenotypes captured. We also present example use cases of the various user interfaces and integrated analytic tools to demonstrate their extensive utility in enabling the extraction of high-quality results at the source, for both genomics experts and those in other disciplines. Overall, the NDKP promotes open science and collaboration, maximizing the potential for discovery from the large-scale datasets researchers and consortia are expending immense resources to produce and resulting in reproducible conclusions to improve diagnostic and therapeutic care for patients with neurodegenerative disease.
Collapse
Affiliation(s)
- Allison A Dilliott
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Maria C Costanzo
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
| | - Bradford Casey
- Michael J. Fox Foundation for Parkinson's Research, New York, NY
| | - Quy Hoang
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- DataTecnica LLC, Washington, DC
| | - Dongkeun Jang
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Jonggeol Jeffrey Kim
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
| | - Hampton L Leonard
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- DataTecnica LLC, Washington, DC
| | - Kristin S Levine
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- DataTecnica LLC, Washington, DC
| | - Mary Makarious
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
| | - Trang T Nguyen
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
| | - Patrick Smadbeck
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - J Solle
- Michael J. Fox Foundation for Parkinson's Research, New York, NY
| | - Dan Vitale
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- DataTecnica LLC, Washington, DC
| | - Mike Nalls
- Center for Alzheimer's and Related Dementias, NIH, Bethesda, MD
- Laboratory of Neurogenetics, NIH, Bethesda, MD
- DataTecnica LLC, Washington, DC
| | - Jason Flannick
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatrics, Boston Children's Hospital, Boston, MA; and
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Noël P Burtt
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Sali M K Farhan
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
32
|
Lorenz A, Sathe A, Zaras D, Yang Y, Durant A, Kim ME, Gao C, Newlin NR, Ramadass K, Kanakaraj P, Khairi NM, Li Z, Yao T, Huo Y, Dumitrescu L, Shashikumar N, Pechman KR, Jackson TB, Workmeister AW, Risacher SL, Beason‐Held LL, An Y, Arfanakis K, Erus G, Davatzikos C, Habes M, Wang D, Tosun D, Toga AW, Thompson PM, Mormino EC, Zhang P, Schilling K, Albert M, Kukull W, Biber SA, Landman BA, Johnson SC, Bendlin B, Schneider J, Barnes LL, Bennett DA, Jefferson AL, Resnick SM, Saykin AJ, Hohman TJ, Archer DB. The effect of Alzheimer's disease genetic factors on limbic white matter microstructure. Alzheimers Dement 2025; 21:e70130. [PMID: 40219815 PMCID: PMC11992597 DOI: 10.1002/alz.70130] [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: 12/04/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION White matter (WM) microstructure is essential for brain function but deteriorates with age and in neurodegenerative conditions such as Alzheimer's disease (AD). Diffusion MRI, enhanced by advanced bi-tensor models accounting for free water (FW), enables in vivo quantification of WM microstructural differences. METHODS To evaluate how AD genetic risk factors affect limbic WM microstructure - crucial for memory and early impacted in disease - we conducted linear regression analyses in a cohort of 2,614 non-Hispanic White aging adults (aged 50.12 to 100.85 years). The study evaluated 36 AD risk variants across 26 genes, the association between AD polygenic scores (PGSs) and WM metrics, and interactions with cognitive status. RESULTS AD PGSs, variants in TMEM106B, PTK2B, WNT3, and apolipoprotein E (APOE), and interactions involving MS4A6A were significantly linked to WM microstructure. DISCUSSION These findings implicate AD-related genetic factors related to neurodevelopment (WNT3), lipid metabolism (APOE), and inflammation (TMEM106B, PTK2B, MS4A6A) that contribute to alternations in WM microstructure in older adults. HIGHLIGHTS AD risk variants in TMEM106B, PTK2B, WNT3, and APOE genes showed distinct associations with limbic FW-corrected WM microstructure metrics. Interaction effects were observed between MS4A6A variants and cognitive status. PGS for AD was associated with higher FW content in the limbic system.
Collapse
|
33
|
Hu H, Mao J, Zhao Y, Zhang Y, Zhuang C, He JH, Yang X. Assessing the causal role of lipid metabolites in Alzheimer's disease: A mendelian randomization study. J Prev Alzheimers Dis 2025; 12:100067. [PMID: 39855965 DOI: 10.1016/j.tjpad.2025.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/02/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND The causal relationship between lipid metabolites and Alzheimer's disease (AD) remains unclear and contradictory. This study aimed to systematically assess the causal relationship between lipid metabolites and AD. METHODS A two-step bidirectional Mendelian Randomization (MR) study was employed. The principal analytical technique used to evaluate causation was inverse variance weighting (IVW). Furthermore, mediation analysis was conducted to evaluate the possible function of lipidomes as mediators in the lipid-AD pathway. RESULTS Among the 213 lipid metabolites analyzed, significant causal associations with AD were identified Cholesterol esters in large LDL(L-LDL-CE) (OR = 1.236, 95 %CI = 1.052-1.453, P = 0.010), Total cholesterol in large LDL(L-LDL-TC) (OR = 1.506, 95 %CI = 1.235-1.835, P < 0.001), Total cholesterol in medium LDL(M-LDL-TC) (OR = 1.378, 95 %CI = 1.132-1.677, P = 0.001). Mediation analysis further revealed ceramide (d42:2) and phosphatidylinositol (PI) (18:1_18:1) as potential mediators in this relationship. CONCLUSION The identification of specific lipid metabolites with causal effects on AD provides new insights into AD pathogenesis and highlights potential targets for preventive strategies.
Collapse
Affiliation(s)
- Haoxiang Hu
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Jiesheng Mao
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Yunhan Zhao
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Yihan Zhang
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Caixiang Zhuang
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Jiang Hai He
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China
| | - Xiaokai Yang
- Postgraduate training base Alliance of Wenzhou Medical University, Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou 325000, China.
| |
Collapse
|
34
|
Shardell M, Chen C. Genetic geroscience and Alzheimer's disease: The pleiotropy is the point! J Alzheimers Dis 2025; 104:1001-1005. [PMID: 40026006 DOI: 10.1177/13872877251321182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Geroscience aims to understand how the biology of aging serves as a shared contributor to multiple age-related health conditions. Genetic variants that influence multiple traits are said to exert pleiotropic effects. The study by Pan and colleagues applied a modern statistical model to identify genetic variants with potentially pleiotropic effects by assessing their joint association with Alzheimer's disease and related dementias and another age-related comorbidity (e.g., coronary heart disease, hyperlipidemia, cancer). Motivated by Pan and colleagues, this commentary introduces the concept of genetic geroscience as a paradigm for identifying genetic variants with potentially pleotropic effects on multiple age-related health conditions.
Collapse
Affiliation(s)
- Michelle Shardell
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chixiang Chen
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
He R, Ren J, Malakhov MM, Pan W. Enhancing nonlinear transcriptome- and proteome-wide association studies via trait imputation with applications to Alzheimer's disease. PLoS Genet 2025; 21:e1011659. [PMID: 40209152 PMCID: PMC12040266 DOI: 10.1371/journal.pgen.1011659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 04/29/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
Genome-wide association studies (GWAS) performed on large cohort and biobank datasets have identified many genetic loci associated with Alzheimer's disease (AD). However, the younger demographic of biobank participants relative to the typical age of late-onset AD has resulted in an insufficient number of AD cases, limiting the statistical power of GWAS and any downstream analyses. To mitigate this limitation, several trait imputation methods have been proposed to impute the expected future AD status of individuals who may not have yet developed the disease. This paper explores the use of imputed AD status in nonlinear transcriptome/proteome-wide association studies (TWAS/PWAS) to identify genes and proteins whose genetically regulated expression is associated with AD risk. In particular, we considered the TWAS/PWAS method DeLIVR, which utilizes deep learning to model the nonlinear effects of expression on disease. We trained transcriptome and proteome imputation models for DeLIVR on data from the Genotype-Tissue Expression (GTEx) Project and the UK Biobank (UKB), respectively, with imputed AD status in UKB participants as the outcome. Next, we performed hypothesis testing for the DeLIVR models using clinically diagnosed AD cases from the Alzheimer's Disease Sequencing Project (ADSP). Our results demonstrate that nonlinear TWAS/PWAS trained with imputed AD outcomes successfully identifies known and putative AD risk genes and proteins. Notably, we found that training with imputed outcomes can increase statistical power without inflating false positives, enabling the discovery of molecular exposures with potentially nonlinear effects on neurodegeneration.
Collapse
Affiliation(s)
- Ruoyu He
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jingchen Ren
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Mykhaylo M. Malakhov
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei Pan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
36
|
Saari TT, Palviainen T, Hiltunen M, Herukka S, Kokkola T, Kärkkäinen S, Urjansson M, Aaltonen A, Palotie A, Runz H, Kaprio J, Julkunen V, Vuoksimaa E. Cross-sectional study of plasma phosphorylated tau 217 in persons without dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70107. [PMID: 40352683 PMCID: PMC12064337 DOI: 10.1002/dad2.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Little is known about plasma phosphorylated tau 217 (p-tau217) in individuals without a clinical diagnosis of Alzheimer's disease (AD). We studied associations of plasma p-tau217 with age, sex, education, and genetic risk; estimated the heritability; and conducted a genome-wide association study (GWAS). METHODS A population-based biobank recall study of 65- to 85-year-old twins (N = 697, mean [SD] age 76.2 [4.6] years; 53% women, 154 full pairs) excluding those with AD based on health registry data. RESULTS Higher p-tau217 level and likelihood of AD neuropathologic change (p-tau217 > 0.42 pg/mL; evident in 39%) were associated with higher age and having an apolipoprotein E (APOE) ε4 allele. Heritability was 0.56 (95% confidence interval [CI]: 0.36-0.79) and GWAS indicated 45 single nucleotide polymorphisms (SNPs) (p < 5 × 10-08) centered around the APOE locus. DISCUSSION Our results elucidate the characteristics and genetic associations of p-tau217 in a population-based setting. We found many 65- to 85-year-olds without a clinical diagnosis of AD to have AD neuropathologic change based on plasma p-tau217. Highlights Plasma phosphorylated tau 217 (p-tau217) is a promising biomarker of Alzheimer's disease (AD).We studied plasma p-tau217 in a population-based sample of 65- to -85-year-olds.We excluded those with a clinical diagnosis of AD.Older age and having an apolipoprotein E (APOE) ε4 allele were associated with higher plasma p-tau217.Heritability of p-tau217 was 56% and a genome-wide association study (GWAS) implicated genes around the APOE region.
Collapse
Affiliation(s)
- Toni T. Saari
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Teemu Palviainen
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Mikko Hiltunen
- Institute of BiomedicineUniversity of Eastern FinlandKuopioFinland
| | - Sanna‐Kaisa Herukka
- Institute of Clinical Medicine/NeurologyUniversity of Eastern FinlandKuopioFinland
- Department of NeurologyNeurocenterKuopio University HospitalKuopioFinland
| | - Tarja Kokkola
- Institute of Clinical Medicine/NeurologyUniversity of Eastern FinlandKuopioFinland
| | - Sari Kärkkäinen
- Institute of Clinical Medicine/NeurologyUniversity of Eastern FinlandKuopioFinland
| | - Mia Urjansson
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Aino Aaltonen
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
- Analytic and Translational Genetics UnitDepartment of MedicineDepartment of Neurology and Department of Psychiatry Massachusetts General HospitalBostonMassachusettsUSA
- The Stanley Center for Psychiatric Research and Program in Medical and Population GeneticsThe Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Heiko Runz
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
- European Molecular Biological Laboratories (EMBL)HeidelbergGermany
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Valtteri Julkunen
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
- Institute of Clinical Medicine/NeurologyUniversity of Eastern FinlandKuopioFinland
- Department of NeurologyNeurocenterKuopio University HospitalKuopioFinland
| | - Eero Vuoksimaa
- Institute for Molecular Medicine Finland (FIMM)Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | |
Collapse
|
37
|
Donkor DM, Marfo E, Bockarie A, Tettevi EJ, Antwi MH, Dogah J, Osei GN, Simpong DL. Genetic and environmental risk factors for dementia in African adults: A systematic review. Alzheimers Dement 2025; 21:e70220. [PMID: 40289851 PMCID: PMC12035544 DOI: 10.1002/alz.70220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/16/2025] [Accepted: 03/29/2025] [Indexed: 04/30/2025]
Abstract
Dementia, a leading cause of global mortality, disproportionately impacts sub-Saharan Africans due to complex genetic and environmental interactions. This systematic review evaluated dementia risk factors among sub-Saharan Africans, identifying significant genetic and environmental influences using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The ATP-binding cassette subfamily A member 7 (ABCA7) gene, linked to dementia in African Americans, and unique genetic variants like those in A-kinase anchor protein 9 (AKAP9) and cytidine deaminase (CDA) genes, emerge as potential contributors. Conversely, apolipoprotein E (APOE) ε4 shows lesser impact in older sub-Saharan Africans. Environmental findings highlight that exposure to air pollution, including nitrogen dioxide and particulate matter increases the likelihood of dementia. These findings highlight the role of genetic and environmental diversity in shaping dementia risk profiles. Strategies such as training health-care professionals, enhancing funding for research, combating stigma through awareness campaigns, and fostering global collaborations are vital to ensure African representation in dementia studies. These efforts aim to improve the knowledge of dementia tailored to sub-Saharan Africa's needs. HIGHLIGHTS: The ATP-binding cassette subfamily A member 7 (ABCA7) gene is strongly associated with dementia risk, particularly in African American populations. Apolipoprotein E (APOE) ε4, a well-established risk factor for Alzheimer's disease in Western populations, has a lesser impact in older sub-Saharan Africans, suggesting unique genetic-environment interactions. Exposure to air pollutants, such as nitrogen dioxide and particulate matter, significantly increases dementia risk. The development of dementia in sub-Saharan Africans is influenced by complex interactions between genetic predispositions and environmental exposures, emphasizing the need for tailored prevention strategies.
Collapse
Affiliation(s)
- David Mawutor Donkor
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| | - Esther Marfo
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| | - Ansumana Bockarie
- Department of Internal Medicine and TherapeuticsSchool of Medical SciencesUniversity of Cape CoastCape CoastGhana
| | - Edward Jenner Tettevi
- Department of BiochemistryCell and Molecular BiologySchool of Biological ScienceUniversity of GhanaAccraGhana
| | - Maxwell Hubert Antwi
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
- Department of Medical Laboratory ScienceFaculty of Health SciencesKoforidua Technical UniversityKoforiduaGhana
| | - John Dogah
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| | - George Nkrumah Osei
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| | - David Larbi Simpong
- Department of Medical Laboratory ScienceSchool of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| |
Collapse
|
38
|
Jeong S, Shivakumar M, Jung S, Won H, Nho K, Huang H, Davatzikos C, Saykin AJ, Thompson PM, Shen L, Kim YJ, Kim B, Lee S, Kim D. Addressing overfitting bias due to sample overlap in polygenic risk scoring. Alzheimers Dement 2025; 21:e70109. [PMID: 40189831 PMCID: PMC11972974 DOI: 10.1002/alz.70109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/10/2024] [Accepted: 02/20/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Numerous studies on Alzheimer's disease polygenic risk scores (PRSs) overlook sample overlap between International Genomics of Alzheimer's Project (IGAP) and target datasets like Alzheimer's Disease Neuroimaging Initiative (ADNI). METHODS To address this, we developed overlap-adjusted PRS (OA PRS) and tested it on simulated data to assess biases from different scenarios by varying training, testing, and overlap proportions. OA PRS was used to adjust for sample bias in simulations; then, we applied OA PRS to IGAP and ADNI datasets and validated through visual diagnosis. RESULTS OA PRS effectively adjusted for sample overlap in all simulation scenarios, as well as for IGAP and ADNI. The original IGAP PRS showed an inflated area under the receiver operating characteristic (AUROC: 0.915) on overlapping samples. OA PRS reduced the AUROC to 0.726, closely aligning with the AUROC of non-overlapping samples (0.712). Further, visual diagnostics confirmed the effectiveness of our adjustments. DISCUSSION With OA PRS, we were able to adjust the IGAP summary-based PRS for the overlapped ADNI samples, allowing the dataset to be fully used without the risk of overfitting. HIGHLIGHTS Sample overlap between large Alzheimer's disease (AD) cohorts poses overfitting bias when using AD polygenic risk scores (PRSs). This study highlighted the effectiveness of overlap-adjusted PRS (OA -PRS) in mitigating overfitting and improving the accuracy of PRS estimations. New PRSs based on adjusted effect sizes showed increased power in association with clinical features.
Collapse
Affiliation(s)
- Seokho Jeong
- Graduate School of Data ScienceSeoul National UniversitySeoulRepublic of Korea
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sang‐Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Medical InformaticsKangwon National University, College of MedicineChuncheonRepublic of Korea
| | - Hong‐Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST)Samsung Medical CenterSungkyunkwan UniversitySeoulRepublic of Korea
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Heng Huang
- Department of Electrical and Computer EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and AnalyticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciencesand Indiana Alzheimer Disease CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Paul M. Thompson
- Imaging Genetics CenterLaboratory of Neuro ImagingDepartment of Neurology & PsychiatryUCLA School of MedicineLos AngelesCaliforniaUSA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Young Jin Kim
- Division of Genome ScienceDepartment of Precision MedicineNational Institute of HealthCheongjuRepublic of Korea
| | - Bong‐Jo Kim
- Division of Genome ScienceDepartment of Precision MedicineNational Institute of HealthCheongjuRepublic of Korea
| | - Seunggeun Lee
- Graduate School of Data ScienceSeoul National UniversitySeoulRepublic of Korea
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Institute for Biomedical InformaticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
39
|
Xavier C, Pinto N. Navigating the blurred boundary: Neuropathologic changes versus clinical symptoms in Alzheimer's disease, and its consequences for research in genetics. J Alzheimers Dis 2025; 104:611-626. [PMID: 39956949 DOI: 10.1177/13872877251317543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
During decades scientists tried to unveil the genetic architecture of Alzheimer's disease (AD), recurring to increasingly larger sample numbers for genome-wide association studies (GWAS) in hope for higher statistical gains. Here, a retrospective look on the most prominent GWAS was performed, focusing on the quality of the diagnosis associated with the used data and databases. Different methods for AD diagnosis (or absence) carry different levels of accuracy and certainty applied to both subsets of cases and controls. Furthermore, the different phenotypes included in these databases were explored, as several incorporate other ageing comorbidities and might be encompassing many confounding agents as well. Age of the samples' donors and origin populations were also investigated as these could be biasing factors in posterior analyses. A tendency for looser diagnostic methods in more recent GWAS was observed, where greater datasets of individuals are analyzed, which may have been hampering the discovery of associated genetic variants. Specifically for AD, a diagnostic method conveying a clinical outcome may be distinct from the disease neuropathological assessment, since the first has a practical perspective that not necessarily needs a confirmation. Due to its properties and complex diagnosis, this work highlights the importance of the neuropathological confirmation of AD (or its absence) in the subjects considered for research purposes to avoid reaching statistically weak and/or misleading conclusions that may trigger further studies with powerless groundwork.
Collapse
Affiliation(s)
- Catarina Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Nádia Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- CMUP - Centro de Matemática da Universidade do Porto, Porto, Portugal
| |
Collapse
|
40
|
Al Massadi O, Labarchède M, de Pins B, Longueville S, Giralt A, Irinopoulou T, Savariradjane M, Subashi E, Ginés S, Caboche J, Mariani LL, Betuing S, Girault JA. PYK2 in the dorsal striatum of Huntington's disease R6/2 mouse model. Neurobiol Dis 2025; 207:106840. [PMID: 39971200 DOI: 10.1016/j.nbd.2025.106840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
Huntington's disease (HD) is a devastating disease due to autosomal dominant mutation in the HTT gene. Its pathophysiology involves multiple molecular alterations including transcriptional defects. We previously showed that in HD patients and mouse model, the protein levels of the non-receptor tyrosine kinase PYK2 were decreased in the hippocampus and that viral expression of PYK2 improved the hippocampal phenotype. Here, we investigated the possible contribution of PYK2 in the striatum, a brain region particularly altered in HD. PYK2 mRNA levels were decreased in the striatum and hippocampus of R6/2 mice, a severe HD model. Striatal PYK2 protein levels were also decreased in R6/2 mice and human patients. PYK2 knockout by itself did not result in motor symptoms observed in HD mouse models. We examined whether PYK2 deficiency participated in the R6/2 mice phenotype by expressing PYK2 in their dorsal striatum using AAV vectors. With an AAV1/Camk2a promoter, we did not observe significant improvement of body weight, clasping, motor activity and coordination (rotarod) alterations observed in R6/2 mice. With an AAV9/SYN1 promoter we found a slightly higher body weight and a trend to better rotarod performance. Both viruses similarly transduced striatal projection neurons and somatostatin-positive interneurons but only AAV9/SYN1 led to PYK2 expression in cholinergic and parvalbumin-positive interneurons. Expression of PYK2 in cholinergic interneurons may contribute to the slight effects observed. We conclude that PYK2 mRNA and protein levels are decreased in the striatum as in hippocampus of HD patients and mouse models. However, in contrast to hippocampus, striatal viral expression of PYK2 has only a minor effect on the R6/2 model striatal phenotype.
Collapse
Affiliation(s)
- Omar Al Massadi
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France.
| | - Mélody Labarchède
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Benoit de Pins
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Sophie Longueville
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Albert Giralt
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Theano Irinopoulou
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Mythili Savariradjane
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France
| | - Enejda Subashi
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Silvia Ginés
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Production and Validation Center of Advanced Therapies (Creatio), University of Barcelona, Barcelona, Spain
| | - Jocelyne Caboche
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Louise-Laure Mariani
- Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France
| | - Sandrine Betuing
- Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Neuroscience Paris Seine, Institut de Biologie Paris-Seine, Paris, France; CNRS UMR8246, Paris, France; INSERM U1130, Paris, France
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, Paris 75005, France; Sorbonne Université, Faculty of Sciences and Engineering, Paris 75005, France; Institut du Fer à Moulin, 17 rue du Fer à Moulin, Paris 75005, France; Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, Sorbonne Université, Paris, France.
| |
Collapse
|
41
|
Achouri-Rassas A, Fray S, Said Z, Ben Sassi S, Ben Ali N, Baraket G. Genetic association study between rs2234253 (p.T96K) variant of TREM2 and Alzheimer's disease in a Tunisian population. Neurol Res 2025; 47:290-295. [PMID: 40043316 DOI: 10.1080/01616412.2025.2472841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of major neurodegenerative cognitive impairment. The risk of developing AD is influenced by a complex interaction of genetic predisposition and environmental factors. Among the genetic risk factors, the APOE ɛ4 allele is the most significant, while variants in the TREM2 (Triggering Receptor Expressed on Myeloid Cells 2) and ABCA7 (ATP-binding cassette transporter A7) genes have also been associated with an increased risk of AD. OBJECTIVE This study aimed to investigate the association of APOE ɛ4, TREM2 gene variants (rs75932628 [p.R47H] and rs2234253 [p.T96K]), and ABCA7 gene variants (rs142076058 and rs115550680) with sporadic AD in the Tunisian population. Methods: A case-control study was conducted including 222 Tunisian patients diagnosed with sporadic AD and 99 cognitively healthy controls. Genotyping was performed to assess the presence and association of the selected genetic variants with AD. Statistical analyses were conducted to determine the significance of genetic associations. RESULTS A significant association was found between the TREM2 rs2234253 (p.T96K) variant and AD, with the T allele identified as a risk factor in the Tunisian population. The APOE ɛ4 allele was also associated with an increased risk of developing AD. However, no significant association was observed for the ABCA7 gene variants or the TREM2 rs75932628 (p.R47H) variant in either the AD or control groups. CONCLUSION Our findings suggest that the TREM2 rs2234253 (p.T96K) variant is a significant genetic risk factor for late-onset AD (LOAD) in the Tunisian population. Further studies with larger cohorts are needed to validate these findings and explore potential gene-gene interactions contributing to AD risk.
Collapse
Affiliation(s)
- Afef Achouri-Rassas
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
| | - Saloua Fray
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
- Neurological Department, Charles Nicolle Hospital, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Zakaria Said
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
- Department of Neurology, National Institute Mongi Ben Hamida of Neurology, Tunis, Tunisia
| | - Samia Ben Sassi
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
- Department of Neurology, National Institute Mongi Ben Hamida of Neurology, Tunis, Tunisia
| | - Nadia Ben Ali
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
- Neurological Department, Charles Nicolle Hospital, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Ghada Baraket
- Faculty of Sciences of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
42
|
Jiang W, Vogelgsang J, Dan S, Durning P, McCoy TH, Berretta S, Klengel T. Association of RDoC dimensions with post mortem brain transcriptional profiles in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70103. [PMID: 40352682 PMCID: PMC12064340 DOI: 10.1002/dad2.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/16/2025] [Accepted: 02/28/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Neuropsychiatric symptoms are common in people with Alzheimer's disease (AD) across all severity stages. Their heterogeneous presentation and variable temporal association with cognitive decline suggest shared and distinct biological mechanisms. We hypothesized that specific patterns of gene expression associate with distinct National Institute of Mental Health Research Domain Criteria (RDoC) domains in AD. METHODS Post-mortem bulk RNA sequencing of the insula and anterior cingulate cortex from 60 brain donors, representing the spectrum of canonical Alzheimer's disease neuropathology, was combined with natural language processing approaches based on the RDoC Clinical Domains to uncover transcriptomic patterns linked to disease progression. RESULTS Distinct sets of >100 genes (P false discovery rate < 0.05) were specifically associated with at least one clinical domain (cognitive, social, negative, positive, arousal). In addition, dysregulation of immune response pathways was shared across domains and brain regions. DISCUSSION Our findings provide evidence for distinct transcriptional profiles associated with RDoC domains suggesting that each dimension is characterized by sets of genes providing insight into the underlying mechanisms. Highlights Post mortem brain tissue investigations are critically important for Alzheimer's disease (AD) research.Neuropsychiatric symptoms in AD are common and an important aspect of AD.Categorical phenotypes are commonly used, but insufficiently describe the heterogenous presentation of AD.Using natural language processing (NLP) of post mortem brain donor health records provides insight into dimensional phenotypes of AD.We provide evidence for distinct RNA expression profiles associated with NLP-derived Research Domain Criteria clinical domain scores.
Collapse
Affiliation(s)
- Weiqian Jiang
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| | - Jonathan Vogelgsang
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| | - Shu Dan
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| | - Peter Durning
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| | - Thomas H. McCoy
- Center for Quantitative HealthMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Sabina Berretta
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| | - Torsten Klengel
- Department of PsychiatryHarvard Medical SchoolMcLean HospitalBelmontMassachusettsUSA
| |
Collapse
|
43
|
Jung ES, Choi H, Mook-Jung I. Decoding microglial immunometabolism: a new frontier in Alzheimer's disease research. Mol Neurodegener 2025; 20:37. [PMID: 40149001 PMCID: PMC11948825 DOI: 10.1186/s13024-025-00825-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) involves a dynamic interaction between neuroinflammation and metabolic dysregulation, where microglia play a central role. These immune cells undergo metabolic reprogramming in response to AD-related pathology, with key genes such as TREM2, APOE, and HIF-1α orchestrating these processes. Microglial metabolism adapts to environmental stimuli, shifting between oxidative phosphorylation and glycolysis. Hexokinase-2 facilitates glycolytic flux, while AMPK acts as an energy sensor, coordinating lipid and glucose metabolism. TREM2 and APOE regulate microglial lipid homeostasis, influencing Aβ clearance and immune responses. LPL and ABCA7, both associated with AD risk, modulate lipid processing and cholesterol transport, linking lipid metabolism to neurodegeneration. PPARG further supports lipid metabolism by regulating microglial inflammatory responses. Amino acid metabolism also contributes to microglial function. Indoleamine 2,3-dioxygenase controls the kynurenine pathway, producing neurotoxic metabolites linked to AD pathology. Additionally, glucose-6-phosphate dehydrogenase regulates the pentose phosphate pathway, maintaining redox balance and immune activation. Dysregulated glucose and lipid metabolism, influenced by genetic variants such as APOE4, impair microglial responses and exacerbate AD progression. Recent findings highlight the interplay between metabolic regulators like REV-ERBα, which modulates lipid metabolism and inflammation, and Syk, which influences immune responses and Aβ clearance. These insights offer promising therapeutic targets, including strategies aimed at HIF-1α modulation, which could restore microglial function depending on disease stage. By integrating metabolic, immune, and genetic factors, this review underscores the importance of microglial immunometabolism in AD. Targeting key metabolic pathways could provide novel therapeutic strategies for mitigating neuroinflammation and restoring microglial function, ultimately paving the way for innovative treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- Eun Sun Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hayoung Choi
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Inhee Mook-Jung
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Korea Dementia Research Center, Seoul, South Korea.
| |
Collapse
|
44
|
Korologou-Linden R, Xu B, Coulthard E, Walton E, Wearn A, Hemani G, White T, Cecil C, Sharp T, Tiemeier H, Banaschewski T, Bokde A, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Paillère Martinot ML, Artiges E, Nees F, Orfanos DP, Paus T, Poustka L, Millenet S, Fröhner JH, Smolka M, Walter H, Winterer J, Whelan R, Schumann G, Howe LD, Ben-Shlomo Y, Davies NM, Anderson EL. Genetics impact risk of Alzheimer's disease through mechanisms modulating structural brain morphology in late life. J Neurol Neurosurg Psychiatry 2025; 96:350-360. [PMID: 38663994 PMCID: PMC7616849 DOI: 10.1136/jnnp-2023-332969] [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: 11/09/2023] [Accepted: 03/11/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Alzheimer's disease (AD)-related neuropathological changes can occur decades before clinical symptoms. We aimed to investigate whether neurodevelopment and/or neurodegeneration affects the risk of AD, through reducing structural brain reserve and/or increasing brain atrophy, respectively. METHODS We used bidirectional two-sample Mendelian randomisation to estimate the effects between genetic liability to AD and global and regional cortical thickness, estimated total intracranial volume, volume of subcortical structures and total white matter in 37 680 participants aged 8-81 years across 5 independent cohorts (Adolescent Brain Cognitive Development, Generation R, IMAGEN, Avon Longitudinal Study of Parents and Children and UK Biobank). We also examined the effects of global and regional cortical thickness and subcortical volumes from the Enhancing NeuroImaging Genetics through Meta-Analysis (ENIGMA) Consortium on AD risk in up to 37 741 participants. RESULTS Our findings show that AD risk alleles have an age-dependent effect on a range of cortical and subcortical brain measures that starts in mid-life, in non-clinical populations. Evidence for such effects across childhood and young adulthood is weak. Some of the identified structures are not typically implicated in AD, such as those in the striatum (eg, thalamus), with consistent effects from childhood to late adulthood. There was little evidence to suggest brain morphology alters AD risk. CONCLUSIONS Genetic liability to AD is likely to affect risk of AD primarily through mechanisms affecting indicators of brain morphology in later life, rather than structural brain reserve. Future studies with repeated measures are required for a better understanding and certainty of the mechanisms at play.
Collapse
Affiliation(s)
- Roxanna Korologou-Linden
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Bing Xu
- The Generation R Study Group, Erasmus MC University Medical Center, Rotterdam, UK
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elizabeth Coulthard
- Bristol Medical School, University of Bristol, Bristol, UK
- North Bristol NHS Trust, Bristol, UK
| | - Esther Walton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Department of Psychology, University of Bath, Bath, UK
| | - Alfie Wearn
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Gibran Hemani
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Tonya White
- The Generation R Study Group, Erasmus MC University Medical Center, Rotterdam, UK
- Department of Radiology and Nuclear Medicine, Erasmus University School of Medicine, Rotterdam, UK
| | - Charlotte Cecil
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamsin Sharp
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Biostatistics and Health Informatics Department, King's College London, Boston, UK
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Social and Behavioral Sciences, Harvard T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
| | - Arun Bokde
- Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Kings College London, Centre for Population Neuroscience and Precision Medicine (PONS), London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, University of Mannheim, Mannheim, Germany
- Psychology, School of Social Sciences, University of Mannheim, Mannheim, Germany
| | | | | | | | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Berlin Institute of Health, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299, Paris, France
- Centre Borelli, Cachan, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, University of Mannheim, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, Kiel University, Kiel, Germany
| | | | - Tomáš Paus
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Montreal, Montreal, Quebec, Canada
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Centre Göttingen, Göttingen, Germany
| | - Sabina Millenet
- Department of Child and Adolescent Psychiatry and Psychotherapy, Heidelberg University, Heidelberg, Germany
| | - Juliane H Fröhner
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Michael Smolka
- Department of Psychiatry, Technische Universität Dresden, Dresden, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charite, Berlin, Germany
| | - Jeanne Winterer
- Department of Psychiatry and Psychotherapy CCM, Berlin Institute of Health, Berlin, Germany
- Department of Education and Psychology, Freie Universität Berlin, Berlin, Germany
| | - Robert Whelan
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Kings College London, Centre for Population Neuroscience and Precision Medicine (PONS), London, UK
- Fudan University, Shanghai, People's Republic of China
- PONS Centre, Dept. of Psychiatry and Clinical Neuroscience, CCM, Berlin, Germany
| | - Laura D Howe
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol, UK
| | - Neil M Davies
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
- University College London Division of Psychiatry, London, UK
| | - Emma Louise Anderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, University of Bristol, Bristol, UK
- University College London Division of Psychiatry, London, UK
| |
Collapse
|
45
|
Sun Y, Liu Z, Zhang Z, Kang Y, Wang X, Zhang Y, Liu Y, Zhao P. Human induced pluripotent stem cell models for Alzheimer's disease research: a bibliometric analysis. Front Hum Neurosci 2025; 19:1548701. [PMID: 40177166 PMCID: PMC11962003 DOI: 10.3389/fnhum.2025.1548701] [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: 12/20/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Alzheimer's disease (AD), the leading cause of dementia, remains without adequate treatment. Current models do not fully replicate human physiology and pathology. The advent of human induced pluripotent stem cell (hiPSC) technology offers a novel approach to studying AD. Methods Our study conducted a bibliometric analysis to assess the application and development of hiPSC technology in AD research. We retrieved 531 articles on hiPSC models of AD from the Web of Science Core Collection, published between January 2010 and June 2024. CiteSpace and VOSviewer were used to analyze authorship, geographic contributions, journal influence, and citation patterns. Results Our findings reveal a steady increase in publications over 14 years, with the United States leading in contributions, followed by China. Li-Huei Tsai from the Massachusetts Institute of Technology is a prominent researcher. PLoS One emerges as the most influential journal. Research trends have focused on inflammation, astrocytes, microglia, apolipoprotein E (ApoE), and tau. Discussion Bibliometric analysis is crucial in identifying research gaps and trends and guiding future studies to address unmet needs in understanding and modeling human physiology and pathology. Leveraging hiPSC models to investigate the molecular mechanisms of familial and sporadic AD is expected to provide a crucial foundation for developing future treatment strategies. Conclusion In summary, the bibliometric findings from this study provide a comprehensive overview of the current research landscape in hiPSC models for AD. It also highlights emerging trends and research gaps, crucial for guiding future research efforts, particularly in exploring novel therapeutic targets and improving understanding of disease mechanisms.
Collapse
Affiliation(s)
- Yuning Sun
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zhilong Liu
- Gansu Provincial People’s Hospital, Lanzhou, China
| | - Zongbo Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yufeng Kang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xinlian Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yiping Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yan Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Pei Zhao
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, China
- Gansu Provincial People’s Hospital, Lanzhou, China
| |
Collapse
|
46
|
Miller JB, Brandon JA, Harmon LM, Sabra HW, Lucido CC, Murcia JDG, Nations KA, Payne SH, Ebbert MTW, Kauwe JSK, Ridge PG. Ramp Sequence May Explain Synonymous Variant Association with Alzheimer's Disease in the Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA). Biomedicines 2025; 13:739. [PMID: 40149715 PMCID: PMC11940050 DOI: 10.3390/biomedicines13030739] [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: 01/16/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: The synonymous variant NC_000007.14:g.100373690T>C (rs2405442:T>C) in the Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA) gene was previously associated with decreased risk for Alzheimer's disease (AD) in genome-wide association studies, but its biological impact is largely unknown. Objective: We hypothesized that rs2405442:T>C decreases mRNA and protein levels by destroying a ramp of slowly translated codons at the 5' end of PILRA. Methods: We assessed rs2405442:T>C predicted effects on PILRA through quantitative polymerase chain reactions (qPCRs) and enzyme-linked immunosorbent assays (ELISAs) using Chinese hamster ovary (CHO) cells. RESULTS: Both mRNA (p = 1.9184 × 10-13) and protein (p = 0.01296) levels significantly decreased in the mutant versus the wildtype in the direction that we predicted based on the destruction of a ramp sequence. Conclusions: We show that rs2405442:T>C alone directly impacts PILRA mRNA and protein expression, and ramp sequences may play a role in regulating AD-associated genes without modifying the protein product.
Collapse
Affiliation(s)
- Justin B. Miller
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - J. Anthony Brandon
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
| | - Lauren M. Harmon
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Hady W. Sabra
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Chloe C. Lucido
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Josue D. Gonzalez Murcia
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Kayla A. Nations
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
| | - Samuel H. Payne
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Mark T. W. Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40506, USA
| | - John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| |
Collapse
|
47
|
Meng X, Li X, Cao M, Dong J, Wang H, Cao W, Liu D, Wang Y. Summarizing attributable factors and evaluating risk of bias of Mendelian randomization studies for Alzheimer's dementia and cognitive status: a systematic review and meta-analysis. Syst Rev 2025; 14:61. [PMID: 40082927 PMCID: PMC11905674 DOI: 10.1186/s13643-025-02792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND No effective treatment is available to delay or reverse the onset and progression of Alzheimer's dementia (AD). Mild cognitive impairment, a clinical state between normal aging and AD, may offer the proper window for AD intervention and treatment. This systematic review aimed to summarize evidence from Mendelian randomization (MR) studies exploring factors attributable to AD and related cognitive status and to assess its credibility. METHODS We searched PubMed, Embase, MEDLINE, and the Cochrane Library to identify MR studies investigating the associations between any factor and AD and related cognitive status. The risk of bias in MR studies was evaluated using nine signaling questions tailored to identify potential biases based on the STROBE-MR guidelines. RESULTS A total of 125 eligible publications were examined, including 106 AD-related MR studies reporting 674 records and 28 cognition-related MR studies reporting 141 records. We identified 185 unique causal risk factors for AD and 49 for cognitive status. More than half of the MR studies reporting AD or cognitive status outcomes exhibited poor methodological quality, with a high risk of bias observed in 59% of the AD-related studies and 64% of the cognitive-related studies. CONCLUSIONS This systematic review summarized modifiable factors and omics signatures, providing a database of MR studies on AD and related cognitive status. The evaluation of bias risk in MR studies serves to raise awareness and improve overall quality. A critical appraisal checklist for assessing the risk of bias may pave the way for the development of a standardized tool. SYSTEMATIC REVIEW REGISTRATION The review protocol was registered with the Prospective Register of Systematic Reviews (PROSPERO) under the registration number CRD42023213990.
Collapse
Affiliation(s)
- Xiaoni Meng
- Department of Clinical Epidemiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Xiaochun Li
- School of Public Health, Capital Medical University, Beijing, 100069, China
- The Medical Department, Xijing Hospital, the Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Meiling Cao
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Jing Dong
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, 100050, China
| | - Haotian Wang
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Weijie Cao
- Centre for Precision Medicine, Edith Cowan University, Perth, WA, 7027, Australia
| | - Di Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, University Town, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, China.
| | - Youxin Wang
- Centre for Precision Medicine, Edith Cowan University, Perth, WA, 7027, Australia.
- School of Public Health, North China University of Science and Technology, 21 Bohaidadao, Caofeidian District, Tangshan, 063210, China.
| |
Collapse
|
48
|
Chaar DL, Li Z, Shang L, Ratliff SM, Mosley TH, Kardia SLR, Zhao W, Zhou X, Smith JA. Multi-Ancestry Transcriptome-Wide Association Studies of Cognitive Function, White Matter Hyperintensity, and Alzheimer's Disease. Int J Mol Sci 2025; 26:2443. [PMID: 40141087 PMCID: PMC11942532 DOI: 10.3390/ijms26062443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Genetic variants increase the risk of neurocognitive disorders in later life, including vascular dementia (VaD) and Alzheimer's disease (AD), but the precise relationships between genetic risk factors and underlying disease etiologies are not well understood. Transcriptome-wide association studies (TWASs) can be leveraged to better characterize the genes and biological pathways underlying genetic influences on disease. To date, almost all existing TWASs on VaD and AD have been conducted using expression studies from individuals of a single genetic ancestry, primarily European. Using the joint likelihood-based inference framework in Multi-ancEstry TRanscriptOme-wide analysis (METRO), we leveraged gene expression data from European ancestry (EA) and African ancestry (AA) samples to identify genes associated with general cognitive function, white matter hyperintensity (WMH), and AD. Regions were fine-mapped using Fine-mapping Of CaUsal gene Sets (FOCUS). We identified 266, 23, 69, and 2 genes associated with general cognitive function, WMH, AD (using EA GWAS summary statistics), and AD (using AA GWAS), respectively (Bonferroni-corrected alpha = p < 2.9 × 10-6), some of which had been previously identified. Enrichment analysis showed that many of the identified genes were in pathways related to innate immunity, vascular dysfunction, and neuroinflammation. Further, the downregulation of ICA1L was associated with a higher WMH and with AD, indicating its potential contribution to overlapping AD and VaD neuropathology. To our knowledge, our study is the first TWAS on cognitive function and neurocognitive disorders that used expression mapping studies for multiple ancestries. This work may expand the benefits of TWASs beyond a single ancestry group and help to identify gene targets for pharmaceuticals or preventative treatments for dementia.
Collapse
Affiliation(s)
- Dima L. Chaar
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Zheng Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (Z.L.); (X.Z.)
| | - Lulu Shang
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Scott M. Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Thomas H. Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Xiang Zhou
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (Z.L.); (X.Z.)
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA; (D.L.C.); (S.M.R.); (S.L.R.K.); (W.Z.)
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
49
|
Mitsumori R, Asanomi Y, Morizono T, Shigemizu D, Niida S, Ozaki K. A genome-wide association study identifies a novel East Asian-specific locus for dementia with Lewy bodies in Japanese subjects. Mol Med 2025; 31:87. [PMID: 40045203 PMCID: PMC11884146 DOI: 10.1186/s10020-025-01115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is the second most common type of degenerative dementia in older patients. As with other multifactorial diseases, the pathogenesis results from interactions of environmental and genetic factors. The genetic basis of DLB is not yet fully understood. Recent genomic analyses of DLB in Caucasian cohorts identified genetic susceptibility loci for DLB, but the comprehensive genomic analysis in Asians was still not performed. METHODS We conducted a genome-wide association study (GWAS) in Japanese subjects (211 DLB cases and 6113 controls) to clarify the genetic architecture of DLB pathogenesis. RESULTS We identified the East Asian-specific DHTKD1 locus (rs138587229) on chromosome 10 with genome-wide significance (GWS; P = 3.27 × 10-8) and the ICOS/PARD3B locus on chromosome 2 with suggestive significance (P = 3.95 × 10-7) as novel DLB genetic risk loci. We also confirmed the APOE locus (rs429358, P < 5.0 × 10-8), a known risk locus for DLB and Alzheimer's disease in Caucasians. The DHTKD1 locus was associated with the gene expression of SEC61A2 and showed a causal relationship with cholinesterase levels. In a trans-ethnic meta-analysis that included Japanese, UK Biobank, and other Caucasian GWAS, we confirmed the risk for DLB at APOE and SNCA loci with GWS. Transcriptome-wide association analysis identified ZNF155 and ZNF284 in the brain cortex and GPRIN3 in the substantia nigra as putative causal genes for DLB. CONCLUSIONS This is the first GWAS for DLB in East Asians, and our findings provide new biological and clinical insights into the pathogenesis of DLB.
Collapse
Affiliation(s)
- Risa Mitsumori
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Yuya Asanomi
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Takashi Morizono
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Daichi Shigemizu
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Shumpei Niida
- National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Kouichi Ozaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-Cho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
50
|
Zelek WM, Bevan RJ, Nimmo J, Dewilde M, De Strooper B, Morgan BP. Brain-penetrant complement inhibition mitigates neurodegeneration in an Alzheimer's disease mouse model. Brain 2025; 148:941-954. [PMID: 39215579 PMCID: PMC11884734 DOI: 10.1093/brain/awae278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Complement activation is implicated in driving brain inflammation, self-cell damage and progression of injury in Alzheimer's disease and other neurodegenerative diseases. Here, we investigate the impact of brain delivery of a complement-blocking antibody on neurodegeneration in an Alzheimer's mouse model. We engineered a brain-penetrant recombinant antibody targeting the pro-inflammatory membrane attack complex. Systemic administration of this antibody in APPNL-G-F mice reduced brain levels of complement activation products, demonstrating successful brain entry and target engagement. Prolonged treatment decreased synapse loss, amyloid burden and brain inflammatory cytokine levels, concomitant with cognitive improvement compared to controls. These results underscore the potential of brain-penetrant complement-inhibiting drugs as promising therapeutics, targeting downstream of amyloid plaques in Alzheimer's disease.
Collapse
Affiliation(s)
- Wioleta M Zelek
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Ryan J Bevan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Jacqui Nimmo
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Maarten Dewilde
- Therapeutic and Diagnostic Antibodies, Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Bart De Strooper
- Centre for Brain and Disease Research, KU Leuven and VIB Leuven, Leuven 3000, Belgium
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Bryan Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|