1
|
Gomez AR, Byun HR, Wu S, Muhammad AKMG, Ikbariyeh J, Chen J, Muro A, Li L, Bernstein KE, Ainsworth R, Tourtellotte WG. Boosting angiotensin-converting enzyme (ACE) in microglia protects against Alzheimer's disease in 5xFAD mice. NATURE AGING 2025:10.1038/s43587-025-00879-1. [PMID: 40490625 DOI: 10.1038/s43587-025-00879-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 04/17/2025] [Indexed: 06/11/2025]
Abstract
Genome-wide association studies have identified many gene polymorphisms associated with an increased risk of developing late-onset Alzheimer's disease (LOAD). Many of these LOAD risk-associated alleles alter disease pathogenesis by influencing innate immune responses and lipid metabolism of microglia (MG). Here we show that boosting the expression of angiotensin-converting enzyme (ACE), a genome-wide association study LOAD risk-associated gene product, specifically in MG, reduces amyloid-β (Aβ) plaque load, preserves vulnerable neurons and excitatory synapses, and significantly reduces learning and memory abnormalities in the 5xFAD amyloid mouse model of AD. ACE-expressing MG surround plaques more frequently and they have increased Aβ phagocytosis, endolysosomal trafficking and spleen tyrosine kinase activation downstream of the major Aβ receptors, triggering receptor expressed on myeloid cells 2 (Trem2) and C-type lectin domain family 7 member A (Clec7a). These findings establish a role for ACE in enhancing microglial immune function and they identify a potential use for ACE-expressing MG as a cell-based therapy to augment endogenous microglial responses to Aβ in AD.
Collapse
Affiliation(s)
- Andrew R Gomez
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hyae Ran Byun
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shaogen Wu
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - A K M Ghulam Muhammad
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jasmine Ikbariyeh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jaelin Chen
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alek Muro
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lin Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard Ainsworth
- Kao Autoimmunity Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Warren G Tourtellotte
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
2
|
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
|
3
|
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 PMCID: PMC12109626 DOI: 10.3390/brainsci15050486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
| | | | | |
Collapse
|
4
|
Liu S, Bush WS, Akinyemi RO, Byrd GS, Caban-Holt AM, Rajabli F, Reitz C, Kunkle BW, Tosto G, Vance JM, Pericak-Vance M, Haines JL, Williams SM, Crawford DC. Alzheimer disease is (sometimes) highly heritable: Drivers of variation in heritability estimates for binary traits, a systematic review. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.29.25326648. [PMID: 40343016 PMCID: PMC12060970 DOI: 10.1101/2025.04.29.25326648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Estimating heritability has been fundamental in understanding the genetic contributions to complex disorders like late-onset Alzheimer's disease (LOAD), and provides a rationale for identifying genetic factors associated with disease susceptibility. While numerous studies have established substantial genetic contribution for LOAD, the interpretation of heritability estimates remains challenging. These challenges are further complicated by the binary nature of LOAD status, where estimation and interpretation require additional considerations. Through a systematic review, we identified LOAD heritability estimates from 6 twin studies and 17 genome-wide association studies, all conducted in populations of European ancestry. We demonstrate that these heritability estimates for LOAD vary considerably. The variation reflects not only differences in study design and methodological approaches but also the underlying study population characteristics. Our findings indicate that commonly cited heritability estimates, often treated as universal values, should be interpreted within specific population contexts and methodological frameworks.
Collapse
Affiliation(s)
- Shiying Liu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rufus Olusola Akinyemi
- Neuroscience and Ageing Research Unit, Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Goldie S Byrd
- Maya Angelou Center for Health Equity, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Allison Mercedes Caban-Holt
- Department of Behavioral Science, College of Medicine and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Farid Rajabli
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christiane Reitz
- Gertrude H. Sergievsky Center, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology Columbia University, Department of Epidemiology, Columbia University, New York, NY, USA
| | - Brian W Kunkle
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University; Department of Neurology, Columbia University Irving Medical Center, Columbia University; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY USA
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Margaret Pericak-Vance
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Dana C Crawford
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
5
|
Gaudio A, Gotta F, Ponti C, Geroldi A, La Barbera A, Mandich P. GWAS by Subtraction to Disentangle RBD Genetic Background from α-Synucleinopathies. Int J Mol Sci 2025; 26:3578. [PMID: 40332088 PMCID: PMC12026788 DOI: 10.3390/ijms26083578] [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: 03/17/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Rapid eye movement (REM) sleep behavior disorder (RBD) is a parasomnia characterized by loss of muscle atonia and abnormal behaviors occurring during REM sleep. Idiopathic RBD (iRBD) is recognized as the strongest prodromal hallmark of α-synucleinopathies, with an established conversion rate to a neurodegenerative condition that reaches up to 96.6% at 15 years of follow-up. Moreover, RBD-converters display a more severe clinical trajectory compared to those that do not present with RBD. However, the extent to which iRBD represents a distinct genetic entity or an early manifestation of neurodegeneration remains unclear. To address this, we applied Genomic Structural Equation Modeling (GenomicSEM) using a GWAS-by-subtraction approach to disentangle the genetic architecture of iRBD from the shared genomic liability across α-synucleinopathies. Our findings highlight the SNCA locus as a key genetic regulator of iRBD susceptibility. While iRBD exhibits a partially distinct genetic signature, residual genomic overlap with neurodegenerative traits suggests that its genetic architecture exists along a continuum of α-synucleinopathy risk. In this scenario, the associations with neuroanatomical correlates may serve as early indicators of a trajectory toward future neurodegeneration. These findings provide a framework for identifying biomarkers that could aid in disease stratification and risk prediction, potentially improving early intervention strategies.
Collapse
Affiliation(s)
- Andrea Gaudio
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Fabio Gotta
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Clarissa Ponti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Alessandro Geroldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| | - Andrea La Barbera
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
| | - Paola Mandich
- IRCCS Ospedale Policlinico San Martino–UOC Genetica Medica, Largo R. Benzi 10, 16132 Genova, Italy; (F.G.); (A.L.B.); (P.M.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genova, Largo P. Daneo 3, 16132 Genova, Italy; (C.P.); (A.G.)
| |
Collapse
|
6
|
Barral S, Yang Z, Phillips N, Barber RC, Brickman AM, Honig LS, Cieza B, Reyes‐Dumeyer D, Mayeux R, Rajabli F, Cuccaro ML, Vance JM, Arango SM, Samper‐Ternent R, Obregon AM, Montesinos R, Soto‐Añari M, Duran JC, Cusicanqui M, Velazquez IZJ, Marca V, Illanes‐Manrique M, Cornejo‐Olivas M, Pericak‐Vance M, Wong R, O'Bryant S, Custodio N, Tosto G. APOE and Alzheimer's disease and related dementias risk among 12,221 Hispanics/Latinos. Alzheimers Dement 2025; 21:e70138. [PMID: 40219824 PMCID: PMC11992591 DOI: 10.1002/alz.70138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Effect of apolipoprotein E (APOE) on Alzheimer's disease and related dementias (ADRD) risk is heterogeneous across populations, with scarce data on Hispanics/Latinos. METHODS APOE genotype was studied in 12,221 Hispanics/Latinos (per cohort and via metanalysis): Caribbean-Hispanics, Mexicans, Mexican-Americans, and Peruvians/Bolivians. A subsample had longitudinal assessment and plasma p-tau. We tested the modifying effects of global and local ancestries. Results were replicated in an independent Peruvian cohort and brain samples. RESULTS APOE ε4 effect was strongest in Peruvians/Bolivians (odds ratio [OR] = 6.13, 95% confidence interval [CI] = 2.71-13.83), followed by Mexicans (OR = 4.31, 95% CI = 1.58-11.74), Mexican-Americans (OR = 3.06, 95% CI = 2.04-4.59), and Caribbean-Hispanics (OR = 2.22, 95% CI = 1.99-2.48). Meta-analyses showed OR = 2.32 (95% CI = 2.09-2.57) and OR = 0.81 (95% CI = 0.68-0.97) for the ε4 and ε2 allele, respectively. The APOE ε4 effect was replicated independently in Peruvians (OR = 5.06, 95% CI = 2.48-10.70). ε4 carriers displayed higher ADRD conversions and p-tau levels. Global and local ancestries did not modify ADRD risk, and they were associated with Braak stage. DISCUSSION APOE shows a heterogeneous effect on ADRD risk in our Hispanics/Latinos sample, the largest to date. HIGHLIGHTS The apolipoprotein E (APOE) ε4 effect is stronger in Peruvians/Bolivians than in other Hispanic/Latino groups. The strong APOE effect size in Peruvians and Bolivians was replicated in a second independent Peruvian cohort. Meta-analysis for ε4 and ε2 confirmed a significant association with Alzheimer's disease and related dementias (ADRD). Global and local ancestry do not modify the association between APOE genotype and ADRD.
Collapse
Affiliation(s)
- Sandra Barral
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Zikun Yang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Nicole Phillips
- Department of MicrobiologyImmunology and Genetics UNT Health Science Center Fort WorthFort WorthTexasUSA
| | - Robert C. Barber
- Institute for Translational Research and Department of Family MedicineUNT Health Science CenterFort WorthTexasUSA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Lawrence S. Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Basilio Cieza
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Dolly Reyes‐Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Farid Rajabli
- John P. Hussman Institute for Human Genomics, Dr. John Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Michael L. Cuccaro
- John P. Hussman Institute for Human Genomics, Dr. John Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jeffery M. Vance
- John P. Hussman Institute for Human Genomics, Dr. John Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Silvia Mejia Arango
- Institute of Neuroscience, School of MedicineUniversity of Texas Rio Grande ValleyHarlingenTexasUSA
| | | | | | - Rosa Montesinos
- Unidad de diagnóstico de deterioro cognitivo y prevención de demenciaInstituto Peruano de NeurocienciasLince LimaPerú
| | | | | | | | - Ivonne Z. Jimenez Velazquez
- Department of Medicine, Medical Sciences CampusUniversity of Puerto Rico School of MedicineSan JuanPuerto Rico
| | - Victoria Marca
- Neurogenetics Working GroupUniversidad Cientifica del SurLimaPeru
| | | | - Mario Cornejo‐Olivas
- Neurogenetics Working GroupUniversidad Cientifica del SurLimaPeru
- Neurogenetics Research CenterInstituto Nacional de Ciencias NeurologicasLimaPeru
| | - Margaret Pericak‐Vance
- John P. Hussman Institute for Human Genomics, Dr. John Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Rebeca Wong
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas HealthSan AntonioTexasUSA
| | - Sid O'Bryant
- Department of MicrobiologyImmunology and Genetics UNT Health Science Center Fort WorthFort WorthTexasUSA
| | - Nilton Custodio
- Unidad de diagnóstico de deterioro cognitivo y prevención de demenciaInstituto Peruano de NeurocienciasLince LimaPerú
- Escuela Profesional de Medicina HumanaUniversidad Privada San Juan BautistaChorrillosPeru
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky Center, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| |
Collapse
|
7
|
He R, Ren J, Malakhov MM, Pan W. Enhancing nonlinear transcriptome- and proteome-wide association studies via trait imputation with applications to Alzheimer's disease. PLoS Genet 2025; 21:e1011659. [PMID: 40209152 PMCID: PMC12040266 DOI: 10.1371/journal.pgen.1011659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 04/29/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
Genome-wide association studies (GWAS) performed on large cohort and biobank datasets have identified many genetic loci associated with Alzheimer's disease (AD). However, the younger demographic of biobank participants relative to the typical age of late-onset AD has resulted in an insufficient number of AD cases, limiting the statistical power of GWAS and any downstream analyses. To mitigate this limitation, several trait imputation methods have been proposed to impute the expected future AD status of individuals who may not have yet developed the disease. This paper explores the use of imputed AD status in nonlinear transcriptome/proteome-wide association studies (TWAS/PWAS) to identify genes and proteins whose genetically regulated expression is associated with AD risk. In particular, we considered the TWAS/PWAS method DeLIVR, which utilizes deep learning to model the nonlinear effects of expression on disease. We trained transcriptome and proteome imputation models for DeLIVR on data from the Genotype-Tissue Expression (GTEx) Project and the UK Biobank (UKB), respectively, with imputed AD status in UKB participants as the outcome. Next, we performed hypothesis testing for the DeLIVR models using clinically diagnosed AD cases from the Alzheimer's Disease Sequencing Project (ADSP). Our results demonstrate that nonlinear TWAS/PWAS trained with imputed AD outcomes successfully identifies known and putative AD risk genes and proteins. Notably, we found that training with imputed outcomes can increase statistical power without inflating false positives, enabling the discovery of molecular exposures with potentially nonlinear effects on neurodegeneration.
Collapse
Affiliation(s)
- Ruoyu He
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jingchen Ren
- School of Statistics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Mykhaylo M. Malakhov
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Wei Pan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
8
|
Xavier C, Pinto N. Navigating the blurred boundary: Neuropathologic changes versus clinical symptoms in Alzheimer's disease, and its consequences for research in genetics. J Alzheimers Dis 2025; 104:611-626. [PMID: 39956949 DOI: 10.1177/13872877251317543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
During decades scientists tried to unveil the genetic architecture of Alzheimer's disease (AD), recurring to increasingly larger sample numbers for genome-wide association studies (GWAS) in hope for higher statistical gains. Here, a retrospective look on the most prominent GWAS was performed, focusing on the quality of the diagnosis associated with the used data and databases. Different methods for AD diagnosis (or absence) carry different levels of accuracy and certainty applied to both subsets of cases and controls. Furthermore, the different phenotypes included in these databases were explored, as several incorporate other ageing comorbidities and might be encompassing many confounding agents as well. Age of the samples' donors and origin populations were also investigated as these could be biasing factors in posterior analyses. A tendency for looser diagnostic methods in more recent GWAS was observed, where greater datasets of individuals are analyzed, which may have been hampering the discovery of associated genetic variants. Specifically for AD, a diagnostic method conveying a clinical outcome may be distinct from the disease neuropathological assessment, since the first has a practical perspective that not necessarily needs a confirmation. Due to its properties and complex diagnosis, this work highlights the importance of the neuropathological confirmation of AD (or its absence) in the subjects considered for research purposes to avoid reaching statistically weak and/or misleading conclusions that may trigger further studies with powerless groundwork.
Collapse
Affiliation(s)
- Catarina Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Nádia Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- CMUP - Centro de Matemática da Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Ferreiro López S, Ferrero R, Blom-Dahl J, Alonso-Bernáldez M, González A, Pérez-Solero G, Tenorio-Castano J. Development of a k-Nearest Neighbors Model for the Prediction of Late-Onset Alzheimer's Risk by Combining Polygenic Risk Scores and Phenotypic Variables. Genes (Basel) 2025; 16:377. [PMID: 40282337 PMCID: PMC12027161 DOI: 10.3390/genes16040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Alzheimer's disease (AD), and more specifically late-onset Alzheimer's disease (LOAD), represents a considerable challenge in terms of early and timely diagnosis and treatment. Early diagnosis is crucial to improve the efficacy of the therapies and patients' quality of life. The current challenge is to accurately identify at-risk individuals before the manifestations of the first symptoms of AD. METHODS AND RESULTS Here, we present an improved model for LOAD risk prediction, which applies the k-nearest neighbors (KNN) algorithm. We have achieved a sensitivity of 0.80 and an area under the curve (AUC) of 0.71, which represents a high performance especially when compared to an AUC of 0.66 reported previously in 2019 using a KNN model. DISCUSSION The application of a mathematical model that combines genetic and clinical covariates showed a good prediction of the AD/LOAD risk, with the higher weight being the polygenic genetic risk, APOE haplotype, and age. Compared to previous studies, our model integrates and correlates genetic prediction together with phenotypic information by fine-tuning the parameters of the model in order to achieve the best performance. This algorithm can be used in the general population and does not require the manifestation of any symptoms for its effective application. Thus, we present here an advanced model for risk prediction of LOAD.
Collapse
Affiliation(s)
- Sandra Ferreiro López
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
| | - Rosana Ferrero
- Center of Applied Ecology and Sustainability (CAPES), Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
| | - Jorge Blom-Dahl
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
| | - Marta Alonso-Bernáldez
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
| | - Adán González
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
| | - Guillermo Pérez-Solero
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
| | - Jair Tenorio-Castano
- ADNTRO Genetics, Carretera Betlem, s/n, Colonia de Sant Pere, 07579 Arta, Spain; (S.F.L.); (J.B.-D.); (M.A.-B.); (A.G.); (G.P.-S.)
- INGEMM, Institute of Medical and Molecular Genetics, La Paz University Hospital, IdiPAZ, 28046 Madrid, Spain
- ITHACA, European Research Network, La Paz University Hospital, 28046 Madrid, Spain
- Network for Biomedical Research on Rare Diseases (CIBERER), Carlos III Health Institute (ISCIII), 28046 Madrid, Spain
| |
Collapse
|
10
|
Miller JB, Brandon JA, Harmon LM, Sabra HW, Lucido CC, Murcia JDG, Nations KA, Payne SH, Ebbert MTW, Kauwe JSK, Ridge PG. Ramp Sequence May Explain Synonymous Variant Association with Alzheimer's Disease in the Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA). Biomedicines 2025; 13:739. [PMID: 40149715 PMCID: PMC11940050 DOI: 10.3390/biomedicines13030739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background: The synonymous variant NC_000007.14:g.100373690T>C (rs2405442:T>C) in the Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA) gene was previously associated with decreased risk for Alzheimer's disease (AD) in genome-wide association studies, but its biological impact is largely unknown. Objective: We hypothesized that rs2405442:T>C decreases mRNA and protein levels by destroying a ramp of slowly translated codons at the 5' end of PILRA. Methods: We assessed rs2405442:T>C predicted effects on PILRA through quantitative polymerase chain reactions (qPCRs) and enzyme-linked immunosorbent assays (ELISAs) using Chinese hamster ovary (CHO) cells. RESULTS: Both mRNA (p = 1.9184 × 10-13) and protein (p = 0.01296) levels significantly decreased in the mutant versus the wildtype in the direction that we predicted based on the destruction of a ramp sequence. Conclusions: We show that rs2405442:T>C alone directly impacts PILRA mRNA and protein expression, and ramp sequences may play a role in regulating AD-associated genes without modifying the protein product.
Collapse
Affiliation(s)
- Justin B. Miller
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - J. Anthony Brandon
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
| | - Lauren M. Harmon
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Hady W. Sabra
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Chloe C. Lucido
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Josue D. Gonzalez Murcia
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Kayla A. Nations
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
| | - Samuel H. Payne
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Mark T. W. Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA (M.T.W.E.)
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40506, USA
| | - John S. K. Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, UT 84602, USA; (L.M.H.); (J.D.G.M.); (J.S.K.K.)
| |
Collapse
|
11
|
Jang B, Bp K, Tokolyi A, Cuddleston WH, Ravi A, Jung SH, Naito T, Kim B, Seo Kim M, Cho M, Park MS, Rosen M, Blanchard J, Humphrey J, Knowles DA, Won HH, Raj T. SingleBrain: A Meta-Analysis of Single-Nucleus eQTLs Linking Genetic Risk to Brain Disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.06.25323424. [PMID: 40093234 PMCID: PMC11908325 DOI: 10.1101/2025.03.06.25323424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Most genetic risk variants for neurological diseases are located in non-coding regulatory regions, where they may often act as expression quantitative trait loci (eQTLs), modulating gene expression and influencing disease susceptibility. However, eQTL studies in bulk brain tissue or specific cell types lack the resolution to capture the brain's cellular diversity. Single-nucleus RNA sequencing (snRNA-seq) offers high-resolution mapping of eQTLs across diverse brain cell types. Here, we performed a meta-analysis, "SingleBrain," integrating publicly available snRNA-seq and genotype data from four cohorts, totaling 5.8 million nuclei from 983 individuals. We mapped cis-eQTLs across major brain cell types and subtypes and employed statistical colocalization and Mendelian randomization to identify genes mediating neurological disease risk. We observed up to a 10-fold increase in cis-eQTLs compared to previous studies and uncovered novel cell type-specific genes linked to Alzheimer's disease, Parkinson's disease, and schizophrenia that were previously undetectable in bulk tissue analyses. Additionally, we prioritized putative causal variants for each locus through fine-mapping and integration with cell type-specific enhancer and promoter regulatory elements. SingleBrain represents a comprehensive single-cell eQTL resource, advancing insights into the genetic regulation of brain disorders.
Collapse
Affiliation(s)
- Beomjin Jang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Kailash Bp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alex Tokolyi
- Departments of Computer Science and Systems Biology, Columbia University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Winston H Cuddleston
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashvin Ravi
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Biological and Medical Informatics PhD Program, University of California, San Francisco, San Francisco, CA
| | - Sang-Hyuk Jung
- Department of Medical Informatics, Kangwon National University College of Medicine, Chuncheon 24341, Republic of Korea
| | - Tatsuhiko Naito
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Beomsu Kim
- Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Min Seo Kim
- Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Minyoung Cho
- Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Mi-So Park
- Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Mikaela Rosen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joel Blanchard
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jack Humphrey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David A Knowles
- Departments of Computer Science and Systems Biology, Columbia University, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Hong-Hee Won
- Department of Digital Health, SAIHST, Sungkyunkwan University, Samsung Medical Center, Seoul 06351, Republic of Korea
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
12
|
Peng J, Tang Q, Li Y, Liu L, Biswal BB, Wang P. Neuromorphic deviations associated with transcriptomic expression and specific cell type in Alzheimer's disease. Sci Rep 2025; 15:7460. [PMID: 40032887 PMCID: PMC11876660 DOI: 10.1038/s41598-025-90872-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: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is known to be associated with cortical anatomical atrophy and neurodegeneration across various brain regions. However, the relationships between brain structural changes in AD and gene expression remain unclear. We perform the morphometric similarity network (MSN) analysis to reveal the consistent cortical structural differences in individuals with AD compared to controls, and investigate the associations between brain-wide gene expression and morphometric changes. Furthermore, we identify abnormally MSN-related genes linked to specific cell types as the major contributors to transcriptomic relationships. MSN-related structural changes are located in the lateral ventral prefrontal cortex, temporal pole and medial prefrontal lobe, which are highly associated with the AD's cognitive decline. Analysis of gene expression shows the spatial correlations between AD-related genes and MSN differences. Examination of cell type-specific signature genes indicates that changes in microglia and neuronal transcriptional profiles largely contribute to AD-specific MSN differences. The study map the disease-specific structural alterations in AD down to the cellular level, offering a novel perspective on the linking surface-level changes to molecular mechanisms.
Collapse
Affiliation(s)
- Jinzhong Peng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Qin Tang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Yilu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Lin Liu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China
| | - Bharat Bhusan Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China.
- Department of Biomedical Engineering, New Jersey Institute of Technology, 607 Fenster Hall, University Height, Newark, NJ, 07102, USA.
| | - Pan Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, 2006 Xiyuan Avenue, Chengdu, 611731, China.
| |
Collapse
|
13
|
Han S, Cho SA, Choi W, Eilbeck K, Coon H, Nho K, Lee Y. Interaction of genetic variants and methylation in transcript-level expression regulation in Alzheimer's disease by multi-omics data analysis. BMC Genomics 2025; 26:170. [PMID: 39979805 PMCID: PMC11844006 DOI: 10.1186/s12864-025-11362-x] [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: 04/05/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) presents a significant public health problem and major cause of dementia. Not only genetic but epigenetic factors contribute to complex and heterogeneous molecular mechanisms underlying AD risk; in particular, single nucleotide polymorphisms (SNPs) and DNA methylation can lead to dysregulation of gene expression in the AD brain. Each of these regulators has been independently studied well in AD progression, however, their interactive roles, particularly when they are located differently, still remains unclear. Here, we aimed to explore the interplay between SNPs and DNA methylation in regulating transcript expression levels in the AD brain through an integrative analysis of whole-genome sequencing, RNA-seq, and methylation data measured from the dorsolateral prefrontal cortex. RESULTS We identified 179 SNP-methylation combination pairs that showed statistically significant interactions associated with the expression of 67 transcripts (63 unique genes), enriched in functional pathways, including immune-related and post-synaptic assembly pathways. Particularly, a number of HLA family genes (HLA-A, HLA-B, HLA-C, HLA-DRB1, HLA-DRB5, HLA-DPA1, HLA-K, HLA-DQB1, and HLA-DMA) were observed as having expression changes associated with the interplay. CONCLUSIONS Our findings especially implicate immune-related pathways as targets of these regulatory interactions. SNP-methylation interactions may thus contribute to the molecular complexity underlying immune-related pathogenies in AD patients. Our study provides a new molecular knowledge in the context of the interplay between genetic and epigenetic regulations, in that it concerns transcript expression status in AD.
Collapse
Affiliation(s)
- Seonggyun Han
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Soo-Ah Cho
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Wongyung Choi
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Karen Eilbeck
- Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Younghee Lee
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
14
|
Chandler HL, Wheeler J, Escott‐Price V, Murphy K, Lancaster TM. Non-APOE variants predominately expressed in smooth muscle cells contribute to the influence of Alzheimer's disease genetic risk on white matter hyperintensities. Alzheimers Dement 2025; 21:e14455. [PMID: 39737667 PMCID: PMC11848156 DOI: 10.1002/alz.14455] [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: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION White matter hyperintensity volumes (WMHVs) are disproportionally prevalent in individuals with Alzheimer's disease (AD), potentially reflecting neurovascular injury. We quantify the association between AD polygenic risk score (AD-PRS) and WMHV, exploring single-nucleotide polymorphisms (SNPs) that are proximal to genes overexpressed in cerebrovascular cell species. METHODS In a UK-Biobank sub-sample (mean age = 64, range = 45-81 years), we associate WMHV with (1) AD-PRS estimated via SNPs across the genome (minus apolipoprotein E [APOE] locus) and (2) AD-PRS estimated with SNPs proximal to specific genes that are overexpressed in cerebrovascular cell species. RESULTS We observed a positive association between non-APOE-AD-PRS and WMHVs. We further demonstrate an association between WMHVs and AD-PRS constructed with SNPs that are proximal to genes over-represented in smooth muscles cells (SMCs; β = 0.135, PFWE < 0.01) and internally replicated (PDISCOVERY+REPLICATION < 0.01). DISCUSSION Common AD genetic risk could explain physiological processes underlying vascular pathology in AD. SMC function may offer a treatment target to prevent WMHV-related AD pathophysiology prior to the onset of symptoms. HIGHLIGHTS Alzheimer's disease (AD) risk factors such as apolipoprotein E (APOE) ε4, link to increased white matter hyperintensity volume (WMHV). WMHVs indicate vascular risk and neurovascular injury in AD. The broader genetic link between AD risk and WMHV is not fully understood. We quantify AD polygenic risk score (PRS) associations with WMHV, excluding APOE. AD-PRS in smooth muscle cells (SMCs) shows a significant association with increased WMHV.
Collapse
Affiliation(s)
- Hannah Louise Chandler
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | - Joshua Wheeler
- School of Clinical SciencesUniversity of BristolBristolUK
- Department of PsychologyUniversity of BathBathUK
| | - Valentina Escott‐Price
- Centre for Neuropsychiatric Genetics and GenomicsDepartment of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUK
| | - Kevin Murphy
- School of Physics and AstronomyCardiff University Brain Research Imaging Centre (CUBRIC)Cardiff UniversityCardiffUK
| | | |
Collapse
|
15
|
Torre M, Zanella CA, Feany MB. The Biological Intersection Between Chemotherapy-Related Cognitive Impairment and Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00026-4. [PMID: 39863251 DOI: 10.1016/j.ajpath.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/27/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Alzheimer disease (AD) is the most common type of dementia and one of the leading causes of death in elderly patients. The number of patients with AD in the United States is projected to double by 2060. Thus, understanding modifiable risk factors for AD is an urgent public health priority. In parallel with the number of patients with AD, the number of cancer survivors is estimated to increase significantly, and up to 80% of cancer patients treated with chemotherapy will develop cognitive deficits, termed chemotherapy-related cognitive impairment. This review discusses biologically plausible pathways underlying both disorders, with the goal of understanding why a proportion of chemotherapy patients may be at higher risk of developing AD. Highlighted are the E4 allele of the apolipoprotein E gene, neuroinflammation, oxidative stress, DNA damage, mitochondrial dysfunction, neuronal and synaptic loss, cellular senescence, brain-derived neurotrophic factor signaling, white matter damage, blood-brain barrier/vascular dysfunction, tau pathology, and transposable element reactivation.
Collapse
Affiliation(s)
- Matthew Torre
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas; Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, Texas.
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Miller JB, Brandon JA, McKinnon LM, Sabra HW, Lucido CC, Gonzalez Murcia JD, Nations KA, Payne SH, Ebbert MT, Kauwe JS, Ridge PG. Ramp sequence may explain synonymous variant association with Alzheimer's disease in the Paired Immunoglobulin-like Type 2 Receptor Alpha ( PILRA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631528. [PMID: 39829933 PMCID: PMC11741268 DOI: 10.1101/2025.01.06.631528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND Synonymous variant NC_000007.14:g.100373690T>C (rs2405442:T>C) in the Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA) gene was previously associated with decreased risk for Alzheimer's disease (AD) in genome-wide association studies, but its biological impact is largely unknown. OBJECTIVE We hypothesized that rs2405442:T>C decreases mRNA and protein levels by destroying a ramp of slowly translated codons at the 5' end of PILRA. METHODS We assessed rs2405442:T>C predicted effects on PILRA through quantitative polymerase chain reactions (qPCR) and enzyme-linked immunosorbent assays (ELISA) using Chinese hamster ovary (CHO) cells. RESULTS Both mRNA (P=1.9184 × 10-13) and protein (P=0.01296) levels significantly decreased in the mutant versus the wildtype in the direction that we predicted based on destroying a ramp sequence. CONCLUSIONS We show that rs2405442:T>C alone directly impacts PILRA mRNA and protein expression, and ramp sequences may play a role in regulating AD-associated genes without modifying the protein product.
Collapse
Affiliation(s)
- Justin B. Miller
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - J. Anthony Brandon
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | | | - Hady W. Sabra
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Chloe C. Lucido
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40506, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
| | | | - Kayla A. Nations
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Samuel H. Payne
- Department of Biology, Brigham Young University, Provo, UT 84602
| | - Mark T.W. Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY 40506, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40506, USA
| | - John S.K. Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602
| | - Perry G. Ridge
- Department of Biology, Brigham Young University, Provo, UT 84602
| |
Collapse
|
17
|
Nazarian A, Morado M, Kulminski AM. Complex genetic interactions affect susceptibility to Alzheimer's disease risk in the BIN1 and MS4A6A loci. GeroScience 2025:10.1007/s11357-024-01477-6. [PMID: 39751715 DOI: 10.1007/s11357-024-01477-6] [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: 10/28/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025] Open
Abstract
Genetics is the second strongest risk factor for Alzheimer's disease (AD) after age. More than 70 loci have been implicated in AD susceptibility so far, and the genetic architecture of AD entails both additive and nonadditive contributions from these loci. To better understand nonadditive impact of single-nucleotide polymorphisms (SNPs) on AD risk, we examined individual, joint, and interacting (SNPxSNP) effects of 139 and 66 SNPs mapped to the BIN1 and MS4A6A AD-associated loci, respectively. The analyses were conducted by fitting three respective dominant allelic-effect models using data from four independent studies. Joint effects were analyzed by considering pairwise combinations of genotypes of the selected SNPs, i.e., compound genotypes (CompG). The individual SNP analyses showed associations of 18 BIN1 SNPs and 4 MS4A6A SNPs with AD. We identified 589 BIN1 and 217 MS4A6A SNP pairs associated with AD in the CompG analysis, although their individual SNPs were not linked to AD independently. Notably, 34 BIN1 and 10 MS4A6A SNP pairs exhibited both significant SNPxSNP interaction effects and significant CompG effects. The vast majority of nonadditive effects were captured through the CompG analysis. These results expand the current understanding of the contributions of the BIN1 and MS4A6A loci to AD susceptibility. The identified nonadditive effects suggest a significant genetic modulation mechanism underlying the genetic heterogeneity of AD in these loci. Our findings highlight the importance of considering nonadditive genetic impacts on AD risk beyond the traditional SNPxSNP approximation, as they may uncover critical mechanisms not apparent when examining SNPs individually.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC, 27705, USA.
| | - Marissa Morado
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC, 27705, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St, Durham, NC, 27705, USA.
| |
Collapse
|
18
|
Eulalio T, Sun MW, Gevaert O, Greicius MD, Montine TJ, Nachun D, Montgomery SB. regionalpcs improve discovery of DNA methylation associations with complex traits. Nat Commun 2025; 16:368. [PMID: 39753567 PMCID: PMC11698866 DOI: 10.1038/s41467-024-55698-6] [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: 04/08/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
We have developed the regionalpcs method, an approach for summarizing gene-level methylation. regionalpcs addresses the challenge of deciphering complex epigenetic mechanisms in diseases like Alzheimer's disease. In contrast to averaging, regionalpcs uses principal components analysis to capture complex methylation patterns across gene regions. Our method demonstrates a 54% improvement in sensitivity over averaging in simulations, providing a robust framework for identifying subtle epigenetic variations. Applying regionalpcs to Alzheimer's disease brain methylation data, combined with cell type deconvolution, we uncover 838 differentially methylated genes associated with neuritic plaque burden-significantly outperforming conventional methods. Integrating methylation quantitative trait loci with genome-wide association studies identified 17 genes with potential causal roles in Alzheimer's disease risk, including MS4A4A and PICALM. Available in the Bioconductor package regionalpcs, our approach facilitates a deeper understanding of the epigenetic landscape in Alzheimer's disease and opens avenues for research into complex diseases.
Collapse
Affiliation(s)
- Tiffany Eulalio
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| | - Min Woo Sun
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Olivier Gevaert
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael D Greicius
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | - Daniel Nachun
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Stephen B Montgomery
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
19
|
Gao S, Zhu P, Wang T, Han Z, Xue Y, Zhang Y, Wang L, Zhang H, Chen Y, Liu G. Alzheimer's disease genome-wide association studies in the context of statistical heterogeneity. Mol Psychiatry 2025; 30:342-348. [PMID: 38965422 DOI: 10.1038/s41380-024-02654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Affiliation(s)
- Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Ping Zhu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Tao Wang
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Zhifa Han
- Center of Respiratory Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Acadamy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, 100029, Beijing, China
| | - Yanli Xue
- School of Biomedical Engineering, Capital Medical University, 10069, Beijing, China
| | - Yan Zhang
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Longcai Wang
- Department of Anesthesiology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Haihua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Yan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu, 241002, Anhui, China
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No.22, Wenchang Road, Wuhu, 241002, Anhui, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, 100069, Beijing, China.
- Chinese Institute for Brain Research, 102206, Beijing, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Wannan Medical College, No. 22, Wenchang Road, Wuhu, 241002, Anhui, China.
- Institute of Chronic Disease Prevention and Control, Wannan Medical College, No.22, Wenchang Road, Wuhu, 241002, Anhui, China.
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Brain Hospital, Shengli Oilfield Central Hospital, Dongying, 257000, Shandong, China.
| |
Collapse
|
20
|
Weymouth L, Smith AR, Lunnon K. DNA Methylation in Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:149-178. [PMID: 39455499 DOI: 10.1007/7854_2024_530] [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: 10/28/2024]
Abstract
To date, DNA methylation is the best characterized epigenetic modification in Alzheimer's disease. Involving the addition of a methyl group to the fifth carbon of the cytosine pyrimidine base, DNA methylation is generally thought to be associated with the silencing of gene expression. It has been hypothesized that epigenetics may mediate the interaction between genes and the environment in the manifestation of Alzheimer's disease, and therefore studies investigating DNA methylation could elucidate novel disease mechanisms. This chapter comprehensively reviews epigenomic studies, undertaken in human brain tissue and purified brain cell types, focusing on global methylation levels, candidate genes, epigenome wide approaches, and recent meta-analyses. We discuss key differentially methylated genes and pathways that have been highlighted to date, with a discussion on how new technologies and the integration of multiomic data may further advance the field.
Collapse
Affiliation(s)
- Luke Weymouth
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Adam R Smith
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
21
|
Güzel Ö, Kehoe PG. The Contribution of the Renin-Angiotensin System to Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:107-127. [PMID: 39543022 DOI: 10.1007/7854_2024_525] [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: 11/17/2024]
Abstract
The renin-angiotensin system (RAS) is becoming increasingly recognised as a biochemical pathway relevant to the development and progression of Alzheimer's disease (AD). RAS involvement in AD was initially linked to AD via numerous genetic association studies and more recent Genome-Wide Association Studies (GWAS), and in some cases in relation to classical hallmarks of AD pathology. Since these initial findings, which will be summarised here, several complementary areas of research are converging in support of what has been proposed as the Angiotensin Hypothesis for Alzheimer's disease. This hypothesis proposes how the RAS and disease-associated changes to the normal balance between opposing regulatory pathways within RAS warrant careful consideration in the pathogenesis of AD and its pathology. We discuss some of these in relation to RAS-targeting therapeutics, originally developed for the treatment of cardiovascular conditions, and how they might be repurposed as interventions for AD.
Collapse
Affiliation(s)
- Özge Güzel
- Cerebrovascular and Dementia Research Group, Bristol Medical School, University of Bristol, Bristol, UK.
- Department of Genetics and Bioengineering, Alanya Alaaddin Keykubat University, Antalya, Türkiye.
| | - Patrick G Kehoe
- Cerebrovascular and Dementia Research Group, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
22
|
Dai L, Jiang C, Cui Q, Huang L, Chen S, Zhang Y, Luo X, Zhang P, Li J, Zhang Y. Cardiovascular health, genetic predisposition, and dementia risk among atherosclerotic cardiovascular disease patients. J Prev Alzheimers Dis 2025; 12:100020. [PMID: 39800454 DOI: 10.1016/j.tjpad.2024.100020] [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: 05/02/2025]
Abstract
BACKGROUND While optimal cardiovascular health (CVH) has been linked to a lower risk of dementia, few studies considered individuals' genetic background. We aimed to examine the interaction between CVH and genetic predisposition on dementia risk among individuals with atherosclerotic cardiovascular disease (ASCVD). METHODS We included 30,818 ASCVD patients from the UK Biobank. CVH was assessed using Life's Essential 8, and genetic predisposition determined by a genetic risk score (GRS) incorporating 85 genetic variants. Cox proportional hazard models were used to estimate hazard ratios (HRs) for all-cause dementia, Alzheimer's disease (AD), and vascular dementia (VaD). RESULTS Over a median follow-up of 13.5 years, 1,360 cases of all-cause dementia were identified, including 489 AD and 440 VaD cases. Higher CVH levels were associated with a reduced risk of all-cause dementia (HR for high vs. low CVH: 0.60; 95 % CI: 0.47-0.77) and VaD (HR for high vs. low CVH: 0.32; 95 % CI: 0.19-0.54), with a stronger association in individuals with lower GRS. Although the overall CVH score was not associated with the risk of dementia in individuals with high GRS, higher levels of sleep and glucose control were associated with a lower risk of VaD. CVH levels showed no association with the risk of AD. CONCLUSION Higher CVH levels were associated with a lower risk of VaD, not AD, with a stronger association in individuals with low GRS. Improvements in specific LE8 components, particularly sleep health and blood glucose management, were associated with reduced VaD risk across various genetic risk strata.
Collapse
Affiliation(s)
- Lingyan Dai
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chunxiang Jiang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Qingmei Cui
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Leen Huang
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Siqi Chen
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yidan Zhang
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoyi Luo
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; School of Public Health, Southern Medical University, Guangzhou, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Li
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China; School of Public Health, Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China; Department of Epidemiology and Center for Global Cardiometabolic Health, School of Public Health, Brown University, Providence, RI, USA.
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
23
|
Breddels EM, Snihirova Y, Pishva E, Gülöksüz S, Blokland GAM, Luykx J, Andreassen OA, Linden DEJ, van der Meer D, For the Alzheimer's Disease Neuroimaging Initiative. Brain morphology mediating the effects of common genetic risk variants on Alzheimer's disease. J Alzheimers Dis Rep 2025; 9:25424823251328300. [PMID: 40144144 PMCID: PMC11938454 DOI: 10.1177/25424823251328300] [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: 08/02/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background Late-onset Alzheimer's disease (LOAD) has been associated with alterations in the morphology of multiple brain structures, and it is likely that disease mechanisms differ between brain regions. Coupling genetic determinants of LOAD with measures of brain morphology could localize and identify primary causal neurobiological pathways. Objective To determine causal pathways from genetic risk variants of LOAD via brain morphology to LOAD. Methods Mediation and Mendelian randomization (MR) analysis were performed using common genetic variation, T1 MRI and clinical data collected by UK Biobank and Alzheimer's Disease Neuroimaging Initiative. Results Thickness of the entorhinal cortex and the volumes of the hippocampus, amygdala and inferior lateral ventricle mediated the effect of APOE ε4 on LOAD. MR showed that a thinner entorhinal cortex, a smaller hippocampus and amygdala, and a larger volume of the inferior lateral ventricles, increased the risk of LOAD as well as vice versa. Conclusions Combining neuroimaging and genetic data can give insight into the causal neuropathological pathways of LOAD.
Collapse
Affiliation(s)
- Esmee M Breddels
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Yelyzaveta Snihirova
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Ehsan Pishva
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Faculty of Health and Life Sciences, Medical School, University of Exeter, Exeter, UK
| | - Sinan Gülöksüz
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Faculty of Health and Life Sciences, Medical School, University of Exeter, Exeter, UK
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Gabriëlla AM Blokland
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jurjen Luykx
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Center, Amsterdam, the Netherlands
- GGZ in Geest Mental Health Care, Amsterdam, The Netherlands
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders Research, Oslo University Hospital & University of Oslo, Oslo, Norway
| | - David EJ Linden
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Dennis van der Meer
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | |
Collapse
|
24
|
Visscher PM, Gyngell C, Yengo L, Savulescu J. Heritable polygenic editing: the next frontier in genomic medicine? Nature 2025; 637:637-645. [PMID: 39779842 PMCID: PMC11735401 DOI: 10.1038/s41586-024-08300-4] [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: 06/30/2022] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Polygenic genome editing in human embryos and germ cells is predicted to become feasible in the next three decades. Several recent books and academic papers have outlined the ethical concerns raised by germline genome editing and the opportunities that it may present1-3. To date, no attempts have been made to predict the consequences of altering specific variants associated with polygenic diseases. In this Analysis, we show that polygenic genome editing could theoretically yield extreme reductions in disease susceptibility. For example, editing a relatively small number of genomic variants could make a substantial difference to an individual's risk of developing coronary artery disease, Alzheimer's disease, major depressive disorder, diabetes and schizophrenia. Similarly, large changes in risk factors, such as low-density lipoprotein cholesterol and blood pressure, could, in theory, be achieved by polygenic editing. Although heritable polygenic editing (HPE) is still speculative, we completed calculations to discuss the underlying ethical issues. Our modelling demonstrates how the putatively positive consequences of gene editing at an individual level may deepen health inequalities. Further, as single or multiple gene variants can increase the risk of some diseases while decreasing that of others, HPE raises ethical challenges related to pleiotropy and genetic diversity. We conclude by arguing for a collectivist perspective on the ethical issues raised by HPE, which accounts for its effects on individuals, their families, communities and society4.
Collapse
Affiliation(s)
- Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Christopher Gyngell
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Loic Yengo
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Julian Savulescu
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Uehiro Oxford Institute, University of Oxford, Oxford, UK.
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
25
|
Ramprasad P, Ren J, Pan W. Enhancing Gene Expression Predictions Using Deep Learning and Functional Annotations. Genet Epidemiol 2025; 49:e22595. [PMID: 39344923 DOI: 10.1002/gepi.22595] [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: 07/17/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
Transcriptome-wide association studies (TWAS) aim to uncover genotype-phenotype relationships through a two-stage procedure: predicting gene expression from genotypes using an expression quantitative trait locus (eQTL) data set, then testing the predicted expression for trait associations. Accurate gene expression prediction in stage 1 is crucial, as it directly impacts the power to identify associations in stage 2. Currently, the first stage of such studies is primarily conducted using linear models like elastic net regression, which fail to capture the nonlinear relationships inherent in biological systems. Deep learning methods have the potential to model such nonlinear effects, but have yet to demonstrably outperform linear methods at this task. To address this gap, we propose a new deep learning architecture to predict gene expression from genotypic variation across individuals. Our method utilizes a learnable input scaling layer in conjunction with a convolutional encoder to capture nonlinear effects and higher-order interactions without compromising on interpretability. We further augment this approach to allow for parameter sharing across multiple networks, enabling us to utilize prior information for individual variants in the form of functional annotations. Evaluations on real-world genomic data show that our method consistently outperforms elastic net regression across a large set of heritable genes. Furthermore, our model statistically significantly improved predictive performance by leveraging functional annotations, whereas elastic net regression failed to show equivalent gains when using the same information, suggesting that our method can capture nonlinear functional information beyond the capability of linear models.
Collapse
Grants
- This research was supported by NIH grants U01 AG073079, R01 AG065636, R01 AG069895, and RF1 AG067924, and by the Minnesota Supercomputing Institute at the University of Minnesota. The Genotype-Tissue Expression (GTEx) Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by NCI, NHGRI, NHLBI, NIDA, NIMH, and NINDS; the GTEx data were obtained from dbGaP Project #26511.
Collapse
Affiliation(s)
- Pratik Ramprasad
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jingchen Ren
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei Pan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
26
|
Chen J, Xie J, Deng F, Cai J, Chen S, Song X, Xia S, Shen Q, Guo X, Tang Y. Expansion of peripheral cytotoxic CD4+ T cells in Alzheimer's disease: New insights from multi-omics evidence. Genomics 2025; 117:110976. [PMID: 39657893 DOI: 10.1016/j.ygeno.2024.110976] [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: 06/14/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The significance of the adaptive immune response in Alzheimer's disease (AD) is increasingly recognized. We analyzed scRNA-Seq data from AD patients, revealing a notable rise in CD4 cytotoxic T cells (CD4-CTLs) in peripheral blood mononuclear cells (PBMCs), validated in vivo and in vitro. This rise correlates with cognitive decline in AD patients. We also identified transcription factors TBX21 and MYBL1 as key drivers of CD4-CTL expansion. Further analyses indicate these cells are terminally differentiated, showing clonal expansion, metabolic changes, and unique communication patterns. Mendelian randomization identified risk genes SRGN and ITGB1, suggesting their genetic regulation in CD4-CTLs may contribute to AD. To summarize, our findings characterize the expansion of CD4-CTLs in the PBMCs of AD patients, providing valuable understanding into the possible mechanisms involved in the expansion of CD4-CTLs in AD.
Collapse
Affiliation(s)
- Jiongxue Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Fuyin Deng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Sitai Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Xinying Guo
- Department of Anesthesiology, Guangzhou Women and Children Medical Center, Guangzhou 510623, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Epigenetics and Gene Regulation of Malignant Tumors, Sun Yat-sen Memorial Hospital, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
27
|
Yuan S, Liu Q, Huang X, Tan S, Bai Z, Yu J, Lei F, Le H, Ye Q, Peng X, Yang J, Ling Y, Lyu J. Development of an individualized dementia risk prediction model using deep learning survival analysis incorporating genetic and environmental factors. Alzheimers Res Ther 2024; 16:278. [PMID: 39736679 DOI: 10.1186/s13195-024-01663-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: 10/13/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND Dementia is a major public health challenge in modern society. Early detection of high-risk dementia patients and timely intervention or treatment are of significant clinical importance. Neural network survival analysis represents the most advanced technology for survival analysis to date. However, there is a lack of deep learning-based survival analysis models that integrate both genetic and clinical factors to develop and validate individualized dynamic dementia risk prediction models. METHODS AND RESULTS This study is based on a large prospective cohort from the UK Biobank, which includes a total of 41,484 participants with an average follow-up period of 12.6 years. Initially, 364 candidate features (predictor variables) were screened. The top 30 key features were then identified by ranking the importance of each predictor variable using the Gradient Boosting Machine (GBM) model. A multi-model comparison strategy was employed to evaluate the predictive performance of four survival analysis models: DeepSurv, DeepHit, Kaplan-Meier estimation, and the Cox proportional hazards model (CoxPH). The results showed that the average Harrell's C-index for the DeepSurv model was 0.743, for the DeepHit model it was 0.633, for the CoxPH model it was 0.749, and for the Kaplan-Meier estimator model it was 0.500. In addition, the average D-Calibration Survival Measure was 6.014, 4408.086, 32274.743, and 1.508, respectively. The Brier score (BS) was used to assess the importance of features for the DeepSurv dementia prediction model, and the relationship between features and dementia was visualized using a partial dependence plot (PDP). To facilitate further research, the team deployed the DeepSurv dementia prediction model on AliCloud servers and designated it as the UKB-DementiaPre Tool. CONCLUSION This study successfully developed and validated the DeepSurv dementia prediction model for individuals aged 60 years and above, integrating both genetic and clinical data. The model was then deployed on AliCloud servers to promote its clinical translation. It is anticipated that this prediction model will provide more accurate decision support for clinical treatment and will serve as a valuable tool for the primary prevention of dementia.
Collapse
Affiliation(s)
- Shiqi Yuan
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Qing Liu
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Xiaxuan Huang
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Shanyuan Tan
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Zihong Bai
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China
| | - Juan Yu
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Fazhen Lei
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Huan Le
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Qingqing Ye
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Xiaoxue Peng
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Juying Yang
- Department of Neurology, The Second People's Hospital of Guiyang (The Affiliated Jinyang Hospital of Guizhou Medical University), Guiyang, Guizhou Province, 550000, China
| | - Yitong Ling
- Department of Neurology, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China.
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, No.613, Huangpu Road West, Guangzhou, Guangdong Province, 510630, China.
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|
28
|
Li T, Fili M, Mohammadiarvejeh P, Dawson A, Hu G, Willette AA. Associations of Coffee and Tea Consumption on Neural Network Connectivity: Unveiling the Role of Genetic Factors in Alzheimer's Disease Risk. Nutrients 2024; 16:4303. [PMID: 39770924 PMCID: PMC11677865 DOI: 10.3390/nu16244303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Coffee and tea are widely consumed beverages, but their long-term effects on cognitive function and aging remain largely unexplored. Lifestyle interventions, particularly dietary habits, offer promising strategies for enhancing cognitive performance and preventing cognitive decline. METHODS This study utilized data from the UK Biobank cohort (n = 12,025) to examine the associations between filtered coffee, green tea, and standard tea consumption and neural network functional connectivity across seven resting-state networks. We focused on networks spanning prefrontal and occipital areas that are linked to complex cognitive and behavioral functions. Linear mixed models were used to assess the main effects of coffee and tea consumption, as well as their interactions with Apolipoprotein E (APOE) genetic risk-the strongest genetic risk factor for Alzheimer's disease (AD). RESULTS Higher filtered coffee consumption was associated with increased functional connectivity in several networks, including Motor Execution, Sensorimotor, Fronto-Cingular, and a Prefrontal + 'What' Pathway Network. Similarly, greater green tea intake was associated with enhanced connectivity in the Extrastriate Visual and Primary Visual Networks. In contrast, higher standard tea consumption was linked to reduced connectivity in networks such as Memory Consolidation, Motor Execution, Fronto-Cingular, and the "What" Pathway + Prefrontal Network. The APOE4 genotype and family history of AD influenced the relationship between coffee intake and connectivity in the Memory Consolidation Network. Additionally, the APOE4 genotype modified the association between standard tea consumption and connectivity in the Sensorimotor Network. CONCLUSIONS The distinct patterns of association between coffee, green tea, and standard tea consumption and resting-state brain activity may provide insights into AD-related brain changes. The APOE4 genotype, in particular, appears to play a significant role in modulating these relationships. These findings enhance our knowledge of how commonly consumed beverages may influence cognitive function and potentially AD risk among older adults.
Collapse
Affiliation(s)
- Tianqi Li
- Genetics and Genomics Program, Iowa State University, Ames, IA 50011, USA;
| | - Mohammad Fili
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
| | - Parvin Mohammadiarvejeh
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
- Department of Industrial and Manufacturing Systems Engineering, Iowa State University, Ames, IA 50011, USA
| | - Alice Dawson
- Chestnut Health Systems, Lighthouse Institute, Chicago, IL 60610, USA;
| | - Guiping Hu
- School of Industrial Engineering and Management, Oklahoma State University, Stillwater, OK 74078, USA; (M.F.); (P.M.); (G.H.)
| | - Auriel A. Willette
- Department of Neurology, Rutgers University, New Brunswick, NJ 08854, USA
| |
Collapse
|
29
|
Kuzma A, Valladares O, Greenfest-Allen E, Nicaretta H, Kirsch M, Ren Y, Katanic Z, White H, Wilk A, Bass L, Brettschneider J, Carter L, Cifello J, Chuang WH, Clark K, Gangadharan P, Haut J, Ho PC, Horng W, Iqbal T, Jin Y, Keskinen P, Rose AL, Moon MK, Manuel J, Qu L, Robbins F, Saravanan N, Sha J, Tate S, Zhao Y, Cantwell L, Gardner J, Chou SY, Tzeng JY, Bush W, Naj A, Kuksa P, Lee WP, Leung YY, Schellenberg G, Wang LS. NIAGADS: A Comprehensive National Data Repository for Alzheimer's Disease and Related Dementia Genetics and Genomics Research. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.07.24315029. [PMID: 39417134 PMCID: PMC11483014 DOI: 10.1101/2024.10.07.24315029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
NIAGADS is the National Institute on Aging (NIA) designated national data repository for human genetics research on Alzheimer's Disease and related dementia (ADRD). NIAGADS maintains a high-quality data collection for ADRD genetic/genomic research and supports genetics data production and analysis. NIAGADS hosts whole genome and exome sequence data from the Alzheimer's Disease Sequencing Project (ADSP) and other genotype/phenotype data, encompassing 209,000 samples. NIAGADS shares these data with hundreds of research groups around the world via the Data Sharing Service, a FISMA moderate compliant cloud-based platform that fully supports the NIH Genome Data Sharing Policy. NIAGADS Open Access consists of multiple knowledge bases with genome-wide association summary statistics and rich annotations on the biological significance of genetic variants and genes across the human genome. NIAGADS stands as a keystone in promoting collaborations to advance the understanding and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Amanda Kuzma
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Otto Valladares
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily Greenfest-Allen
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Heather Nicaretta
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Maureen Kirsch
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Youli Ren
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zivadin Katanic
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Heather White
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Andrew Wilk
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lauren Bass
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jascha Brettschneider
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Luke Carter
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jeffrey Cifello
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wei-Hsuan Chuang
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kaylyn Clark
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Prabhakaran Gangadharan
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jacob Haut
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pei-Chuan Ho
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenhwai Horng
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Taha Iqbal
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yumi Jin
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter Keskinen
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexis Lerro Rose
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michelle K Moon
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph Manuel
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liming Qu
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Flawless Robbins
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Naveensri Saravanan
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jin Sha
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sam Tate
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yi Zhao
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Laura Cantwell
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jake Gardner
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Shin-Yi Chou
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Economics, Lehigh University, Bethlehem, PA, United States
- National Bureau of Economic Research, Cambridge, MA, United States
| | - Jung-Ying Tzeng
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Bioinformatics Research Center, North Carolina State University, NC, USA
- Department of Statistics, North Carolina State University, NC, USA
| | - William Bush
- Cleveland Institute for Computational Biology, Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adam Naj
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pavel Kuksa
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wan-Ping Lee
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuk Yee Leung
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Gerard Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
30
|
Pedersen EM, Wimberley T, Vilhjálmsson BJ. A cautionary tale for Alzheimer's disease GWAS by proxy. Nat Genet 2024; 56:2590-2591. [PMID: 39623102 DOI: 10.1038/s41588-024-02023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Affiliation(s)
- Emil M Pedersen
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Theresa Wimberley
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark
| | - Bjarni J Vilhjálmsson
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark.
- Bioinformatics Research Centre, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
- The Novo Nordisk Foundation Centre for Genomics Mechanisms of Disease, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
31
|
Wu Y, Sun Z, Zheng Q, Miao J, Dorn S, Mukherjee S, Fletcher JM, Lu Q. Pervasive biases in proxy genome-wide association studies based on parental history of Alzheimer's disease. Nat Genet 2024; 56:2696-2703. [PMID: 39496879 PMCID: PMC11929606 DOI: 10.1038/s41588-024-01963-9] [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: 10/26/2023] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
Almost every recent Alzheimer's disease (AD) genome-wide association study (GWAS) has performed meta-analysis to combine studies with clinical diagnosis of AD with studies that use proxy phenotypes based on parental disease history. Here, we report major limitations in current GWAS-by-proxy (GWAX) practices due to uncorrected survival bias and nonrandom participation in parental illness surveys, which cause substantial discrepancies between AD GWAS and GWAX results. We demonstrate that the current AD GWAX provide highly misleading genetic correlations between AD risk and higher education, which subsequently affects a variety of genetic epidemiological applications involving AD and cognition. Our study sheds light on potential issues in the design and analysis of middle-aged biobank cohorts and underscores the need for caution when interpreting genetic association results based on proxy-reported parental disease history.
Collapse
Affiliation(s)
- Yuchang Wu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongxuan Sun
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Qinwen Zheng
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Stephen Dorn
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Jason M Fletcher
- Department of Population Health Sciences, University of Wisconsin-Madison, Madison, WI, USA
- La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Ghose U, Sproviero W, Winchester L, Amin N, Zhu T, Newby D, Ulm BS, Papathanasiou A, Shi L, Liu Q, Fernandes M, Adams C, Albukhari A, Almansouri M, Choudhry H, van Duijn C, Nevado-Holgado A. Genome-wide association neural networks identify genes linked to family history of Alzheimer's disease. Brief Bioinform 2024; 26:bbae704. [PMID: 39775791 PMCID: PMC11707606 DOI: 10.1093/bib/bbae704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Augmenting traditional genome-wide association studies (GWAS) with advanced machine learning algorithms can allow the detection of novel signals in available cohorts. We introduce "genome-wide association neural networks (GWANN)" a novel approach that uses neural networks (NNs) to perform a gene-level association study with family history of Alzheimer's disease (AD). In UK Biobank, we defined cases (n = 42 110) as those with AD or family history of AD and sampled an equal number of controls. The data was split into an 80:20 ratio of training and testing samples, and GWANN was trained on the former followed by identifying associated genes using its performance on the latter. Our method identified 18 genes to be associated with family history of AD. APOE, BIN1, SORL1, ADAM10, APH1B, and SPI1 have been identified by previous AD GWAS. Among the 12 new genes, PCDH9, NRG3, ROR1, LINGO2, SMYD3, and LRRC7 have been associated with neurofibrillary tangles or phosphorylated tau in previous studies. Furthermore, there is evidence for differential transcriptomic or proteomic expression between AD and healthy brains for 10 of the 12 new genes. A series of post hoc analyses resulted in a significantly enriched protein-protein interaction network (P-value < 1 × 10-16), and enrichment of relevant disease and biological pathways such as focal adhesion (P-value = 1 × 10-4), extracellular matrix organization (P-value = 1 × 10-4), Hippo signaling (P-value = 7 × 10-4), Alzheimer's disease (P-value = 3 × 10-4), and impaired cognition (P-value = 4 × 10-3). Applying NNs for GWAS illustrates their potential to complement existing algorithms and methods and enable the discovery of new associations without the need to expand existing cohorts.
Collapse
Affiliation(s)
- Upamanyu Ghose
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
| | - William Sproviero
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Laura Winchester
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Najaf Amin
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Taiyu Zhu
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Centre for Statistics in Medicine, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Brittany S Ulm
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Department of Translational Medicine, Nxera Pharma UK Limited, Cambridge, United Kingdom
| | - Qiang Liu
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- School of Engineering Mathematics and Technology University of Bristol, Ada Lovelace Building, Bristol, United Kingdom
| | - Marco Fernandes
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Cassandra Adams
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ashwag Albukhari
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majid Almansouri
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hani Choudhry
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Cornelia van Duijn
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Alejo Nevado-Holgado
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- King Abdulaziz University and the University of Oxford Centre for Artificial Intelligence in Precision Medicine (KO-CAIPM), Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Pugalenthi PV, He B, Xie L, Nho K, Saykin AJ, Yan J. Deciphering the tissue-specific functional effect of Alzheimer risk SNPs with deep genome annotation. BioData Min 2024; 17:50. [PMID: 39538253 PMCID: PMC11558841 DOI: 10.1186/s13040-024-00400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a highly heritable brain dementia, along with substantial failure of cognitive function. Large-scale genome-wide association studies (GWASs) have led to a set of SNPs significantly associated with AD and related traits. GWAS hits usually emerge as clusters where a lead SNP with the highest significance is surrounded by other less significant neighboring SNPs. Although functionality is not guaranteed even with the strongest associations in GWASs, lead SNPs have historically been the focus of the field, with the remaining associations inferred to be redundant. Recent deep genome annotation tools enable the prediction of function from a segment of a DNA sequence with significantly improved precision, which allows in-silico mutagenesis to interrogate the functional effect of SNP alleles. In this project, we explored the impact of top AD GWAS hits around APOE region on chromatin functions and whether it will be altered by the genetic context (i.e., alleles of neighboring SNPs). Our results showed that highly correlated SNPs in the same LD block could have distinct impacts on downstream functions. Although some GWAS lead SNPs showed dominant functional effects regardless of the neighborhood SNP alleles, several other SNPs did exhibit enhanced loss or gain of function under certain genetic contexts, suggesting potential additional information hidden in the LD blocks.
Collapse
Affiliation(s)
- Pradeep Varathan Pugalenthi
- Department of Biomedical Engineering and Informatics, Indiana University Indianapolis, 420 University Blvd, Indianapolis, IN, 46202, USA
| | - Bing He
- Department of Biomedical Engineering and Informatics, Indiana University Indianapolis, 420 University Blvd, Indianapolis, IN, 46202, USA
| | - Linhui Xie
- Department of Electrical and Computer Engineering, Purdue University Indianapolis, 420 University Blvd, Indianapolis, IN, 46202, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 University Blvd, Indianapolis, IN, 46202, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 University Blvd, Indianapolis, IN, 46202, USA
| | - Jingwen Yan
- Department of Biomedical Engineering and Informatics, Indiana University Indianapolis, 420 University Blvd, Indianapolis, IN, 46202, USA.
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 University Blvd, Indianapolis, IN, 46202, USA.
| |
Collapse
|
34
|
Jeon S, Salvo MA, Alia AO, Popovic J, Zagardo M, Chandra S, Nassan M, Gate D, Vassar R, Cuddy LK. Neuronal ACE1 knockout disrupts the hippocampal renin angiotensin system leading to memory impairment and vascular loss in normal aging. Neurobiol Dis 2024; 202:106729. [PMID: 39515529 DOI: 10.1016/j.nbd.2024.106729] [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: 06/18/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Angiotensin I converting enzyme (ACE1) maintains blood pressure homeostasis by converting angiotensin I into angiotensin II in the renin-angiotensin system (RAS). ACE1 is expressed in the brain, where an intrinsic RAS regulates complex cognitive functions including learning and memory. ACE1 has been implicated in neurodegenerative disorders including Alzheimer's disease and Parkinson's disease, but the mechanisms remain incompletely understood. Here, we performed single-nucleus RNA sequencing to characterize the expression of RAS genes in the hippocampus and discovered that Ace is mostly expressed in CA1 region excitatory neurons. To gain a deeper understanding of the function of neuronal ACE1, we generated ACE1 conditional knockout (cKO) mice lacking ACE1 expression specifically in hippocampal and cortical excitatory neurons. ACE1 cKO mice exhibited hippocampus-dependent memory impairment in the Morris water maze, y-maze, and fear conditioning tests. Total ACE1 level was significantly reduced in the cortex and hippocampus of ACE1 cKO mice showing that excitatory neurons are the predominant cell type expressing ACE1 in the forebrain. Despite similar reductions in total ACE1 level in both the hippocampus and cortex, the RAS pathway was dysregulated in the hippocampus only. Importantly, ACE1 cKO mice exhibited age-related capillary loss selectively in the hippocampus. Here, we show selective vulnerability of the hippocampal microvasculature and RAS pathway to neuronal ACE1 knockout. Our results provide important insights into the function of ACE1 in the brain and demonstrate a connection between neuronal ACE1 and cerebrovascular function in the hippocampus.
Collapse
Affiliation(s)
- Sohee Jeon
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Miranda A Salvo
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Alia O Alia
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Jelena Popovic
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Mitchell Zagardo
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Sidhanth Chandra
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Malik Nassan
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - David Gate
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Robert Vassar
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| | - Leah K Cuddy
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States.
| |
Collapse
|
35
|
Salas IH, Paumier A, Tao T, Derevyanko A, Switzler C, Burgado J, Movsesian M, Metanat S, Dawoodtabar T, Asbell Q, Fassihi A, Allen NJ. Astrocyte transcriptomic analysis identifies glypican 5 downregulation as a contributor to synaptic dysfunction in Alzheimer's disease models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621182. [PMID: 39554197 PMCID: PMC11565880 DOI: 10.1101/2024.10.30.621182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Synaptic dysfunction is an early feature in Alzheimer's disease (AD) and correlates with cognitive decline. Astrocytes are essential regulators of synapses, impacting synapse formation, maturation, elimination and function. To understand if synapse-supportive functions of astrocytes are altered in AD, we used astrocyte BacTRAP mice to generate a comprehensive dataset of hippocampal astrocyte transcriptional alterations in two mouse models of Alzheimer's pathology (APPswe/PS1dE9 and Tau P301S), characterizing sex and age-dependent changes. We found that astrocytes from both models downregulate genes important for synapse regulation and function such as the synapse-maturation factor Glypican 5. This transcriptional signature is shared with human post-mortem AD patients. Manipulating a key component of this signature by in vivo overexpression of Glypican 5 in astrocytes is sufficient to prevent early synaptic dysfunction and improve spatial learning in APPswe/PS1dE9 mice. These findings open new avenues to target astrocytic factors to mitigate AD synaptic dysfunction.
Collapse
|
36
|
Reus LM, Jansen IE, Tijms BM, Visser PJ, Tesi N, van der Lee SJ, Vermunt L, Peeters CFW, De Groot LA, Hok-A-Hin YS, Chen-Plotkin A, Irwin DJ, Hu WT, Meeter LH, van Swieten JC, Holstege H, Hulsman M, Lemstra AW, Pijnenburg YAL, van der Flier WM, Teunissen CE, del Campo Milan M. Connecting dementia risk loci to the CSF proteome identifies pathophysiological leads for dementia. Brain 2024; 147:3522-3533. [PMID: 38527854 PMCID: PMC11449142 DOI: 10.1093/brain/awae090] [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: 09/04/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
Genome-wide association studies have successfully identified many genetic risk loci for dementia, but exact biological mechanisms through which genetic risk factors contribute to dementia remains unclear. Integrating CSF proteomic data with dementia risk loci could reveal intermediate molecular pathways connecting genetic variance to the development of dementia. We tested to what extent effects of known dementia risk loci can be observed in CSF levels of 665 proteins [proximity extension-based (PEA) immunoassays] in a deeply-phenotyped mixed memory clinic cohort [n = 502, mean age (standard deviation, SD) = 64.1 (8.7) years, 181 female (35.4%)], including patients with Alzheimer's disease (AD, n = 213), dementia with Lewy bodies (DLB, n = 50) and frontotemporal dementia (FTD, n = 93), and controls (n = 146). Validation was assessed in independent cohorts (n = 99 PEA platform, n = 198, mass reaction monitoring-targeted mass spectroscopy and multiplex assay). We performed additional analyses stratified according to diagnostic status (AD, DLB, FTD and controls separately), to explore whether associations between CSF proteins and genetic variants were specific to disease or not. We identified four AD risk loci as protein quantitative trait loci (pQTL): CR1-CR2 (rs3818361, P = 1.65 × 10-8), ZCWPW1-PILRB (rs1476679, P = 2.73 × 10-32), CTSH-CTSH (rs3784539, P = 2.88 × 10-24) and HESX1-RETN (rs186108507, P = 8.39 × 10-8), of which the first three pQTLs showed direct replication in the independent cohorts. We identified one AD-specific association between a rare genetic variant of TREM2 and CSF IL6 levels (rs75932628, P = 3.90 × 10-7). DLB risk locus GBA showed positive trans effects on seven inter-related CSF levels in DLB patients only. No pQTLs were identified for FTD loci, either for the total sample as for analyses performed within FTD only. Protein QTL variants were involved in the immune system, highlighting the importance of this system in the pathophysiology of dementia. We further identified pQTLs in stratified analyses for AD and DLB, hinting at disease-specific pQTLs in dementia. Dissecting the contribution of risk loci to neurobiological processes aids in understanding disease mechanisms underlying dementia.
Collapse
Affiliation(s)
- Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA 90095 CA, USA
| | - Iris E Jansen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Niccoló Tesi
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Lisa Vermunt
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Carel F W Peeters
- Mathematical and Statistical Methods group (Biometris), Wageningen University and Research, Wageningen, 6708 PB Wageningen, The Netherlands
| | - Lisa A De Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Rutgers-RWJ Medical School, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Marc Hulsman
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Marta del Campo Milan
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, 28003 Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08005 Barcelona, Spain
| |
Collapse
|
37
|
Koetsier J, Cavill R, Reijnders R, Harvey J, Homann J, Kouhsar M, Deckers K, Köhler S, Eijssen LMT, van den Hove DLA, Demuth I, Düzel S, for the Alzheimer's Disease Neuroimaging Initiative, Smith RG, Smith AR, Burrage J, Walker EM, Shireby G, Hannon E, Dempster E, Frayling T, Mill J, Dobricic V, Johannsen P, Wittig M, Franke A, Vandenberghe R, Schaeverbeke J, Freund‐Levi Y, Frölich L, Scheltens P, Teunissen CE, Frisoni G, Blin O, Richardson JC, Bordet R, Engelborghs S, de Roeck E, Martinez‐Lage P, Tainta M, Lleó A, Sala I, Popp J, Peyratout G, Verhey F, Tsolaki M, Andreasson U, Blennow K, Zetterberg H, Streffer J, Vos SJB, Lovestone S, Visser P, Lill CM, Bertram L, Lunnon K, Pishva E. Blood-based multivariate methylation risk score for cognitive impairment and dementia. Alzheimers Dement 2024; 20:6682-6698. [PMID: 39193899 PMCID: PMC11633365 DOI: 10.1002/alz.14061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION The established link between DNA methylation and pathophysiology of dementia, along with its potential role as a molecular mediator of lifestyle and environmental influences, positions blood-derived DNA methylation as a promising tool for early dementia risk detection. METHODS In conjunction with an extensive array of machine learning techniques, we employed whole blood genome-wide DNA methylation data as a surrogate for 14 modifiable and non-modifiable factors in the assessment of dementia risk in independent dementia cohorts. RESULTS We established a multivariate methylation risk score (MMRS) for identifying mild cognitive impairment cross-sectionally, independent of age and sex (P = 2.0 × 10-3). This score significantly predicted the prospective development of cognitive impairments in independent studies of Alzheimer's disease (hazard ratio for Rey's Auditory Verbal Learning Test (RAVLT)-Learning = 2.47) and Parkinson's disease (hazard ratio for MCI/dementia = 2.59). DISCUSSION Our work shows the potential of employing blood-derived DNA methylation data in the assessment of dementia risk. HIGHLIGHTS We used whole blood DNA methylation as a surrogate for 14 dementia risk factors. Created a multivariate methylation risk score for predicting cognitive impairment. Emphasized the role of machine learning and omics data in predicting dementia. The score predicts cognitive impairment development at the population level.
Collapse
Affiliation(s)
- Jarno Koetsier
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences (DACS)Faculty of Science and Engineering (FSE)Maastricht UniversityMaastrichtThe Netherlands
| | - Rick Reijnders
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Joshua Harvey
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Jan Homann
- Institute of Epidemiology and Social MedicineUniversity of MünsterMünsterGermany
| | - Morteza Kouhsar
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Kay Deckers
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Lars M. T. Eijssen
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
- Department of Bioinformatics ‐ BiGCaTResearch Institute of Nutrition and Translational Research in Metabolism (NUTRIM)Faculty of HealthMedicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Daniel L. A. van den Hove
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
- Department of PsychiatryPsychosomatics and Psychotherapy, University of Wuerzburg, Dr. Manuel NagelWürzburgGermany
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism)Charité – Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- BCRT ‐ Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| | - Sandra Düzel
- Center for Lifespan Psychology, Max Planck Institute for Human DevelopmentBerlinGermany
| | | | - Rebecca G. Smith
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Adam R. Smith
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Joe Burrage
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Emma M. Walker
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Gemma Shireby
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Eilis Hannon
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Emma Dempster
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Tim Frayling
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Jonathan Mill
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of LübeckLübeckGermany
| | - Peter Johannsen
- Danish Dementia Research Centre, RigshospitaletCopenhagenDenmark
| | - Michael Wittig
- Institute of Clinical Molecular Biology, Christian‐Albrechts‐University of KielKielGermany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian‐Albrechts‐University of KielKielGermany
| | | | | | - Yvonne Freund‐Levi
- Department of Clinical Science and EducationSödersjukhuset, Karolinska InstitutetStockholmSweden
- School of Medical SciencesÖrebro UniversityÖrebroSweden
- Department of GeriatricsSödertälje HospitalSödertäljeSweden
| | - Lutz Frölich
- Department of Geriatric PsychiatryCentral Institute of Mental Health; Medical Faculty Mannheim/Heidelberg UniversityMannheimGermany
| | - Philip Scheltens
- Department of NeurologyAlzheimer Center AmsterdamAmsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry LaboratoryDepartment of Laboratory MedicineAmsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMCAmsterdamThe Netherlands
| | - Giovanni Frisoni
- Memory CenterGeneva University and University Hospitals; on behalf of the AMYPAD ConsortiumGenèveSwitzerland
| | | | - Jill C. Richardson
- Neuroscience Therapeutic Area, GlaxoSmithKline R&DStevenageHertfordshireUK
| | | | - Sebastiaan Engelborghs
- Department of Biomedical SciencesUniversity of AntwerpAntwerpenBelgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB)Jette, BrusselsBelgium
| | - Ellen de Roeck
- Department of Biomedical SciencesUniversity of AntwerpAntwerpenBelgium
| | - Pablo Martinez‐Lage
- Center for Research and Advanced TherapiesCITA‐Alzheimer FoundationGipuzkoaSpain
| | - Mikel Tainta
- Center for Research and Advanced TherapiesCITA‐Alzheimer FoundationGipuzkoaSpain
| | - Alberto Lleó
- Neurology DepartmentCentro de Investigación en Red en enfermedades neurodegenerativas (CIBERNED), Hospital Sant Pau, Sant Antoni Maria ClaretBarcelonaSpain
| | - Isabel Sala
- Neurology DepartmentCentro de Investigación en Red en enfermedades neurodegenerativas (CIBERNED), Hospital Sant Pau, Sant Antoni Maria ClaretBarcelonaSpain
| | - Julius Popp
- University Hospital of Psychiatry ZürichUniversity of ZürichZürichSwitzerland
| | - Gwendoline Peyratout
- Department of PsychiatryUniversity Hospital of Lausanne (CHUV)LausanneSwitzerland
| | - Frans Verhey
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Magda Tsolaki
- 1st Department of NeurologySchool of Medicine`Laboratory of Neurodegenerative DiseasesCenter for Interdisciplinary Research and InnovationAristotle University of Thessaloniki, and Alzheimer Hellas, Macedonia, Balkan CenterThessalonikiGreece
| | - Ulf Andreasson
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
| | - Kaj Blennow
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Clinical Neurochemistry LabSahlgrenska University HospitalGöteborgSweden
- Paris Brain InstituteICM, Pitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiP.R. China
| | - Henrik Zetterberg
- Institute of Neuroscience and PhysiologyDepartment of Psychiatry and NeurochemistryThe Sahlgrenska Academy at University of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCL, Maple HouseLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayShatin, N.T.Hong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Johannes Streffer
- AC Immune SA, formerly Janssen R&D, LLC. Beerse, Belgium at the time of study conductLausanneSwitzerland
| | - Stephanie J. B. Vos
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
| | - Simon Lovestone
- University of OxfordOxford, United Kingdom; Currently at Johnson & Johnson Innovative MedicinesBeerseBelgium
| | - Pieter‐Jelle Visser
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
- Department of GeriatricsSödertälje HospitalSödertäljeSweden
| | - Christina M. Lill
- Institute of Epidemiology and Social MedicineUniversity of MünsterMünsterGermany
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College, South Kensington CampusLondonUK
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of LübeckLübeckGermany
| | - Katie Lunnon
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Ehsan Pishva
- Department of Psychiatry and NeuropsychologySchool for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht UniversityMaastrichtThe Netherlands
- Medical SchoolFaculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
38
|
Ling Y, Yuan S, Huang X, Tan S, Cheng H, Li L, Li S, Huang L, Xu A, Lyu J. Association between probable sarcopenia and dementia risk: a prospective cohort study with mediation analysis. Transl Psychiatry 2024; 14:398. [PMID: 39353910 PMCID: PMC11445531 DOI: 10.1038/s41398-024-03131-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
The role of alcohol consumption as a mediator in the risk between sarcopenia and dementia remains inadequately studied. There is currently limited research on whether the association between sarcopenia and the risk of Alzheimer's disease (AD) is influenced by genetic susceptibility. Our study incorporated a total of 483,637 baseline non-dementia participants, who were classified into groups of individuals with no sarcopenia and those with probable sarcopenia based on the definition. We employed Cox proportional hazards models to evaluate the association between probable sarcopenia and the risk of all cause dementia (ACD), AD, and vascular dementia (VD). We conducted mediation analysis to explore the role of alcohol consumption in the association between probable sarcopenia and the risk of ACD, AD, and VD. During the median follow-up period of 13.6 years, we documented 9000 new cases of ACD (including 4061 AD and 2025 VD). Fully adjusted multivariate model revealed a significant correlation between probable sarcopenia and elevated risk for ACD (HR = 1.54, 95% CI: 1.46-1.62, p < 0.001), AD (HR = 1.32, 95% CI: 1.21-1.43, p < 0.001), and VD (HR = 1.69, 95% CI: 1.52-1.89, p < 0.001). Mediation analysis elucidates that alcohol consumption explained 12.8%, 15.2%, and 11.1% of the associations of probable sarcopenia with the risk of ACD, AD, and VD, respectively. An interactive relationship prevails between probable sarcopenia and genetic factors (p for interaction <0.001), and regardless of the degree of genetic risk, probable sarcopenia correlates with an elevated AD risk. Our study reveals a significant association between probable sarcopenia and an increased risk of dementia, with alcohol consumption playing a mediating role in this association. There is an interaction between probable sarcopenia and genetic susceptibility related to the risk of AD.
Collapse
Affiliation(s)
- Yitong Ling
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shiqi Yuan
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiaxuan Huang
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shanyuan Tan
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Hongtao Cheng
- School of Nursing, Jinan University, Guangzhou, Guangdong, China
| | - Li Li
- Department of Clinical Research, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shuna Li
- Department of Clinical Research, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Liying Huang
- Department of Clinical Research, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Anding Xu
- Department of Neurology, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Jun Lyu
- Department of Clinical Research, Jinan University First Affiliated Hospital, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China.
| |
Collapse
|
39
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
40
|
Kim YS, Choi SH, Kim KY, Navia-Pelaez JM, Perkins GA, Choi S, Kim J, Nazarenkov N, Rissman RA, Ju WK, Ellisman MH, Miller YI. AIBP controls TLR4 inflammarafts and mitochondrial dysfunction in a mouse model of Alzheimer's disease. J Neuroinflammation 2024; 21:245. [PMID: 39342323 PMCID: PMC11439205 DOI: 10.1186/s12974-024-03214-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024] Open
Abstract
Microglia-driven neuroinflammation plays an important role in the development of Alzheimer's disease. Microglia activation is accompanied by the formation and chronic expression of TLR4 inflammarafts, defined as enlarged and cholesterol-rich lipid rafts serving as an assembly platform for TLR4 dimers and complexes of other inflammatory receptors. The secreted apoA-I binding protein (APOA1BP or AIBP) binds TLR4 and selectively targets cholesterol depletion machinery to TLR4 inflammaraft-expressing inflammatory, but not homeostatic microglia. Here we demonstrated that amyloid-beta (Aβ) induced formation of TLR4 inflammarafts in microglia in vitro and in the brain of APP/PS1 mice. Mitochondria in Apoa1bp-/- APP/PS1 microglia were hyperbranched and cupped, which was accompanied by increased reactive oxygen species and the dilated endoplasmic reticulum. The size and number of Aβ plaques and neuronal cell death were significantly increased, and the animal survival was decreased in Apoa1bp-/-APP/PS1 compared to APP/PS1 female mice. These results suggest that AIBP exerts control of TLR4 inflammarafts and mitochondrial dynamics in microglia and plays a protective role in Alzheimer's disease associated oxidative stress and neurodegeneration.
Collapse
Affiliation(s)
- Yi Sak Kim
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Juliana M Navia-Pelaez
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Seunghwan Choi
- Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center and Shiley Eye Institute, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Jungsu Kim
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Nicolaus Nazarenkov
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Won-Kyu Ju
- Viterbi Family Department of Ophthalmology, Hamilton Glaucoma Center and Shiley Eye Institute, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, 92093, USA
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
41
|
Joseph PV, Abbas M, Goodney G, Diallo A, Gaye A. Genomic study of taste perception genes in African Americans reveals SNPs linked to Alzheimer's disease. Sci Rep 2024; 14:21560. [PMID: 39284855 PMCID: PMC11405524 DOI: 10.1038/s41598-024-71669-9] [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: 03/05/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
While previous research has shown the potential links between taste perception pathways and brain-related conditions, the area involving Alzheimer's disease remains incompletely understood. Taste perception involves neurotransmitter signaling, including serotonin, glutamate, and dopamine. Disruptions in these pathways are implicated in neurodegenerative diseases. The integration of olfactory and taste signals in flavor perception may impact brain health, evident in olfactory dysfunction as an early symptom in neurodegenerative conditions. Shared immune response and inflammatory pathways may contribute to the association between altered taste perception and conditions like neurodegeneration, present in Alzheimer's disease. This study consists of an exploration of expression-quantitative trait loci (eQTL), utilizing whole-blood transcriptome profiles, of 28 taste perception genes, from a combined cohort of 475 African American subjects. This comprehensive dataset was subsequently intersected with single-nucleotide polymorphisms (SNPs) identified in Genome-Wide Association Studies (GWAS) of Alzheimer's Disease (AD). Finally, the investigation delved into assessing the association between eQTLs reported in GWAS of AD and the profiles of 741 proteins from the Olink Neurological Panel. The eQTL analysis unveiled 3,547 statistically significant SNP-Gene associations, involving 412 distinct SNPs that spanned all 28 taste genes. In 17 GWAS studies encompassing various traits, a total of 14 SNPs associated with 12 genes were identified, with three SNPs consistently linked to Alzheimer's disease across four GWAS studies. All three SNPs demonstrated significant associations with the down-regulation of TAS2R41, and two of them were additionally associated with the down-regulation of TAS2R60. In the subsequent pQTL analysis, two of the SNPs linked to TAS2R41 and TAS2R60 genes (rs117771145 and rs10228407) were correlated with the upregulation of two proteins, namely EPHB6 and ADGRB3. Our investigation introduces a new perspective to the understanding of Alzheimer's disease, emphasizing the significance of bitter taste receptor genes in its pathogenesis. These discoveries set the stage for subsequent research to delve into these receptors as promising avenues for both intervention and diagnosis. Nevertheless, the translation of these genetic insights into clinical practice requires a more profound understanding of the implicated pathways and their pertinence to the disease's progression across diverse populations.
Collapse
Affiliation(s)
- Paule Valery Joseph
- Sensory Science and Metabolism Unit, Biobehavioral Branch, National Institute On Alcohol Abuse and Alcoholism, National Institue of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Malak Abbas
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gabriel Goodney
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ana Diallo
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, School of Graduate Studies, Meharry Medical College, Nashville, TN, USA.
| |
Collapse
|
42
|
Hu T, Parrish RL, Dai Q, Buchman AS, Tasaki S, Bennett DA, Seyfried NT, Epstein MP, Yang J. Omnibus proteome-wide association study identifies 43 risk genes for Alzheimer disease dementia. Am J Hum Genet 2024; 111:1848-1863. [PMID: 39079537 PMCID: PMC11393696 DOI: 10.1016/j.ajhg.2024.07.001] [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: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 09/08/2024] Open
Abstract
Transcriptome-wide association study (TWAS) tools have been applied to conduct proteome-wide association studies (PWASs) by integrating proteomics data with genome-wide association study (GWAS) summary data. The genetic effects of PWAS-identified significant genes are potentially mediated through genetically regulated protein abundance, thus informing the underlying disease mechanisms better than GWAS loci. However, existing TWAS/PWAS tools are limited by considering only one statistical model. We propose an omnibus PWAS pipeline to account for multiple statistical models and demonstrate improved performance by simulation and application studies of Alzheimer disease (AD) dementia. We employ the Aggregated Cauchy Association Test to derive omnibus PWAS (PWAS-O) p values from PWAS p values obtained by three existing tools assuming complementary statistical models-TIGAR, PrediXcan, and FUSION. Our simulation studies demonstrated improved power, with well-calibrated type I error, for PWAS-O over all three individual tools. We applied PWAS-O to studying AD dementia with reference proteomic data profiled from dorsolateral prefrontal cortex of postmortem brains from individuals of European ancestry. We identified 43 risk genes, including 5 not identified by previous studies, which are interconnected through a protein-protein interaction network that includes the well-known AD risk genes TOMM40, APOC1, and APOC2. We also validated causal genetic effects mediated through the proteome for 27 (63%) PWAS-O risk genes, providing insights into the underlying biological mechanisms of AD dementia and highlighting promising targets for therapeutic development. PWAS-O can be easily applied to studying other complex diseases.
Collapse
Affiliation(s)
- Tingyang Hu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Division of Biostatistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Randy L Parrish
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Qile Dai
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA 30322, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Epstein
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jingjing Yang
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
43
|
Hu T, Liu Q, Dai Q, Parrish RL, Buchman AS, Tasaki S, Seyfried NT, Wang Y, Bennett DA, De Jager PL, Epstein MP, Yang J. Proteome-wide association studies using summary pQTL data of three tissues identified 30 risk genes of Alzheimer's disease dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.28.24305044. [PMID: 38585769 PMCID: PMC10996749 DOI: 10.1101/2024.03.28.24305044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background Proteome-wide association study (PWAS) integrating proteomic data with genome-wide association study (GWAS) summary data is a powerful tool for studying Alzheimer's disease (AD) dementia. Existing PWAS analyses of AD often rely on the availability of individual-level proteomic and genetic data of a reference panel. Leveraging summary protein quantitative trait loci (pQTL) reference data of multiple AD-relevant tissues is expected to improve PWAS findings of AD dementia. Methods We conducted PWAS of AD dementia by integrating publicly available summary pQTL data of brain, cerebrospinal fluid (CSF), and plasma tissues, with the latest GWAS summary data of AD dementia. For each target protein per tissue, we employed our recently published OTTERS tool to obtain omnibus PWAS p-value, to test whether the genetically regulated protein abundance in the corresponding tissue is associated with AD dementia. Protein-protein interactions and enriched pathways of identified significant PWAS risk genes were analyzed by STRING. The potential causal effects of these PWAS risk genes were assessed by probabilistic Mendelian randomization analyses. Results We identified 30 unique significant PWAS risk genes for AD dementia, including 11 for brain, 9 for CSF, and 16 for plasma tissues. Four of these were shared by at least two tissues, and gene MAPK3 was found in all three tissues. We found that 11 of these PWAS risk genes were associated with AD or AD pathological hall marks as shown in GWAS Catalog; 18 of these were detected by transcriptome-wide association studies (TWAS); and 25 of these, including 8 out of 9 novel genes, were interconnected within a protein-protein interaction network involving the well-known AD risk gene APOE. Especially, these PWAS risk genes were enriched in immune response, glial cell proliferation, and high-density lipoprotein particle clearance pathways. Mediated causal effects were validated for 13 PWAS risk genes (43.3%). Conclusions Our findings provide novel insights into the genetic mechanisms of AD dementia in brain, CSF, and plasma tissues, and targets for developing therapeutic interventions. We also demonstrated the effectiveness of integrating summary pQTL and GWAS data for mapping risk genes of complex human diseases.
Collapse
Affiliation(s)
- Tingyang Hu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Biostatistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Qiang Liu
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Qile Dai
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Randy L. Parrish
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, 30322, USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Shinya Tasaki
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY10032, USA
| | - Michael P. Epstein
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jingjing Yang
- Center for Computational and Quantitative Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
44
|
The Mega Vascular Cognitive Impairment and Dementia (MEGAVCID) consortium. A genome-wide association meta-analysis of all-cause and vascular dementia. Alzheimers Dement 2024; 20:5973-5995. [PMID: 39046104 PMCID: PMC11497727 DOI: 10.1002/alz.14115] [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: 06/09/2023] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Dementia is a multifactorial disease with Alzheimer's disease (AD) and vascular dementia (VaD) pathologies making the largest contributions. Yet, most genome-wide association studies (GWAS) focus on AD. METHODS We conducted a GWAS of all-cause dementia (ACD) and examined the genetic overlap with VaD. Our dataset includes 800,597 individuals, with 46,902 and 8702 cases of ACD and VaD, respectively. Known AD loci for ACD and VaD were replicated. Bioinformatic analyses prioritized genes that are likely functionally relevant and shared with closely related traits and risk factors. RESULTS For ACD, novel loci identified were associated with energy transport (SEMA4D), neuronal excitability (ANO3), amyloid deposition in the brain (RBFOX1), and magnetic resonance imaging markers of small vessel disease (SVD; HBEGF). Novel VaD loci were associated with hypertension, diabetes, and neuron maintenance (SPRY2, FOXA2, AJAP1, and PSMA3). DISCUSSION Our study identified genetic risks underlying ACD, demonstrating overlap with neurodegenerative processes, vascular risk factors, and cerebral SVD. HIGHLIGHTS We conducted the largest genome-wide association study of all-cause dementia (ACD) and vascular dementia (VaD). Known genetic variants associated with AD were replicated for ACD and VaD. Functional analyses identified novel loci for ACD and VaD. Genetic risks of ACD overlapped with neurodegeneration, vascular risk factors, and cerebral small vessel disease.
Collapse
|
45
|
Iqbal MS, Belal Bin Heyat M, Parveen S, Ammar Bin Hayat M, Roshanzamir M, Alizadehsani R, Akhtar F, Sayeed E, Hussain S, Hussein HS, Sawan M. Progress and trends in neurological disorders research based on deep learning. Comput Med Imaging Graph 2024; 116:102400. [PMID: 38851079 DOI: 10.1016/j.compmedimag.2024.102400] [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: 01/02/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024]
Abstract
In recent years, deep learning (DL) has emerged as a powerful tool in clinical imaging, offering unprecedented opportunities for the diagnosis and treatment of neurological disorders (NDs). This comprehensive review explores the multifaceted role of DL techniques in leveraging vast datasets to advance our understanding of NDs and improve clinical outcomes. Beginning with a systematic literature review, we delve into the utilization of DL, particularly focusing on multimodal neuroimaging data analysis-a domain that has witnessed rapid progress and garnered significant scientific interest. Our study categorizes and critically analyses numerous DL models, including Convolutional Neural Networks (CNNs), LSTM-CNN, GAN, and VGG, to understand their performance across different types of Neurology Diseases. Through particular analysis, we identify key benchmarks and datasets utilized in training and testing DL models, shedding light on the challenges and opportunities in clinical neuroimaging research. Moreover, we discuss the effectiveness of DL in real-world clinical scenarios, emphasizing its potential to revolutionize ND diagnosis and therapy. By synthesizing existing literature and describing future directions, this review not only provides insights into the current state of DL applications in ND analysis but also covers the way for the development of more efficient and accessible DL techniques. Finally, our findings underscore the transformative impact of DL in reshaping the landscape of clinical neuroimaging, offering hope for enhanced patient care and groundbreaking discoveries in the field of neurology. This review paper is beneficial for neuropathologists and new researchers in this field.
Collapse
Affiliation(s)
- Muhammad Shahid Iqbal
- Department of Computer Science and Information Technology, Women University of Azad Jammu & Kashmir, Bagh, Pakistan.
| | - Md Belal Bin Heyat
- CenBRAIN Neurotech Center of Excellence, School of Engineering, Westlake University, Hangzhou, Zhejiang, China.
| | - Saba Parveen
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen 518060, China.
| | | | - Mohamad Roshanzamir
- Department of Computer Engineering, Faculty of Engineering, Fasa University, Fasa, Iran.
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation, Deakin University, VIC 3216, Australia.
| | - Faijan Akhtar
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China.
| | - Eram Sayeed
- Kisan Inter College, Dhaurahara, Kushinagar, India.
| | - Sadiq Hussain
- Department of Examination, Dibrugarh University, Assam 786004, India.
| | - Hany S Hussein
- Electrical Engineering Department, Faculty of Engineering, King Khalid University, Abha 61411, Saudi Arabia; Electrical Engineering Department, Faculty of Engineering, Aswan University, Aswan 81528, Egypt.
| | - Mohamad Sawan
- CenBRAIN Neurotech Center of Excellence, School of Engineering, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
46
|
Oh S, Kim S, Lee JE, Park BY, Hye Won J, Park H. Multimodal analysis of disease onset in Alzheimer's disease using Connectome, Molecular, and genetics data. Neuroimage Clin 2024; 43:103660. [PMID: 39197213 PMCID: PMC11393605 DOI: 10.1016/j.nicl.2024.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Alzheimer's disease (AD) and its related age at onset (AAO) are highly heterogeneous, due to the inherent complexity of the disease. They are affected by multiple factors, such as neuroimaging and genetic predisposition. Multimodal integration of various data types is necessary; however, it has been nontrivial due to the high dimensionality of each modality. We aimed to identify multimodal biomarkers of AAO in AD using an extended version of sparse canonical correlation analysis, in which we integrated two imaging modalities, functional magnetic resonance imaging (fMRI) and positron emission tomography (PET), and genetic data in the form of single-nucleotide polymorphisms (SNPs) obtained from the Alzheimer's disease neuroimaging initiative database. These three modalities cover low-to-high-level complementary information and offer multiscale insights into the AAO. We identified multivariate markers of AAO in AD using fMRI, PET, and SNP. Furthermore, the markers identified were largely consistent with those reported in the existing literature. In particular, our serial mediation analysis suggests that genetic variants influence the AAO in AD by indirectly affecting brain connectivity by mediation of amyloid-beta protein accumulation, supporting a plausible path in existing research. Our approach provides comprehensive biomarkers related to AAO in AD and offers novel multimodal insights into AD.
Collapse
Affiliation(s)
- Sewook Oh
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunghun Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Jong-Eun Lee
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Ji Hye Won
- Department of Computer Engineering, Pukyong National University, Busan, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea.
| |
Collapse
|
47
|
Garcia MF, Retallick-Townsley K, Pruitt A, Davidson E, Dai Y, Fitzpatrick SE, Sen A, Cohen S, Livoti O, Khan S, Dossou G, Cheung J, Deans PJM, Wang Z, Huckins L, Hoffman E, Brennand K. Dynamic convergence of autism disorder risk genes across neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609190. [PMID: 39229156 PMCID: PMC11370590 DOI: 10.1101/2024.08.23.609190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Over a hundred risk genes underlie risk for autism spectrum disorder (ASD) but the extent to which they converge on shared downstream targets to increase ASD risk is unknown. To test the hypothesis that cellular context impacts the nature of convergence, here we apply a pooled CRISPR approach to target 29 ASD loss-of-function genes in human induced pluripotent stem cell (hiPSC)-derived neural progenitor cells, glutamatergic neurons, and GABAergic neurons. Two distinct approaches (gene-level and network-level analyses) demonstrate that convergence is greatest in mature glutamatergic neurons. Convergent effects are dynamic, varying in strength, composition, and biological role between cell types, increasing with functional similarity of the ASD genes examined, and driven by cell-type-specific gene co-expression patterns. Stratification of ASD genes yield targeted drug predictions capable of reversing gene-specific convergent signatures in human cells and ASD-related behaviors in zebrafish. Altogether, convergent networks downstream of ASD risk genes represent novel points of individualized therapeutic intervention.
Collapse
Affiliation(s)
- Meilin Fernandez Garcia
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Kayla Retallick-Townsley
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - April Pruitt
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
| | - Elizabeth Davidson
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Yi Dai
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Sarah E Fitzpatrick
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
| | - Annabel Sen
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Sophie Cohen
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Olivia Livoti
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Suha Khan
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Grace Dossou
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Jen Cheung
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - P J Michael Deans
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
| | - Zuoheng Wang
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Laura Huckins
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ellen Hoffman
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Child Study Center, Yale University School of Medicine, New Haven, CT 06511
| | - Kristen Brennand
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Department of Genetics, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06511
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06511
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
48
|
Islam MR, Rabbi MA, Hossain T, Sultana S, Uddin S. Mechanistic Approach to Immunity and Immunotherapy of Alzheimer's Disease: A Review. ACS Chem Neurosci 2024. [PMID: 39173186 DOI: 10.1021/acschemneuro.4c00360] [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: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative condition characterized by progressive cognitive decline and memory loss, affecting millions of people worldwide. Traditional treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, offer limited symptomatic relief without addressing the underlying disease mechanisms. These limitations have driven the development of more potent and effective therapies. Recent advances in immunotherapy present promising avenues for AD treatment. Immunotherapy strategies, including both active and passive approaches, harness the immune system to target and mitigate AD-related pathology. Active immunotherapy stimulates the patient's immune response to produce antibodies against AD-specific antigens, while passive immunotherapy involves administering preformed antibodies or immune cells that specifically target amyloid-β (Aβ) or tau proteins. Monoclonal antibodies, such as aducanumab and lecanemab, have shown potential in reducing Aβ plaques and slowing cognitive decline in clinical trials, despite challenges related to adverse immune responses and the need for precise targeting. This comprehensive review explores the role of the immune system in AD, evaluates the current successes and limitations of immunotherapeutic approaches, and discusses future directions for enhancing the treatment efficacy.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md Afser Rabbi
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tanbir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Sadia Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Shihab Uddin
- Department of Bioengineering, King Fahad University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Bio Systems and Machines, King Fahad University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| |
Collapse
|
49
|
Kikuchi M, Viet J, Nagata K, Sato M, David G, Audic Y, Silverman MA, Yamamoto M, Akatsu H, Hashizume Y, Takeda S, Akamine S, Miyamoto T, Uozumi R, Gotoh S, Mori K, Ikeda M, Paillard L, Morihara T. Gene-gene functional relationships in Alzheimer's disease: CELF1 regulates KLC1 alternative splicing. Biochem Biophys Res Commun 2024; 721:150025. [PMID: 38768546 DOI: 10.1016/j.bbrc.2024.150025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024]
Abstract
The causes of Alzheimer's disease (AD) are poorly understood, although many genes are known to be involved in this pathology. To gain insights into the underlying molecular mechanisms, it is essential to identify the relationships between individual AD genes. Previous work has shown that the splice variant E of KLC1 (KLC1_vE) promotes AD, and that the CELF1 gene, which encodes an RNA-binding protein involved in splicing regulation, is at a risk locus for AD. Here, we identified a functional link between CELF1 and KLC1 in AD pathogenesis. Transcriptomic data from human samples from different ethnic groups revealed that CELF1 mRNA levels are low in AD brains, and the splicing pattern of KLC1 is strongly correlated with CELF1 expression levels. Specifically, KLC1_vE is negatively correlated with CELF1. Depletion and overexpression experiments in cultured cells demonstrated that the CELF1 protein down-regulates KLC1_vE. In a cross-linking and immunoprecipitation sequencing (CLIP-seq) database, CELF1 directly binds to KLC1 RNA, following which it likely modulates terminal exon usage, hence KLC1_vE formation. These findings reveal a new pathogenic pathway where a risk allele of CELF1 is associated with reduced CELF1 expression, which up-regulates KLC1_vE to promote AD.
Collapse
Affiliation(s)
- Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Justine Viet
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, F-35000, Rennes, France
| | - Kenichi Nagata
- Department of Functional Anatomy and Neuroscience, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masahiro Sato
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Geraldine David
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, F-35000, Rennes, France
| | - Yann Audic
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, F-35000, Rennes, France
| | - Michael A Silverman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, Canada
| | - Mitsuko Yamamoto
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiroyasu Akatsu
- Department of Community-based Medical Education, Graduate School of Medicine, Nagoya City University, Nagoya, Japan; Choju Medical/Neuropathological Institute, Fukushimura Hospital, Toyohashi, Japan
| | | | - Shuko Takeda
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Suita, Japan; Osaka Psychiatric Medical Center, Osaka Psychiatric Research Center, Hirakata, Japan
| | - Shoshin Akamine
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tesshin Miyamoto
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ryota Uozumi
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shiho Gotoh
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kohji Mori
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Luc Paillard
- Université de Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, F-35000, Rennes, France.
| | - Takashi Morihara
- Department of Psychiatry, Graduate School of Medicine, Osaka University, Suita, Japan; Toyonaka Municipal Hospital, Toyonaka, Japan.
| |
Collapse
|
50
|
Joseph PV, Abbas M, Goodney G, Diallo A, Gaye A. Genomic Study of Taste Perception Genes in African Americans Reveals SNPs Linked to Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607452. [PMID: 39372803 PMCID: PMC11451608 DOI: 10.1101/2024.08.10.607452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Background While previous research has shown the potential links between taste perception pathways and brain-related conditions, the area involving Alzheimer's disease remains incompletely understood. Taste perception involves neurotransmitter signaling, including serotonin, glutamate, and dopamine. Disruptions in these pathways are implicated in neurodegenerative diseases. The integration of olfactory and taste signals in flavor perception may impact brain health, evident in olfactory dysfunction as an early symptom in neurodegenerative conditions. Shared immune response and inflammatory pathways may contribute to the association between altered taste perception and conditions like neurodegeneration, present in Alzheimer's disease. Methods This study consists of an exploration of expression-quantitative trait loci (eQTL), utilizing whole-blood transcriptome profiles, of 28 taste perception genes, from a combined cohort of 475 African American subjects. This comprehensive dataset was subsequently intersected with single-nucleotide polymorphisms (SNPs) identified in Genome-Wide Association Studies (GWAS) of Alzheimer's Disease (AD). Finally, the investigation delved into assessing the association between eQTLs reported in GWAS of AD and the profiles of 741 proteins from the Olink Neurological Panel. Results The eQTL analysis unveiled 3,547 statistically significant SNP-Gene associations, involving 412 distinct SNPs that spanned all 28 taste genes. In 17 GWAS studies encompassing various traits, a total of 14 SNPs associated with 12 genes were identified, with three SNPs consistently linked to Alzheimer's disease across four GWAS studies. All three SNPs demonstrated significant associations with the down-regulation of TAS2R41, and two of them were additionally associated with the down-regulation of TAS2R60. In the subsequent pQTL analysis, two of the SNPs linked to TAS2R41 and TAS2R60 genes (rs117771145 and rs10228407) were correlated with the upregulation of two proteins, namely EPHB6 and ADGRB3. Conclusions Our investigation introduces a new perspective to the understanding of Alzheimer's disease, emphasizing the significance of bitter taste receptor genes in its pathogenesis. These discoveries set the stage for subsequent research to delve into these receptors as promising avenues for both intervention and diagnosis. Nevertheless, the translation of these genetic insights into clinical practice requires a more profound understanding of the implicated pathways and their pertinence to the disease's progression across diverse populations.
Collapse
Affiliation(s)
- Paule Valery Joseph
- National Institute on Alcohol Abuse and Alcoholism, National Institue of Nursing Research, Sensory Science and Metabolism Unit, Biobehavioral Branch, National Institutes of Health, Bethesda, MD, USA
| | - Malak Abbas
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gabriel Goodney
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ana Diallo
- Department of Pharmacotherapy & Outcomes Science, Virginia Commonwealth University, Richmond, VA
| | - Amadou Gaye
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|