1
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. Nat Commun 2025; 16:4441. [PMID: 40393985 PMCID: PMC12092734 DOI: 10.1038/s41467-025-59654-w] [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: 10/09/2024] [Accepted: 04/28/2025] [Indexed: 05/22/2025] Open
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, remain incompletely understood. To define relevant pathways, we conducted an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila. We also measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's genetic variants that modify gene expression in disease-vulnerable neurons in humans. We then used a network model to integrate these data with previously published Alzheimer's disease proteomics, lipidomics and genomics. Here, we computationally predict and experimentally confirm how HNRNPA2B1 and MEPCE enhance toxicity of the tau protein, a pathological feature of Alzheimer's disease. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human stem cell-derived neural progenitor cells. Our study identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Stephen and Denise Adams Center of Yale School of Medicine, New Haven, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
2
|
Giannakis A, Konitsiotis S. Anti-Amyloid Agents: A Self-Fulfilling prophecy. J Clin Neurosci 2025; 137:111338. [PMID: 40393205 DOI: 10.1016/j.jocn.2025.111338] [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: 03/27/2025] [Revised: 05/15/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
The introduction of anti-amyloid antibodies has ushered in a new era in the treatment of Alzheimer's disease (AD), coinciding with the revision of its diagnostic criteria, which now focus on the biological definition of AD, with amyloid beta at its core. However, despite being fully aligned with these criteria-and therefore with how we define the disease-amyloid-targeting therapies have not yielded the expected results. How can a treatment targeting the very core of the disease be ineffective? Perhaps because AD, as we have defined it, is not actually the disease that afflicts millions of patients worldwide. Patients with conditions related to AD, such as apolipoprotein ε4 allele (APOE4) homozygotes, patients receiving anticoagulant therapy for atrial fibrillation, and those with microhemorrhages, are excluded from treatment. Several other pathogenetic mechanisms continue to arise, including neuroinflammation, cerebrovascular disease, and metal ion dysregulation. At the same time, Alzheimer's pathology frequently coexists with other brain pathologies in AD patients, the roles and interactions of which remain largely unknown. Thus, AD should be redefined as a multifactorial neurodegenerative disorder, in which various processes contribute to amyloid accumulation or independently drive neurodegeneration.
Collapse
Affiliation(s)
- Alexandros Giannakis
- Department of Neurology, University of Ioannina, University Campus, Stavrou Niarchou Av., Ioannina, Greece.
| | - Spiridon Konitsiotis
- Department of Neurology, University of Ioannina, University Campus, Stavrou Niarchou Av., Ioannina, Greece
| |
Collapse
|
3
|
Schneider PG, Liu S, Bullinger L, Ostendorf BN. BEscreen: a versatile toolkit to design base editing libraries. Nucleic Acids Res 2025:gkaf406. [PMID: 40384567 DOI: 10.1093/nar/gkaf406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025] Open
Abstract
Base editing enables the high-throughput screening of genetic variants for phenotypic effects. Base editing screens require the design of single guide RNA (sgRNA) libraries to enable either gene- or variant-centric approaches. While computational tools supporting the design of sgRNAs exist, no solution offers versatile and scalable library design enabling all major use cases. Here, we introduce BEscreen, a comprehensive base editing guide design tool provided as a web server (bescreen.ostendorflab.org) and as a command line tool. BEscreen provides variant-, gene-, and region-centric modes to accommodate various screening approaches. The variant mode accepts genomic coordinates, amino acid changes, or rsIDs as input. The gene mode designs near-saturation libraries covering the entire coding sequence of given genes or transcripts, and the region mode designs all possible guides for given genomic regions. BEscreen enables selection of guides by biological consequence, it features comprehensive customization of base editor characteristics, and it offers optional annotation using Ensembl's Variant Effect Predictor. In sum, BEscreen is a highly versatile tool to design base editing screens for a wide range of use cases with seamless scalability from individual variants to large, near-saturation libraries.
Collapse
Affiliation(s)
- Philipp G Schneider
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Shuang Liu
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site, 13353 Berlin, Germany
| | - Benjamin N Ostendorf
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Berlin Institute of Health, 10178 Berlin, Germany
| |
Collapse
|
4
|
Pal S, Dolai S, Deepa S, Garai K. Investigation of Domain Interaction in the Apolipoprotein E Isoforms by HDX-MS. J Mol Biol 2025; 437:169036. [PMID: 40010433 DOI: 10.1016/j.jmb.2025.169036] [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: 12/23/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Involvement of apoE4 in the pathology of Alzheimer's disease (AD) is hypothesized to arise from its unique structural properties, most importantly the interactions between the N- and C-terminal domains. However, structural understanding of the domain interaction is still lacking. Here, we use Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS) to study domain interactions by measuring the effect of the C-terminal domain (CTD) on the solvent accessibility of the N-terminal domain (NTD) in both apoE3 and apoE4. Our results indicate that the presence of CTD enhances the solvent accessibility of all the four helices in the NTD in apoE4, but only two helices, specifically Helix-1 and 4 in apoE3. Therefore, the allosteric changes in the conformational ensemble of the NTD induced by the CTD is more extensive in apoE4 than in apoE3. Moreover, strong pH dependence suggests role of the salt bridges in the interdomain interactions. Since the NTD harbors the receptor binding region, the destabilizing effect of CTD on it provides a structural basis for the role of interdomain interactions on the pathological functions of apoE4. Furthermore, we propose HDX-MS as a methodology for screening and assessing the efficacy of 'structure corrector' molecules targeting apoE4 to mitigate its pathological effects in AD.
Collapse
Affiliation(s)
- Sudip Pal
- Tata Institute of Fundamental Research, 36/P, Gopanpally Village, Serilingampally Mandal, Hyderabad 500046, India
| | - Subhrajyoti Dolai
- Tata Institute of Fundamental Research, 36/P, Gopanpally Village, Serilingampally Mandal, Hyderabad 500046, India
| | - S Deepa
- Tata Institute of Fundamental Research, 36/P, Gopanpally Village, Serilingampally Mandal, Hyderabad 500046, India
| | - Kanchan Garai
- Tata Institute of Fundamental Research, 36/P, Gopanpally Village, Serilingampally Mandal, Hyderabad 500046, India.
| |
Collapse
|
5
|
Butovsky O, Rosenzweig N. Alzheimer's disease and age-related macular degeneration: Shared and distinct immune mechanisms. Immunity 2025; 58:1120-1139. [PMID: 40324382 DOI: 10.1016/j.immuni.2025.04.013] [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: 01/22/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) and age-related macular degeneration (AMD) represent the leading causes of dementia and vision impairment in the elderly, respectively. The retina is an extension of the brain, yet these two central nervous system (CNS) compartments are often studied separately. Despite affecting cognition vs. vision, AD and AMD share neuroinflammatory pathways. By comparing these diseases, we can identify converging immune mechanisms and potential cross-applicable therapies. Here, we review immune mechanisms highlighting the shared and distinct aspects of these two age-related neurodegenerative conditions, focusing on responses to hallmark disease manifestations, the opposite role of overlapping immune risk loci, and potential unified therapeutic approaches. We also discuss unique tissue requirements that may dictate different outcomes of conserved immune mechanisms and how we can reciprocally utilize lessons from AD therapeutics to AMD. Looking forward, we suggest promising directions for research, including the exploration of regenerative medicine, gene therapies, and innovative diagnostics.
Collapse
Affiliation(s)
- Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Neta Rosenzweig
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Mejía-Ortiz P, Genis-Mendoza AD, Ramírez Villanueva R, López Ramírez S, Guzmán Sánchez R, Fernández T, Sigg-Alonso J, Nicolini-Sánchez H. Shorter Telomere Length in Individuals with Neurocognitive Disorder and APOE ε4 Genotype. Int J Mol Sci 2025; 26:4577. [PMID: 40429722 DOI: 10.3390/ijms26104577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Neurocognitive disorders (NCD) are neurodegenerative diseases characterized by decline or loss of cognitive functions. Aging and the APOE genotype have been identified as major risk factors. Telomere length (TL) has been proposed as a biomarker of aging, with shorter TL associated with cognitive decline. This study investigated the relationship between TL and the APOE genotype in individuals with cognitive impairments (CIs). A total of 170 participants aged >55 years were included. Cognitive function was assessed using the MMSE and MoCA tests. Relative telomere quantification and APOE genotype were determined by real-time PCR. A significant association was observed between shorter TL and an increased risk of CI (p < 0.001). Although APOE ε4 is a known genetic risk factor, its association with CI was less clear in this study population, as a considerable proportion of ε4 carriers did not present cognitive impairment (p < 0.05). However, ε4 carriers with CI tended to have shorter TL than those with non-cognitive impairment (NCI-SMC). Furthermore, fewer years of education were strongly correlated with higher CI risk (p < 0.0001). Overall, individuals with both shorter telomeres and lower educational levels exhibited the highest risk of CI. APOE ε4 may contribute to telomere shortening.
Collapse
Affiliation(s)
- Paola Mejía-Ortiz
- Posgrado en Ciencias (Neurobiología), Unidad de Enseñanza Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla 3001, Querétaro C.P. 76230, Mexico
- Laboratorio de Genómica de las Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Ciudad de México C.P. 14610, Mexico
| | - Alma Delia Genis-Mendoza
- Laboratorio de Genómica de las Enfermedades Psiquiátricas y Neurodegenerativas, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Ciudad de México C.P. 14610, Mexico
- Hospital Psiquiátrico Infantil Dr. Juan N. Navarro, Servicios de Atención Psiquiátrica, Secretaria de Salud, Periferico sur 4809, Tlalpan CDMX, Ciudad de México C.P. 14610, Mexico
| | - Ramon Ramírez Villanueva
- Servicio de Geriatría y Gerontología, ISSSTE Clínica de Medicina Familiar Dr. Ignacio Chávez, Oriental 10, Coapa, Coyoacán, Ciudad de México C.P. 04800, Mexico
| | - Susana López Ramírez
- Servicio de Geriatría y Gerontología, ISSSTE Clínica de Medicina Familiar Dr. Ignacio Chávez, Oriental 10, Coapa, Coyoacán, Ciudad de México C.P. 04800, Mexico
| | - Rafael Guzmán Sánchez
- Servicio de Geriatría y Gerontología, ISSSTE Clínica de Medicina Familiar Dr. Ignacio Chávez, Oriental 10, Coapa, Coyoacán, Ciudad de México C.P. 04800, Mexico
| | - Thalia Fernández
- Laboratorio de Psicofisiología, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla 3001, Querétaro C.P. 76230, Mexico
| | - Jorge Sigg-Alonso
- Laboratorio de Psicofisiología, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla 3001, Querétaro C.P. 76230, Mexico
| | - Humberto Nicolini-Sánchez
- Hospital Psiquiátrico Infantil Dr. Juan N. Navarro, Servicios de Atención Psiquiátrica, Secretaria de Salud, Periferico sur 4809, Tlalpan CDMX, Ciudad de México C.P. 14610, Mexico
| |
Collapse
|
7
|
Harada R, Kudo K, Carmona Soto G, Iwata R, Yanai K, Taki Y, Kudo Y, Furumoto S, Okamura N. Visualization of Apolipoprotein E-Binding Amyloid Plaques in Postmortem Alzheimer's Disease Brains Using a Novel Fluorescent Probe THK-5320. ACS Chem Neurosci 2025; 16:1804-1814. [PMID: 40292539 DOI: 10.1021/acschemneuro.4c00882] [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: 04/30/2025] Open
Abstract
(E)-2-(4-(dimethylamino)styryl)-N,N-dimethylquinolin-6-amine) (THK-5320) is a unique fluorescent compound that recognizes apolipoprotein E (ApoE)-binding amyloid plaques in postmortem human brain sections. To understand the distinctive characteristics of THK-5320 chemically and biologically, its fluorescence properties were investigated, and the association of the fluorescence wavelength with plaque subtypes and amyloid isoforms was explored. Blue plaques visualized with THK-5320 were consistent with those with anti-amyloid-β1-16/amyloid-βN3pE-stained antibodies, whereas red plaques visualized with THK-5320 were consistent with those with an ApoE-stained antibody in postmortem brain sections from patients with Alzheimer's disease. In contrast, the amyloid positron emission tomography (PET) tracer PiB and its fluorescent derivative did not show significant signals in ApoE-binding plaques, whereas the signals correlated well with those of amyloid-βN3pE-positive plaques. Thus, THK-5320 may detect ApoE-binding amyloid plaques that conventional amyloid PET probes cannot detect. Multispectral fluorescence imaging with THK-5320 could be a useful tool to better understand the role of ApoE in amyloid pathology.
Collapse
Affiliation(s)
- Ryuichi Harada
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai 980-8578, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8577, Japan
- Research Center for Accelerator and Radioisotope Science (RARiS), Tohoku University, Sendai 982-0826, Japan
| | - Kaede Kudo
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8577, Japan
| | - Gonzalo Carmona Soto
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai 980-8578, Japan
| | - Ren Iwata
- Research Center for Accelerator and Radioisotope Science (RARiS), Tohoku University, Sendai 982-0826, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai 980-8578, Japan
- Research Center for Accelerator and Radioisotope Science (RARiS), Tohoku University, Sendai 982-0826, Japan
| | - Yasuyuki Taki
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8577, Japan
| | - Yukitsuka Kudo
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8577, Japan
| | - Shozo Furumoto
- Research Center for Accelerator and Radioisotope Science (RARiS), Tohoku University, Sendai 982-0826, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai 983-8536, Japan
- Division of Brain Science, Department of Aging Research and Geriatrics Medicine, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8577, Japan
- Research Center for Accelerator and Radioisotope Science (RARiS), Tohoku University, Sendai 982-0826, Japan
| |
Collapse
|
8
|
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
|
9
|
Timofeeva AM, Aulova KS, Nevinsky GA. Modeling Alzheimer's Disease: A Review of Gene-Modified and Induced Animal Models, Complex Cell Culture Models, and Computational Modeling. Brain Sci 2025; 15:486. [PMID: 40426657 DOI: 10.3390/brainsci15050486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/30/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Alzheimer's disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer's disease and facilitate therapeutic development. Mammals, especially mice, are the most common models for studying the pathogenesis of this disease in vivo. To date, the scientific literature has documented more than 280 mouse models exhibiting diverse aspects of Alzheimer's disease pathogenesis. Other mammalian species, including rats, pigs, and primates, have also been utilized as models. Selected aspects of Alzheimer's disease have also been modeled in simpler model organisms, such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio. It is possible to model Alzheimer's disease not only by creating genetically modified animal lines but also by inducing symptoms of this neurodegenerative disease. This review discusses the main methods of creating induced models, with a particular focus on modeling Alzheimer's disease on cell cultures. Induced pluripotent stem cell (iPSC) technology has facilitated novel investigations into the mechanistic underpinnings of diverse diseases, including Alzheimer's. Progress in culturing brain tissue allows for more personalized studies on how drugs affect the brain. Recent years have witnessed substantial advancements in intricate cellular system development, including spheroids, three-dimensional scaffolds, and microfluidic cultures. Microfluidic technologies have emerged as cutting-edge tools for studying intercellular interactions, the tissue microenvironment, and the role of the blood-brain barrier (BBB). Modern biology is experiencing a significant paradigm shift towards utilizing big data and omics technologies. Computational modeling represents a powerful methodology for researching a wide array of human diseases, including Alzheimer's. Bioinformatic methodologies facilitate the analysis of extensive datasets generated via high-throughput experimentation. It is imperative to underscore the significance of integrating diverse modeling techniques in elucidating pathogenic mechanisms in their entirety.
Collapse
Affiliation(s)
- Anna M Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| | - Kseniya S Aulova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| | - Georgy A Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| |
Collapse
|
10
|
Ellis D, Watanabe K, Wilmanski T, Lustgarten MS, Korat AVA, Glusman G, Hadlock J, Fiehn O, Sebastiani P, Price ND, Hood L, Magis AT, Evans SJ, Pflieger L, Lovejoy JC, Gibbons SM, Funk CC, Baloni P, Rappaport N. APOE genotype and biological age impact inter-omic associations related to bioenergetics. Aging (Albany NY) 2025; 17:206243. [PMID: 40323280 DOI: 10.18632/aging.206243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Apolipoprotein E (APOE) modifies human aging; specifically, the ε2 and ε4 alleles are among the strongest genetic predictors of longevity and Alzheimer's disease (AD) risk, respectively. However, detailed mechanisms for their influence on aging remain unclear. In the present study, we analyzed multi-omic association patterns across APOE genotypes, sex, and biological age (BA) axes in 2,229 community dwelling individuals. Our analysis, supported by validation in an independent cohort, identified diacylglycerols as the top APOE-associated plasma metabolites. However, despite the known opposing aging effects of the allele variants, both ε2- and ε4-carriers showed higher diacylglycerols compared to ε3-homozygotes. 'Omics association patterns of ε2-carriers and increased biological age were also counter-intuitively similar, displaying significantly increased associations between insulin resistance markers and energy-generating pathway metabolites. These results demonstrate the context-dependence of the influence of APOE, with ε2 potentially strengthening insulin resistance-like pathways in the decades prior to imparting its longevity benefits. Additionally, they provide an atlas of APOE-related 'omic associations and support the involvement of bioenergetic pathways in mediating the impact of APOE on aging.
Collapse
Affiliation(s)
- Dylan Ellis
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Kengo Watanabe
- Institute for Systems Biology, Seattle, WA 98109, USA
- Present address: Department of Medical Artificial Intelligence and Data Science, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | | | - Michael S Lustgarten
- Metabolism and Basic Biology of Aging, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | - Andres V Ardisson Korat
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| | | | - Jennifer Hadlock
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA 95616, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Nathan D Price
- Thorne HealthTech, New York, NY 10019, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Phenome Health, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Simon J Evans
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Lance Pflieger
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jennifer C Lovejoy
- Institute for Systems Biology, Seattle, WA 98109, USA
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- eScience Institute, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Priyanka Baloni
- Institute for Systems Biology, Seattle, WA 98109, USA
- Present address: School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Noa Rappaport
- Institute for Systems Biology, Seattle, WA 98109, USA
- Phenome Health, Seattle, WA 98109, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| |
Collapse
|
11
|
Chun MY, Park YH, Kim HJ, Na DL, Kim JP, Seo SW, Jang H. Distinct Characteristics of Suspected Non-Alzheimer Pathophysiology in Relation to Cognitive Status and Cerebrovascular Burden. Clin Nucl Med 2025; 50:368-380. [PMID: 40025666 PMCID: PMC11969373 DOI: 10.1097/rlu.0000000000005793] [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: 11/18/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF THE REPORT This study investigated the prevalence and clinical characteristics of suspected non-Alzheimer disease pathophysiology (SNAP) across varying cognitive statuses and cerebral small vessel disease (CSVD) burden. PATIENTS AND METHODS We included 1992 participants with cognitive status categorized as cognitively unimpaired, mild cognitive impairment, or dementia. β-amyloid (Aβ, A) positivity was assessed by Aβ PET, and neurodegeneration (N) positivity was determined through hippocampal volume. Participants were further divided by the presence or absence of severe CSVD. The clinical and imaging characteristics of A-N+ (SNAP) group were compared with those of the A-N- and A+N+ groups. RESULTS SNAP participants were older and had more vascular risk factors compared with A-N- and A+N+ in the CSVD(-) cohort. SNAP and A+N+ showed similar cortical thinning. At the dementia stage, SNAP had a cognitive trajectory similar to A+N+ in the CSVD(-) cohort. However, SNAP exhibited less cognitive decline than A+N+ in the CSVD(+) cohort. CONCLUSIONS SNAP is characterized by distinct clinical and imaging characteristics; however, it does not necessarily indicate a benign prognosis, particularly at the dementia stage. These findings highlight the need to assess SNAP in relation to the cognitive stage and CSVD presence to better understand its progression and guide interventions.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine
- Department of Neurology, Yonsei University College of Medicine
| | - Yu Hyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Gangnam-gu
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-gu, Seoul, South Korea
| |
Collapse
|
12
|
van Olst L, Simonton B, Edwards AJ, Forsyth AV, Boles J, Jamshidi P, Watson T, Shepard N, Krainc T, Argue BM, Zhang Z, Kuruvilla J, Camp L, Li M, Xu H, Norman JL, Cahan J, Vassar R, Chen J, Castellani RJ, Nicoll JA, Boche D, Gate D. Microglial mechanisms drive amyloid-β clearance in immunized patients with Alzheimer's disease. Nat Med 2025; 31:1604-1616. [PMID: 40050704 DOI: 10.1038/s41591-025-03574-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease (AD) therapies utilizing amyloid-β (Aβ) immunization have shown potential in clinical trials. Yet, the mechanisms driving Aβ clearance in the immunized AD brain remain unclear. Here, we use spatial transcriptomics to explore the effects of both active and passive Aβ immunization in the AD brain. We compare actively immunized patients with AD with nonimmunized patients with AD and neurologically healthy controls, identifying distinct microglial states associated with Aβ clearance. Using high-resolution spatial transcriptomics alongside single-cell RNA sequencing, we delve deeper into the transcriptional pathways involved in Aβ removal after lecanemab treatment. We uncover spatially distinct microglial responses that vary by brain region. Our analysis reveals upregulation of the triggering receptor expressed on myeloid cells 2 (TREM2) and apolipoprotein E (APOE) in microglia across immunization approaches, which correlate positively with antibody responses and Aβ removal. Furthermore, we show that complement signaling in brain myeloid cells contributes to Aβ clearance after immunization. These findings provide new insights into the transcriptional mechanisms orchestrating Aβ removal and shed light on the role of microglia in immune-mediated Aβ clearance. Importantly, our work uncovers potential molecular targets that could enhance Aβ-targeted immunotherapies, offering new avenues for developing more effective therapeutic strategies to combat AD.
Collapse
Affiliation(s)
- Lynn van Olst
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brooke Simonton
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alex J Edwards
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Anne V Forsyth
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jake Boles
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pouya Jamshidi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Thomas Watson
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nate Shepard
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Talia Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benney Mr Argue
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyang Zhang
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joshua Kuruvilla
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lily Camp
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mengwei Li
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hang Xu
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Jeanette L Norman
- Clinical Neurosciences, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Joshua Cahan
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert Vassar
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jinmiao Chen
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore, Singapore
- Centre for Computational Biology and Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Immunology Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rudolph J Castellani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - James Ar Nicoll
- Clinical Neurosciences, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Cellular Pathology, University Hospital Southampton National Health Service Trust, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - David Gate
- Abrams Research Center on Neurogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
13
|
Dasadhikari S, Ghosh S, Pal S, Knowles TPJ, Garai K. A single fibril study reveals that ApoE inhibits the elongation of Aβ42 fibrils in an isoform-dependent manner. Commun Chem 2025; 8:133. [PMID: 40307479 PMCID: PMC12044155 DOI: 10.1038/s42004-025-01524-z] [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: 10/11/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
ApoE-ε4 is the strongest genetic risk factor for late-onset Alzheimer's disease (AD), linked to increased amyloid-β (Aβ) deposition in the brain. In AD mouse models, microglial expression of apoE3 reduces amyloid plaque burden through enhanced phagocytosis, whereas apoE4 is associated with impaired Aβ clearance. However, the isoform-specific interactions of apoE with Aβ aggregates and the molecular mechanisms by which these isoforms influence Aβ aggregation and clearance remain poorly understood, which is critical for developing potential therapeutic interventions. Here, we employed TIRFM, superresolution microscopy, and single-molecule photobleaching techniques to investigate the isoform-specific effects of apoE on the rate constants of Aβ42 aggregation at the single-fibril level, as well as to quantify the binding affinity and specificity of apoE isoforms to individual Aβ fibril ends. Our results show that apoE4 is ca. 4-5 times less effective than apoE3 and apoE2 in inhibiting fibril elongation, while secondary nucleation is largely unaffected by any of the isoforms. Furthermore, apoE3 exhibits stronger and more specific binding to fibril ends compared to apoE4. These findings suggest that apoE4's reduced affinity for growing fibril ends may impair microglial clearance and increase amyloid deposition through a higher elongation rate in the brain of ApoE-ε4 carriers.
Collapse
Affiliation(s)
| | - Shamasree Ghosh
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, SE-90187, Sweden
| | - Sudip Pal
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | - Kanchan Garai
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India.
| |
Collapse
|
14
|
Wendt S, Lin AJ, Ebert SN, Brennan DJ, Cai W, Bai Y, Kong DY, Sorrentino S, Groten CJ, Lee C, Frew J, Choi HB, Karamboulas K, Delhaye M, Mackenzie IR, Kaplan DR, Miller FD, MacVicar BA, Nygaard HB. A 3D human iPSC-derived multi-cell type neurosphere system to model cellular responses to chronic amyloidosis. J Neuroinflammation 2025; 22:119. [PMID: 40275379 PMCID: PMC12023538 DOI: 10.1186/s12974-025-03433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive amyloid beta (Aβ) deposition in the brain, with eventual widespread neurodegeneration. While the cell-specific molecular signature of end-stage AD is reasonably well characterized through autopsy material, less is known about the molecular pathways in the human brain involved in the earliest exposure to Aβ. Human model systems that not only replicate the pathological features of AD but also the transcriptional landscape in neurons, astrocytes and microglia are crucial for understanding disease mechanisms and for identifying novel therapeutic targets. METHODS In this study, we used a human 3D iPSC-derived neurosphere model to explore how resident neurons, microglia and astrocytes and their interplay are modified by chronic amyloidosis induced over 3-5 weeks by supplementing media with synthetic Aβ1 - 42 oligomers. Neurospheres under chronic Aβ exposure were grown with or without microglia to investigate the functional roles of microglia. Neuronal activity and oxidative stress were monitored using genetically encoded indicators, including GCaMP6f and roGFP1, respectively. Single nuclei RNA sequencing (snRNA-seq) was performed to profile Aβ and microglia driven transcriptional changes in neurons and astrocytes, providing a comprehensive analysis of cellular responses. RESULTS Microglia efficiently phagocytosed Aβ inside neurospheres and significantly reduced neurotoxicity, mitigating amyloidosis-induced oxidative stress and neurodegeneration following different exposure times to Aβ. The neuroprotective effects conferred by the presence of microglia was associated with unique gene expression profiles in astrocytes and neurons, including several known AD-associated genes such as APOE. These findings reveal how microglia can directly alter the molecular landscape of AD. CONCLUSIONS Our human 3D neurosphere culture system with chronic Aβ exposure reveals how microglia may be essential for the cellular and transcriptional responses in AD pathogenesis. Microglia are not only neuroprotective in neurospheres but also act as key drivers of Aβ-dependent APOE expression suggesting critical roles for microglia in regulating APOE in the AD brain. This novel, well characterized, functional in vitro platform offers unique opportunities to study the roles and responses of microglia to Aβ modelling key aspects of human AD. This tool will help identify new therapeutic targets, accelerating the transition from discovery to clinical applications.
Collapse
Affiliation(s)
- Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Sarah N Ebert
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Declan J Brennan
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Wenji Cai
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Yanyang Bai
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Da Young Kong
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Stefano Sorrentino
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher Lee
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Jonathan Frew
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Opalia Co, Montreal, QC, H2X 3Y7, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
| | - Mathias Delhaye
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Ian R Mackenzie
- Department of Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David R Kaplan
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1 A8, Canada
| | - Freda D Miller
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| |
Collapse
|
15
|
Campagna J, Chandra S, Teter B, Cohn W, Pham J, Kim YS, Jagodzinska B, Vadivel K, Alam P, Bilousova T, Young M, Elias C, Marcucci J, Flacau I, Jackman A, Padder S, Wi D, Zhu C, Spilman P, Jung ME, Bredesen DE, John V. Discovery of an ApoE4-targeted small-molecule SirT1 enhancer for the treatment of Alzheimer's disease. Sci Rep 2025; 15:14028. [PMID: 40269061 PMCID: PMC12019328 DOI: 10.1038/s41598-025-96131-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
Decreased expression of sirtuin 1 (SirT1) has been implicated in Alzheimer's disease (AD), and as we previously reported, is related to transcriptional repression by the major risk factor for sporadic AD, apolipoprotein E4 (ApoE4). Herein we describe the discovery of an orally brain-permeable small-molecule, DDL-218, that enhanced SirT1 in ApoE4-expressing neuronal cells and a murine AD model. DDL-218 increased the transcription factor NFYb resulting in upregulation of PRMT5. Mechanistic and modeling studies show that binding of ApoE4 to the SirT1 gene promoter can be displaced by PRMT5 leading to increased SirT1 transcription. DDL-218 treatment elicited improvement in memory in the AD model, suggesting that DDL-218 enhancement of neurotrophic SirT1 in the brain has potential to modulate neuronal activity that may clinically provide an improvement in cognitive function and complement the current anti-Aβ antibody monotherapy. Our findings support further development of DDL-218 as a novel ApoE4-targeted therapeutic candidate for AD.
Collapse
Affiliation(s)
- Jesus Campagna
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Sujyoti Chandra
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Bruce Teter
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Whitaker Cohn
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Johnny Pham
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA, 90095, USA
| | - Young-Sug Kim
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA, 90095, USA
| | - Barbara Jagodzinska
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Kanagasabai Vadivel
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Parvez Alam
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Tina Bilousova
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Malaney Young
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Chris Elias
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Juan Marcucci
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Ilinca Flacau
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Ainsley Jackman
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Samar Padder
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Dongwook Wi
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Chunni Zhu
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Patricia Spilman
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Michael E Jung
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, 650 Charles E. Young Drive, Los Angeles, CA, 90095, USA
| | - Dale E Bredesen
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA
| | - Varghese John
- The Drug Discovery Lab, Department of Neurology, David Geffen School of Medicine, 710 Westwood Plaza, Los Angeles, CA, 90095, USA.
| |
Collapse
|
16
|
Yang X, Chi L, Qiao M, Huang A, Wu H, Chen S, Fan J, Lin X, Chen J. The effect of C-reactive protein and interleukin-3 on mild cognitive impairment with APOE ɛ4. J Alzheimers Dis 2025:13872877251333149. [PMID: 40261294 DOI: 10.1177/13872877251333149] [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: 04/24/2025]
Abstract
BackgroundThe apolipoprotein E ε4 allele (APOE ε4) and inflammation are associated with Alzheimer's disease (AD) pathology. Mild cognitive impairment (MCI) is considered the preclinical and early stage of AD. However, the comprehensive effects of APOE ε4 and inflammatory mediators on MCI patients with specific APOE ε4 genotypes remain poorly understood.ObjectiveOur study aimed to explore how different numbers of the APOE ε4 alleles affect plasma C-reactive protein (CRP) and interleukin-3 (IL-3) levels and their associations with brain structure.MethodsA total of 339 MCI patients from the Alzheimer's Disease Neuroimaging Initiative study were enrolled. We compared their plasma concentrations of CRP and IL-3, cognitive performance, and cerebrospinal fluid (CSF) AD biomarkers levels across different APOE ε4 genotypes. Structural magnetic resonance imaging was utilized to measure gray matter volume outcomes. Pearson correlation analysis was used to explore the associations between the above indicators.ResultsPlasma CRP levels increased in the APOE ε4 carriers, but IL-3 expression notably decreased, and the homozygous state is the most significant. A negative correlation between CRP and several cognitive abilities was observed only in APOE ε4 homozygotes. Additionally, a positive correlation between IL-3, cognitive scores, and CSF biomarker levels was confirmed only in APOE ε4 homozygotes. Imaging data demonstrated that the gray matter volume of the right middle frontal gyrus was associated with CRP only in APOE ε4 non-carriers.ConclusionsOur study demonstrated that peripheral inflammatory mediators' effect on cognitive function and brain structure in MCI patients differs based on their APOE ε4 allele carrier status.
Collapse
Affiliation(s)
- Xinyi Yang
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Chi
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Meizhao Qiao
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Anxing Huang
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Huimin Wu
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Shanshan Chen
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jia Fan
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Xingjian Lin
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
17
|
Rajendra A, Bondonno NP, Murray K, Zhong L, Rainey-Smith SR, Gardener SL, Blekkenhorst LC, Doré V, Villemagne VL, Laws SM, Brown BM, Taddei K, Masters CL, Rowe CC, Martins RN, Hodgson JM, Bondonno CP. Baseline habitual dietary nitrate intake and Alzheimer's Disease related neuroimaging biomarkers in the Australian Imaging, Biomarkers and Lifestyle study of ageing. J Prev Alzheimers Dis 2025:100161. [PMID: 40221237 DOI: 10.1016/j.tjpad.2025.100161] [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: 10/23/2024] [Revised: 02/27/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Dietary nitrate, as a nitric oxide (NO) precursor, may support brain health and protect against dementia. OBJECTIVE Our primary aim was to investigate whether dietary nitrate is associated with neuroimaging markers of brain health linked with Alzheimer's disease (AD). PARTICIPANTS Study participants were cognitively unimpaired individuals from the Australian Imaging, Biomarkers and Lifestyle Study of Ageing (AIBL) who had β-amyloid positron emission tomography (PET) scans (n = 554) and magnetic resonance imaging (MRI) scans (n = 335) and had completed a Food Frequency Questionnaire at baseline. METHODS Source-specific nitrate intakes were estimated using comprehensive nitrate food composition databases. Rates of cerebral β-amyloid (Aβ) deposition, measured using PET, and rates of brain atrophy, measured using MRI, were assessed between baseline and 126-months follow-up, at intervals of 18 months. Multivariable-adjusted linear mixed effect models were used to examine associations between baseline source-specific nitrate intake and rates of (i) cerebral Aβ deposition and (ii) brain atrophy, over the 126 months of follow-up. Analyses were carried out following stratification of the sample by established dementia Alzheimer's disease (AD) risk factors including sex and presence or absence of the apolipoprotein E (APOE) ε4 allele. RESULTS In women carriers of the APOE ε4 allele, higher plant sourced nitrate intake (median intake 121 mg/day), was associated with a slower rate of cerebral Aβ deposition [β: 4.47 versus 8.99 Centiloid (CL) /18 months, p < 0.05] and right hippocampal atrophy [-0.01 versus -0.03 mm3 /18 months, p < 0.01], after multivariable adjustments. Moderate intake showed protective associations in men carriers and in both men and women non-carriers of APOE ε4. CONCLUSIONS Associations were observed between plant-derived nitrate intake and cerebral Aβ deposition, particularly in high-risk populations (women and APOE ε4 carriers). Associations were also observed for brain volume atrophy, however these exhibited subgroup variability without clear patterns relative to sex and APOE ε4 allele carriage. These findings suggest a potential link between plant-sourced nitrate and AD related neuroimaging markers of brain health improved brain health, but further validation in larger studies is required.
Collapse
Affiliation(s)
- Anjana Rajendra
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Nicola P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; The Danish Cancer Institute, Copenhagen, Denmark
| | - Kevin Murray
- School of Population and Global Health, University of Western Australia, Perth, Western Australia, Australia
| | - Liezhou Zhong
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Perth, Western Australia, Australia
| | - Samantha L Gardener
- Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Australian Alzheimer's Research Foundation, Nedlands, Western Australia, Australia
| | - Lauren C Blekkenhorst
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; For a full list of the AIBL Research Group see aibl.org.au
| | - Vincent Doré
- Australian E-Health Research Centre, CSIRO, 351 Royal Parade, Parkville, Victoria, Australia; Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia; Department of Psychiatry, University of Pittsburgh, Thomas Detre Hall, 3811 O'Hara Street, Pittsburgh, PA, USA; Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia; Collaborative Genomics and Translation Group, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia; Curtin Medical School, Curtin University, Kent Street, Bentley, Western Australia, Australia
| | - Belinda M Brown
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Lifestyle Approaches Towards Cognitive Health Research Group, Murdoch University, Murdoch, Western Australia, Australia; Collaborative Genomics and Translation Group, Edith Cowan University, 270 Joondalup Drive, Joondalup, Western Australia, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy, Austin Health, 145 Studley Road, Heidelberg, Victoria, Australia; The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Jonathan M Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia
| | - Catherine P Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia; Medical School, The University of Western Australia, Royal Perth Hospital Research Foundation, Perth, Western Australia, Australia.
| |
Collapse
|
18
|
Xu IRL, Danzi MC, Raposo J, Züchner S. The continued promise of genomic technologies and software in neurogenetics. J Neuromuscul Dis 2025:22143602251325345. [PMID: 40208247 DOI: 10.1177/22143602251325345] [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: 04/11/2025]
Abstract
The continued evolution of genomic technologies over the past few decades has revolutionized the field of neurogenetics, offering profound insights into the genetic underpinnings of neurological disorders. Identification of causal genes for numerous monogenic neurological conditions has informed key aspects of disease mechanisms and facilitated research into critical proteins and molecular pathways, laying the groundwork for therapeutic interventions. However, the question remains: has this transformative trend reached its zenith? In this review, we suggest that despite significant strides in genome sequencing and advanced computational analyses, there is still ample room for methodological refinement. We anticipate further major genetic breakthroughs corresponding with the increased use of long-read genomes, variant calling software, AI tools, and data aggregation databases. Genetic progress has historically been driven by technological advancements from the commercial sector, which are developed in response to academic research needs, creating a continuous cycle of innovation and discovery. This review explores the potential of genomic technologies to address the challenges of neurogenetic disorders. By outlining both established and modern resources, we aim to emphasize the importance of genetic technologies as we enter an era poised for discoveries.
Collapse
Affiliation(s)
- Isaac R L Xu
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jacquelyn Raposo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
19
|
Ramsden CE, Cutler RG, Li X, Keyes GS. HYPOTHESIS: Lipid-protecting disulfide bridges are the missing molecular link between ApoE4 and sporadic Alzheimer's disease in humans. Prostaglandins Leukot Essent Fatty Acids 2025; 205:102681. [PMID: 40209641 DOI: 10.1016/j.plefa.2025.102681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
As the principal lipid transporter in the human brain, apolipoprotein E (ApoE) is tasked with transport and protection of highly vulnerable lipids that are required to support and remodel neuronal membranes, in a process that is dependent on ApoE receptors. APOE allele variants that encode proteins differing only in the number of cysteine (Cys)-to-arginine (Arg) exchanges (ApoE2 [2 Cys], ApoE3 [1 Cys], ApoE4 [0 Cys]) comprise the strongest genetic risk factor for sporadic Alzheimer's disease (AD); however, the specific molecular feature(s) and resultant mechanisms that underlie these isoform-dependent effects are unknown. One signature feature of Cys is the capacity to form disulfide (Cys-Cys) bridges, which are required to form disulfide-linked dimers and multimers. Here we propose the overarching hypothesis that super-ability (for ApoE2), intermediate ability (for ApoE3) or inability (for ApoE4) to form lipid-protecting intermolecular disulfide bridges, is the central molecular determinant accounting for the disparate effects of APOE alleles on AD risk and amyloid-β and Tau pathologies in humans. We posit that presence and abundance of Cys in human ApoE3 and ApoE2 respectively, conceal and protect vulnerable lipids transported by ApoE from peroxidation by enabling formation of disulfide-linked homo- and heteromeric ApoE complexes. We thus propose that inability to form intermolecular disulfide bridges makes ApoE4-containing lipoproteins uniquely vulnerable to peroxidation and its downstream consequences. Consistent with our model, we found that brain-enriched polyunsaturated fatty acid-containing phospholipids induce disulfide-dependent dimerization and multimerization of ApoE3 and ApoE2 (but not ApoE4). By contrast, incubation with the peroxidation-resistant lipid DMPC or cholesterol alone had minimal effects on dimerization. These novel concepts and findings are integrated into our unifying model implicating peroxidation of ApoE-containing lipoproteins, with consequent ApoE receptor-ligand disruption, as initiating molecular events that ultimately lead to AD in humans.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA; NIH, Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD 20892, USA.
| | - Roy G Cutler
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD 21224, USA
| |
Collapse
|
20
|
Kerr EM, Ailshire JA, Crimmins E, Walsemann KM. Sex variation in the relationship between APOE ε4, cognitive decline, and dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2025; 17:e70053. [PMID: 40322470 PMCID: PMC12047073 DOI: 10.1002/dad2.70053] [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: 07/31/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 05/08/2025]
Abstract
INTRODUCTION We examine if the relationship between apolipoprotein E (APOE) ε4 and cognitive decline and dementia onset differs by sex in non-Hispanic White and Black respondents from the Health and Retirement Study. METHODS We used race-stratified linear mixed models to estimate cognitive decline and Cox proportional hazards models to estimate time to dementia onset. Sex differences were estimated using interaction terms. RESULTS APOE ε4 was associated with cognitive decline (b = -0.4) and dementia onset (hazard ratio [HR] = 1.48) in White adults, and cognitive decline (b = -0.5) in Black adults. The relationship between APOE ε4 and cognitive decline or dementia onset did not differ by sex in either group. DISCUSSION Our findings question a key hypothesis in the field-that female APOE ε4 carriers experience faster cognitive decline and earlier dementia onset than their male counterparts-and highlight the importance of using probability samples to reduce survivor and participation bias commonly found in genetics research. Highlights White apolipoprotein E ε4 allele (APOE ε4) carriers had faster cognitive decline and earlier dementia onset.Black APOE ε4 carriers had faster cognitive decline.These patterns did not vary by sex for either Black or White adults.
Collapse
Affiliation(s)
- Eleanor M. Kerr
- School of Public PolicyUniversity of MarylandCollege ParkMarylandUSA
| | - Jennifer A. Ailshire
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Eileen Crimmins
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | |
Collapse
|
21
|
Shi Q, Gutierrez RA, Bhat MA. Microglia, Trem2, and Neurodegeneration. Neuroscientist 2025; 31:159-176. [PMID: 38769824 PMCID: PMC11576490 DOI: 10.1177/10738584241254118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Microglia are a specialized type of neuroimmune cells that undergo morphological and molecular changes through multiple signaling pathways in response to pathological protein aggregates, neuronal death, tissue injury, or infections. Microglia express Trem2, which serves as a receptor for a multitude of ligands enhancing their phagocytic activity. Trem2 has emerged as a critical modulator of microglial activity, especially in many neurodegenerative disorders. Human TREM2 mutations are associated with an increased risk of developing Alzheimer disease (AD) and other neurodegenerative diseases. Trem2 plays dual roles in neuroinflammation and more specifically in disease-associated microglia. Most recent developments on the molecular mechanisms of Trem2, emphasizing its role in uptake and clearance of amyloid β (Aβ) aggregates and other tissue debris to help protect and preserve the brain, are encouraging. Although Trem2 normally stimulates defense mechanisms, its dysregulation can intensify inflammation, which poses major therapeutic challenges. Recent therapeutic approaches targeting Trem2 via agonistic antibodies and gene therapy methodologies present possible avenues for reducing the burden of neurodegenerative diseases. This review highlights the promise of Trem2 as a therapeutic target, especially for Aβ-associated AD, and calls for more mechanistic investigations to understand the context-specific role of microglial Trem2 in developing effective therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Qian Shi
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Raul A. Gutierrez
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
22
|
Zhong R, Chernick D, Hottman D, Tan Y, Kim M, Narayanan M, Li L. The HDL-Mimetic Peptide 4F Mitigates Vascular and Cortical Amyloid Pathology and Associated Neuroinflammation in a Transgenic Mouse Model of Cerebral Amyloid Angiopathy and Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04859-9. [PMID: 40120042 DOI: 10.1007/s12035-025-04859-9] [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: 09/01/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Despite recent advances, more effective and safer treatment options for AD are needed. Cerebral amyloid angiopathy (CAA) is one of the key pathological hallmarks of AD characterized by amyloid-β (Aβ) deposition in the cerebral vasculature and is associated with intracerebral hemorrhage, cerebrovascular dysfunction, and cognitive impairment. CAA is also considered to underlie the main adverse effect of recently FDA-approved anti-Aβ immunotherapies, namely the amyloid-related imaging abnormalities (ARIA). Substantial evidence has shown that elevated levels of high-density lipoprotein (HDL) and its main protein component, APOA-I, are associated with reduced CAA and superior cognitive function. 4F is an APOA-I/HDL-mimetic peptide and its clinical safety and activity have been demonstrated in human trials for cardiovascular diseases. The present study investigates whether treatment with 4F modulates CAA and associated cognitive deficits and neuropathologies in the well-established Tg-SwDI mouse model of CAA/AD. Age/sex-matched Tg-SwDI mice received daily treatments of 4F or vehicle (PBS), respectively, by intraperitoneal injections for 12 weeks. The results showed that 4F treatment reduced overall Aβ plaque deposition and CAA, and attenuated CAA-associated microgliosis, without significantly affecting total levels of Aβ, astrocytosis, and behavioral function. Unbiased transcriptomic analysis revealed a heightened inflammatory state in the brain of SwDI mice and that 4F treatment reversed the overactivation of vascular cells, in particular vascular smooth muscle cells, relieving cerebrovascular inflammation in CAA/AD mice. Our study provides experimental evidence for the therapeutic potential of 4F to mitigate CAA and associated pathologies in AD.
Collapse
Affiliation(s)
- Rui Zhong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Dustin Chernick
- Graduate Program in Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David Hottman
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yejun Tan
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Minwoo Kim
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Manojkumar Narayanan
- Graduate Program in Comparative and Molecular Biosciences, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Graduate Program in Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
23
|
Honda K, Awazu A. Potential multiple disease progression pathways in female patients with Alzheimer's disease inferred from transcriptome and epigenome data of the dorsolateral prefrontal cortex. PLoS One 2025; 20:e0313733. [PMID: 40100818 PMCID: PMC11918443 DOI: 10.1371/journal.pone.0313733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/07/2025] [Indexed: 03/20/2025] Open
Abstract
Late-onset Alzheimer's disease (AD) is a typical type of dementia for which therapeutic strategies have not yet been established. The database of the Rush Alzheimer's Disease study by the ENCODE consortium contains transcriptome and various epigenome data. Although the Rush AD database may contain a satisfactory amount of data for women, the amount of data for men remains insufficient. Here, based on an analysis of publicly available data from female patients, this study found that AD pathology appears to be nonuniform; AD patients were divided into several groups with differential gene expression patterns, including those related to cognitive function. First, cluster analysis was performed on individuals diagnosed with "No Cognitive Impairment (NCI)," "Mild Cognitive Impairment (MCI)," and "Alzheimer's Disease (AD)" stages in clinical trials using gene expression, and multiple substages were identified across AD progression. The epigenome data, in particular genome-wide H3k4me3 distribution data, also supported the existence of multiple AD substages. However, APOE gene polymorphisms of individuals seemed to not correlate with disease stage. An inference of adjacency networks among substages, evaluated via partition-based graph abstraction using the gene expression profiles of individuals, suggested the possibility of multiple typical disease progression pathways from NCI to different AD substages through various MCI substages. These findings could refine biomarker discovery or inform personalized therapeutic approaches.
Collapse
Affiliation(s)
- Kousei Honda
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Akinori Awazu
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Hiroshima, Japan
- Research Center for the Mathematics on Chromatin Live Dynamics, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| |
Collapse
|
24
|
Nakamya MF, Hu K, Jiang C, Chong Z, Liu RM. Age- and ApoE Genotype-Dependent Transcriptomic Responses to O 3 in the Hippocampus of Mice. Int J Mol Sci 2025; 26:2407. [PMID: 40141051 PMCID: PMC11942628 DOI: 10.3390/ijms26062407] [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: 01/16/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is a leading cause of dementia in the elderly, with late-onset AD (LOAD) accounting for 95% of the cases. The etiology underlying LOAD, however, remains unclear. Using a humanized mouse model, we showed previously that exposure to ozone (O3), a potential environment risk factor, in a cyclic exposure protocol that mimics a human exposure scenario, accelerated AD-like neuropathophysiology in old humanized male ApoE3 (E3) but not ApoE4 (E4) mice. Using RNA sequencing (RNA-seq) techniques, we further demonstrate here that the ApoE genotype has the greatest influence on transcriptional changes, followed by age and O3 exposure. Notably, AD-related genes were expressed even at baseline and in young mice, but the differences in the expression levels are obvious in old age. Importantly, although both E3 and E4 mice exhibited some AD-related transcriptomic alterations, old E3 mice exposed to O3, which showed memory impairment, experienced more pronounced disruptions in the expression of genes related to redox balance, neurogenesis, neuroinflammation, and cellular senescence in the hippocampus, compared with O3-exposed old E4 mice. These results provide new insights into the molecular mechanisms underlying memory loss in O3-exposed old E3 male mice and emphasize the complexity of interactions between gene, environment, and aging in AD pathophysiology.
Collapse
Affiliation(s)
- Mary F. Nakamya
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| | - Kaili Hu
- Department of Biomedical Informatics and Data Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Chunsun Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| | - Zechen Chong
- Department of Biomedical Informatics and Data Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Rui-Ming Liu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.F.N.); (C.J.)
| |
Collapse
|
25
|
Wang N, Pan Y, Starling SC, Haskell DH, Quintero AC, Kawatani K, Inoue Y, Shue F, Ma X, Aikawa T, Martens YA, Kurti A, Parsons TM, Perkerson RB, Roy B, Raulin A, Ren Y, DeTure M, Dickson DW, Bao H, Han X, Bu G, Kanekiyo T. Neuronal ABCA7 deficiency aggravates mitochondrial dysfunction and neurodegeneration in Alzheimer's disease. Alzheimers Dement 2025; 21:e70112. [PMID: 40145325 PMCID: PMC11947734 DOI: 10.1002/alz.70112] [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: 01/08/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION Loss-of-function variants of the ABCA7 gene are associated with an increased risk of Alzheimer's disease (AD). How neuronal ABCA7 contributes to AD pathogenesis is unknown. METHODS Using neuron-specific Abca7 KO mice (nAbca7-/-) with or without 5×FAD amyloid model background and post mortem AD brains, we investigated AD-related phenotypes through comprehensive approaches including transcriptomics and lipidomics. RESULTS Lipidomics analysis detected altered lipid profiles in the brains and synaptosomes of 5×FAD; nAbca7-/- mice compared to controls. Transcriptomics profiling revealed that neuronal ABCA7 deficiency altered the expression of genes and pathways related to mitochondrial homeostasis and apoptosis, particularly in excitatory neurons. Consistently, synaptosomes isolated from 5×FAD; nAbca7-/- mice showed diminished mitochondria respiration and reduced synaptic protein levels, which is further supported by results from human AD brains. DISCUSSION Our findings reveal that neuronal ABCA7 plays a critical role in mitochondrial homeostasis important for neuronal function and survival in the presence of AD pathology. HIGHLIGHTS Neuronal ABCA7 deficiency exacerbates Aβ pathology and neuronal damage in 5×FAD mice. Neuronal ABCA7 deficiency alters brain transcriptomes and lipidomes of 5×FAD mice. Neuronal ABCA7 deficiency disturbs mitochondria functions in synaptosomes from 5×FAD mice. Neuronal ABCA7 expression associates with genes and pathways related to mitochondrial homeostasis in AD brains.
Collapse
Affiliation(s)
- Ni Wang
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yining Pan
- Department of Public HealthUniversity of North FloridaJacksonvilleFloridaUSA
| | | | | | | | - Keiji Kawatani
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yasuteru Inoue
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Francis Shue
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Xiaoye Ma
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Yuka A. Martens
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- SciNeuro PharmaceuticalsRockvilleMarylandUSA
| | - Aishe Kurti
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Bhaskar Roy
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Yingxue Ren
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Michael DeTure
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Hanmei Bao
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Division of Life ScienceThe Hong Kong University of Science and TechnologyClear Water Bay Hong KongChina
| | | |
Collapse
|
26
|
Andersen JV. The Glutamate/GABA-Glutamine Cycle: Insights, Updates, and Advances. J Neurochem 2025; 169:e70029. [PMID: 40066661 PMCID: PMC11894596 DOI: 10.1111/jnc.70029] [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: 11/16/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Synaptic homeostasis of the principal neurotransmitters glutamate and GABA is tightly regulated by an intricate metabolic coupling between neurons and astrocytes known as the glutamate/GABA-glutamine cycle. In this cycle, astrocytes take up glutamate and GABA from the synapse and convert these neurotransmitters into glutamine. Astrocytic glutamine is subsequently transferred to neurons, serving as the principal precursor for neuronal glutamate and GABA synthesis. The glutamate/GABA-glutamine cycle integrates multiple cellular processes, including neurotransmitter release, uptake, synthesis, and metabolism. All of these processes are deeply interdependent and closely coupled to cellular energy metabolism. Astrocytes display highly active mitochondrial oxidative metabolism and several unique metabolic features, including glycogen storage and pyruvate carboxylation, which are essential to sustain continuous glutamine release. However, new roles of oligodendrocytes and microglia in neurotransmitter recycling are emerging. Malfunction of the glutamate/GABA-glutamine cycle can lead to severe synaptic disruptions and may be implicated in several brain diseases. Here, I review central aspects and recent advances of the glutamate/GABA-glutamine cycle to highlight how the cycle is functionally connected to critical brain functions and metabolism. First, an overview of glutamate, GABA, and glutamine transport is provided in relation to neurotransmitter recycling. Then, central metabolic aspects of the glutamate/GABA-glutamine cycle are reviewed, with a special emphasis on the critical metabolic roles of glial cells. Finally, I discuss how aberrant neurotransmitter recycling is linked to neurodegeneration and disease, focusing on astrocyte metabolic dysfunction and brain lipid homeostasis as emerging pathological mechanisms. Instead of viewing the glutamate/GABA-glutamine cycle as individual biochemical processes, a more holistic and integrative approach is needed to advance our understanding of how neurotransmitter recycling modulates brain function in both health and disease.
Collapse
Affiliation(s)
- Jens V. Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
27
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [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: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
28
|
Kim SG, Keum M, Choe YM, Suh GH, Lee BC, Kim HS, Lee JH, Hwang J, Yi D, Kim JW. Selenium and Episodic Memory: The Moderating Role of Apolipoprotein E ε4. Nutrients 2025; 17:595. [PMID: 39940451 PMCID: PMC11819958 DOI: 10.3390/nu17030595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Selenium (Se), a vital trace element, plays a neuroprotective role by mitigating oxidative stress through selenoproteins and regulating metal balance. The apolipoprotein E ε4 allele (APOE4), a significant genetic risk factor for Alzheimer's disease (AD), has been linked to reduced Se levels and weakened antioxidant capacity. This research explores the association between serum Se concentrations and cognitive performance, with an emphasis on how APOE4 status influences this relationship. Methods: This study included 196 older adults from community and memory clinic settings, who underwent assessments for episodic memory, global cognition, and non-memory functions using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) neuropsychological battery, with serum selenium levels analyzed via inductively coupled plasma-mass spectrometry (ICP-MS) and APOE genotyping conducted to determine allele status. Results: Higher serum Se levels were associated with better episodic memory score (EMS) (B = 0.065, 95% CI = 0.020-0.110, p = 0.005) and CERAD total score (TS) (B = 0.119, 95% CI = 0.046-0.193, p = 0.002). However, the interaction between Se and APOE4 status significantly affected EMS (B = -0.074, 95% CI = -0.109 to -0.039, p < 0.001), with significant benefits observed in APOE4-negative participants. Conclusions: This study highlights the genotype-specific impact of Se on cognitive health, emphasizing the need for personalized nutritional interventions targeting Se levels, particularly for APOE4-negative individuals. Future research should further elucidate the mechanisms of Se's effects and assess its therapeutic potential in aging populations.
Collapse
Affiliation(s)
- Shin Gyeom Kim
- Department of Neuropsychiatry, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Musung Keum
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
| | - Young Min Choe
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| | - Guk-Hee Suh
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| | - Boung Chul Lee
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
- Department of Neuropsychiatry, Hallym University Hangang Sacred Heart Hospital, Seoul 07247, Republic of Korea
| | - Hyun Soo Kim
- Department of Laboratory Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea;
| | - Jun Hyung Lee
- Department of Laboratory Medicine, Green Cross Laboratories (GC Labs), Yongin 16924, Gyeonggi, Republic of Korea;
| | - Jaeuk Hwang
- Department of Psychiatry, Soonchunhyang University Hospital Seoul, Seoul 04401, Republic of Korea;
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul 03080, Republic of Korea;
| | - Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong 18450, Gyeonggi, Republic of Korea; (M.K.); (Y.M.C.); (G.-H.S.)
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon 24252, Gangwon, Republic of Korea;
| |
Collapse
|
29
|
Dang W, Hao T, Li N, Zhang H, Li Z, Yu H, Wen Y, Zheng D, Liu L. Investigating shared risk variants and genetic etiology between Alzheimer's disease and three stress-related psychiatric disorders: a large-scale genome-wide cross-trait analysis. FRONTIERS IN AGING 2025; 6:1488528. [PMID: 39975850 PMCID: PMC11837265 DOI: 10.3389/fragi.2025.1488528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/13/2025] [Indexed: 02/21/2025]
Abstract
Introduction Observational studies have reported that patients with Alzheimer's disease (AD) have a greater burden of comorbidities typically associated with stress-related psychiatric disorders. However, the contribution of hereditary factors to this comorbidity remains unclear. We evaluated phenotypic associations using observational data from the UK Biobank. Method Our study focused on investigating the shared risk variants and genetic etiology underlying AD and three stress-related psychiatric disorders: post-traumatic stress disorder, anxiety disorder, and major depressive disorder. By leveraging summary statistics from genome-wide association studies, we investigated global genetic correlations using linkage disequilibrium score regression, genetic covariance analysis, and high-definition likelihood. Genome-wide cross-trait analysis with association analysis based on subsets and cross-phenotype association were performed to discover genome-wide significant risk variants shared between AD and the three stress-related psychiatric disorders. Results A significant positive genetic correlation was observed between AD and major depressive disorder using linkage disequilibrium score regression (rg = 0.231; P = 0.018), genetic covariance analysis (rg = 0.138; P < 0.001), and high-definition likelihood (rg = 0.188; P < 0.001). Association analysis based on subsets and cross-phenotype association revealed thirteen risk variants in six genes shared between AD and post-traumatic stress disorder; seven risk variants in four genes shared between AD and anxiety disorder; and 23 risk variants in four genes shared between AD and major depressive disorder. Functional annotation and gene-set enrichment analysis indicated that 12 genes for comorbidity shared between patients with AD and all three stress-related psychiatric disorders were enriched in the spleen, pancreas, and whole blood. Conclusion These results advance our knowledge of the shared genetic origins of comorbidities and pave the way for advancements in the diagnosis, management, and prevention of stress-related AD.
Collapse
Affiliation(s)
- Weijia Dang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tianqi Hao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ning Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Hualin Zhang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ziqi Li
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongmei Yu
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yalu Wen
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Long Liu
- Department of Health Statistics, School of Public Health, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
30
|
Jiang WI, Cao Y, Xue Y, Ji Y, Winer BY, Chandra R, Zhang XF, Zhang M, Singhal NS, Pierce JT, Chen S, Ma DK. Suppressing APOE4-induced neural pathologies by targeting the VHL-HIF axis. Proc Natl Acad Sci U S A 2025; 122:e2417515122. [PMID: 39874294 PMCID: PMC11804744 DOI: 10.1073/pnas.2417515122] [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: 08/29/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
The ε4 variant of human apolipoprotein E (APOE4) is a key genetic risk factor for neurodegeneration in Alzheimer's disease and elevated all-cause mortality in humans. Understanding the factors and mechanisms that can mitigate the harmful effects of APOE4 has significant implications. In this study, we find that inactivating the VHL-1 (Von Hippel-Lindau) protein can suppress mortality, neural and behavioral pathologies caused by transgenic human APOE4 in Caenorhabditis elegans. The protective effects of VHL-1 deletion are recapitulated by stabilized HIF-1 (hypoxia-inducible factor), a transcription factor degraded by VHL-1. HIF-1 activates a genetic program that safeguards against mitochondrial dysfunction, oxidative stress, proteostasis imbalance, and endolysosomal rupture-critical cellular events linked to neural pathologies and mortality. Furthermore, genetic inhibition of Vhl reduces cerebral vascular injury and synaptic lesions in APOE4 mice, suggesting an evolutionarily conserved mechanism. Thus, we identify the VHL-HIF axis as a potent modulator of APOE4-induced neural pathologies and propose that targeting this pathway in nonproliferative tissues may curb cellular damage, protect against neurodegeneration, and reduce tissue injuries and mortality.
Collapse
Affiliation(s)
- Wei I. Jiang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Yiming Cao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yue Xue
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yichun Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Benjamin Y. Winer
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- HHMI, Chevy Chase, MD20815
| | - Rashmi Chandra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Xingyuan Fischer Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Mengqi Zhang
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Neel S. Singhal
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Jonathan T. Pierce
- Department of Neuroscience, The Center for Learning and Memory, Waggoner Center for Alcohol and Addiction Research, Institute of Neuroscience, University of Texas at Austin, Austin, TX78712
| | - Song Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Department of Physiology, University of California San Francisco, San Francisco, CA94158
- Innovative Genomics Institute, University of California, Berkeley, CA94720
| |
Collapse
|
31
|
Wroblewski TH, Ajmal E, Ononogbu-Uche F, Lerner DP, Bigdeli TB, Divers J, Barthélemy EJ. Molecular Biomarkers Associated with Traumatic Brain Injury Outcome in Individuals of Black Racial Identity or African Ancestry: A Narrative Review. World Neurosurg 2025; 194:123620. [PMID: 39732452 DOI: 10.1016/j.wneu.2024.123620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide and a major global health concern. In the United States, individuals of Black or African American racial identity experience disproportionately higher rates of TBI and suffer from worse postinjury outcomes. Contemporary research agendas have largely overlooked or excluded Black populations, resulting in the continued marginalization of Black patient populations in TBI studies, thereby limiting the generalizability of ongoing research to patients in the United States and around the world. This review aims to highlight what is currently known, and identify knowledge gaps, in research on molecular biomarkers associated with TBI in Black populations. A PubMed literature search was conducted to identify studies that investigate molecular biomarkers associated with TBI outcomes that include participants of Black racial identity and those of African ancestry. Studies identified for this review investigate biomarkers associated with TBI outcomes through a lens that specifically examines race, ethnicity, or ancestry. Most studies focused on blood- or cerebrospinal fluid-derived protein biomarkers. Studies identified statistical variation in S100ß, ubiquitin C-terminal hydrolase-L1, amyloid-ß, and tau across participant race, either at baseline or following TBI. Additionally, several studies identified genetic polymorphisms associated with TBI outcomes related to apolipoprotein E, ANKK1, and COMT polymorphism and TBI outcome and identified allele frequency variation across population ancestry. The role of race and ancestry on biomarkers associated with TBI outcome remains indeterminate and subsequent work is still required to understand the implications for patients with TBI.
Collapse
Affiliation(s)
- Tadeusz H Wroblewski
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Global Neurosurgery Laboratory, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; MD-PhD Program, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Erum Ajmal
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Global Neurosurgery Laboratory, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Favour Ononogbu-Uche
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Global Neurosurgery Laboratory, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - David P Lerner
- Department of Neurology, One Brooklyn Health/Brookdale University Hospital and Medical Center, Brooklyn, New York, USA; Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Tim B Bigdeli
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; VA New York Harbor Healthcare System, Brooklyn, New York, USA
| | - Jasmin Divers
- Division of Health Services Research, Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, NYU Langone Health, New York, New York, USA
| | - Ernest J Barthélemy
- College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Global Neurosurgery Laboratory, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Department of Neurology, One Brooklyn Health/Brookdale University Hospital and Medical Center, Brooklyn, New York, USA; Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Division of Neurosurgery, Department of Surgery, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Department of Community Health Sciences, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, New York, USA; Department of Surgery, One Brooklyn Health/Brookdale University Hospital and Medical Center, Brooklyn, New York, USA.
| |
Collapse
|
32
|
Karagas N, Young JE, Blue EE, Jayadev S. The Spectrum of Genetic Risk in Alzheimer Disease. Neurol Genet 2025; 11:e200224. [PMID: 39885961 PMCID: PMC11781270 DOI: 10.1212/nxg.0000000000200224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer disease (AD), the most common dementing syndrome in the United States, is currently established by the presence of amyloid-β and tau protein biomarkers in the setting of clinical cognitive impairment. These straightforward diagnostic parameters belie an immense complexity of genetic architecture underlying risk and presentation in AD. In this review, we provide a focused overview of the current state of AD genetics. We discuss the discovery of familial autosomal dominant genes, the identification of candidate genes associated with AD, and genetic variants conferring higher risk of developing AD compared with the general population. In particular, we discuss important features of AD risk due to the APOE ε4 allele. In addition to risk, we describe how the field has made headway understanding genetic factors that may protect from AD. The biological implications and practical limitations of information gleaned from genome-wide association studies in AD over the years are also discussed. The readers will have an up-to-date understanding of where we are in our efforts to understand the layers of genetic complexity in AD.
Collapse
Affiliation(s)
- Nicholas Karagas
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| | - Jessica E Young
- Department of Lab Medicine and Pathology, University of Washington, Seattle; and
| | - Elizabeth E Blue
- Division Medical Genetics, Department of Medicine, University of Washington, Seattle
| | - Suman Jayadev
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| |
Collapse
|
33
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
34
|
He S, Xu Z, Han X. Lipidome disruption in Alzheimer's disease brain: detection, pathological mechanisms, and therapeutic implications. Mol Neurodegener 2025; 20:11. [PMID: 39871348 PMCID: PMC11773937 DOI: 10.1186/s13024-025-00803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Alzheimer's disease (AD) is among the most devastating neurodegenerative disorders with limited treatment options. Emerging evidence points to the involvement of lipid dysregulation in the development of AD. Nevertheless, the precise lipidomic landscape and the mechanistic roles of lipids in disease pathology remain poorly understood. This review aims to highlight the significance of lipidomics and lipid-targeting approaches in the diagnosis and treatment of AD. We summarized the connection between lipid dysregulation in the human brain and AD at both genetic and lipid species levels. We briefly introduced lipidomics technologies and discussed potential challenges and areas of future advancements in the lipidomics field for AD research. To elucidate the central role of lipids in converging multiple pathological aspects of AD, we reviewed the current knowledge on the interplay between lipids and major AD features, including amyloid beta, tau, and neuroinflammation. Finally, we assessed the progresses and obstacles in lipid-based therapeutics and proposed potential strategies for leveraging lipidomics in the treatment of AD.
Collapse
Affiliation(s)
- Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA
| | - Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA.
| |
Collapse
|
35
|
Cacabelos R, Martínez-Iglesias O, Cacabelos N, Carrera J, Rodríguez D, Naidoo V. The impact of genetic variability on Alzheimer's therapies: obstacles for pharmacogenetic progress. Expert Opin Drug Metab Toxicol 2025:1-28. [PMID: 39835706 DOI: 10.1080/17425255.2024.2433626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/20/2024] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Genetic load influences the therapeutic response to conventional drugs in Alzheimer's disease (AD). Pharmacogenetics (PGx) is the best option to reduce drug-drug interactions and adverse drug reactions in patients undergoing polypharmacy regimens. However, there are important limitations that make it difficult to incorporate pharmacogenetics into routine clinical practice. AREAS COVERED This article analyzes the pharmacogenetic apparatus made up of pathogenic, mechanistic, metabolic, transporter, and pleiotropic genes responsible for the efficacy and safety of pharmacological treatment, the impact of genetic load on the outcome of multifactorial treatments, and practical aspects for the effective use of PGx. EXPERT OPINION Over 120 genes are closely associated with AD. There is an accumulation of cerebrovascular (CVn) and neurodegenerative (ADn) genes in AD. APOE-4 carriers accumulate more deleterious genetic load related to other CVn and ADn genes, develop the disease earlier, and are at a biological disadvantage compared to APOE-4 non-carriers. CYP2D6-PMs and APOE-4 carriers are the worst responders to anti-dementia drugs. Some limitations hinder the implementation of PGx in clinical practice, including lack of pharmacogenetic information for many drugs, low number of genes in PGx screening protocols, and educational deficiencies in the medical community regarding PGx and genomic medicine.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| | - Olaia Martínez-Iglesias
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| | - Natalia Cacabelos
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| | - Jairo Carrera
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| | - Daniel Rodríguez
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| | - Vinogran Naidoo
- EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine, Bergondo, Corunna, Spain
| |
Collapse
|
36
|
Ramsden CE, Cutler RG, Li X, Keyes GS. Lipid-protecting disulfide bridges are the missing molecular link between ApoE4 and sporadic Alzheimer's disease in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633633. [PMID: 39868210 PMCID: PMC11761642 DOI: 10.1101/2025.01.17.633633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
As the principal lipid transporter in the human brain, apolipoprotein E (ApoE) is tasked with the transport and protection of highly vulnerable lipids required to support and remodel neuronal membranes, in a process that is dependent on ApoE receptors. Human APOE allele variants that encode proteins differing only in the number of cysteine (Cys)-to-arginine (Arg) exchanges (ApoE2 [2 Cys], ApoE3 [1 Cys], ApoE4 [0 Cys]) comprise the strongest genetic risk factor for sporadic Alzheimer's disease (AD); however, the specific molecular feature(s) and resultant mechanisms that underlie these isoform-dependent effects are unknown. One signature feature of Cys is the capacity to form disulfide (Cys-Cys) bridges, which are required to form disulfide bridge-linked dimers and multimers. Here we propose the overarching hypothesis that the super-ability (for ApoE2), intermediate ability (for ApoE3) or inability (for ApoE4) to form lipid-protecting intermolecular disulfide bridges, is the central molecular determinant accounting for the disparate effects of APOE alleles on AD risk and amyloid-β and Tau pathologies in humans. We posit that presence and abundance of Cys in human ApoE3 and ApoE2 respectively, conceal and protect vulnerable lipids transported by ApoE from peroxidation by enabling formation of ApoE homo-dimers/multimers and heteromeric ApoE complexes such as ApoE-ApoJ and ApoE-ApoD. We thus propose that the inability to form intermolecular disulfide bridges makes ApoE4-containing lipoproteins uniquely vulnerable to peroxidation and its downstream consequences. Consistent with our model, we found that brain-enriched polyunsaturated fatty acid-containing phospholipids induce disulfide-dependent dimerization and multimerization of ApoE3 and ApoE2 (but not ApoE4). By contrast, incubation with the peroxidation-resistant lipid DMPC or cholesterol alone had minimal effects on dimerization. These novel concepts and findings are integrated into our unifying model implicating peroxidation of ApoE-containing lipoproteins, with consequent ApoE receptor-ligand disruption, as the initiating molecular events that ultimately lead to AD in humans.
Collapse
Affiliation(s)
- Christopher E. Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Roy G. Cutler
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gregory S. Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| |
Collapse
|
37
|
Tortajada-Pérez J, Carranza ADV, Trujillo-del Río C, Collado-Pérez M, Millán JM, García-García G, Vázquez-Manrique RP. Lipid Oxidation at the Crossroads: Oxidative Stress and Neurodegeneration Explored in Caenorhabditis elegans. Antioxidants (Basel) 2025; 14:78. [PMID: 39857412 PMCID: PMC11762898 DOI: 10.3390/antiox14010078] [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: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Lipid metabolism plays a critical role in maintaining cellular integrity, especially within the nervous system, where lipids support neuronal structure, function, and synaptic plasticity. However, this essential metabolic pathway is highly susceptible to oxidative stress, which can lead to lipid peroxidation, a damaging process induced by reactive oxygen species. Lipid peroxidation generates by-products that disrupt many cellular functions, with a strong impact on proteostasis. In this review, we explore the role of lipid oxidation in protein folding and its associated pathological implications, with a particular focus on findings in neurodegeneration from Caenorhabditis elegans studies, an animal model that remains underutilized. Additionally, we highlight the effectiveness of different methodologies applied in this nematode to deepen our understanding of this intricate process. In the nervous system of any animal, including mammals and invertebrates, lipid oxidation can disturb the delicate balance of cellular homeostasis, leading to oxidative stress, the build-up of toxic by-products, and protein misfolding, key factors in neurodegenerative diseases. This disruption contributes to the pathogenesis of neurodegenerative disorders such as Alzheimer's, Parkinson's, or Huntington's disease. The findings from Caenorhabditis elegans studies offer valuable insights into these complex processes and highlight potential avenues for developing targeted therapies to mitigate neurodegenerative disease progression.
Collapse
Affiliation(s)
- Julia Tortajada-Pérez
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
| | - Andrea del Valle Carranza
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
| | - Cristina Trujillo-del Río
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
| | - Mar Collado-Pérez
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
| | - José María Millán
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Gema García-García
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Pascual Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain; (J.T.-P.); (C.T.-d.R.); (M.C.-P.); (J.M.M.); (G.G.-G.)
- Joint Unit for Rare Diseases IIS La Fe—CIPF, 46026 Valencia, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
38
|
Dunn J, Moore C, Kim NS, Gao T, Cheng Z, Jin P, Ming GL, Qian J, Su Y, Song H, Zhu H. Transcription Factor-Wide Association Studies to Identify Functional SNPs in Alzheimer's Disease. J Neurosci 2025; 45:e1800242024. [PMID: 39622643 PMCID: PMC11714347 DOI: 10.1523/jneurosci.1800-24.2024] [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: 09/20/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with profound global impact. While genome-wide association studies (GWAS) have revealed genomic variants linked to AD, their translational impact has been limited due to challenges in interpreting the identified genetic associations. To address this challenge, we have devised a novel approach termed transcription factor-wide association studies (TF-WAS). By integrating the GWAS, expression quantitative trait loci, and transcriptome analyses, we selected 30 AD single nucleotide polymorphisms (SNPs) in noncoding regions that are likely to be functional. Using human transcription factor (TF) microarrays, we have identified 90 allele-specific TF interactions with 53 unique TFs. We then focused on several interactions involving SMAD4 and further validated them using electrophoretic mobility shift assay, luciferase, and chromatin immunoprecipitation on engineered genetic backgrounds (female cells). This approach holds promise for unraveling the intricacies of not just AD, but any complex disease with available GWAS data, providing insight into underlying molecular mechanisms and clues toward potential therapeutic targets.
Collapse
Affiliation(s)
- Jessica Dunn
- Department of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205
| | - Cedric Moore
- Department of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205
| | - Nam-Shik Kim
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tianshun Gao
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland 21205
| | - Zhiqiang Cheng
- Department of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland 21205
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Heng Zhu
- Department of Pharmacology, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
39
|
Verma S, Paliwal S, Paramanick D, Narayan CV, Saini M. Connecting the Dots: Gender, Sexuality, and Societal Influences on Cognitive Aging and Alzheimer's Disease. Curr Aging Sci 2025; 18:14-28. [PMID: 38899350 DOI: 10.2174/0118746098299754240530111755] [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: 12/21/2023] [Revised: 02/19/2024] [Accepted: 03/21/2024] [Indexed: 06/21/2024]
Abstract
Alzheimer's disease (AD) has many etiologies and the impact of gender on AD changes throughout time. As a consequence of advancements in precision medical procedures and methodology, Alzheimer's disease is now better understood and treated. Several risk factors may be addressed to lower one's chances of developing Alzheimer's disease or associated dementia (ADRD). The presence of amyloid-α protein senile plaques, intracellular tau protein neurofibrillary tangles (NfTs), neurodegeneration, and neuropsychiatric symptoms (NPS) characterizes Alzheimer's disease. NPS is common in persons with Alzheimer's disease dementia, although its presentation varies widely. Gender differences might explain this clinical variability. The fundamental goal of this review is to 1) emphasize the function of old age, sex, and gender in the development of Alzheimer's disease, dementia, and ADRD, and 2) explain the importance of sexual hormones, education, and APOE (Apolipoprotein E) status. This is a narrative summary of new ideas and concepts on the differences in the chance of developing dementia or Alzheimer's disease between men and women. A more thorough examination of risk and protective variables in both men and women might hasten research into the epidemiology of neurological illnesses such as dementia and Alzheimer's disease. Similarly, future preventive efforts should target men and women separately.
Collapse
Affiliation(s)
- Swati Verma
- Department of Pharmacy, ITS College of Pharmacy, Muradnagar, Ghaziabad, India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Debashish Paramanick
- Department of Pharmacy, School of Medical and Allied Science, KR Mangalam University, Gurugram, Haryana, India
| | | | - Manasvi Saini
- Department of Pharmacy, ITS College of Pharmacy, Muradnagar, Ghaziabad, India
| |
Collapse
|
40
|
Safiri S, Ghaffari Jolfayi A, Fazlollahi A, Morsali S, Sarkesh A, Daei Sorkhabi A, Golabi B, Aletaha R, Motlagh Asghari K, Hamidi S, Mousavi SE, Jamalkhani S, Karamzad N, Shamekh A, Mohammadinasab R, Sullman MJM, Şahin F, Kolahi AA. Alzheimer's disease: a comprehensive review of epidemiology, risk factors, symptoms diagnosis, management, caregiving, advanced treatments and associated challenges. Front Med (Lausanne) 2024; 11:1474043. [PMID: 39736972 PMCID: PMC11682909 DOI: 10.3389/fmed.2024.1474043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Background Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired reasoning. It is the leading cause of dementia in older adults, marked by the pathological accumulation of amyloid-beta plaques and neurofibrillary tangles. These pathological changes lead to widespread neuronal damage, significantly impacting daily functioning and quality of life. Objective This comprehensive review aims to explore various aspects of Alzheimer's disease, including its epidemiology, risk factors, clinical presentation, diagnostic advancements, management strategies, caregiving challenges, and emerging therapeutic interventions. Methods A systematic literature review was conducted across multiple electronic databases, including PubMed, MEDLINE, Cochrane Library, and Scopus, from their inception to May 2024. The search strategy incorporated a combination of keywords and Medical Subject Headings (MeSH) terms such as "Alzheimer's disease," "epidemiology," "risk factors," "symptoms," "diagnosis," "management," "caregiving," "treatment," and "novel therapies." Boolean operators (AND, OR) were used to refine the search, ensuring a comprehensive analysis of the existing literature on Alzheimer's disease. Results AD is significantly influenced by genetic predispositions, such as the apolipoprotein E (APOE) ε4 allele, along with modifiable environmental factors like diet, physical activity, and cognitive engagement. Diagnostic approaches have evolved with advances in neuroimaging techniques (MRI, PET), and biomarker analysis, allowing for earlier detection and intervention. The National Institute on Aging and the Alzheimer's Association have updated diagnostic criteria to include biomarker data, enhancing early diagnosis. Conclusion The management of AD includes pharmacological treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, which provide symptomatic relief but do not slow disease progression. Emerging therapies, including amyloid-beta and tau-targeting treatments, gene therapy, and immunotherapy, offer potential for disease modification. The critical role of caregivers is underscored, as they face considerable emotional, physical, and financial burdens. Support programs, communication strategies, and educational interventions are essential for improving caregiving outcomes. While significant advancements have been made in understanding and managing AD, ongoing research is necessary to identify new therapeutic targets and enhance diagnostic and treatment strategies. A holistic approach, integrating clinical, genetic, and environmental factors, is essential for addressing the multifaceted challenges of Alzheimer's disease and improving outcomes for both patients and caregivers.
Collapse
Affiliation(s)
- Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaffari Jolfayi
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Morsali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Aila Sarkesh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Daei Sorkhabi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Golabi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Aletaha
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kimia Motlagh Asghari
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Hamidi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Tabriz USERN Office, Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Jamalkhani
- Cardiovascular Research Center, Rajaie Cardiovascular, Medical, and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional, Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J. M. Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Chen G, Xi E, Gu X, Wang H, Tang Q. The study on cuproptosis in Alzheimer's disease based on the cuproptosis key gene FDX1. Front Aging Neurosci 2024; 16:1480332. [PMID: 39759399 PMCID: PMC11696982 DOI: 10.3389/fnagi.2024.1480332] [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: 08/13/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory and cognitive impairments. Previous studies have shown neuronal death in the brains of AD patients, but the role of cuproptosis and its associated genes in AD neurons remains unclear. Methods Intersection analysis was conducted using the AD transcriptome dataset GSE63060, neuron dataset GSE147528, and reported cuproptosis-related genes to identify the cuproptosis key gene FDX1 highly expressed in AD. Subsequently, cell experiments were performed by treating SH-SY5Y cells with Aβ25-35 to establish AD cell model. The real-time reverse transcriptase-polymerase chain reaction (RT-qPCR) and western blotting (WB) assays were employed to detect the expression levels of FDX1, DLAT, and DLST. Cell proliferation was analyzed by counting Kit-8 (CCK8), mitochondrial ROS levels were analyzed using flow cytometry. shRNA was used to downregulate FDX1 expression, followed by repetition of the aforementioned experiments. Clinical experiments utilized qPCR to detect FDX1 mRNA levels in peripheral venous blood of patients, and analyzed FDX1 expression differences in different APOE genotypes of AD patients. Finally, a protein-protein interaction (PPI) network of FDX1 was constructed based on the GeneMANIA database, immune infiltration analysis was conducted using R language, and transcription factors prediction for FDX1 was performed based on the ENCODE database. Results The cuproptosis key gene FDX1 showed significantly higher expression in peripheral blood and neuron models of AD compared to non-AD individuals, with significantly higher expression in APOE ε4/ε4 genotype than other APOE genotype of AD patients. Knockdown of FDX1 expression reduced the lipidation levels of DLAT and DLST in neurons, alleviated ROS accumulation in mitochondria, improved cell viability, and mitigated cuproptosis. Immune infiltration analysis results indicated a high enrichment of peripheral blood γδ-T lymphocytes in AD, and FDX1 was significantly associated with the infiltration of four immune cells and may be regulated by three transcription factors. Conclusion The cuproptosis key gene FDX1 is highly expressed in AD and may promote cuproptosis in AD neurons by regulating the lipidation levels of DLAT and DLST, thereby participating in the onset and development of AD. This provides a potential target for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Guilin Chen
- Department of Neurology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Erwei Xi
- Department of Neurology, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiaozhen Gu
- Institute of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Huili Wang
- Institute of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
42
|
Khan MA. Targeting Iron Responsive Elements (IREs) of APP mRNA into Novel Therapeutics to Control the Translation of Amyloid-β Precursor Protein in Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:1669. [PMID: 39770511 PMCID: PMC11677800 DOI: 10.3390/ph17121669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The hallmark of Alzheimer's disease (AD) is the buildup of amyloid-β (Aβ), which is produced when the amyloid precursor protein (APP) misfolds and deposits as neurotoxic plaques in the brain. A functional iron responsive element (IRE) RNA stem loop is encoded by the APP 5'-UTR and may be a target for regulating the production of Alzheimer's amyloid precursor protein. Since modifying Aβ protein expression can give anti-amyloid efficacy and protective brain iron balance, targeted regulation of amyloid protein synthesis through modulation of 5'-UTR sequence function is a novel method for the prospective therapy of Alzheimer's disease. Numerous mRNA interference strategies target the 2D RNA structure, even though messenger RNAs like tRNAs and rRNAs can fold into complex, three-dimensional structures, adding even another level of complexity. The IRE family is among the few known 3D mRNA regulatory elements. This review seeks to describe the structural and functional aspects of IREs in transcripts, including that of the amyloid precursor protein, that are relevant to neurodegenerative diseases, including AD. The mRNAs encoding the proteins involved in iron metabolism are controlled by this family of similar base sequences. Like ferritin IRE RNA in their 5'-UTR, iron controls the production of APP in their 5'-UTR. Iron misregulation by iron regulatory proteins (IRPs) can also be investigated and contrasted using measurements of the expression levels of tau production, Aβ, and APP. The development of AD is aided by iron binding to Aβ, which promotes Aβ aggregation. The development of small chemical therapeutics to control IRE-modulated expression of APP is increasingly thought to target messenger RNAs. Thus, IRE-modulated APP expression in AD has important therapeutic implications by targeting mRNA structures.
Collapse
Affiliation(s)
- Mateen A Khan
- Department of Life Science, College of Science and General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
43
|
Weinstock M. Therapeutic agents for Alzheimer's disease: a critical appraisal. Front Aging Neurosci 2024; 16:1484615. [PMID: 39717349 PMCID: PMC11663918 DOI: 10.3389/fnagi.2024.1484615] [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: 09/12/2024] [Accepted: 10/31/2024] [Indexed: 12/25/2024] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Mutations in genes and precursors of β amyloid (Aβ) are found in the familial form of the disease. This led to the evaluation of seven monoclonal antibodies against Aβ in subjects with AD, two of which were approved for use by the FDA. They caused only a small improvement in cognitive function, probably because they were given to those with much more prevalent sporadic forms of dementia. They also have potentially serious adverse effects. Oxidative stress and elevated pro-inflammatory cytokines are present in all subjects with AD and are well correlated with the degree of memory impairment. Drugs that affect these processes include TNFα blocking antibodies and MAPK p38 inhibitors that reduce cognitive impairment when given for other inflammatory conditions. However, their adverse effects and inability to penetrate the brain preclude their use for dementia. Rosiglitazone is used to treat diabetes, a risk factor for AD, but failed in a clinical trial because it was given to subjects that already had dementia. Ladostigil reduces oxidative stress and suppresses the release of pro-inflammatory cytokines from activated microglia without blocking their effects. Chronic oral administration to aging rats prevented the decline in memory and suppressed overexpression of genes adversely affecting synaptic function in relevant brain regions. In a phase 2 trial, ladostigil reduced the decline in short-term memory and in whole brain and hippocampal volumes in human subjects with mild cognitive impairment and had no more adverse effects than placebo.
Collapse
Affiliation(s)
- Marta Weinstock
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
44
|
Günaydin C, Sondhi D, Kaminsky SM, Lephart HC, Leopold PL, Hackett NR, Khanna R, Crystal RG. AAVrh.10 delivery of novel APOE2-Christchurch variant suppresses amyloid and Tau pathology in Alzheimer's disease mice. Mol Ther 2024; 32:4303-4318. [PMID: 39511891 PMCID: PMC11638875 DOI: 10.1016/j.ymthe.2024.11.003] [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: 06/26/2024] [Revised: 10/11/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024] Open
Abstract
Gene therapy to treat hereditary disorders conventionally delivers the normal allele to compensate for loss-of-function mutations. More effective gene therapy may be achieved using a gain-of-function variant. We tested the hypothesis that AAVrh.10-mediated CNS delivery of the human APOE2 allele with the Christchurch mutation (R136S) (E2Ch) will provide superior protection against APOE4-associated Alzheimer's disease (AD) in mice compared to the unmodified APOE2 allele (E2). The vectors were assessed in two mouse strains with humanized APOE4: APP.PSEN1/TRE4 "amyloid mice" and P301S/TRE4, "tau mice." Both the E2Ch and E2 vectors prevented Aβ42 and Aβ40 accumulation and decreased β-amyloid aggregates in amyloid mice, but only the E2Ch vector suppressed tau tangles in tau mice. Microglial activation and reactive astrocytes were significantly suppressed by both vectors in amyloid mice but only the E2Ch vector mediated significant suppression of Iba1 and glial fibrillary acidic protein (GFAP) in tau mice. In four behavioral assays, the E2 and E2Ch vectors had similar benefits in amyloid mice, but E2Ch outperformed E2 in tau mice. In summary, while E2 is effective in suppressing amyloid pathology, the novel E2 variant E2Ch more effectively treats both the amyloid and tau pathology of murine AD in APOE4 background, supporting the development of AAVrh.10APOE2Ch as a therapy for APOE4-associated AD.
Collapse
Affiliation(s)
- Caner Günaydin
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Hailey C Lephart
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Philip L Leopold
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil R Hackett
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
45
|
Carling GK, Fan L, Foxe NR, Norman K, Wong MY, Zhu D, Corona C, Razzoli A, Yu F, Yarahmady A, Ye P, Chen H, Huang Y, Amin S, Sereda R, Lopez-Lee C, Zacharioudakis E, Chen X, Xu J, Cheng F, Gavathiotis E, Cuervo AM, Holtzman DM, Mok SA, Sinha SC, Sidoli S, Ratan RR, Luo W, Gong S, Gan L. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. Neuron 2024; 112:3877-3896.e8. [PMID: 39353433 PMCID: PMC11624100 DOI: 10.1016/j.neuron.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
The strongest risk factors for late-onset sporadic Alzheimer's disease (AD) include the ε4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H (R47H) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting detrimental disease mechanisms. We find that R47H induces neurodegeneration in 9- to 10-month-old female APOE4 tauopathy mice. The combination of APOE4 and R47H (APOE4-R47H) worsened hyperphosphorylated tau pathology in the frontal cortex and amplified tauopathy-induced microglial cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling and downstream interferon response. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.
Collapse
Affiliation(s)
- Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nessa R Foxe
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kendra Norman
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daphne Zhu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carlo Corona
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Agnese Razzoli
- Transfusion Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia 42122, Italy; Clinical and Experimental PhD Program, University of Modena and Reggio Emilia, Modena 41121, Italy
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hao Chen
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Structural Biology, Cell Biology, Developmental Biology, and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rebecca Sereda
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emmanouil Zacharioudakis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Simone Sidoli
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Rajiv R Ratan
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
46
|
Preman P, Moechars D, Fertan E, Wolfs L, Serneels L, Shah D, Lamote J, Poovathingal S, Snellinx A, Mancuso R, Balusu S, Klenerman D, Arranz AM, Fiers M, De Strooper B. APOE from astrocytes restores Alzheimer's Aβ-pathology and DAM-like responses in APOE deficient microglia. EMBO Mol Med 2024; 16:3113-3141. [PMID: 39528861 PMCID: PMC11628604 DOI: 10.1038/s44321-024-00162-7] [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: 04/24/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The major genetic risk factor for Alzheimer's disease (AD), APOE4, accelerates beta-amyloid (Aβ) plaque formation, but whether this is caused by APOE expressed in microglia or astrocytes is debated. We express here the human APOE isoforms in astrocytes in an Apoe-deficient AD mouse model. This is not only sufficient to restore the amyloid plaque pathology but also induces the characteristic transcriptional pathological responses in Apoe-deficient microglia surrounding the plaques. We find that both APOE4 and the protective APOE2 from astrocytes increase fibrillar plaque deposition, but differentially affect soluble Aβ aggregates. Microglia and astrocytes show specific alterations in function of APOE genotype expressed in astrocytes. Our experiments indicate a central role of the astrocytes in APOE mediated amyloid plaque pathology and in the induction of associated microglia responses.
Collapse
Affiliation(s)
- Pranav Preman
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Daan Moechars
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Leen Wolfs
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Lutgarde Serneels
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Disha Shah
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Jochen Lamote
- VIB FACS Expertise Center, Center for Cancer Biology, Leuven, Belgium
| | | | - An Snellinx
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB-UAntwerp, Centre for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sriram Balusu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Amaia M Arranz
- Laboratory of Humanized Models of Disease, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
- UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
47
|
He K, Zhao Z, Zhang J, Li D, Wang S, Liu Q. Cholesterol Metabolism in Neurodegenerative Diseases. Antioxid Redox Signal 2024; 41:1051-1072. [PMID: 38842175 DOI: 10.1089/ars.2024.0674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Significance: Cholesterol plays a crucial role in the brain, where it is highly concentrated and tightly regulated to support normal brain functions. It serves as a vital component of cell membranes, ensuring their integrity, and acts as a key regulator of various brain processes. Dysregulation of cholesterol metabolism in the brain has been linked to impaired brain function and the onset of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and Huntington's disease. Recent Advances: A significant advancement has been the identification of astrocyte-derived apoliprotein E as a key regulator of de novo cholesterol biosynthesis in neurons, providing insights into how extracellular signals influence neuronal cholesterol levels. In addition, the development of antibody-based therapies, particularly for AD, presents promising opportunities for therapeutic interventions. Critical Issues: Despite significant research, the association between cholesterol and neurodegenerative diseases remains inconclusive. It is crucial to distinguish between plasma cholesterol and brain cholesterol, as these pools are relatively independent. This differentiation should be considered when evaluating statin-based treatment approaches. Furthermore, assessing not only the total cholesterol content in the brain but also its distribution among different types of brain cells is essential. Future Direction: Establishing a causal link between changes in brain/plasma cholesterol levels and the onset of brain dysfunction/neurodegenerative diseases remains a key objective. In addition, conducting cell-specific analyses of cholesterol homeostasis in various types of brain cells under pathological conditions will enhance our understanding of cholesterol metabolism in neurodegenerative diseases. Manipulating cholesterol levels to restore homeostasis may represent a novel approach for alleviating neurological symptoms. Antioxid. Redox Signal. 41, 1051-1072.
Collapse
Affiliation(s)
- Keqiang He
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Sheng Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qiang Liu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| |
Collapse
|
48
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [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: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
49
|
Yakubu A, Adedeji I, Maduka OC, Jegede A, Adebamowo C. Ethical issues in genomics research in persons with Alzheimer's Disease/Alzheimer's Disease-related dementia (AD/ADRD): a systematic review. BMC Med Ethics 2024; 25:138. [PMID: 39587628 PMCID: PMC11587778 DOI: 10.1186/s12910-024-01141-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
INTRODUCTION Given the growing number of Alzheimer's Disease and Alzheimer's Disease Related Dementias (AD/ADRD) genomics research projects and the vulnerabilities of study participants, it is critical to evaluate the literature on the ethical challenges in such studies to ensure high ethical standards. METHODS We conducted a systematic review of the literature on ethical issues in AD/ADRD genomics research. We searched Embase, PsycINFO, CiNAHL, Scopus, and Ovid Medline for empirical and normative papers published in peer-reviewed journals on the ethical issues involved in conducting genomics research among persons with AD/ADRD. We used ethical principles from an existing framework as a priori codes to categorize the ethical issues and adapted another framework of Dementia Research Ethical Issues (DREI) as subcategories for our synthesis. We used the 2021 PRISMA guidelines to guide our study. RESULTS We screened 5,509 papers and included 27 of these papers in the systematic review after deduplication, title, and full-text review. The papers contained 109 ethical issues that were mapped against 42 out of 75 relevant DREIs. The highest number of DREIs were mapped to "respect for persons and communities", "favorable risk-benefit ratio", "informed consent" and "scientific validity". The least mapped principles to the DREIs were "fair participant selection", "independent review", "social value", and "collaborative partnership". CONCLUSION Our review showed that there is a dearth of literature on the ethical principles of "fair participant selection", "independent review", "social value" and "collaborative partnership" in genomics research on AD/ADRDs. It is difficult to draw firm conclusions from the distribution of attention paid to specific principles because these may only reflect the concerns of AD/ADRD genomics research ethicists in high-income countries. There is need for more research on the ethics of AD/ADRD genomics research in low and middle-income countries for a more balanced account of the important ethical considerations in this field.
Collapse
Affiliation(s)
- Aminu Yakubu
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Bioethics, Faculty of Multi-Disciplinary Studies, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Isaac Adedeji
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Sociology, Olabisi Onabanjo University, Ago-Iwoye, Ogun State, Nigeria
| | - Oluchi C Maduka
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Bioethics, Faculty of Multi-Disciplinary Studies, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Ayodele Jegede
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria
- Department of Sociology, Faculty of the Social Sciences, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Clement Adebamowo
- Center for Bioethics and Research, 102 Basorun Rd, Akobo, Ibadan, Oyo State, 200285, Nigeria.
- Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Martorell-Marugán J, López-Domínguez R, Villatoro-García JA, Toro-Domínguez D, Chierici M, Jurman G, Carmona-Sáez P. Explainable deep neural networks for predicting sample phenotypes from single-cell transcriptomics. Brief Bioinform 2024; 26:bbae673. [PMID: 39814561 PMCID: PMC11735047 DOI: 10.1093/bib/bbae673] [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: 07/04/2024] [Revised: 11/14/2024] [Indexed: 01/18/2025] Open
Abstract
Recent advances in single-cell RNA-Sequencing (scRNA-Seq) technologies have revolutionized our ability to gather molecular insights into different phenotypes at the level of individual cells. The analysis of the resulting data poses significant challenges, and proper statistical methods are required to analyze and extract information from scRNA-Seq datasets. Sample classification based on gene expression data has proven effective and valuable for precision medicine applications. However, standard classification schemas are often not suitable for scRNA-Seq due to their unique characteristics, and new algorithms are required to effectively analyze and classify samples at the single-cell level. Furthermore, existing methods for this purpose have limitations in their usability. Those reasons motivated us to develop singleDeep, an end-to-end pipeline that streamlines the analysis of scRNA-Seq data training deep neural networks, enabling robust prediction and characterization of sample phenotypes. We used singleDeep to make predictions on scRNA-Seq datasets from different conditions, including systemic lupus erythematosus, Alzheimer's disease and coronavirus disease 2019. Our results demonstrate strong diagnostic performance, validated both internally and externally. Moreover, singleDeep outperformed traditional machine learning methods and alternative single-cell approaches. In addition to prediction accuracy, singleDeep provides valuable insights into cell types and gene importance estimation for phenotypic characterization. This functionality provided additional and valuable information in our use cases. For instance, we corroborated that some interferon signature genes are consistently relevant for autoimmunity across all immune cell types in lupus. On the other hand, we discovered that genes linked to dementia have relevant roles in specific brain cell populations, such as APOE in astrocytes.
Collapse
Affiliation(s)
- Jordi Martorell-Marugán
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain
- Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero (FIBAO), Avenida de Madrid 15, Granada 18012, Spain
| | - Raúl López-Domínguez
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain
| | - Juan Antonio Villatoro-García
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain
- Department of Statistics and Operational Research, University of Granada, Avenida de la Fuente Nueva S/N, Granada 18071, Spain
| | - Daniel Toro-Domínguez
- Unit of Inflammatory Diseases, Department of Environmental Medicine, Karolinska Institutet, Nobels väg 13, Solna 171 77, Sweden
| | - Marco Chierici
- Data Science for Health Research Unit, Fondazione Bruno Kessler, Via Sommarive 18, Trento 38123, Italy
| | - Giuseppe Jurman
- Data Science for Health Research Unit, Fondazione Bruno Kessler, Via Sommarive 18, Trento 38123, Italy
| | - Pedro Carmona-Sáez
- GENYO, Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain
- Department of Statistics and Operational Research, University of Granada, Avenida de la Fuente Nueva S/N, Granada 18071, Spain
| |
Collapse
|