1
|
Karagas N, Young JE, Blue EE, Jayadev S. The Spectrum of Genetic Risk in Alzheimer Disease. Neurol Genet 2025; 11:e200224. [PMID: 39885961 PMCID: PMC11781270 DOI: 10.1212/nxg.0000000000200224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/13/2024] [Indexed: 02/01/2025]
Abstract
Alzheimer disease (AD), the most common dementing syndrome in the United States, is currently established by the presence of amyloid-β and tau protein biomarkers in the setting of clinical cognitive impairment. These straightforward diagnostic parameters belie an immense complexity of genetic architecture underlying risk and presentation in AD. In this review, we provide a focused overview of the current state of AD genetics. We discuss the discovery of familial autosomal dominant genes, the identification of candidate genes associated with AD, and genetic variants conferring higher risk of developing AD compared with the general population. In particular, we discuss important features of AD risk due to the APOE ε4 allele. In addition to risk, we describe how the field has made headway understanding genetic factors that may protect from AD. The biological implications and practical limitations of information gleaned from genome-wide association studies in AD over the years are also discussed. The readers will have an up-to-date understanding of where we are in our efforts to understand the layers of genetic complexity in AD.
Collapse
Affiliation(s)
- Nicholas Karagas
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| | - Jessica E Young
- Department of Lab Medicine and Pathology, University of Washington, Seattle; and
| | - Elizabeth E Blue
- Division Medical Genetics, Department of Medicine, University of Washington, Seattle
| | - Suman Jayadev
- Department of Neurology, Adjunct Medicine, Division Medical Genetics, University of Washington, Seattle
| |
Collapse
|
2
|
Sampatakakis SN, Mourtzi N, Charisis S, Mamalaki E, Ntanasi E, Hatzimanolis A, Ramirez A, Lambert JC, Yannakoulia M, Kosmidis MH, Dardiotis E, Hadjigeorgiou G, Megalou M, Sakka P, Scarmeas N. Walking time and genetic predisposition for Alzheimer's disease: Results from the HELIAD study. Clin Neuropsychol 2025; 39:83-99. [PMID: 38741352 DOI: 10.1080/13854046.2024.2344869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
Objective: Our study aimed to explore whether physical condition might affect the association between genetic predisposition for Alzheimer's Disease (AD) and AD incidence. Methods: The sample of participants consisted of 561 community-dwelling adults over 64 years old, without baseline dementia (508 cognitively normal and 53 with mild cognitive impairment), deriving from the HELIAD, an ongoing longitudinal study with follow-up evaluations every 3 years. Physical condition was assessed at baseline through walking time (WT), while a Polygenic Risk Score for late onset AD (PRS-AD) was used to estimate genetic predisposition. The association between WT and PRS-AD with AD incidence was evaluated with Cox proportional hazard models adjusted for age, sex, education years, global cognition score and APOE ε-4 genotype. Then, the association between WT and AD incidence was investigated after stratifying participants by low and high PRS-AD. Finally, we examined the association between PRS-AD and AD incidence after stratifying participants by WT. Results: Both WT and PRS-AD were connected with increased AD incidence (p < 0.05), after adjustments. In stratified analyses, in the slow WT group participants with a greater genetic risk had a 2.5-fold higher risk of developing AD compared to participants with lower genetic risk (p = 0.047). No association was observed in the fast WT group or when participants were stratified based on PRS-AD. Conclusions: Genetic predisposition for AD is more closely related to AD incidence in the group of older adults with slow WT. Hence, physical condition might be a modifier in the relationship of genetic predisposition with AD incidence.
Collapse
Affiliation(s)
- Stefanos N Sampatakakis
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Niki Mourtzi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sokratis Charisis
- Department of Neurology, UT Health San Antonio, San Antonio, TX, USA
| | - Eirini Mamalaki
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Eva Ntanasi
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alex Hatzimanolis
- Department of Psychiatry, National and Kapodistrian University of Athens Medical School, Aiginition Hospital, Athens, Greece
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
- Department of Psychiatry, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, USA
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jean-Charles Lambert
- U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liés au vieillissement, Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary H Kosmidis
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | | | | | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, Marousi, Greece
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| |
Collapse
|
3
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
4
|
Gunasekaran TI, Reyes‐Dumeyer D, Faber KM, Goate A, Boeve B, Cruchaga C, Pericak‐Vance M, Haines JL, Rosenberg R, Tsuang D, Mejia DR, Medrano M, Lantigua RA, Sweet RA, Bennett DA, Wilson RS, Alba C, Dalgard C, Foroud T, Vardarajan BN, Mayeux R. Missense and loss-of-function variants at GWAS loci in familial Alzheimer's disease. Alzheimers Dement 2024; 20:7580-7594. [PMID: 39233587 PMCID: PMC11567820 DOI: 10.1002/alz.14221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/10/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Few rare variants have been identified in genetic loci from genome-wide association studies (GWAS) of Alzheimer's disease (AD), limiting understanding of mechanisms, risk assessment, and genetic counseling. METHODS Using genome sequencing data from 197 families in the National Institute on Aging Alzheimer's Disease Family Based Study and 214 Caribbean Hispanic families, we searched for rare coding variants within known GWAS loci from the largest published study. RESULTS Eighty-six rare missense or loss-of-function (LoF) variants completely segregated in 17.5% of families, but in 91 (22.1%) families Apolipoprotein E (APOE)-𝜀4 was the only variant segregating. However, in 60.3% of families, APOE 𝜀4, missense, and LoF variants were not found within the GWAS loci. DISCUSSION Although APOE 𝜀4and several rare variants were found to segregate in both family datasets, many families had no variant accounting for their disease. This suggests that familial AD may be the result of unidentified rare variants. HIGHLIGHTS Rare coding variants from GWAS loci segregate in familial Alzheimer's disease. Missense or loss of function variants were found segregating in nearly 7% of families. APOE-𝜀4 was the only segregating variant in 29.7% in familial Alzheimer's disease. In Hispanic and non-Hispanic families, different variants were found in segregating genes. No coding variants were found segregating in many Hispanic and non-Hispanic families.
Collapse
Affiliation(s)
- Tamil Iniyan Gunasekaran
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Dolly Reyes‐Dumeyer
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Kelley M. Faber
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD), 410 W. 10th St., HS 4000. Indiana University School of MedicineIndianapolisIndianaUSA
| | - Alison Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's diseaseIcahn School of Medicine at Mount SinaiIcahn Bldg., One Gustave L. Levy PlaceNew YorkNew YorkUSA
| | - Brad Boeve
- Department of Neurology, Mayo ClinicRochesterMinnesotaUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St. Louis, Rand Johnson Building, 600 S Euclid Ave., Wohl Hospital BuildingSt. LouisMissouriUSA
| | - Margaret Pericak‐Vance
- John P Hussman Institute for Human GenomicsDr. John T Macdonald Foundation Department of Human GeneticsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Sciences and Cleveland Institute for Computational Biology. Case Western Reserve UniversityClevelandOhioUSA
| | - Roger Rosenberg
- Department of NeurologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Debby Tsuang
- Department of Psychiatry and Behavioral SciencesUniversity of Washington, GRECC VA Puget Sound, 1660 South Columbian WaySeattleWashingtonUSA
| | - Diones Rivera Mejia
- Los Centros de Diagnóstico y Medicina Avanzada y de Conferencias Médicas y TelemedicinaCEDIMAT, Arturo LogroñoPlaza de la Salud, Dr. Juan Manuel Taveras Rodríguez, C. Pepillo Salcedo esqSanto DomingoDominican Republic
- Universidad Pedro Henríquez Urena, Av. John F. Kennedy Km. 7‐1/2 Santo Domingo 1423Santo DomingoDominican Republic
| | - Martin Medrano
- Pontíficia Universidad Católica Madre y Maestra (PUCMM), Autopista Duarte Km 1 1/2Santiago de los CaballerosDominican Republic
| | - Rafael A. Lantigua
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Department of MedicineVagelos College of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Robert A. Sweet
- Departments of Psychiatry and NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical Center, 1750, West Harrison StChicagoIllinoisUSA
| | - Robert S. Wilson
- Rush Alzheimer's Disease CenterRush University Medical Center, 1750, West Harrison StChicagoIllinoisUSA
| | - Camille Alba
- Department of AnatomyPhysiology and GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Clifton Dalgard
- Department of AnatomyPhysiology and GeneticsUniformed Services University of the Health SciencesBethesdaMarylandUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD), 410 W. 10th St., HS 4000. Indiana University School of MedicineIndianapolisIndianaUSA
| | - Badri N. Vardarajan
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Richard Mayeux
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
5
|
Frick EA, Emilsson V, Jonmundsson T, Steindorsdottir AE, Johnson ECB, Puerta R, Dammer EB, Shantaraman A, Cano A, Boada M, Valero S, García-González P, Gudmundsson EF, Gudjonsson A, Pitts R, Qiu X, Finkel N, Loureiro JJ, Orth AP, Seyfried NT, Levey AI, Ruiz A, Aspelund T, Jennings LL, Launer LJ, Gudmundsdottir V, Gudnason V. Serum proteomics reveal APOE-ε4-dependent and APOE-ε4-independent protein signatures in Alzheimer's disease. NATURE AGING 2024; 4:1446-1464. [PMID: 39169269 PMCID: PMC11485263 DOI: 10.1038/s43587-024-00693-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
A deeper understanding of the molecular processes underlying late-onset Alzheimer's disease (LOAD) could aid in biomarker and drug target discovery. Using high-throughput serum proteomics in the prospective population-based Age, Gene/Environment Susceptibility-Reykjavik Study (AGES) cohort of 5,127 older Icelandic adults (mean age, 76.6 ± 5.6 years), we identified 303 proteins associated with incident LOAD over a median follow-up of 12.8 years. Over 40% of these proteins were associated with LOAD independently of APOE-ε4 carrier status, were implicated in neuronal processes and overlapped with LOAD protein signatures in brain and cerebrospinal fluid. We identified 17 proteins whose associations with LOAD were strongly dependent on APOE-ε4 carrier status, with mostly consistent associations in cerebrospinal fluid. Remarkably, four of these proteins (TBCA, ARL2, S100A13 and IRF6) were downregulated by APOE-ε4 yet upregulated due to LOAD, a finding replicated in external cohorts and possibly reflecting a response to disease onset. These findings highlight dysregulated pathways at the preclinical stages of LOAD, including those both independent of and dependent on APOE-ε4 status.
Collapse
Affiliation(s)
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | - Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Raquel Puerta
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
| | - Eric B Dammer
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Anantharaman Shantaraman
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Amanda Cano
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Agustin Ruiz
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Thor Aspelund
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland.
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
6
|
Tábuas-Pereira M, Bernardes C, Durães J, Lima M, Nogueira AR, Saraiva J, Tábuas T, Coelho M, Paquette K, Westra K, Kun-Rodrigues C, Almeida MR, Baldeiras I, Brás J, Guerreiro R, Santana I. Exploring first-degree family history in a cohort of Portuguese Alzheimer's disease patients: population evidence for X-chromosome linked and recessive inheritance of risk factors. J Neurol 2024; 271:6983-6990. [PMID: 39235525 PMCID: PMC11447147 DOI: 10.1007/s00415-024-12673-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: 07/22/2024] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) heritability is estimated to be around 70-80%. Yet, much of it remains to be explained. Studying transmission patterns may help in understanding other factors contributing to the development of AD. OBJECTIVE In this study, we aimed to search for evidence of autosomal recessive or X- and Y-linked inheritance of risk factors in a large cohort of Portuguese AD patients. METHODS We collected family history from patients with AD and cognitively healthy controls over 75 years of age. We compared the proportions of maternal and paternal history in male and female patients and controls (to search for evidence of X-linked and Y-linked inherited risk factors). We compared the risk of developing AD depending on parents' birthplace (same vs. different), as a proxy of remote consanguinity. We performed linear regressions to study the association of these variables with different endophenotypes. RESULTS We included 3090 participants, 2183 cognitively healthy controls and 907 patients with AD. Men whose mother had dementia have increased odds of developing AD comparing to women whose mother had dementia. In female patients with a CSF biomarker-supported diagnosis of AD, paternal history of dementia is associated with increased CSF phosphorylated Tau levels. People whose parents are from the same town have higher risk of dementia. In multivariate analysis, this proxy is associated with a lower age of onset and higher CSF phosphorylated tau. CONCLUSIONS Our study gives evidence supporting an increased risk of developing AD associated with an X-linked inheritance pattern and remote consanguinity.
Collapse
Affiliation(s)
- Miguel Tábuas-Pereira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal.
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal.
| | - Catarina Bernardes
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - João Durães
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Marisa Lima
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | | | - Jorge Saraiva
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Teresa Tábuas
- Instituto Politécnico de Bragança, Bragança, Portugal
| | - Mariana Coelho
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| | - Kimberly Paquette
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Kaitlyn Westra
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Célia Kun-Rodrigues
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Maria Rosário Almeida
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3004-561, Coimbra, Portugal
| |
Collapse
|
7
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PB, Perkins B, Soni D, Ingraham C, Lee‐Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. Alzheimers Dement 2024; 20:4970-4984. [PMID: 38687251 PMCID: PMC11247676 DOI: 10.1002/alz.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. HIGHLIGHTS A novel approach to validate genetic risk factors for late-onset AD (LOAD) is presented. LOAD risk variants were knocked in to conserved mouse loci. Variant effects were assayed by transcriptional analysis. Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease. This approach should generate more translationally relevant animal models.
Collapse
Affiliation(s)
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Asli Uyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | | | | | - Adrian L. Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | | | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| |
Collapse
|
8
|
Gouveia Roque C, Phatnani H, Hengst U. The broken Alzheimer's disease genome. CELL GENOMICS 2024; 4:100555. [PMID: 38697121 PMCID: PMC11099344 DOI: 10.1016/j.xgen.2024.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/25/2024] [Accepted: 04/07/2024] [Indexed: 05/04/2024]
Abstract
The complex pathobiology of late-onset Alzheimer's disease (AD) poses significant challenges to therapeutic and preventative interventions. Despite these difficulties, genomics and related disciplines are allowing fundamental mechanistic insights to emerge with clarity, particularly with the introduction of high-resolution sequencing technologies. After all, the disrupted processes at the interface between DNA and gene expression, which we call the broken AD genome, offer detailed quantitative evidence unrestrained by preconceived notions about the disease. In addition to highlighting biological pathways beyond the classical pathology hallmarks, these advances have revitalized drug discovery efforts and are driving improvements in clinical tools. We review genetic, epigenomic, and gene expression findings related to AD pathogenesis and explore how their integration enables a better understanding of the multicellular imbalances contributing to this heterogeneous condition. The frontiers opening on the back of these research milestones promise a future of AD care that is both more personalized and predictive.
Collapse
Affiliation(s)
- Cláudio Gouveia Roque
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA; The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| | - Hemali Phatnani
- Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY 10013, USA; Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY 10032, USA
| | - Ulrich Hengst
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
9
|
Lepiarz-Raba I, Hidayat T, Hannan AJ, Jawaid A. Potential Alzheimer's disease drug targets identified through microglial biology research. Expert Opin Drug Discov 2024; 19:587-602. [PMID: 38590098 DOI: 10.1080/17460441.2024.2335210] [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: 01/20/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Microglia, the primary immune cells in the brain, play multifaceted roles in Alzheimer's disease (AD). Microglia can potentially mitigate the pathological progression of AD by clearing amyloid beta (Aβ) deposits in the brain and through neurotrophic support. In contrast, disproportionate activation of microglial pro-inflammatory pathways, as well as excessive elimination of healthy synapses, can exacerbate neurodegeneration in AD. The challenge, therefore, lies in discerning the precise regulation of the contrasting microglial properties to harness their therapeutic potential in AD. AREAS COVERED This review examines the evidence relevant to the disease-modifying effects of microglial manipulators in AD preclinical models. The deleterious pro-inflammatory effects of microglia in AD can be ameliorated via direct suppression or indirectly through metabolic manipulation, epigenetic targeting, and modulation of the gut-brain axis. Furthermore, microglial clearance of Aβ deposits in AD can be enhanced via strategically targeting microglial membrane receptors, lysosomal functions, and metabolism. EXPERT OPINION Given the intricate and diverse nature of microglial responses throughout the course of AD, therapeutic interventions directed at microglia warrant a tactical approach. This could entail employing therapeutic regimens, which concomitantly suppress pro-inflammatory microglial responses while selectively enhancing Aβ phagocytosis.
Collapse
Affiliation(s)
- Izabela Lepiarz-Raba
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Taufik Hidayat
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ali Jawaid
- Laboratory for Translational Research in Exposures and Neuropsychiatric Disorders (TREND), Braincity: Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| |
Collapse
|
10
|
Villa M, Wu J, Hansen S, Pahnke J. Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System. Cells 2024; 13:740. [PMID: 38727275 PMCID: PMC11083179 DOI: 10.3390/cells13090740] [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: 02/28/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
ATP-binding cassette (ABC) transporters play a crucial role for the efflux of a wide range of substrates across different cellular membranes. In the central nervous system (CNS), ABC transporters have recently gathered significant attention due to their pivotal involvement in brain physiology and neurodegenerative disorders, such as Alzheimer's disease (AD). Glial cells are fundamental for normal CNS function and engage with several ABC transporters in different ways. Here, we specifically highlight ABC transporters involved in the maintenance of brain homeostasis and their implications in its metabolic regulation. We also show new aspects related to ABC transporter function found in less recognized diseases, such as Huntington's disease (HD) and experimental autoimmune encephalomyelitis (EAE), as a model for multiple sclerosis (MS). Understanding both their impact on the physiological regulation of the CNS and their roles in brain diseases holds promise for uncovering new therapeutic options. Further investigations and preclinical studies are warranted to elucidate the complex interplay between glial ABC transporters and physiological brain functions, potentially leading to effective therapeutic interventions also for rare CNS disorders.
Collapse
Affiliation(s)
- Maria Villa
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Jingyun Wu
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Stefanie Hansen
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
- Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia
- School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Tel Aviv IL-6997801, Israel
| |
Collapse
|
11
|
Gudmundsdottir V, Frick E, Emilsson V, Jonmundsson T, Steindorsdottir A, Johnson ECB, Puerta R, Dammer E, Shantaraman A, Cano A, Boada M, Valero S, Garcia-Gonzalez P, Gudmundsson E, Gudjonsson A, Pitts R, Qiu X, Finkel N, Loureiro J, Orth A, Seyfried N, Levey A, Ruiz A, Aspelund T, Jennings L, Launer L, Gudnason V. Serum proteomics reveals APOE dependent and independent protein signatures in Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-3706206. [PMID: 38260284 PMCID: PMC10802738 DOI: 10.21203/rs.3.rs-3706206/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The current demand for early intervention, prevention, and treatment of late onset Alzheimer's disease (LOAD) warrants deeper understanding of the underlying molecular processes which could contribute to biomarker and drug target discovery. Utilizing high-throughput proteomic measurements in serum from a prospective population-based cohort of older adults (n = 5,294), we identified 303 unique proteins associated with incident LOAD (median follow-up 12.8 years). Over 40% of these proteins were associated with LOAD independently of APOE-ε4 carrier status. These proteins were implicated in neuronal processes and overlapped with protein signatures of LOAD in brain and cerebrospinal fluid. We found 17 proteins which LOAD-association was strongly dependent on APOE-ε4 carrier status. Most of them showed consistent associations with LOAD in cerebrospinal fluid and a third had brain-specific gene expression. Remarkably, four proteins in this group (TBCA, ARL2, S100A13 and IRF6) were downregulated by APOE-ε4 yet upregulated as a consequence of LOAD as determined in a bi-directional Mendelian randomization analysis, reflecting a potential response to the disease onset. Accordingly, the direct association of these proteins to LOAD was reversed upon APOE-ε4 genotype adjustment, a finding which we replicate in an external cohort (n = 719). Our findings provide an insight into the dysregulated pathways that may lead to the development and early detection of LOAD, including those both independent and dependent on APOE-ε4. Importantly, many of the LOAD-associated proteins we find in the circulation have been found to be expressed - and have a direct link with AD - in brain tissue. Thus, the proteins identified here, and their upstream modulating pathways, provide a new source of circulating biomarker and therapeutic target candidates for LOAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Merce Boada
- Research Center and Memory Clinic of Fundació ACE, Institut Català de Neurociències Aplicades-UIC, Barcelona
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lenore Launer
- National Institute on Aging, National Institutes of Health
| | | |
Collapse
|
12
|
Monteil A, Guérineau NC, Gil-Nagel A, Parra-Diaz P, Lory P, Senatore A. New insights into the physiology and pathophysiology of the atypical sodium leak channel NALCN. Physiol Rev 2024; 104:399-472. [PMID: 37615954 DOI: 10.1152/physrev.00014.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/13/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cell excitability and its modulation by hormones and neurotransmitters involve the concerted action of a large repertoire of membrane proteins, especially ion channels. Unique complements of coexpressed ion channels are exquisitely balanced against each other in different excitable cell types, establishing distinct electrical properties that are tailored for diverse physiological contributions, and dysfunction of any component may induce a disease state. A crucial parameter controlling cell excitability is the resting membrane potential (RMP) set by extra- and intracellular concentrations of ions, mainly Na+, K+, and Cl-, and their passive permeation across the cell membrane through leak ion channels. Indeed, dysregulation of RMP causes significant effects on cellular excitability. This review describes the molecular and physiological properties of the Na+ leak channel NALCN, which associates with its accessory subunits UNC-79, UNC-80, and NLF-1/FAM155 to conduct depolarizing background Na+ currents in various excitable cell types, especially neurons. Studies of animal models clearly demonstrate that NALCN contributes to fundamental physiological processes in the nervous system including the control of respiratory rhythm, circadian rhythm, sleep, and locomotor behavior. Furthermore, dysfunction of NALCN and its subunits is associated with severe pathological states in humans. The critical involvement of NALCN in physiology is now well established, but its study has been hampered by the lack of specific drugs that can block or agonize NALCN currents in vitro and in vivo. Molecular tools and animal models are now available to accelerate our understanding of how NALCN contributes to key physiological functions and the development of novel therapies for NALCN channelopathies.
Collapse
Affiliation(s)
- Arnaud Monteil
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Antonio Gil-Nagel
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Paloma Parra-Diaz
- Department of Neurology, Epilepsy Program, Hospital Ruber Internacional, Madrid, Spain
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- LabEx "Ion Channel Science and Therapeutics," Montpellier, France
| | - Adriano Senatore
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
13
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PBC, Perkins B, Soni D, Ingraham C, Lee-Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572849. [PMID: 38187758 PMCID: PMC10769393 DOI: 10.1101/2023.12.21.572849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Introduction Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. Methods Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE4 and Trem2*R47H. Potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. Results We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. Discussion These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics.
Collapse
Affiliation(s)
- Michael Sasner
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | | | - Ravi S Pandey
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| | - Asli Uyar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| | - Dylan Garceau
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | | | | | - Adrian L Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Audrey Lee-Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana University, NB Building, 320 W 15th St #414, Indianapolis, IN 46202
| | - Gareth R Howell
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
| | - Gregory W Carter
- The Jackson Laboratory, 600 Main St, Bar Harbor, ME, 04609 USA
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032 USA
| |
Collapse
|
14
|
Zhang J, Wang Y, Zhang Y, Yao J. Genome-wide association study in Alzheimer's disease: a bibliometric and visualization analysis. Front Aging Neurosci 2023; 15:1290657. [PMID: 38094504 PMCID: PMC10716290 DOI: 10.3389/fnagi.2023.1290657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/08/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Thousands of research studies concerning genome-wide association studies (GWAS) in Alzheimer's disease (AD) have been published in the last decades. However, a comprehensive understanding of the current research status and future development trends of GWAS in AD have not been clearly shown. In this study, we tried to gain a systematic overview of GWAS in AD by bibliometric and visualization analysis. METHODS The literature search terms are: ("genome-wide analysis" or "genome-wide association study" or "whole-genome analysis") AND ("Alzheimer's Disease" or "Alzheimer Disease"). Relevant publications were extracted from the Web of Science Core Collection (WoSCC) database. Collected data were further analyzed using VOSviewer, CiteSpace and R package Bibliometrix. The countries, institutions, authors and scholar collaborations were investigated. The co-citation analysis of publications was visualized. In addition, research hotspots and fronts were examined. RESULTS A total of 1,350 publications with 59,818 citations were identified. The number of publications and citations presented a significant rising trend since 2013. The United States was the leading country with an overwhelming number of publications (775) and citations (42,237). The University of Washington and Harvard University were the most prolific institutions with 101 publications each. Bennett DA was the most influential researcher with the highest local H-index. Neurobiology of Aging was the journal with the highest number of publications. Aβ, tau, immunity, microglia and DNA methylation were research hotspots. Disease and causal variants were research fronts. CONCLUSION The most frequently studied AD pathogenesis and research hotspots are (1) Aβ and tau, (2) immunity and microglia, with TREM2 as a potential immunotherapy target, and (3) DNA methylation. The research fronts are (1) looking for genetic similarities between AD and other neurological diseases and syndromes, and (2) searching for causal variants of AD. These hotspots suggest noteworthy directions for future studies on AD pathogenesis and genetics, in which basic research regarding immunity is promising for clinical conversion. The current under-researched directions are (1) GWAS in AD biomarkers based on large sample sizes, (2) studies of causal variants of AD, and (3) GWAS in AD based on non-European populations, which need to be strengthened in the future.
Collapse
Affiliation(s)
- Junyao Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinuo Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingying Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyan Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Frick EA, Emilsson V, Jonmundsson T, Steindorsdottir AE, Johnson ECB, Puerta R, Dammer EB, Shantaraman A, Cano A, Boada M, Valero S, García-González P, Gudmundsson EF, Gudjonsson A, Loureiro JJ, Orth AP, Seyfried NT, Levey AI, Ruiz A, Aspelund T, Jennings LL, Launer LJ, Gudmundsdottir V, Gudnason V. Serum proteomics reveals APOE dependent and independent protein signatures in Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.08.23298251. [PMID: 37986771 PMCID: PMC10659486 DOI: 10.1101/2023.11.08.23298251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The current demand for early intervention, prevention, and treatment of late onset Alzheimer's disease (LOAD) warrants deeper understanding of the underlying molecular processes which could contribute to biomarker and drug target discovery. Utilizing high-throughput proteomic measurements in serum from a prospective population-based cohort of older adults (n=5,294), we identified 303 unique proteins associated with incident LOAD (median follow-up 12.8 years). Over 40% of these proteins were associated with LOAD independently of APOE-ε4 carrier status. These proteins were implicated in neuronal processes and overlapped with protein signatures of LOAD in brain and cerebrospinal fluid. We found 17 proteins which LOAD-association was strongly dependent on APOE-ε4 carrier status. Most of them showed consistent associations with LOAD in cerebrospinal fluid and a third had brain-specific gene expression. Remarkably, four proteins in this group (TBCA, ARL2, S100A13 and IRF6) were downregulated by APOE-ε4 yet upregulated as a consequence of LOAD as determined in a bi-directional Mendelian randomization analysis, reflecting a potential response to the disease onset. Accordingly, the direct association of these proteins to LOAD was reversed upon APOE-ε4 genotype adjustment, a finding which we replicate in an external cohort (n=719). Our findings provide an insight into the dysregulated pathways that may lead to the development and early detection of LOAD, including those both independent and dependent on APOE-ε4. Importantly, many of the LOAD-associated proteins we find in the circulation have been found to be expressed - and have a direct link with AD - in brain tissue. Thus, the proteins identified here, and their upstream modulating pathways, provide a new source of circulating biomarker and therapeutic target candidates for LOAD.
Collapse
Affiliation(s)
| | - Valur Emilsson
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | | | - Erik C B Johnson
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Raquel Puerta
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
| | - Eric B Dammer
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Anantharaman Shantaraman
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Amanda Cano
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Mercè Boada
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Sergi Valero
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Pablo García-González
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | | | | | | | | | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Allan I Levey
- Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, 30329, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, 30329, GA, USA
| | - Agustin Ruiz
- Research Center and Memory Clinic. Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, 08028, Spain, Barcelona
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, 28029, Spain
| | - Thor Aspelund
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, 20892, MD, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, 200, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| |
Collapse
|
16
|
Prigge CL, Dembla M, Sharma A, El-Quessny M, Kozlowski C, Paisley CE, Miltner AM, Johnson TM, Della Santina L, Feller MB, Kay JN. Rejection of inappropriate synaptic partners in mouse retina mediated by transcellular FLRT2-UNC5 signaling. Dev Cell 2023; 58:2080-2096.e7. [PMID: 37557174 PMCID: PMC10615732 DOI: 10.1016/j.devcel.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/26/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
During nervous system development, neurons choose synaptic partners with remarkable specificity; however, the cell-cell recognition mechanisms governing rejection of inappropriate partners remain enigmatic. Here, we show that mouse retinal neurons avoid inappropriate partners by using the FLRT2-uncoordinated-5 (UNC5) receptor-ligand system. Within the inner plexiform layer (IPL), FLRT2 is expressed by direction-selective (DS) circuit neurons, whereas UNC5C/D are expressed by non-DS neurons projecting to adjacent IPL sublayers. In vivo gain- and loss-of-function experiments demonstrate that FLRT2-UNC5 binding eliminates growing DS dendrites that have strayed from the DS circuit IPL sublayers. Abrogation of FLRT2-UNC5 binding allows mistargeted arbors to persist, elaborate, and acquire synapses from inappropriate partners. Conversely, UNC5C misexpression within DS circuit sublayers inhibits dendrite growth and drives arbors into adjacent sublayers. Mechanistically, UNC5s promote dendrite elimination by interfering with FLRT2-mediated adhesion. Based on their broad expression, FLRT-UNC5 recognition is poised to exert widespread effects upon synaptic partner choices across the nervous system.
Collapse
Affiliation(s)
- Cameron L Prigge
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Mayur Dembla
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Arsha Sharma
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Malak El-Quessny
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher Kozlowski
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Caitlin E Paisley
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Adam M Miltner
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Tyler M Johnson
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Luca Della Santina
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX 77204, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeremy N Kay
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA.
| |
Collapse
|
17
|
Sun Z, Zhao C, Liu X, Zhang P, Wang X, Man X, Li Z, Du Y, Che X, Xiang Y. Mutation analysis of the ECE1 gene in late-onset Alzheimer's disease. Neurobiol Aging 2023; 129:58-61. [PMID: 37271044 DOI: 10.1016/j.neurobiolaging.2023.05.002] [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: 01/12/2023] [Revised: 04/22/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023]
Abstract
We recently identified a rare coding mutation (R186C) in the ECE2 gene in a late-onset AD (LOAD) family, and demonstrated ECE2 is a risk gene for AD development. ECE1 is a homologous enzyme that shares catalytic activity with ECE2. Although ECE1 has been regarded as a potential candidate gene for AD, few studies have investigated the role of ECE1 variants in patients with AD. In this study, we aimed to investigate rare variants in ECE1 in a cohort of 610 patients with LOAD (age of onset ≥65 years). The summary data of ECE1 variants from ChinaMAP database were used as controls (n = 10,588). We found four rare variants (p.R50W, p.A166=, p.R650Q, and p.P751=) in the patients with sporadic LOAD, while we identified a large number of controls carrying rare variants in ECE1. Moreover, there was no significant association between LOAD and non-synonymous rare damaging variants at the gene level. Our results suggest rare coding variants of ECE1 might not play an important role in AD risk in the Chinese population.
Collapse
Affiliation(s)
- Zhanfang Sun
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cheng Zhao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xueming Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Peng Zhang
- iCarbonX (Zhuhai) Company Limited, Shenzhen, China
| | - Xiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiao Man
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhen Li
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangqian Che
- Department of Neurology & Neuroscience Institute, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuanyuan Xiang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
18
|
Nott A, Holtman IR. Genetic insights into immune mechanisms of Alzheimer's and Parkinson's disease. Front Immunol 2023; 14:1168539. [PMID: 37359515 PMCID: PMC10285485 DOI: 10.3389/fimmu.2023.1168539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 06/28/2023] Open
Abstract
Microglia, the macrophages of the brain, are vital for brain homeostasis and have been implicated in a broad range of brain disorders. Neuroinflammation has gained traction as a possible therapeutic target for neurodegeneration, however, the precise function of microglia in specific neurodegenerative disorders is an ongoing area of research. Genetic studies offer valuable insights into understanding causality, rather than merely observing a correlation. Genome-wide association studies (GWAS) have identified many genetic loci that are linked to susceptibility to neurodegenerative disorders. (Post)-GWAS studies have determined that microglia likely play an important role in the development of Alzheimer's disease (AD) and Parkinson's disease (PD). The process of understanding how individual GWAS risk loci affect microglia function and mediate susceptibility is complex. A rapidly growing number of publications with genomic datasets and computational tools have formulated new hypotheses that guide the biological interpretation of AD and PD genetic risk. In this review, we discuss the key concepts and challenges in the post-GWAS interpretation of AD and PD GWAS risk alleles. Post-GWAS challenges include the identification of target cell (sub)type(s), causal variants, and target genes. Crucially, the prediction of GWAS-identified disease-risk cell types, variants and genes require validation and functional testing to understand the biological consequences within the pathology of the disorders. Many AD and PD risk genes are highly pleiotropic and perform multiple important functions that might not be equally relevant for the mechanisms by which GWAS risk alleles exert their effect(s). Ultimately, many GWAS risk alleles exert their effect by changing microglia function, thereby altering the pathophysiology of these disorders, and hence, we believe that modelling this context is crucial for a deepened understanding of these disorders.
Collapse
Affiliation(s)
- Alexi Nott
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Inge R. Holtman
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
19
|
Young-Pearse TL, Lee H, Hsieh YC, Chou V, Selkoe DJ. Moving beyond amyloid and tau to capture the biological heterogeneity of Alzheimer's disease. Trends Neurosci 2023; 46:426-444. [PMID: 37019812 PMCID: PMC10192069 DOI: 10.1016/j.tins.2023.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 04/05/2023]
Abstract
Alzheimer's disease (AD) manifests along a spectrum of cognitive deficits and levels of neuropathology. Genetic studies support a heterogeneous disease mechanism, with around 70 associated loci to date, implicating several biological processes that mediate risk for AD. Despite this heterogeneity, most experimental systems for testing new therapeutics are not designed to capture the genetically complex drivers of AD risk. In this review, we first provide an overview of those aspects of AD that are largely stereotyped and those that are heterogeneous, and we review the evidence supporting the concept that different subtypes of AD are important to consider in the design of agents for the prevention and treatment of the disease. We then dive into the multifaceted biological domains implicated to date in AD risk, highlighting studies of the diverse genetic drivers of disease. Finally, we explore recent efforts to identify biological subtypes of AD, with an emphasis on the experimental systems and data sets available to support progress in this area.
Collapse
Affiliation(s)
- Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Hyo Lee
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yi-Chen Hsieh
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vicky Chou
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Wittrahm R, Takalo M, Kuulasmaa T, Mäkinen PM, Mäkinen P, Končarević S, Fartzdinov V, Selzer S, Kokkola T, Antikainen L, Martiskainen H, Kemppainen S, Marttinen M, Jeskanen H, Rostalski H, Rahunen E, Kivipelto M, Ngandu T, Natunen T, Lambert JC, Tanzi RE, Kim DY, Rauramaa T, Herukka SK, Soininen H, Laakso M, Pike I, Leinonen V, Haapasalo A, Hiltunen M. Protective Alzheimer's disease-associated APP A673T variant predominantly decreases sAPPβ levels in cerebrospinal fluid and 2D/3D cell culture models. Neurobiol Dis 2023; 182:106140. [PMID: 37120095 DOI: 10.1016/j.nbd.2023.106140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/01/2023] Open
Abstract
The rare A673T variant was the first variant found within the amyloid precursor protein (APP) gene conferring protection against Alzheimer's disease (AD). Thereafter, different studies have discovered that the carriers of the APP A673T variant show reduced levels of amyloid beta (Aβ) in the plasma and better cognitive performance at high age. Here, we analyzed cerebrospinal fluid (CSF) and plasma of APP A673T carriers and control individuals using a mass spectrometry-based proteomics approach to identify differentially regulated targets in an unbiased manner. Furthermore, the APP A673T variant was introduced into 2D and 3D neuronal cell culture models together with the pathogenic APP Swedish and London mutations. Consequently, we now report for the first time the protective effects of the APP A673T variant against AD-related alterations in the CSF, plasma, and brain biopsy samples from the frontal cortex. The CSF levels of soluble APPβ (sAPPβ) and Aβ42 were significantly decreased on average 9-26% among three APP A673T carriers as compared to three well-matched controls not carrying the protective variant. Consistent with these CSF findings, immunohistochemical assessment of cortical biopsy samples from the same APP A673T carriers did not reveal Aβ, phospho-tau, or p62 pathologies. We identified differentially regulated targets involved in protein phosphorylation, inflammation, and mitochondrial function in the CSF and plasma samples of APP A673T carriers. Some of the identified targets showed inverse levels in AD brain tissue with respect to increased AD-associated neurofibrillary pathology. In 2D and 3D neuronal cell culture models expressing APP with the Swedish and London mutations, the introduction of the APP A673T variant resulted in lower sAPPβ levels. Concomitantly, the levels of sAPPα were increased, while decreased levels of CTFβ and Aβ42 were detected in some of these models. Our findings emphasize the important role of APP-derived peptides in the pathogenesis of AD and demonstrate the effectiveness of the protective APP A673T variant to shift APP processing towards the non-amyloidogenic pathway in vitro even in the presence of two pathogenic mutations.
Collapse
Affiliation(s)
- Rebekka Wittrahm
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Teemu Kuulasmaa
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Petra M Mäkinen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Petri Mäkinen
- A.I. Virtanen Institute for Molecular Sciences, 70211 Kuopio, Finland.
| | | | | | - Stefan Selzer
- Proteome Sciences GmbH & Co. KG, 60438 Frankfurt, Germany.
| | - Tarja Kokkola
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Leila Antikainen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Mikael Marttinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany.
| | - Heli Jeskanen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Hannah Rostalski
- A.I. Virtanen Institute for Molecular Sciences, 70211 Kuopio, Finland.
| | - Eija Rahunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Miia Kivipelto
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland; Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; The Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Theme Aging, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Tiia Ngandu
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland; Division of Clinical Geriatrics, Department of Neurobiology, Center for Alzheimer Research, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Jean-Charles Lambert
- U1167, University of Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital, 70211 Kuopio, Finland; Unit of Pathology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Sanna-Kaisa Herukka
- Department of Neurology, University of Eastern Finland, 70210 Kuopio, Finland; NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland.
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland, 70210 Kuopio, Finland.
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland.
| | - Ian Pike
- Proteome Sciences plc, Hamilton House, London, WC1H 9BB, UK.
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, and Institute of Clinical Medicine, Unit of Neurosurgery, University of Eastern Finland, Kuopio, Finland.
| | | | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland.
| |
Collapse
|
21
|
Andrews SJ, Renton AE, Fulton-Howard B, Podlesny-Drabiniok A, Marcora E, Goate AM. The complex genetic architecture of Alzheimer's disease: novel insights and future directions. EBioMedicine 2023; 90:104511. [PMID: 36907103 PMCID: PMC10024184 DOI: 10.1016/j.ebiom.2023.104511] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder and the most common form of dementia. AD is highly heritable, with heritability estimates of ∼70% from twin studies. Progressively larger genome-wide association studies (GWAS) have continued to expand our knowledge of AD/dementia genetic architecture. Until recently these efforts had identified 39 disease susceptibility loci in European ancestry populations. RECENT DEVELOPMENTS Two new AD/dementia GWAS have dramatically expanded the sample sizes and the number of disease susceptibility loci. The first increased total sample size to 1,126,563-with an effective sample size of 332,376-by predominantly including new biobank and population-based dementia datasets. The second, expands on an earlier GWAS from the International Genomics of Alzheimer's Project (IGAP) by increasing the number of clinically-defined AD cases/controls in addition to incorporating biobank dementia datasets, resulting in a total sample size to 788,989 and an effective sample size of 382,472. Collectively both GWAS identified 90 independent variants across 75 AD/dementia susceptibility loci, including 42 novel loci. Pathway analyses indicate the susceptibility loci are enriched for genes involved in amyloid plaque and neurofibrillary tangle formation, cholesterol metabolism, endocytosis/phagocytosis, and the innate immune system. Gene prioritization efforts for the novel loci identified 62 candidate causal genes. Many of the candidate genes from known and newly discovered loci play key roles in macrophages and highlight phagocytic clearance of cholesterol-rich brain tissue debris by microglia (efferocytosis) as a core pathogenetic hub and putative therapeutic target for AD. WHERE NEXT?: While GWAS in European ancestry populations have substantially enhanced our understanding of AD genetic architecture, heritability estimates from population based GWAS cohorts are markedly smaller than those from twin studies. While this missing heritability is likely due to a combination of factors, it highlights that our understanding of AD genetic architecture and genetic risk mechanisms remains incomplete. These knowledge gaps result from several underexplored areas in AD research. First, rare variants remain understudied due to methodological issues in identifying them and the cost of generating sufficiently powered whole exome/genome sequencing datasets. Second, sample sizes of non-European ancestry populations in AD GWAS remain small. Third, GWAS of AD neuroimaging and cerebrospinal fluid endophenotypes remains limited due to low compliance and high costs associated with measuring amyloid-β and tau levels and other disease-relevant biomarkers. Studies generating sequencing data, including diverse populations, and incorporating blood-based AD biomarkers are set to substantially improve our knowledge of AD genetic architecture.
Collapse
Affiliation(s)
- Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
| | - Alan E Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fulton-Howard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Podlesny-Drabiniok
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
22
|
Anderson AG, Rogers BB, Loupe JM, Rodriguez-Nunez I, Roberts SC, White LM, Brazell JN, Bunney WE, Bunney BG, Watson SJ, Cochran JN, Myers RM, Rizzardi LF. Single nucleus multiomics identifies ZEB1 and MAFB as candidate regulators of Alzheimer's disease-specific cis-regulatory elements. CELL GENOMICS 2023; 3:100263. [PMID: 36950385 PMCID: PMC10025452 DOI: 10.1016/j.xgen.2023.100263] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/06/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Cell type-specific transcriptional differences between brain tissues from donors with Alzheimer's disease (AD) and unaffected controls have been well documented, but few studies have rigorously interrogated the regulatory mechanisms responsible for these alterations. We performed single nucleus multiomics (snRNA-seq plus snATAC-seq) on 105,332 nuclei isolated from cortical tissues from 7 AD and 8 unaffected donors to identify candidate cis-regulatory elements (CREs) involved in AD-associated transcriptional changes. We detected 319,861 significant correlations, or links, between gene expression and cell type-specific transposase accessible regions enriched for active CREs. Among these, 40,831 were unique to AD tissues. Validation experiments confirmed the activity of many regions, including several candidate regulators of APP expression. We identified ZEB1 and MAFB as candidate transcription factors playing important roles in AD-specific gene regulation in neurons and microglia, respectively. Microglia links were globally enriched for heritability of AD risk and previously identified active regulatory regions.
Collapse
Affiliation(s)
| | - Brianne B. Rogers
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacob M. Loupe
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | | | - Lauren M. White
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | | | - William E. Bunney
- Department of Psychiatry and Human Behavior, College of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Blynn G. Bunney
- Department of Psychiatry and Human Behavior, College of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Stanley J. Watson
- Mental Health Research Institute, University of Michigan, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
23
|
Miyashita A, Kikuchi M, Hara N, Ikeuchi T. Genetics of Alzheimer's disease: an East Asian perspective. J Hum Genet 2023; 68:115-124. [PMID: 35641666 PMCID: PMC9968656 DOI: 10.1038/s10038-022-01050-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/20/2022] [Accepted: 05/16/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is an age-related multifactorial neurodegenerative disorder. Advances in genome technology, including next generation sequencing have uncovered complex genetic effects in AD by analyzing both common and rare functional variants. Multiple lines of evidence suggest that the pathogenesis of AD is influenced by multiple genetic components rather than single genetic factor. Previous genetic studies on AD have predominantly included European ancestry cohorts; hence, the non-European population may be underrepresented, potentially leading to reduced diversity in AD genetic research. Additionally, ethnic diversity may result in dissimilar effects of genetic determinants in AD. APOE genotypes are a well-established genetic risk factor in AD, with the East Asian population having a higher risk of AD associated with the APOE ε4 allele. To date, seven genome-wide association studies (GWAS) have been conducted in East Asians, which report a total of 26 AD-associated loci. Several rare variants, including the p.H157Y variant in TREM2, and the p.G186R and p.R274W variants in SHARPIN are associated with risk of AD in East Asians. Extending genetic studies to diverse populations, including East Asians is necessary, which could yield more comprehensive insights into AD, and here we review the recent findings regarding the genetic determinants of AD from an East Asian perspective.
Collapse
Affiliation(s)
- Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
24
|
Bartoletti-Stella A, Tarozzi M, Mengozzi G, Asirelli F, Brancaleoni L, Mometto N, Stanzani-Maserati M, Baiardi S, Linarello S, Spallazzi M, Pantieri R, Ferriani E, Caffarra P, Liguori R, Parchi P, Capellari S. Dementia-related genetic variants in an Italian population of early-onset Alzheimer’s disease. Front Aging Neurosci 2022; 14:969817. [PMID: 36133075 PMCID: PMC9484406 DOI: 10.3389/fnagi.2022.969817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Early-onset Alzheimer’s disease (EOAD) is the most common form of early-onset dementia. Although three major genes have been identified as causative, the genetic contribution to the disease remains unsolved in many patients. Recent studies have identified pathogenic variants in genes representing a risk factor for developing Alzheimer’s disease (AD) and in causative genes for other degenerative dementias as responsible for EOAD. To study them further, we investigated a panel of candidate genes in 102 Italian EOAD patients, 45.10% of whom had a positive family history and 21.74% with a strong family history of dementia. We found that 10.78% of patients carried pathogenic or likely pathogenic variants, including a novel variant, in PSEN1, PSEN2, or APP, and 7.84% showed homozygosity for the ε4 APOE allele. Additionally, 7.84% of patients had a moderate risk allele in PSEN1, PSEN2, or TREM2 genes. Besides, we observed that 12.75% of our patients carried only a variant in genes associated with other neurodegenerative diseases. The combination of these variants contributes to explain 46% of cases with a definite familiarity and 32% of sporadic forms. Our results confirm the importance of extensive genetic screening in EOAD for clinical purposes, to select patients for future treatments and to contribute to the definition of overlapping pathogenic mechanisms between AD and other forms of dementia.
Collapse
Affiliation(s)
- Anna Bartoletti-Stella
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Martina Tarozzi
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Giacomo Mengozzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Francesca Asirelli
- Department of Medical Science and Surgery (DIMEC), University of Bologna, Bologna, Italy
| | - Laura Brancaleoni
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Neurologia e Rete Stroke Metropolitana, Ospedale Maggiore, Bologna, Italy
| | - Nicola Mometto
- UOC Neurologia, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | | | - Simone Baiardi
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Simona Linarello
- Programma Cure Intermedie - Azienda USL di Bologna, Bologna, Italy
| | - Marco Spallazzi
- U.O. di Neurologia, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Roberta Pantieri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Elisa Ferriani
- UOC Psicologia Clinica Ospedaliera, Ospedale Bellaria, Azienda USL di Bologna, Bologna, Italy
| | - Paolo Caffarra
- Unità di Neuroscienze, Università di Parma, Parma, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
- *Correspondence: Sabina Capellari,
| |
Collapse
|
25
|
Kalkan H, Akkaya UM, Inal-Gültekin G, Sanchez-Perez AM. Prediction of Alzheimer’s Disease by a Novel Image-Based Representation of Gene Expression. Genes (Basel) 2022; 13:genes13081406. [PMID: 36011317 PMCID: PMC9407775 DOI: 10.3390/genes13081406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Early intervention can delay the progress of Alzheimer’s Disease (AD), but currently, there are no effective prediction tools. The goal of this study is to generate a reliable artificial intelligence (AI) model capable of detecting the high risk of AD, based on gene expression arrays from blood samples. To that end, a novel image-formation method is proposed to transform single-dimension gene expressions into a discriminative 2-dimensional (2D) image to use convolutional neural networks (CNNs) for classification. Three publicly available datasets were pooled, and a total of 11,618 common genes’ expression values were obtained. The genes were then categorized for their discriminating power using the Fisher distance (AD vs. control (CTL)) and mapped to a 2D image by linear discriminant analysis (LDA). Then, a six-layer CNN model with 292,493 parameters were used for classification. An accuracy of 0.842 and an area under curve (AUC) of 0.875 were achieved for the AD vs. CTL classification. The proposed method obtained higher accuracy and AUC compared with other reported methods. The conversion to 2D in CNN offers a unique advantage for improving accuracy and can be easily transferred to the clinic to drastically improve AD (or any disease) early detection.
Collapse
Affiliation(s)
- Habil Kalkan
- Department of Computer Engineering, Gebze Technical University, 41400 Kocaeli, Turkey
- Correspondence: (H.K.); (A.M.S.-P.)
| | - Umit Murat Akkaya
- Department of Computer Engineering, Gebze Technical University, 41400 Kocaeli, Turkey
| | - Güldal Inal-Gültekin
- Department of Physiology, Faculty of Medicine, Istanbul Okan University, 34959 Istanbul, Turkey
| | - Ana Maria Sanchez-Perez
- Faculty of Health Science and Institute of Advanced Materials (INAM), University Jaume I, 12071 Castellon, Spain
- Correspondence: (H.K.); (A.M.S.-P.)
| |
Collapse
|
26
|
Santana DA, Bedrat A, Puga RD, Turecki G, Mechawar N, Faria TC, Gigek CO, Payão SL, Smith MA, Lemos B, Chen ES. The role of H3K9 acetylation and gene expression in different brain regions of Alzheimer's disease patients. Epigenomics 2022; 14:651-670. [PMID: 35588246 DOI: 10.2217/epi-2022-0096] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: To evaluate H3K9 acetylation and gene expression profiles in three brain regions of Alzheimer's disease (AD) patients and elderly controls, and to identify AD region-specific abnormalities. Methods: Brain samples of auditory cortex, hippocampus and cerebellum from AD patients and controls underwent chromatin immunoprecipitation sequencing, RNA sequencing and network analyses. Results: We found a hyperacetylation of AD cerebellum and a slight hypoacetylation of AD hippocampus. The transcriptome revealed differentially expressed genes in the hippocampus and auditory cortex. Network analysis revealed Rho GTPase-mediated mechanisms. Conclusions: These findings suggest that some crucial mechanisms, such as Rho GTPase activity and cytoskeletal organization, are differentially dysregulated in brain regions of AD patients at the epigenetic and transcriptomic levels, and might contribute toward future research on AD pathogenesis.
Collapse
Affiliation(s)
- Daliléia A Santana
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Amina Bedrat
- Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| | - Renato D Puga
- Hermes Pardini Institute, São Paulo, SP, 04038-030, Brazil
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Hospital Research Center, McGill University, Montreal, QC, H4H1R3, Canada
| | - Naguib Mechawar
- Department of Psychiatry, Douglas Hospital Research Center, McGill University, Montreal, QC, H4H1R3, Canada
| | - Tathyane C Faria
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Carolina O Gigek
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, 04023-062, Brazil
| | - Spencer Lm Payão
- Department of Genetics, Blood Center, Faculdade de Medicina de Marília (FAMEMA), Marília, SP, 17519-050, Brazil
| | - Marília Ac Smith
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil
| | - Bernardo Lemos
- Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| | - Elizabeth S Chen
- Department of Morphology & Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo,SP, 04023-062, Brazil.,Department of Environmental Health & Molecular & Integrative Physiological Sciences Program, Harvard TH Chan School of Public Health, Boston, MA 02115-5810, USA
| |
Collapse
|